08.10.2015 Views

Chapter 81

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

<strong>81</strong><br />

CHAPTER<br />

Pediatric Features of<br />

Malignant Hyperthermia<br />

Renée Krivosic-Horber<br />

INTRODUCTION<br />

Malignant hyperthermia (MH) was first described in 1960 as a fatal<br />

complication of anesthesia occurring in members of the same<br />

family. 1 A higher incidence in children younger than 18 years was<br />

observed from the beginning and has been a constant finding<br />

in all epidemiologic studies of MH. This explains the emotional<br />

reaction to a “silent disease” that killed many young people.<br />

However, it is important to note that this higher incidence concerns<br />

only the triggering of an MH episode by anesthesia and not the<br />

other phenotypes (i.e., positive in vitro contracture test [IVCT],<br />

subclinical signs of myopathy, the increase in creatine kinase [CK]<br />

plasmatic levels, and the rare clinical myopathy called central core<br />

disease [CCD]). MH is a potentially lethal state of paroxysmal<br />

hypercatabolism induced in striated muscle by halogenated volatile<br />

anesthetic agents and/or the depolarizing curare succinylcholine<br />

in individuals suffering from a specific muscle abnormality, inher -<br />

ited as autosomal dominant. The designation “MH” persisted over<br />

time even though hyperthermia is a late symptom of the disease.<br />

This pharmacogenetic disorder of skeletal muscle is generally<br />

associated with mutations in the ryanodine receptor isoform<br />

1 (RyR1) of sarcoplasmic reticulum (SR). MH crises develop not<br />

only in humans but also in other species, particularly pigs, that have<br />

been a valuable source for scientific research. Reactions have also<br />

been described in horses, dogs, and other animals. 2<br />

EPIDEMIOLOGY<br />

The evaluation of the incidence of MH crisis is dependent on many<br />

factors, especially the criteria for diagnosis of the MH crisis and<br />

the techniques and anesthetic drugs used. Epidemiologic studies<br />

should be critically analyzed according to the period observed<br />

and the means of diagnosis: clinical signs and determination of<br />

the phenotype by an in vitro contracture test (IVCT) or the geno -<br />

type by the discovery of a causative MH mutation. 3 The estimated<br />

prevalence of the MH genetic abnormalities in the general<br />

population may be as great as 1 in 3000 individuals (range 1:3000–<br />

1:8500). 4<br />

The MH crisis has been observed among all races, regardless of<br />

gender, and all ages. The impact of the MH crisis is highest among<br />

children, with a peak among adolescents, whereas it becomes rarer<br />

after 40 years of age. Although MH is not a sex-linked trait, males<br />

are more commonly affected (male-to-female ratio 1:5). 5,6 The first<br />

case published in France in 1971 was a fulminant fatal crisis in a<br />

girl aged 13 years old. 7 In the first survey published in Canada in<br />

1970, the incidence of MH was estimated at 1 in 15,000 pediatric<br />

anesthesia, much higher than the estimated 1 in 50,000 in adults. 5<br />

In 1985, Ording and coworkers could gather results based on<br />

information about 386,250 anesthetics in Denmark and 154 cases<br />

of suspected MH. 8,9 All cases of MH occurred during general anes -<br />

thesia, and more than 75% during anesthesia with a combination<br />

of potent inhalation agents and succinylcholine. The incidence of<br />

fulminant MH was low: l in 250,000 total anesthetic procedures,<br />

but 1 in 62,000 anesthetic procedures with a combination of potent<br />

inhalation agents and succinylcholine. Masseter spasm (MS)<br />

occurred in 1 of 12,000 anesthetic procedures in which succinyl -<br />

choline was administered. Suspicion of MH was raised in 1 of<br />

16,000 anesthetics total, but in 1 of 4200 anesthetics with the pre -<br />

viously mentioned combination of agents. 8,9<br />

Information concerning probands whose malignant hyper -<br />

thermia susceptibility (MHS) has been proved by IVCT is avail -<br />

able. 10,11 The proband is defined as the first member of a family<br />

to have a suspected MH reaction. The majority (61%) of the<br />

197 MHS probands were children and adolescents aged between<br />

0 and 19 years. In every instance, they were apparently American<br />

Society of Anesthesiologists (ASA) 1or 2 preoperatively and death<br />

(24%) was completely unexpected. In 1993, Strazis and Fox<br />

investigated the epidemiology of MH by analysis of 336 publi -<br />

cations. 12 Five hundred three cases of MH were reported. The<br />

patients’ ages ranged from newborn (reaction to cesarean section<br />

anesthesia) to 73 years. The mean age was 18.3 years. The pediatric<br />

age group was a much larger proportion of the population studied<br />

(52.1%, age < 15 y) than the general surgical population (5% in<br />

the United States). Male gender (65.8%) of MH patients exceeds<br />

the general surgical population. Congenital defects and muscu -<br />

loskeletal surgical procedures were clearly associated with MH.<br />

Previous uneventful anesthesia (20.9%) and absence of positive<br />

family history (75.9%) were common. Case fatality rates have<br />

decreased over time from 80% before 1965 to 16% after 1980. One<br />

limit of this study is the absence of information concerning the<br />

confirmation of MH by IVCT.<br />

MH cardiac arrests still concern young people, as was shown<br />

by Larach and colleagues who analyzed the American database for<br />

AMRA (adverse metabolic/musculoskeletal reaction to anes -<br />

thesia) with inclusion criteria as follows: event date between<br />

January 1, 1987, and December 31, 2006; “very likely” or “almost<br />

certain” MH as ranked by MH clinical grading scale. 13 The median<br />

age of the 8 cases of cardiac arrest (2.7%), of whom 4 (1.4%) died<br />

was 20 years (range 2–31 y), 12 years for the survivors and 22 years<br />

for the deaths. By contrast with these classical data, an incidence<br />

of only 18% pediatric patients (


CHAPTER <strong>81</strong> ■ Pediatric Features of Malignant Hyperthermia 1363<br />

TABLE <strong>81</strong>-1. Published Cases of Anesthetic Malignant Hyperthermia in Newborns and Infants<br />

Associated<br />

Reference Age/Sex/Death Triggering/Dantrolene Abnormalities IVCT Genetics<br />

Peltz, 1975 15 19 mo/M/no Halothane and No No No<br />

succinylcholine<br />

Mayhew, 1978 16 6 mo<br />

Faust, 1979 17 6 mo/no No/no No Yes, both No<br />

parents MHN<br />

Sewall, 1980 18 Neonate/ No (cesarean No No No<br />

M/no<br />

section)/no<br />

Bailey, 1987 19 3 mo/F/no Halothan/yes Arthrogryposis No No<br />

Wilhoit, 1989 20<br />

Dubrow, 1989 21 8 MHS Halothane No Yes, positive No<br />

children<br />

Krivosic, 1990 22 , 19 < 3 y Halothane 15 no/1 Becker’s/1 Yes, in patients 5 mutations<br />

personal data 5/+succinylcholine Duchenne’s/3 or family 11 RYR1<br />

8/sevoflurane 6. syndromic MHS/8 MHN<br />

Hinkle, 1993 23 Newborn Succinylcholine CCD No No<br />

to the mother<br />

with MS/no<br />

Semmler, 1994 24 14 mo/M/no Halothane/yes No Yes, father No<br />

MHS<br />

Pennington, 1996 25 3 mo/M/no Halothane/no No No No<br />

Chamley, 2000 26 6 mo Halothane/no No Yes, 13 y Yes<br />

Kinouchi, 2001 27 9 mo/M/no Sevoflurane/yes King’s syndrome No No<br />

Bonciu, 2007 28 19 mo/M/no Sevoflurane/no No Yes, father No mutation<br />

MHS<br />

CCD = central core disease; IVCT = in vitro contracture test; MHN = malignant hyperthermia nonsusceptible; MHS = malignant hyperthermia susceptible;<br />

MS = master spasm.<br />

inpatient database in the United States, was used to identify<br />

patients discharged with a diagnosis of MH during the years 2000<br />

to 2005. The occurrence of MH increased from 10.2 to 13.3<br />

patients per million hospital discharges (372–521/y; P < .001).<br />

Mortality rates from MH ranged from 6.5% in 2005 to 16.9% in<br />

2001 (P < .0001). The median age of patients with MH was<br />

39 (interquartile range 23–54 y). Only 17.8% of the patients were<br />

children, who had lower mortality than adults (0.7% vs 14.1%,<br />

P < .0001). However, there are many biases in the study that may<br />

lead to doubt if these numbers reflect the true incidence of MH<br />

episode triggered by anesthesia. The authors included all hospital<br />

discharge records with a diagnosis of MH since October 1997, the<br />

International Classification of Diseases, 9th edition, Clinical<br />

Modification coding system has provided a specific diagnosis code<br />

for MH as a result of anesthetics. 14 They excluded other conditions<br />

associated with hyperthermia. Yet, they could not get any infor -<br />

mation concerning the triggering anesthetics and the use of<br />

dantrolene sodium (DS). Many pediatric cases could be missing<br />

because the study analyzed only the inpatients, most of the<br />

children are anesthetized on an outpatient basis, and the authors<br />

are not sure they have covered all the children’s hospitals. The<br />

lower mortality rate found in pediatric patients (0.7%) compared<br />

with in adults (14.1%) was similar to that found in the North<br />

American Malignant Hyperthermia Registry (NAMHR) study.<br />

The main limitations of this study are related to the total absence<br />

of confirmation of the diagnosis of MH either by IVCT or by<br />

genetic analysis or at least an MH clinical grading scale score. The<br />

association of hyperthermia and anesthesia is not a proof of MH.<br />

The fulminant MH crises associated with hyperthermia were<br />

already the less frequent presentation of MH in 2000 and the<br />

following years. The predominance of an MH anesthetic reaction<br />

in adolescents and young adults is obvious but has not been<br />

explained until now. A possible higher use of the triggering agents<br />

could be a cause associated with a higher level of sympathetic tone.<br />

By contrast, the existence of MH crisis in newborns and infants<br />

remains controversial.<br />

Some of the most informed published reports of MH are<br />

gathered in Table <strong>81</strong>–1. 15–28 The only newborn case was published<br />

as a severe muscular rigidity in a premature male infant born by<br />

cesarean section under general anesthesia. A probable diagnosis of<br />

MH was supported by the clinical symptoms of muscular rigidity<br />

and cyanosis, a creatinine phosphokinase of 24,630 IU. The muscle<br />

tone and laboratory values slowly returned to normal over a<br />

period of days. 18 In the case of a 6-month-old infant reported by<br />

Faust and associates, there was no triggering agent used and both<br />

of the child’s parents had normal creatine phosphokinase (CPK)<br />

values and negative caffeine-halothane stimulation tests. As far as<br />

we know, there are very few infant cases proved by the identi -<br />

fication of a novel RYR1 mutation. 17<br />

PHYSIOPATHOLOGY<br />

Understanding the normal muscle contraction is a necessary<br />

step to understand what happens when the muscle with an MH<br />

mutation is in contact with a volatile anesthetic. Normally, the<br />

depolarization of the sarcolemma is induced by the transmission<br />

of nerve impulses through the motor plate. Acetylcholine is<br />

released into synaptic cleft and binds to receptors on postsynaptic


1364 PART 4 ■ Special Monitoring and Resuscitation Techniques<br />

neuron. Action potential is transmitted in sarcolemma and along<br />

the T-tubule causing the release of calcium from cisternae of SR<br />

due to the opening of the so-called calcium released channel or<br />

ryanodine receptors RYR1. The RYR to which the plant alkaloid<br />

ryanodine specifically binds is the major channel for Ca 2+ release<br />

from intracellular stores in skeletal muscle. It mediates the<br />

T-tubular depolarization-induced Ca 2+ release from the SR. The<br />

increase of the sarcoplasmic level of calcium initiates the contrac -<br />

tion cycle through the action on thick and thin filaments (i.e.,<br />

calcium binds to troponin, troponin moves, displacing the trop -<br />

omyosin complex and exposing actin at the active site, myosin<br />

head forms a cross-bridge and bends toward the H zone, the<br />

adenosine triphosphate [ATP] allows release of the cross-bridge).<br />

Relaxation occurs when the process is stopped at the neuro -<br />

muscular junction, there are no more action potential and no<br />

more excitation contraction coupling. The calcium ions return<br />

into the SR, using ATP (Figure <strong>81</strong>–1).<br />

The muscle contraction uses energy coming, first, from the<br />

creatine phosphate with the action of the enzyme CK, and very<br />

quickly from the increase of activity of the mitochondriae. This<br />

aerobic metabolism is very effective, producing 36 ATP molecules<br />

for 1 molecule of glucose. The increase of O 2<br />

con sumption and CO 2<br />

excretion is important and immediate. A percentage of the energy<br />

is lost in heat, leading to an increase in the body temperature.<br />

If the production of energy by the aerobic metabolism is not<br />

sufficient, because the muscle activity is too high (sprint) or too<br />

long (marathon), the anaerobic metabolism is “solicited.” It is much<br />

less effective, producing only 3 ATP molecules per 1 molecule of<br />

glucose, and produces lactic acid. In MH, the mutated calcium<br />

release channels open abnormally in the presence of an anesthetic<br />

volatile agent (AVA), producing the same phenomenon. However,<br />

there is no relaxation as long as the AVA is present (Figure <strong>81</strong>–2).<br />

CLINICAL DESCRIPTION<br />

The characteristic feature of MH is a hypermetabolic response to<br />

potent inhalation agents and/or succinylcholine characterized by<br />

increased CO 2<br />

production, O 2<br />

consumption, and muscle mem -<br />

brane breakdown.<br />

Figure <strong>81</strong>-1. Physiology of the muscle contraction.<br />

Pharmacologic Triggers of MH Crisis<br />

All halogenated agents are MH triggers: halothane, 22 methoxy -<br />

flurane, enflurane, isoflurane, sevoflurane, 28 and desflurane. 29 New<br />

halogenated agents trigger weaker muscle contracture than<br />

halothane, on human muscle in vitro as well as in the pig model in<br />

Figure <strong>81</strong>-2. Physiopathology of the malignant<br />

hyperthermia (MH) crisis.


CHAPTER <strong>81</strong> ■ Pediatric Features of Malignant Hyperthermia 1365<br />

vivo. 29,30 These data seem to confirm in vivo observations of more<br />

delayed and pernicious crises that could make diagnosis more<br />

difficult. An MHS patient presents variable responses to triggering<br />

agents and the responses may depend on many other factors.<br />

Thus, she or he may be a victim of a fulminant MH crisis even if<br />

she or he was previously exposed to the triggering agent without<br />

any problem. 31 History without anesthetic particularities of a<br />

patient should never challenge a hypothetical diagnosis of MH.<br />

The depolarizing curare (succinylcholine) encourages and<br />

accelerates the MH crisis clinically in the context of MS 32 and<br />

experimentally on the porcine model. 33 There is currently no<br />

documented report on the outbreak of a fulminant crisis by<br />

MH depolarizing curare in the absence of halogenated agent<br />

in a patient whose HM susceptibility was proved by IVCT.<br />

Succinylcholine could open the mutated RYR1 channels during<br />

the depolarizing phase, inducing an increase in muscle tone,<br />

especially the masseter, but only for less than 3 minutes. By con -<br />

trast, the nontriggering power of the other anesthetic agents or<br />

other additives of anesthesia seems now acquired. 34 One report<br />

suggested that, in patients known to be MHS, the incidence of<br />

triggering after exposure to a nontriggering anesthetic is not<br />

zero. 35 In their study of 2214 patients undergoing muscle biopsy to<br />

rule out MH, 1082 had a positive contracture tests. Of those<br />

patients found to be positive, 5 developed moderate signs of MH<br />

in the perioperative period, yielding an incidence rate of 0.46%<br />

(confidence interval [CI] 0.058–0.866). However, this report is not<br />

a reason to reconsider the safety of anesthesia without triggering<br />

agents in MHS patients (Table <strong>81</strong>–2).<br />

Clinical Signs of MH Crisis<br />

The clinical signs can be gathered in three groups of symptoms<br />

according to the physiopathology: (1) hypermetabolism, (2) mus -<br />

cular rigidity, and (3) rhabdomyolysis. The clinical and biologic<br />

signs are summarized in Figures <strong>81</strong>–3 and <strong>81</strong>–4. The time to onset<br />

of the crisis, in the absence of injection of succinylcholine, is highly<br />

variable after the start of inhalation of halogenated agents. It seems<br />

unlikely that a crisis can begin in the postoperative period,<br />

TABLE <strong>81</strong>-2. Safe and Unsafe Drugs<br />

Unsafe Drugs<br />

Depolarizing muscle relaxants: succinylcholine<br />

Volatile halogenated anesthetics: halothane, isoflurane,<br />

enflurane, desflurane, sevoflurane<br />

Safe Drugs<br />

Barbiturates<br />

Propofol<br />

Benzodiazepines<br />

Droperidol<br />

Nitrous oxide<br />

Local anesthetics amides and esters<br />

Nondepolarizating muscle relaxants<br />

Morphine and morphinomimetics<br />

Ketamine<br />

Antibiotics<br />

Antihistamines<br />

Antipyretics<br />

Propranolol<br />

Vasoactive drugs<br />

Figure <strong>81</strong>-3. Clinical signs of the MH crisis.<br />

after the end of administration of the triggering agents 36,37 (see<br />

Figure <strong>81</strong>–2).<br />

Hypermetabolism<br />

Signs of hypermetabolism are early. The first sign is a significant<br />

increase of arterial carbon dioxide pressure (PaCO 2<br />

) and end-tidal<br />

carbon dioxide pressure (PETCO 2<br />

) visualized on the capnometer 38,39<br />

(Figure <strong>81</strong>–5). The increase in oxygen consumption (VO 2<br />

) is<br />

suggested by the decrease of Fractional concentration of end-tidal<br />

O2 (FETO 2<br />

). If mixed venous oxygen saturation (SVO 2<br />

) monitoring<br />

is available, it will show a dramatic decrease (Figure <strong>81</strong>–6). The<br />

MH crisis does not cause detectable desaturation of arterial blood,<br />

because of the high fractional con centration of oxygen in inspired<br />

gas (FI O2 ) most often used during anesthesia. However, attention<br />

must be drawn by cyanosis of venous blood in the operative field,<br />

reflecting a considerable increase in muscle VO 2<br />

and resulting in<br />

the collapse of SvO 2<br />

. The tachypnea can be evocative in a patient<br />

with spontaneous venti lation of a response to a hyperproduction of<br />

CO 2<br />

. In the case of controlled ventilation, the sudden increase of<br />

PETCO 2<br />

would also be an indication of this hyperproduction.<br />

Acidosis is initially purely hypercapnic; however, it evolves rapidly<br />

in a mixed acidosis owing to severe anerobic metabolism (see<br />

Figure <strong>81</strong>–4). Tachycardia is almost always observed; however,<br />

because it is commonplace in anesthesia in children, it is often not<br />

Figure <strong>81</strong>-4. Biologic signs of the MH crisis.


1366 PART 4 ■ Special Monitoring and Resuscitation Techniques<br />

Figure <strong>81</strong>-5. End-tidal carbon dioxide<br />

pressure (PETCO 2<br />

).<br />

considered specific. The ventricular arrhythmias due to sympa -<br />

thetic hyperactivity and hyperkalemia (from rhabdomyolysis) are<br />

frequent but not constant. Signs of circulatory failure appear lately,<br />

because initially, increased cardiac output is proportional to the<br />

induced hyper metabolism. Hyperthermia is a response to the<br />

production of heat by the muscular contracture. It is usually<br />

delayed because of the fact that the whole body must be heated<br />

before observing a significant raise in temperature. This sign<br />

occurs earlier in children due to the lower body mass; however, it<br />

remains a late sign in most cases. The elevation of body tempera -<br />

ture can reach 43°C in fulminant forms shortly preceding death.<br />

Muscular Rigidity<br />

Muscular rigidity is a specific sign, but late and not constant.<br />

It may be early in patient in whom anesthesia was induced with a<br />

volatile agent and muscle relaxation was induced with injection of<br />

succinylcholine. It manifests itself as a severe MS. The reasons for<br />

this specific localization are the masseter muscles are the strongest<br />

muscles of the organism and often the first evidence noticed by<br />

the anesthesiologist while taking care of the airway. If the MH<br />

episode develops further, the muscle rigidity involves the lower<br />

limbs and, subsequently, the whole body. In the fulminant MH<br />

crisis, it is impossible to bend the arms and legs. The rigidity<br />

remains after death but, unlike the rigor mortis, does not dis -<br />

appear after mobilization (Figure <strong>81</strong>–7).<br />

Rhadomyolysis<br />

The muscle contracture induces a suffering of the muscle mem -<br />

branes, leading to the leakage from the sarcoplasm to the blood of<br />

intracellular components. Rhabdomyolysis is confirmed by the<br />

determination of high serum levels of potassium, myoglobin, and<br />

CK. The time to maximum increase is in relation with the molec -<br />

ular weight. The hyperkalemia is immediate and can cause sudden<br />

cardiac arrest; however, it is not constant. It is sometimes replaced<br />

by hypokalemia. Myoglobinuria, red color urine, is maximal within<br />

2 to 4 hours. The increase in CK is more delayed with a maximum<br />

between 12 and 24 hours. Elevated CK is variable depending on<br />

the duration and severity of the crisis. It can reach up to 1000 times<br />

the normal value. The results should be inter preted taking into ac -<br />

count the type of surgery, the position, and possible traumatic con -<br />

text. It is essential to repeat CK blood levels regularly during the<br />

first 24 hours, because an increase could reflect a delayed resum -<br />

ption of rhabdomyolysis. If the MH episode is not stopped early<br />

enough, cardiovascular shock will occur in association with wide -<br />

spread vital organ dysfunction, disseminated intravascular coagu -<br />

lation (DIC), and irreversible cellular damage leading to death. 40<br />

MANAGEMENT AND TREATMENT<br />

Despite significant progress made in the diagnosis and treatment<br />

of this disease, the MH crisis remains potentially fatal if the ad -<br />

ministration of the triggering agent is continued and treatment is<br />

not instituted. This medical condition is very rare and statistically<br />

might be experienced less than once in a lifetime. The fact that it<br />

occurs often during anesthesia for emergency surgery is unfa -<br />

vorable to optimal care. The treatment should be initiated as<br />

soon as the diagnosis is evoked. Stopping the administration of<br />

halogenated agent and starting the intravenous injection of DS<br />

are the two therapeutic measures whose success depends on the<br />

immediate and early decision. The diversity of tasks involves<br />

the use of all personnel immediately available, coordinated by the<br />

anesthesiologist.<br />

Figure <strong>81</strong>-6. Mixed venous oxygen saturation (SVO 2<br />

).<br />

Figure <strong>81</strong>-7. Contracture of the quadriceps during the MH crisis.


CHAPTER <strong>81</strong> ■ Pediatric Features of Malignant Hyperthermia 1367<br />

Figure <strong>81</strong>-8. Dantrolene.<br />

DS is a hydantoïc compound that has long been used to fight<br />

against muscle spasticity from pyramidal origin (Figure <strong>81</strong>–8).<br />

It is used in humans since the mid-1970s as a treatment for MH<br />

crisis. It is a muscle relaxant acting directly on the skeletal muscle,<br />

most probably at the calcium channel RyR1. The sole mechanism<br />

of action currently known is the inhibition of the release of<br />

calcium from the SR toward the sarcoplasm. 41 DS has no effect<br />

on the neuromuscular transmission. It is also efficient on the<br />

smooth muscles. For example, it provokes uterine relaxation<br />

during labor and/or vomiting owing to relaxation of the gastroin -<br />

testinal muscles. No cardiodepressive effect has been demon -<br />

strated at therapeutic doses. In the first report of clinical use in<br />

human MH reactions, DS therapy at a dose of 2.5 mg/kg resulted<br />

in a statistically significantly lower mortality rate than would have<br />

normally been expected in MH patients. The study was supported<br />

by animal data, suggesting that DS was specific in reversing MH<br />

cellular process. 42 Peak DS plasma concentrations of 4.2 mg/L were<br />

obtained 3 hours after administering bolus injections of 0.1 mg/kg<br />

until a twitch depression plateau was reached (2.2- to 2.5-mg/kg<br />

cumulative dose). 43 Shime and coworkers have shown that<br />

the administration of DS was safe in 20 pregnant MHS vomen. 44<br />

The mean maternal predelivery DS level was 0.99 ± 0.5 µg/mL,<br />

and the mean neonatal cord blood DS level 0.68 ± 0.3 µg/mL. The<br />

half-life of DS in the neonatal circulation was 20 hours. 44,45<br />

The combination of DS and verapamil results, in the pig model,<br />

in severe arrhythmias and possible ventricular fibrillation. The<br />

simultaneous use of DS and a calcium antagonist in the treatment<br />

of the MH crisis is strongly discouraged because it is known to be<br />

potentially dangerous. In France, the availability of DS is under<br />

the responsibility of the director and the pharmacist of the health<br />

facility. Eighteen vials (5 mg/kg) and the quantity of distilled water<br />

needed for injection (60 mL/vial) should be immediately available<br />

in all areas where anesthesia is provided. The access to 36 vials<br />

(10 mg/kg), which may be necessary to treat an MH crisis, must<br />

be well organized and readily available. A poster indicating the<br />

therapeutic algorithm recommendations in cases of MH should<br />

be displayed (see Figure <strong>81</strong>–4). These guidelines consist of:<br />

1. Stop the triggering agent and maintain the anesthesia with<br />

nontriggering anesthetics, such as propofol, opioids, and<br />

benzodiazepines.<br />

2. Increase minute ventilation with 100% O 2<br />

to lower PETCO 2<br />

.<br />

3. Get help.<br />

4. Mix, shake, and administer intravenous DS 2.5 mg/kg initial<br />

dose (for a 70-kg adult: 175mg = 9 vials for a total volume of<br />

540 mL; for a 30-kg 11-y-old child: 75 mg = 4 vials or a total<br />

volume of 225 mL).<br />

● Titrate DS while following clinical signs such as tachycardia<br />

and hypercarbia.<br />

● 10 mg/kg is the suggested upper limit.<br />

5. Administer bicarbonate 1 to 2 mEq/kg in case of severe<br />

metabolic acidosis.<br />

6. Treat arrhythmias as needed. Do not use calcium channel<br />

blockers.<br />

7. Begin cooling measures and stop them when the central<br />

temperature is under 38°C.<br />

8. Secure blood gases, electrolytes, CK, blood, and urine for<br />

myoglobin.<br />

● Coagulation profile check values every 6 to 12 hours.<br />

9. Treat hyperkalemia with hyperventilation, glucose, and<br />

insulin as needed.<br />

10. Continue DS at 1 mg/kg every 4 to 8 hours for 24 to 48 hours.<br />

Mechanical ventilation is mandatory to compensate for the<br />

respiratory depression induced by DS.<br />

11. Monitor urinary output to prevent acute tubular necrosis and<br />

the presence of myoglobinuria. Administer extra mannitol<br />

(because it is also present in the DS solution) and saline in<br />

case of myoglobinuria.<br />

12. Observe the patient in the intensive care unit for at least<br />

36 hours. Twenty percent of patients will experience a<br />

recrudescence of the syndrome. 40 Do not forget to take into<br />

account the potential respiratory depression induced by the<br />

myorelaxant effect of DS. The plasma elimination half-life is<br />

10 to 13 hours in adults and in children. 45<br />

13. Refer the patient and family to the MH testing center for<br />

contracture and DNA testing.<br />

Most patients with MH can be treated successfully with this<br />

protocol; however, recurrence of the clinical signs and symptoms<br />

of MH can occur hours after resolution of the initial event. 40 With<br />

data obtained from AMRA reports in the NAMHR, it was possible<br />

to show that recurrences happened in 20% of patients. The mean<br />

time from initial reaction to a second break in the symptoms was<br />

13 hours (standard deviation [sd] 13 h). Organ failure was more<br />

frequent in the group subjected to a recurrence of the symptoms<br />

(32% vs 12%; P < .001). The more severe the MH reaction, the<br />

more likely is the probability of a second attack of the symptoms.<br />

These results support the need to manage the patient in the<br />

intensive care unit after a severe MH reaction. Death may still<br />

occur. 13 However, brain damage is very rarely reported as sequelae.<br />

Interestingly, talipes equinus deformity of bilateral lower limbs, a<br />

condition similar to compartment syndrome, has been reported<br />

after MH in a previously healthy pediatric patient. 46<br />

DIFFERENTIAL DIAGNOSIS<br />

Intraoperative<br />

Among the signs of an MH crisis, the increase of PETCO 2<br />

shown<br />

on the capnograph is the most sensitive (Figure <strong>81</strong>–9). 38 The<br />

clinical diagnosis of MH cannot be certain except in a fulminant<br />

form with generalized rigidity, hyperthermia exceeding 40°C, and<br />

hypercapnia exceeding 70 mmHg. This form, often irreversible,<br />

tends to disappear with an early diagnosis because of the sys -<br />

tematic use of capnography and a better appreciation of the early<br />

signs. The analysis of 402 patient charts demonstrated that it is


1368 PART 4 ■ Special Monitoring and Resuscitation Techniques<br />

Figure <strong>81</strong>-9. Differential diagnosis.<br />

often difficult to establish immediately and with certainty the<br />

diagnosis of MH. It is essential to deal with any clinical suspicion. 11<br />

The diagnosis is supported by an immediate arterial and venous<br />

puncture to measure PaCO 2<br />

, pH, arterial oxygen pressure<br />

(PaO 2<br />

), lactates, electrolytes, myoglobin, and CK. This approach<br />

has helped to diagnose more MH of incomplete or aborted<br />

presentation as well as those in which the prognosis was much<br />

better.<br />

The development of a scoring system helping to determine<br />

a diagnostic probability was proposed in 1987 by Larach and<br />

colleagues. 47 The aim of this score is more to compare patient<br />

groups on the basis of scientific purposes rather than to help to<br />

treat a patient suspected to present an MH crisis. Any rise of<br />

postoperative CK plasma concentration can contribute to the<br />

evaluation of patients who present with the possibility of MH,<br />

anesthesia-induced rhabdomyolysis, and underlying muscle<br />

disease. To know the usual profile, 71 patients aged 1 month to<br />

l 7 years were studied. The median CK elevation (range) for the<br />

major and minor surgery groups was 43 IU/L (1–647) and 10 IU/L<br />

(28–122), respectively. The research protocol was similar for<br />

both groups with halothane for induction and isoflurane for<br />

maintenance of anesthesia. Owing to its possible effect on CK,<br />

succinylcholine was avoided during the study. 48<br />

Masseter Spasm<br />

The MS describes a lack of relaxation of the jaw muscles after<br />

injection of a depolarizing curare. Its presence was reported in the<br />

first description of MH during the initial stage of an MH crisis.<br />

The link between MS and MH has long been a source of contro -<br />

versies. In 1984 49 and 1987, 33 a 1% incidence of MS was found in<br />

children receiving a combination of halothane and succinylcholine<br />

during anesthesia. These children were classified as MHS<br />

by the authors at the time, but the use of proper evaluation denied<br />

the diagnosis. An 18-month retrospective chart review of all<br />

patients undergoing general anesthesia at the Children’s Hospital<br />

of Pittsburgh (N = 14,112) was conducted to assess the incidence<br />

of MS and its management. 50 In the otolaryngology service, the<br />

incidence of developing MS was observed in 2 out of 206 (1%)<br />

children who were anesthetized with halothane and received<br />

succinylcholine. In France, unlike the Anglo-Saxon countries, MS<br />

appears far less frequently and no MS was reported in a series of<br />

1055 children intubated after anesthetic induction with halothane<br />

and succinylcholine. 51 Van der Spek and associates suggested in<br />

1990 that, in the general population, the depolarizing curare<br />

caused an increase in the tonus of the muscles of the jaw during<br />

administration. 52 In 1986 53 and 1989, 54 an incidence of 50% of<br />

positive IVCT was reported in a population of children having<br />

MS. More recently, a study reported the results of caffeinehalothane<br />

muscle contracture testing by using the current North<br />

American Malignant Hyperthermia Group (NAMHG) protocol,<br />

on 70 pediatric patients (50 boys and 20 girls) referred for biopsy<br />

between 1986 and 1991. 55 This was based on evidence of muscle<br />

masseter rigidity (MMR) after succinylcholine between 1975 and<br />

1991. The clinical scenario was described as MMR alone or MMR<br />

followed by signs of MH, including PaCO 2<br />

tension lower than<br />

50 mmHg. It was observed in 83% (58 of 70) of anesthetics in<br />

which halothane-succinylcholine was used. Fifty-nine percent<br />

(41 of 70) were classified as MHS, and among them, 5 developed<br />

signs of clinical MH. These results reaffirm the high incidence of<br />

MHS in patients associated with MMR. 55 In summary, MS should<br />

be considered a potentially strong indicator of MH.<br />

MH-Like Syndromes Outside<br />

the Operating Room<br />

Neuroleptic Malignant Syndrome<br />

The neuroleptic malignant syndrome (NMS) is a potentially fatal<br />

hyperthermic syndrome that occurs as a result of ingestion of<br />

drugs used in the treatment of mental conditions such as<br />

schizophrenia. The signs of NMS include muscle rigidity, acidosis,<br />

high fever reactive to DS, and severe rhabdomyolysis. The<br />

prognosis is poor, but survival without sequelae can occur after<br />

2 to 3 weeks of intensive care therapy. The similarities between<br />

MH and NMS made some doctors suspect the existence of<br />

a common disorder of the calcium dysregulation of skeletal<br />

muscle. However, many differences exist between NMS and MH,<br />

including triggering drugs (neuroleptics vs halogenated agents),<br />

duration of evolution (a few hours for the MH crisis vs a few days<br />

for the NMS), and the absence of a hereditary factor in NMS.<br />

Patients who presented NMS seem today, and according to data<br />

from literature, not at risk of MH. The largest published series<br />

do not seem to accredit the thesis of a common mechanism. 56<br />

The mechanism of NMS is still unknown. It could result from<br />

dopamine receptor blockade associated with a possible toxic<br />

action of neuroleptic on the muscle fiber. Dehydration and hypo -<br />

volemia are factors predisposing to NMS. DS use is recommended<br />

only for symptomatic treatment of severe hyperthermia and<br />

muscular rigidity.<br />

Stress-Related MH<br />

It is well known that the MH pigs present the porcine stress<br />

syndrome identified as an “awake” MH episode. The reason why<br />

the MH humans do not have this risk is unknown. None of the<br />

few reports of MH reactions in awake patients or patients given


CHAPTER <strong>81</strong> ■ Pediatric Features of Malignant Hyperthermia 1369<br />

trigger-free anesthetics was totally convincing. Tobin and cowork -<br />

ers reported a fatal episode in a 13-year-old boy who had expe -<br />

rienced a clinical episode of MH and developed signs of MH after<br />

exercise some months later. 57 He and other family members were<br />

found to have a causative RYR1 mutation. In our own experience,<br />

we observed a boy with the same clinical features whose autopsy<br />

showed the existence of a viral cardiomyopathy.<br />

Exertional heat stroke arises from prolonged and intense<br />

muscular exercise, usually in a hot and humid atmosphere. It is<br />

characterized by a hyperthermic response at 40°C, disturbance of<br />

consciousness, rhabdomyolysis, and cardiovascular collapse. It is<br />

not possible at present to prove the existence of an association with<br />

MH. Contracture tests were performed on patients with exerciseinduced<br />

rhabdomyolysis, and positive IVCT were observed in<br />

11 out of 12 patients tested and a RYR1 mutation was found in<br />

3 patients. The authors concluded by recommending that patients<br />

with exercise-induced rhabdomyolysis should have a muscle<br />

biopsy performed for histologic study and implementation of<br />

halothane contracture test/caffeine. 58<br />

This hypothesis should most probably be evoked in children.<br />

Recently, the case of a 2-year and 9-month-old child, who was left<br />

unattended in a car and died of heat stroke, was reported. 59<br />

Postmortem mutation analysis revealed that the child possessed<br />

two distinct RYR1 mutations. Because each mutation had pre -<br />

viously been identified in a separate MHS patient, MHS with<br />

overresponse to the environmental high temperature might have<br />

occurred in this child with these RYR1 mutations.<br />

CONFIRMATION OF<br />

THE DIAGNOSIS OF MH<br />

Not Validated Tests<br />

The existence of elevated CK in susceptible MH individuals was<br />

first reported in 1970. However, a team found in 1976 that this<br />

elevation was present in only 45% of subjects. 60 An evaluation of<br />

the sensitivity and specificity of the assay of CK in the screening<br />

of individual MHS showed in our center a sensitivity of 38% and<br />

a specificity of 88%. 61 It was shown that CK assay for screening<br />

could not be used. However, any rise incidentally discovered,<br />

which was not explained by a muscular dystrophy, illness, or<br />

medication, and persistent in spite of taking care of muscular rest,<br />

absence of trauma, and intramuscular injection suggested a<br />

increased risk of MHS and encouraged the use of contracture tests.<br />

Several teams have found, in these circumstances, positive tests. 62<br />

Many other diagnostic tests have been proposed, seeking a higher<br />

level of calcium in muscular or blood cells, spontaneously or<br />

induced by halothane, but none of them was validated. The search<br />

for abnormalities of muscle metabolism studied by nuclear mag -<br />

netic resonance spectroscopy has also been proposed. Despite<br />

some correlation with contracture tests on muscle biopsy,<br />

sensitivity and specificity are insufficient to make a diagnosis<br />

of MHS.<br />

In Vitro Halothane-Caffeine Contracture Test<br />

Ten years after the description of MH, Kalow and colleagues<br />

described an exaggerated response to caffeine of muscle sus -<br />

ceptible to MH. 63 Ellis and Harriman showed the appearance<br />

of a contracture with halothane. 64 The use of IVCT was born. In a<br />

patient suspected of having presented an attack of MH during<br />

anesthesia, or a family member, the certainty of susceptibility to<br />

MH is achieved by the implementation of the IVCT. Strict<br />

protocols were established on both sides of the Atlantic Ocean by<br />

the European Malignant Hyperpyrexia Group (EMHG), 65 and the<br />

North American Malignant Hyperthermia Group NAMHG. 66 The<br />

muscle biopsy is performed in the vastus lateralis muscle, under<br />

locoregional anesthesia of the femoral nerve and cutaneous lateral<br />

nerve of the thigh (Figure <strong>81</strong>–10A). Local infiltration must be<br />

excluded. The sample is placed immediately in a solution of Krebs-<br />

Ringer, at ambient laboratory temperature, and buffered and<br />

oxygenated by carbogen. The muscle fragments (15–20 mm long,<br />

3 mm in diameter, 100–300 mg) are placed in an insulated testbath<br />

and stimulated directly (see Figure <strong>81</strong>–10B). It is then subjected to<br />

increasing concentrations of halothane and caffeine. A different<br />

muscle bundle is used for each test. The protocols vary slightly<br />

between Europe and North America, but the basic principle is<br />

similar (Figure <strong>81</strong>–11). The twitch responses and the baseline<br />

tensions are recorded. The threshold value is the minimal concen -<br />

tration of halothane or caffeine at which a sustained baseline<br />

contracture of 0.2 g or greater occurs (see Figure <strong>81</strong>–10C–E).<br />

In the EMHG protocol, each test is positive if this threshold is<br />

2% halothane or 2 mM caffeine or less. The test results led to<br />

classifying the subjects into three diagnostic groups: (1) MHS (i.e.,<br />

caffeine and halothane positive); 2) MH nonsusceptible (MHN;<br />

i.e., caffeine and halothane negative); and (3) equivocal halothane<br />

or caffeine (MHE; i.e., when only one test is positive, these patients<br />

are considered clinically MHS-positive). On the normal muscle,<br />

halothane potentiates the twitch but does not induce any con -<br />

tracture and caffeine induces contracture at a higher concen -<br />

tration. Tests conducted according to the protocol of the European<br />

Group can get results with sensitivity of 100% and specificity close<br />

to 94%. 67 Using the NAMHG protocol, an individual is diagnosed<br />

as MHS-positive when either the halothane (using a bolus of<br />

3vol% during 10 min) or the caffeine test is positive and MHN<br />

when both tests are negative. Ryanodine and chlorocresol are two<br />

molecules that may appear useful for certain diagnoses. They seem<br />

to offer better specificity than halothane and caffeine. However,<br />

because of the difficulty of establishing precise thresholds and<br />

pending comparative multicenter studies, these two molecules are<br />

not included in the protocol of the EMH. The IVCT is expensive<br />

and confined to specialized testing centers. It requires a surgical<br />

procedure and can yield equivocal as well as false-positive results.<br />

However, it remains the “gold standard” for diagnosis of MH.<br />

It shows a phenotype that is considered as a proof of a genetic<br />

abnormality. It can be expressed as an MH crisis when the patient<br />

is exposed to the triggering anesthetic agents.<br />

IVCT in Children<br />

The policy of the EMHG established by Ellis indicates a lower age<br />

limit of 10 years because of the inconsistency in muscle responses<br />

in young children. 65 This is possibly related to immature muscle<br />

tissue, but also to the difficulty of obtaining viable muscle bundles<br />

in sufficient quantity. In 1990, Krivosic-Horber and associates<br />

questioned the realization and reliability of the halothane-caffeine<br />

contracture tests in children to detect the susceptibility to MH. 22<br />

The study concerned 26 children aged 2 to 13 years (mean<br />

9.5 ± 1.3 y) who were tested either because of a personal symp -<br />

tomatology (14 cases) or as a member of a susceptible MH family<br />

(12 cases). Half of the children had a positive test (MHS and


1370 PART 4 ■ Special Monitoring and Resuscitation Techniques<br />

A<br />

B<br />

C<br />

D<br />

E<br />

Figure <strong>81</strong>-10. A: Muscular<br />

biopsy of the vastus lateralis<br />

under locoregional anesthesia.<br />

B: Preparation of the muscle bundles.<br />

C: Laboratory device for the<br />

in vitro contracture test (IVCT).<br />

D: The recorder. E: Recordings of<br />

halothane test in normal and malignant<br />

hyperthermia susceptible<br />

(MHS) muscle and caffeine test in<br />

normal and MHS muscle.


CHAPTER <strong>81</strong> ■ Pediatric Features of Malignant Hyperthermia 1371<br />

Figure <strong>81</strong>-11. Comparison of<br />

testing protocols for MH.<br />

MHE) as found in adults. Comparison of threshold concentrations<br />

of halothane and caffeine as well as the 32 nmol caffeine-induced<br />

contractures did not show any significant difference related to<br />

age. These results supported the possibility to perform under<br />

good conditions and with good reliability the diagnostic test of<br />

susceptibility to MH in children from the age of 2 years. Femoral<br />

and lateral femoral cutaneous block anesthesia with light to<br />

moderate sedation is well tolerated in children undergoing<br />

anterior thigh procedures. This was demonstrated in a study of<br />

179 children, aged younger than 18 years (10 ± 3y). 68 In 1997,<br />

Ummenhofer and coworkers reported the results of the IVCT<br />

performed in 24 children between the ages of 6 and 14 years. 69<br />

Seventeen patients out of 24 were diagnosed as MHS according to<br />

the protocol of the EMHG and 7 children as MHN. Twenty-one<br />

children were evaluated postoperatively. Minor side effects of<br />

wound healing occurred, but none of the patients showed any<br />

abnormalities of muscle contracture or movement performance.<br />

Considering the high risk of fatal complications in the presence<br />

of MHS, muscle biopsy of the upper leg for in vitro diagnosis is a<br />

justified procedure that is acceptable to children and their parents.<br />

However, the attitude has changed since the possibility to more<br />

precisely analyze the sensitivity of the IVCT compared with the<br />

results of genetic testing. The very few discordant results between<br />

a negative IVCT and the presence of a causative MH mutation<br />

analyzed in the EMHG was in most cases related to a poor quality<br />

of the muscle studied. 70 The only case observed in our laboratory<br />

while studying 832 IVCT was related to a 4-year-old sister of a boy<br />

tested with MHS when he was 9 years old and who presented in<br />

1986 with typical signs of MH after an induction with enflurane<br />

and succinylcholine. The linkage analysis supported the fact that<br />

the MHS mother had given the same allele of the RYR1 gene to<br />

both her children. Since that observation, the policy regarding<br />

muscle biopsy in our institution was changed. Muscle biopsy is<br />

not performed in children who are younger than 12 years and less<br />

than 30 kg. The recommendation is to test both parents of the<br />

young proband. If the two of them are MHN, they are not at risk<br />

and cannot transmit any MH mutation to their children. The<br />

investigation in the family is stopped. However, if the diagnosis<br />

cannot be eliminated in the proband, the child will be tested by<br />

IVCT when he or she will be old enough to have this intervention<br />

done according to the protocol. Only negative IVCT can eliminate<br />

the diagnosis of MH in individuals at risk. The existence of a<br />

neomutation, although rare, remains possible. 71<br />

Genetic Testing<br />

Mendelian Inheritance<br />

The inherited character of MH has been recognized since its<br />

original description in 1960. 1 Transmission is usually autosomal<br />

dominant. This means that the presence of a mutation in one allele<br />

of the gene is sufficient to cause the phenotype MH. Heterozygous<br />

subjects are at risk of MH. They can transmit the mutated allele to<br />

each child with a probability of 50%. The majority of susceptible<br />

individuals are heterozygous, but there are some homozygous<br />

individuals in whom both parents carry the same or different<br />

mutations in the RYR1 gene mutation. These individuals may<br />

show no more clinical symptoms than the heterozygous ones. 72<br />

Despite the high probability that an MHS subject has inherited an<br />

MH mutation only from one parent, most laboratories routinely<br />

offer IVCT to both parents according to the protocol. An inci -<br />

dence of 5% of positive contracture tests in both parents has<br />

been reported. 73<br />

Molecular Genetics of MH<br />

Since 1990, developments in genetics have allowed the location<br />

and characterization of the gene of the MH at the chromosome<br />

19, in the region q13,1. 74 Thanks to linkage studies, the MH locus


1372 PART 4 ■ Special Monitoring and Resuscitation Techniques<br />

could be located in pigs. In humans, the ryanodine receptor<br />

(RyR1) gene encoding the calcium channel of sarcoplasmic retic -<br />

ulum has also been identified. 75 The location of this gene corre -<br />

sponds to the physiopathology of MH. The existence of an<br />

inversion of amino acids in the calcium channel renders it sus -<br />

ceptible to impregnation by the halogenated agents. and its<br />

opening causes an increase in the rate of sarcoplasmic calcium,<br />

leading to symptoms of the MH crisis. Considering the existence<br />

of a unique mutation in MHS pigs in the RyR1 gene, regardless of<br />

race, one might have expected the possibility of a simple diagnosis<br />

by DNA analysis in humans. However, there is no dominant<br />

mutation in humans. Further research reported that MH has a<br />

high level of locus heterogeneity. As much as five loci, called<br />

MHS1 to MHS6, have been published. A single mutation has<br />

been reported in the CACNA1S gene in two unrelated families.<br />

The CACNA1S encodes the a 1<br />

-subunit of the L-type Ca21 channel<br />

isoform that is expressed in skeletal muscle sarcolemma and is<br />

known as the dihydropyridine receptor. Both channels are involved<br />

in excitation-contraction coupling. 76<br />

The RYR1 is the main candidate gene (Figure <strong>81</strong>–12). Its<br />

structure is known and divided in three domains called MHS1,<br />

MHS2 and MHS3. It is known to be a polymorphic gene with<br />

more than 170 missense mutations, several of these having been<br />

identified as polymorphisms rather than causative mutations.<br />

A novel mutation must not be used diagnostically before it has<br />

been proved to be causative, in extensive pedigree analysis com -<br />

paring molecular genetic results with contracture testing data<br />

and/or in using experimental techniques. 77 Currently, 29 RYR1<br />

mutations have been proven to be causative for MH (http://<br />

www.emhg.org), but to date, more than 60 mutations have been<br />

identified in the MHS population. Although most MHS mutations<br />

map to the MH1 and MH2 domains, a few MHS mutations map<br />

to the C-terminal MH3 domain of RYR1. Complete screening of<br />

the entire coding regions of RYR1 has, however, revealed that<br />

mutations occur in almost all regions of the gene. Owing to the<br />

high level of locus heterogeneity, the diagnosis of MH suscep -<br />

tibility cannot be made using a simple genetic test (EMHG 78 ). It is<br />

important to avoid false MHN diagnoses because of the potential<br />

risk of MH during general anesthesia for these patients and their<br />

offspring. However, there may be situations in which genetic data<br />

provide additional diagnostic information or contribute infor -<br />

mation independent of IVCT. It is possible to undertake screening<br />

in a family, only after confirmation that the suspected proband is<br />

really MHS by performing IVCT, and that she or he bears a known<br />

causative MH mutation. The genetic study should not be offered<br />

initially in an individual suspected of having presented an MH<br />

crisis, because laboratories, having tried this approach, have<br />

obtained a yield of less than 5%.<br />

The discovery of a causative MH mutation in an MHS subject<br />

allows the search of this mutation for diagnosis of MH sensitivity<br />

in the family (Figure <strong>81</strong>–13). This determination should<br />

be organized while taking into account the requirements and<br />

Figure <strong>81</strong>-12. Gene and protein RYR1.


CHAPTER <strong>81</strong> ■ Pediatric Features of Malignant Hyperthermia 1373<br />

interest and requires further investigation, but this patient’s MH<br />

status remains unclear until contracture testing is performed.<br />

Absence of such mutations does not exclude MHS and warrants<br />

eventual IVCT. It is generally not recommended to look system -<br />

atically for an MH mutation in infants and young children, except<br />

when they need a general anesthesia and a causative mutation has<br />

been identified in the family history. It is preferred to wait for the<br />

child to be able to understand the diagnosis. One important reason<br />

is that the insurance companies do not always reimburse the<br />

genetic test that can cost more than 1,000 in the United States.<br />

RISK OF MH-LIKE EPISODE<br />

IN MYOPATHIC OR<br />

SYNDROMIC CHILDREN<br />

The hypothesis of an association of various diseases of striated<br />

muscle with MH (strabismus, scoliosis) was issued in the 1970s.<br />

Since then, various studies have not led to such conclusions,<br />

and the specificity of contracture testing in patients with neuro -<br />

muscular diseases (NMDs) has been questioned. 8 Now it seems<br />

that only the CCD and the King Denborough syndrome (KDS)<br />

can be linked to MH. 80<br />

1. KDS.<br />

2. Dystrophinopathies.<br />

3. Other myopathies.<br />

4. Sudden infant death syndrome (SIDS).<br />

5. Other syndromes.<br />

Figure <strong>81</strong>-13. Suggested route for MH susceptibility testing.<br />

implementation of examinations of the genetic characteristics of a<br />

person for medical purposes. It is clarified that the requirement<br />

of an examination of genetic characteristics can take place only<br />

within a multidisciplinary team comprising clinical and genetic<br />

experts. The proof of the information should appear in the form<br />

of a certificate established by the responsible physician. The<br />

genetic tests should be done in accredited laboratories. A national<br />

advi sory committee in the examination of genetic characteristics<br />

for medical purposes must be established. It is essential to remain<br />

cautious with the use of genetics for the purpose of diagnosis in<br />

patients with MH susceptibility, given the importance of anes -<br />

thetic security issues. In particular, the absence in an individual<br />

of the mutation responsible for MH in his or her family is not<br />

enough to classify him or her as MHN. This person could bear<br />

another abnormality producing an MH crisis in the presence of<br />

the triggering anesthetic agents. One can be classified as MHN<br />

by IVCT only. Sensitivity of genetic testing is estimated to be<br />

approximately 25%.<br />

The only report of MHS in a newborn by molecular genetic<br />

testing of umbilical cord blood was published in 2006. 79 Sampling<br />

of umbilical blood is noninvasive. A possible concern might be<br />

the potential contamination of umbilical cord blood with maternal<br />

nucleated cells. However, the concentration of maternal cells was<br />

found to be 10 –4 to 10 –5 times lower than neonatal nucleated cells,<br />

and therefore, the identical signal intensity of both alleles in the<br />

analyses represent the neonatal MH mutation. For verification,<br />

contamination with maternal DNA was excluded by short tandem<br />

repeat profiling. The genetic variant identified in the case is novel<br />

and, thus, of no diagnostic value at all. This result is of scientific<br />

Central Core Disease 80,<strong>81</strong><br />

CCD is an inherited neuromuscular disorder characterized by<br />

central cores on muscle biopsy and clinical features of a congenital<br />

myopathy. It is probably more common than other congenital<br />

myopathies. The CCD typically presents in infancy with hypotonia<br />

and motor developmental delay. It is characterized by predomi -<br />

nantly proximal weakness pronounced in the hip girdle, and<br />

orthopedic complications are common. In the majority of patients,<br />

weakness is static or only slowly progressive, with a favorable longterm<br />

outcome. The CCD and MHS are allelic conditions both due<br />

to mutations in the RYR1 gene. Altered excitability and/or changes<br />

in calcium homeostasis within muscle cells due to mutationinduced<br />

conformational changes of the RYR protein are con -<br />

sidered the main pathogenetic mechanism(s). The diagnosis of<br />

CCD is based on the presence of suggestive clinical features and<br />

central cores on muscle biopsy. Mutational analysis of the RYR1<br />

gene may provide genetic confirmation of the diagnosis. Manage -<br />

ment is mainly supportive and has to anticipate susceptibility<br />

to MH. The IVCT should be discussed on an individual basis,<br />

because discordant results have been observed within the same<br />

families.<br />

Native American myopathy is a putative autosomal recessive<br />

congenital myopathy that was reported to be frequently associated<br />

with MH. 82 Until now, no IVCT was performed and no MH caus -<br />

ative mutation was found<br />

King Denborough Syndrome<br />

KDS was first reported by King and Denborough in 1973 when<br />

the authors documented 18 males with MH, 5 of whom had


1374 PART 4 ■ Special Monitoring and Resuscitation Techniques<br />

congenital progressive myopathy, short stature, cryptorchidism,<br />

pectus carinatum, lumbar lordosis, and thoracic kyphosis. 83 Three<br />

of the 5 had similar facial features with ptosis, downslanting<br />

palpebral fissures, micrognathia, crowded teeth, apparently<br />

low-set ears, and a short webbed neck. The inheritance of KDS is<br />

unknown, and there is little evidence to conclude that it is a single<br />

disorder. There are also no strict diagnostic criteria for this syn -<br />

drome. It may be actually a phenotype resulting from a hetero -<br />

geneous collection of congenital myopathies with different<br />

microscopic appearances. 27,84 There was no proof of the link<br />

between KDS and MH until the recent report of a KDS caused by<br />

a novel mutation in the ryanodine receptor gene. 85<br />

Dystrophinopathies<br />

Acute rhabdomyolysis leading to hyperkalemic cardiac arrest has<br />

been reported in association with Duchenne muscular dystrophies<br />

(DMDs) and has been understandably confused with MH. Like<br />

MH, most reported cases of rhabdomyolysis have been associated<br />

with the use of halothane and/or succinylcholine.<br />

DMD and Becker muscular dystrophy (BMD) are neuro -<br />

muscular disorders characterized by dystrophin deficiency<br />

(dystrophinopathies). DMD is more frequent, occurs earlier, and<br />

is more severe than BMD. The prevalence of DMD is 1 in 3300<br />

male births. The prevalence of BMD varies from 1 in 18,000 to<br />

1 in 31,000 male births. They affect not only the striated muscle<br />

but also the heart muscle and sometimes smooth muscle. Diag -<br />

nosis is generally made at the age of 5 when children present with<br />

waddling gait and talipes equines with calf hypertrophy (positive<br />

Gowers’ sign). Inability to walk appears by the age of 10 to 12.<br />

Scoliosis, cardiomyopathy, and restrictive respiratory failure<br />

progressively appear. 86 CPK levels are 50- to 200-fold (DMD)<br />

or 10- to 35-fold (BMD) higher than standard values. Muscular<br />

biopsy shows dystrophic features (necrotic and regenerative<br />

fibers). Immunohistochemical studies show a total absence of<br />

dystrophin (DMD) or altered quantity and/or quality (BMD).<br />

Molecular analysis most frequently shows deletions of the DMD<br />

gene, which is located at Xp21.2 and encodes several isoforms.<br />

In 1992, the Malignant Hyperthermia Association of the<br />

United States and the NAMHR received reports of cardiac arrest<br />

in apparently healthy children given succinylcholine. 87 Using data<br />

from 1990 to 1993, this study analyzed: (1) etiology of all reported<br />

pediatric arrests (age < 18 y) occurring within 24 hours of anes -<br />

thesia; (2) whether survival was associated with certain patient or<br />

treatment variables; and (3) presence of myopathy. Twenty-five<br />

patients (92% male, median 45 mo old) had a cardiac arrests;<br />

23 of 25 (92%) were scheduled for minor surgery. Before receiving<br />

a potent inhalational anesthetic (92%) and/or succinylcholine<br />

(72%), these patients were evaluated by the anesthesiologist as<br />

being healthy with no personal or family history of myopathy.<br />

Serum potassium during cardiac arrest was measured in 18 of<br />

25 (72%) patients; hyperkalemia (mean [K + ] = 7.4 ± 2.8, median<br />

7.5 mmol/L) was detected in 13 of 18 (72%) patients. Postarrest<br />

resuscitations lasted a median of 42 minutes (range 10–296). Ten<br />

(40%) patients died, 1 (4%) remained neurologically impaired, and<br />

14 (56%) returned to baseline neurologic function. A previously<br />

unrecognized DMD (N = 8) or unspecified myopathy (N = 4) was<br />

diagnosed in 12 (48%) patients. Eight of these 12 patients’ cardiac<br />

arrests were associated with hyperkalemia. Ten (40%) patients had<br />

no postarrest evaluation to exclude occult myopathy. No patient or<br />

treatment variables were statistically associated with survival. The<br />

authors concluded that, whenever possible, pediatricians should<br />

evaluate their patients (especially male infants and children)<br />

preoperatively for the presence of occult myopathy. During<br />

perianesthetic resuscitations, the pediatric advanced life support<br />

protocol should be modified to detect and treat hyperkalemia, a<br />

potentially reversible state even after prolonged resuscitation<br />

efforts. After anesthetic deaths, pathologists should examine<br />

body fluid electrolytes and skeletal muscle for myopathy and the<br />

presence of dystrophin. If a preanesthetic CK screen for myopathy<br />

in male patients and restrictions on succinylcholine had been<br />

used, 64% of cardiac arrests and 60% of deaths might have been<br />

prevented. A formal prospective risk/benefit analysis for preven -<br />

tive measures is needed.<br />

In 1996 review of the literature revealed 66 pediatric cases<br />

(56 boys and 10 girls) of anesthesia-associated rhabdomyolysis. 88<br />

Forty-nine (74%) cases were caused by an underlying, mostly<br />

unrecognized congenital muscle disease, and 14 (21%) cases were<br />

caused by MHS. Hyperkalemia (23 patients), cardiac arrhythmias<br />

(38 patients), renal failure (4 patients), and death (11 patients)<br />

were the most serious complications of anesthesia-associated<br />

rhabdomyolysis. The neuromuscular blocking agent succinyl -<br />

choline had been used in at least 43 of the patients reported in the<br />

literature. Breucking and colleagues carried out a prospective<br />

epidemiologic study (between 1983 and 2000) on a population of<br />

families including muscular cases of dystrophies: 147 families<br />

DMD and 53 families BMD. 89 A total of 219 male and 9 female<br />

underwent 444 anesthesias; 6 cardiac arrests were reported (1.3%,<br />

much more than the frequency of the cardiac arrests in the<br />

pediatric population 0.1–0.3%) occurring only in the 45 families<br />

without diagnosis at the time of the anesthesia. Schulte-Sasse and<br />

associates reported, in 9 children, on the occurrence of cardiac<br />

arrests within the minutes after the administration of succinyl -<br />

choline. 90 It was later shown that all of them had occult NMD. Five<br />

of the children did not survive the catastrophic event. The clear<br />

association between succinylcholine and rhabdomyolysis led the<br />

U.S. Food and Drug Administration to advise against the use of<br />

succinylcholine in children in 1994. 91<br />

In conclusion, halogenated agents and succinylcholine should<br />

be avoided in patients with muscular dystrophy, even if no genetic<br />

relation and no common mechanism exist with MH. The fragility<br />

of the muscle of these patients, which is characterized by chronic<br />

rhabdomyolysis and the presence of a regenerating muscle, carries<br />

the risk of acute rhabdomyolysis with hyperkalemia when exposed<br />

to halothane and/or succinylcholine. 92<br />

Other Myopathies<br />

Flick and associates examined the risk of MH in children exposed<br />

to a triggering anesthetic while undergoing muscle biopsy for<br />

suspected NMD. 93 Between 1992 and 2005, the medical records<br />

of 351 children younger than 21 years were identified as having<br />

undergone muscle biopsy for suspected NMD. Among them,<br />

274 patients received a volatile anesthetic agent and only 3 patients<br />

were administered succinylcholine. No patient exhibited signs or<br />

symptoms of MH or rhabdomyolysis. The author concluded that<br />

the estimated risk of MH or rhabdomyolysis is less than 1% in a<br />

diverse population of children with suspected NMD.<br />

McArdle’s disease, an isolated deficiency in glycogen degrada -<br />

tion in skeletal muscles, has the potential of creating perioperative


CHAPTER <strong>81</strong> ■ Pediatric Features of Malignant Hyperthermia 1375<br />

problems such as hypoglycemia, rhabdomyolysis, myoglobinuria,<br />

acute renal failure, and possibly MH. 94 A recent review concluded<br />

that no clinical association with MH has been established. 92 How -<br />

ever, it is suggested that halogenated agents and succinylcholine<br />

should be avoided to help to prevent rhabdomyolysis.<br />

Sudden Infant Death Syndrome<br />

Ellis and coworkers report three investigations about the potential<br />

relationship SIDS and MH. 95 In the first study, 151 MHS families<br />

completed a questionnaire designed to identify the incidence<br />

of SIDS within their own pedigree. In the second study, 106 SIDS<br />

families completed a questionnaire designed to identify the<br />

incidence of anesthetic-related problems. In the third study,<br />

14 SIDS parents were subjected to muscle biopsy and IVCT and<br />

caffeine contracture screening for susceptibility to MH. They<br />

concluded that there is no association between SIDS and MH.<br />

Other Conditions Suspected<br />

to Be at Risk of MH<br />

Some published case reports relate the observation of signs similar<br />

to MH during anesthesia with or without triggering agents in<br />

children with rare syndrome. However, MHS was not confirmed<br />

by IVCT or genetic analyses. Noonan’s syndrome, characterized<br />

by short stature, typical facial dysmorphism, and congenital<br />

heart defects presents a special interest because of the physical<br />

similarities with KDS. Lee and colleagues reported one patient<br />

scheduled for spinal arthrodesis whose operation was deferred in<br />

because MH developed during the induction of anesthesia. 96 It is<br />

reasonable to propose trigger-free anesthesia to these patients.<br />

Patients affected with osteogenesis imperfecta are known to have<br />

a temperature control problem due to a hypothalamic dysfunction;<br />

however, no relation with MH has been demonstrated. 97<br />

A new fatal syndrome has been reported in adolescent<br />

males affected with early diabetes mellitus. The features included<br />

hyperglycemic hyperosmolar coma complicated by an MH-like<br />

picture with fever, rhabdomyolysis, and severe cardiovascular<br />

instability. The authors suggest that a genetic predisposition to<br />

MH could be an explanation and that DS should be used in this<br />

situation. There is actually no evidence to support this position. 98,99<br />

ANESTHESIA OF THE PATIENT<br />

WITH A RISK OF MH<br />

Patients who are known to be MHS may be anesthetized with<br />

regional anesthesia or local anesthesia without any problem. If<br />

general anesthesia or sedation is required, the potent volatile<br />

anesthetic agents and succinylcholine should be avoided. It is not<br />

conceivable today to refuse anesthesia to a patient at risk for<br />

MH. 100 When precautions are taken, the administration of anes -<br />

thesia is considered safe. Patients at risk should be identified in<br />

the preoperative anesthesia consultation (Table <strong>81</strong>–3). Referral to<br />

a specialized anesthesia center should always be considered if the<br />

patient’s condition allows. It is the responsibility of the anesthe -<br />

siologist to organize all the investigations necessary, IVCT, and/or<br />

DNA analysis to precise the exact status of the patient concerning<br />

MH (Table <strong>81</strong>–4).<br />

Ambulatory surgery and anesthesia are authorized.<br />

TABLE <strong>81</strong>-3. Categories of Risk of Malignant<br />

Hyperthemia Susceptibility<br />

Risk of MH Susceptibility<br />

Categories of patients at risk of MH in the preoperative<br />

consultation<br />

1. Card MHS by positive IVCT MH precautions<br />

2. Card MHN by negative IVCT<br />

● No exclusion of the triggering agents<br />

● No risk for the children<br />

3. Not tested, relatives of suspected MH MH precautions<br />

4. Member of a family with an MH mutation and positive for<br />

this mutation MH precautions<br />

5. Member of a family with an MH mutation and negative for<br />

this mutation MH precautions until proven MHN by<br />

IVCT<br />

6. Proband MH not tested (anesthesia, exercise) and relatives<br />

MH precautions<br />

IVCT = in vitro contracture test; MH = malignant hyperthermia; MHN =<br />

malignant hyperthermia nonsusceptible; MHS = malignant hyperthermia<br />

susceptible.<br />

The prophylactic administration of DS to prevent MH crisis<br />

was recommended by Gronert, who had proved its effectiveness<br />

in the pig model. 41 Different protocol requirements were subse -<br />

TABLE <strong>81</strong>-4. Management of Malignant Hyperthermia–<br />

Susceptible Patients<br />

1. Appreciate the MH risk by preoperative evaluation<br />

A necessary anesthesia should never be cancelled in an<br />

MH-risk patient.<br />

2. Consider completing the investigation if the anesthesia is<br />

not urgent:<br />

● Details of the previous triggering anesthesia.<br />

● Family tree to calculate the risk of a relative of an MHS<br />

patient.<br />

● Order IVCT or DNA analysis if indicated.<br />

3. Discuss the anesthetic alternatives with the surgeon and the<br />

patient:<br />

● Monitored anesthetic care with sedation and local<br />

anesthesia.<br />

● Regional anesthesia.<br />

● General anesthetic using nontriggering agents.<br />

4. Clean machine; remove vaporizers; replace CO 2<br />

canisters,<br />

bellows, and gas hose, check the monitoring (PETCO 2<br />

++).<br />

5. Flush machine for 20 min with oxygen 10L/min.<br />

6. Bring the MH cart in the operating room (this cart contains<br />

dantrolene; stock all the supplies to resuscitate a patient<br />

with MH).<br />

7. Schedule the patient as the first case of the day, and notify<br />

the postanesthesia care unit to be prepared to provide the<br />

necessary manpower.<br />

8. After surgery measure central temperature and CK, look at<br />

the color of the urines, and monitor patient in an appropriate<br />

setting.<br />

CK = creatine kinase; IVCT = in vitro contracture test; MH = malignant<br />

hyperthermia; MHS = malignant hyperthermia susceptible; PETCO 2<br />

= endtidal<br />

carbon dioxide pressure.


1376 PART 4 ■ Special Monitoring and Resuscitation Techniques<br />

quently proposed, including oral or intravenous administration.<br />

Nowadays, because of the many side effects of DS (hypoventilation<br />

by hypotonia of striated muscles, nausea, vomiting, or uterine<br />

bleeding by hypotonia of the smooth muscle), and the absence of<br />

reports of MH without triggering agents, it is not advisable to<br />

propose a pretreatment by the DS. Patients at risk should not be<br />

given triggering anesthetic agents (i.e., potent volatile anesthetic<br />

agents such as halothane, sevoflurane, desflurane, enflurane,<br />

isoflurane, and the muscle relaxant succinylcholine). All intra -<br />

venous agents and nondepolarizing relaxants are safe to use. 34 The<br />

anesthesia machine must be flushed of all halogenated anesthetics<br />

for a minimum period of 20 minutes (which can be longer<br />

depending on the characteristics of the anesthesia machine used),<br />

and a fresh anesthesia circuit must be used before administering<br />

anesthesia to patients susceptible to MH. The recommendations<br />

for the new ventilators are to flow 100% oxygen through the<br />

machine at 10 L/min for at least 20 minutes. 101,102 All patients must<br />

have their PETCO 2<br />

monitored. In children, the core temperature<br />

measurement is mandatory. It is highly recommended to have on<br />

hand enough DS (at least 18 vials).<br />

The absence of any sign of MH must be checked pre- and<br />

postoperatively. Central temperature and blood CK should be<br />

measured, and looking at the presence of light urine (taking into<br />

account the type of surgery) should be done before the outcome of<br />

the patient. No MH crisis studies have been published regarding<br />

these recommendations.<br />

CONCLUSION<br />

MH has nowadays a proteiform presentation that makes the<br />

diagnosis very difficult. It is necessary to assert the MHS by<br />

determination of the phenotype by IVCTs and confirmation of<br />

the genotype by exploration of the RYR1 gene. To have a better<br />

predictive value, these explorations should be done in the genetic<br />

proband and, if positive, extended to family members in accord -<br />

ance with the family tree according to the higher to the lower level<br />

of risk (i.e., beginning with the father, mother, and siblings,<br />

followed by the relatives).<br />

It is well demonstrated in epidemiologic studies that halo -<br />

genated volatile anesthetic agents can trigger MH. The depolar -<br />

izing neuromuscular blocker reinforces the crisis and can induce<br />

a strong MS. For unexplained reasons, MH crisis occurs more<br />

often in teenagers. However, one must remember that it may<br />

be observed at all stages of life. The prognosis of MH should be<br />

excellent if competent anesthesiologists administer the anesthesia.<br />

Proper monitoring must include PETCO 2<br />

and temperature. The<br />

operating room facility should always have immediate availability<br />

of DS, cold intravenous solutions, and intensive care modalities.<br />

Although MHS has been proved in patients presenting with<br />

exercise heat stroke, exercise and heat exposure are not contra -<br />

indicated in susceptible people.<br />

The existence of mutations in the RYR1 gene can be responsible<br />

for a congenital myopathy, CCD. These patients must be con -<br />

sidered MHS even if there is no sign of clinical myopathy in the<br />

majority of individuals bearing this MH mutation. Pediatric anes -<br />

thesiologists should be aware of the sudden risk of hyperkalemiainduced<br />

cardiac arrest after the administration of succinylcholine<br />

and volatile anesthetic agents in infants suffering from muscular<br />

dystrophies. Unfortunately, this clinical presentation is often the<br />

first indication of this medical condition and can precede the<br />

clinical diagnosis. Anesthesia is always possible in MHS patients<br />

providing that special precautions are taken, of which the most<br />

important is the total removal of all volatile halogenated anes -<br />

thetics agents and elimination of the depolarizing neuromuscular<br />

blocking medications.<br />

REFERENCES<br />

1. Denborough MA, Lovell RRH. Anesthetic deaths in a family. Lancet.<br />

1960;2:45.<br />

2. Rosenberg H, Davis M, James D, et al. Malignant hyperthermia. Orphanet<br />

J Rare Dis. 2007;24:21.<br />

3. Dépret T, Krivosic-Horber R. Malignant hyperthermia: new develop -<br />

ments in diagnosis and clinical management. Ann Fr Anesth Réanim.<br />

2001;20:838–852.<br />

4. Monnier N, Krivosic-Horber R, Payen JF, et al. Presence of two different<br />

genetic traits in malignant hyperthermia families: implication for genetic<br />

analysis, diagnosis, and incidence of malignant hyperthermia suscep -<br />

tibility. Anesthesiology. 2002;97:1067–1074.<br />

5. Britt B, Kalow W. Malignant hyperthermia: a statistical review. Can<br />

Anaesth Soc J. 1970;17:293–315.<br />

6. Britt BA, Nalda Felipe MA, Gottmann S, Khambatta HJ, (eds). Hereditary<br />

and epidemiologic aspects of malignant hyper thermia. In: Malignant<br />

Hyperthermia. Current Concepts. Bad Homburg. Normed Verlag; 1988.<br />

pp. 19–40.<br />

7. Krivosic-Horber R. Hyperthermie à propos d’un cas. Ann Fr Anesth<br />

Réanim. 1971;28:597–600.<br />

8. Ording H, Ranklev E, Fletcher R. Investigation of malignant hyper -<br />

thermia in Denmark and Sweden. Br J Anaesth. 1984;56:1183–1190.<br />

9. Ording H. Incidence of malignant hyperthermia in Denmark. Anesth<br />

Analg. 1985;64:700–704.<br />

10. Ellis FR, Halsall PJ, Harriman DG. The work of the Leeds malignant<br />

hyperpyrexia unit, 1971–1784. Anaesthesia. 1986;41:809–<strong>81</strong>5.<br />

11. Ellis FR, Halsall PJ, Christian AS. Clinical presentation of suspected<br />

malignant hyperthermia during anaesthesia in 402 probands. Anaesthesia.<br />

1990;45:838–841.<br />

12. Strazis KP, Fox AW. Malignant hyperthermia: a review of published cases.<br />

Anesth Analg. 1993;77:297–304.<br />

13. Larach MG, Brandom BW, Allen GC, et al. Cardiac arrests and deaths<br />

associated with malignant hyperthermia in North America from 1987 to<br />

2006: a report from the North American Malignant Hyperthermia<br />

Registry of the Malignant Hyperthermia Association of the United States.<br />

Anesthesiology. 2008;108:603–611.<br />

14. Rosero EB, Adesanya AO, Timaran CH, et al. Trends and outcomes of<br />

malignant hyperthermia in the United States, 2000 to 2005. Anesthesiology.<br />

2009;110:89–94.<br />

15. Peltz B, Carstens J. An unusual case of malignant hyperpyrexia. First<br />

reported case in a South African negro. Anaesthesia. 1975;30:346–350.<br />

16. Mayhew JF, Rudolph J, Tobey RE. Malignant hyperthermia in a sixmonth-old<br />

infant: a case report. Anesth Analg. 1978;57:262–264.<br />

17. Faust DK, Gergis SD, Sokoll MD. Management of suspected malignant<br />

hyperpyrexia in an infant. Anesth Analg. 1979;58:33–35.<br />

18. Sewall K, Flowerdew RM, Bromberger P. Severe muscular rigidity at<br />

birth: malignant hyperthermia syndrome? Can Anaesth Soc J. 1980;27:<br />

279–282.<br />

19. Bailey AC, Bloch EC. Malignant hyperthermia in a three-month-old<br />

American Indian infant. Anesth Analg. 1987;66:1043–1045.<br />

20. Wilhoit RD, Brown RE, Bauman LA. Possible malignant hyperthermia in<br />

a 7-week-old infant. Anesth Analg. 1989;68:688–691.<br />

21. Dubrow TJ, Wackym PA, Abdul-Rasool IH, et al. Malignant hyper -<br />

thermia: experience in the prospective management of eight children.<br />

J Pediatr Surg. 1989;24:163–166.<br />

22. Krivosic-Horber R, Adnet P, Krivosic I, et al. Diagnosis of susceptibility<br />

to malignant hyperthermia in children. Arch Fr Pediatr. 1990;47:421–424.<br />

23. Hinkle AJ, Dorsch JA. Maternal masseter muscle rigidity and neonatal<br />

fasciculations after induction for emergency cesarean section. Anesthe -<br />

siology. 1993;79:175–177.<br />

24. Semmler A, Rieger C, Lesinski R. A case report of malignant hyper -<br />

thermia in a 14-month-old boy. Anaesthesiol Reanim. 1994;19:21–22.<br />

25. Pennington GP, Joeris L. Malignant hyperthermia in a 3-month-old<br />

infant: a case report. J Med Assoc Ga. 1996;85:162–163.


CHAPTER <strong>81</strong> ■ Pediatric Features of Malignant Hyperthermia 1377<br />

26. Chamley D, Pollock NA, Stowell KM, et al. Malignant hyperthermia in<br />

infancy and identification of novel RYR1 mutation. Br J Anaesth. 2000;<br />

84:500–504.<br />

27. Kinouchi K, Okawa M, Fukumitsu K, et al. Two pediatric cases<br />

of malignant hyperthermia caused by sevoflurane. Masui. 2001;50:<br />

1232–1235.<br />

28. Bonciu M, de la Chapelle A, Delpech H, et al. Minor increase of endtidal<br />

CO 2<br />

during sevoflurane-induced malignant hyperthermia. Paediatr<br />

Anaesth. 2007;17:180–182.<br />

29. Ducart A, Adnet P, Renaud B, et al. Malignant hyperthermia during<br />

sevoflurane administration. Anesth Analg. 1995;80:609–611.<br />

30. Wedel DJ, Gammel SA, Milde JH, et al. Delayed onset of malignant<br />

hyperthermia induced by isoflurane and desflurane compared with<br />

halothane in susceptible swine. Anesthesiology. 1993;78:1138–1144.<br />

31. Kunst G, Kunst G, Graf BM, et al. Differential effects of sevoflurane,<br />

isoflurane, and halothane on Ca 2+ release from the sarcoplasmic reticulum<br />

of skeletal muscle. Anesthesiology. 1999;91:179–186.<br />

32. Bendixen D, Skovgaard LT, Ording H. Analysis of anaesthesia in patients<br />

suspected to be susceptible to malignant hyperthermia before diagnostic<br />

in vitro contracture test. Acta Anaesthesiol Scand. 1997;41:480–484.<br />

33. Carroll J. Increased incidence of masseter spasm in children with stra -<br />

bismus anesthetized with halothane and succinylcholine. Anesthesiology.<br />

1987;67:559–561.<br />

34. Krivosic-Horber R, Reyford H, Becq MC, et al. Effect of propofol on the<br />

malignant hyperthermia susceptible pig model. Br J Anaesth. 1989;62:<br />

691–693.<br />

35. Carr AS, Lerman J, Cunliffe M, et al. Incidence of malignant hyper -<br />

thermia reactions in 2,214 patients undergoing muscle biopsy. Can J<br />

Anaesth. 1995;42:2<strong>81</strong>–286.<br />

36. Halsall P, Cain P, Ellis F. Does postoperative pyrexia indicate malignant<br />

hyperthermia susceptibility? Br J Anaesth. 1992;68:209–210.<br />

37. Litman RS, Flood CD, Kaplan RF, et al. Postoperative malignant hyperthermia:<br />

an analysis of cases from the North American Malignant<br />

Hyperthermia Registry. Anesthesiology. 2008;109:825–829.<br />

38. Pollock AN, Langton EE, Couchman K, et al. Suspected malignant<br />

hyperthermia reactions in New Zealand. Anaesth Intensive Care. 2002;<br />

30:453–461.<br />

39. Krivosic-Horber R, Theunynck D, Adnet P. Capnography and anestheticinduced<br />

malignant hyperthermia. Agressologie. 1985;26:929–932.<br />

40. Burkman JM, Posner KL, Domino KB. Analysis of the clinical variables<br />

associated with recrudescence after malignant hyperthermia reactions.<br />

Anesthesiology. 2007;106:901–906.<br />

41. Gronert GA. Malignant hyperthermia. Anesthesiology. 1980;53:395–423.<br />

42. Kolb ME, Horne ML, Martz R. Dantrolene in human malignant hyper -<br />

thermia Anesthesiology. 1982;56:254–262.<br />

43. Flewellen EH, Nelson TE, Jones WP, et al. Dantrolene dose response in<br />

awake man: implications for management of malignant hyperthermia.<br />

Anesthesiology. 1983;59:275– 80.<br />

44. Shime J, Gare D, Andrews J, et al. Dantrolene in pregnancy: lack of<br />

adverse effects on the fetus and newborn infant. Am J Obstet Gynecol.<br />

1988;159:831–834.<br />

45. Lerman J, McLeod ME, Strong HA. Pharmacokinetics of intravenous<br />

dantrolene in children. Anesthesiology. 1989;70:625–629.<br />

46. Takehana K, Kawaguchi Y, Kuroda K, et al. Transient talipes equinus<br />

deformity of bilateral lower limbs following malignant hyperthermia: a<br />

case report and review of literature. J Orthop Sci. 2004;9:657–661.<br />

47. Larach MG, Localio AR, Allen GC, et al. A clinical grading scale to predict<br />

malignant hyperthermia susceptibility. Anesthesiology. 1994;80:771–779.<br />

48. Yousef MA, Vaida S, Somri M, et al. Changes in creatine phosphokinase<br />

(CK) concentrations after minor and major surgeries in children. Br J<br />

Anaesth. 2006;96:786–789.<br />

49. Schwartz L, Rockoff M, Koka B. Masseter spasm with anesthesia: inci -<br />

dence and implications. Anesthesiolgy. 1984;61:772–777.<br />

50. Kosko JR, Brandom BW, Chan KH. Masseter spasm and malignant<br />

hyperthermia: a retrospective review of a hospital-based pediatric<br />

otolaryngology practice. Int J Pediatr Otorhinolaryngol. 1992;23:45–50.<br />

51. Krivosic-Horber R, Sauvage M, Calmes M. Notre expérience à propos de<br />

6,000 anesthésies à l’halothane chez l’enfant. Ann Chir Inf. 1973;14:91–96.<br />

52. Van Der Spek AF, Reynolds PI, Fang WB, et al. Changes in resistance to<br />

mouth opening induced by depolarizing and non-depolarizing neuro -<br />

muscular relaxants. Br J Anaesth. 1990;64:21–27.<br />

53. Rosenberg H, Fletcher J. Masseter muscle rigidity and malignant hyperthermia<br />

susceptibility. Anesth Analg. 1986;654:161–164.<br />

54. Christian S, Ellis F, Halsall P. Is there a relationship between masseteric<br />

muscle spasm and malignant hyperpyrexia? Br J Anaesth. 1989;62:<br />

540–544.<br />

55. O’Flynn RP, Shutack JG, Rosenberg H, et al. Masseter muscle rigidity and<br />

malignant hyperthermia susceptibility in pediatric patients. An update<br />

on management and diagnosis. Anesthesiology. 1994;80:1228–1233.<br />

56. Adnet P, Krivosic-Horber R, Adamantidis M, et al. The association between<br />

the neuroleptic malignant syndrome and malignant hyperthermia. Acta<br />

Anaesthesiol Scand. 1989;33:676–680.<br />

57. Tobin JR, Jason DR, Challa VR, et al. Malignant hyperthermia and<br />

apparent heat stroke. JAMA. 2001;286:168–169.<br />

58. Wappler F, Fiege M, Steinfath M, et al. Evidence for susceptibility to<br />

malignant hyperthermia in patients with exercise-induced rhabdomyolysis.<br />

Anesthesiology. 2001;94:95–100.<br />

59. Nishio H, Sato T, Fukunishi S, et al. Identification of malignant hyperthermia-susceptible<br />

ryanodine receptor type 1 gene (RYR1) mutations in<br />

a child who died in a car after exposure to a high environ mental<br />

temperature. Leg Med (Tokyo). 2009;11:142–143.<br />

60. Britt B, Endrenyi L, Peters PL, et al. Screening of malignant hyperthermia<br />

susceptible families by creatine phosphokinase measurement and other<br />

clinical investigations. Can Anaesth Soc J. 1976;23(Suppl 3):263–284.<br />

61. Krivosic-Horber R, Reyford H, Adnet PJ. Unexplained increase in serum<br />

creatine kinase levels: its relation to malignant hyperthermia sus -<br />

ceptibility. Minerva Anesthesiol. 1994;60(Suppl 3):107–110.<br />

62. Weglinski MR, Wedel DJ, Engel AG. Malignant hyperthermia testing in<br />

patients with persistently increased serum creatine kinase levels. Anesth<br />

Analg. 1997;84:1038–1041.<br />

63. Kalow W, Britt BA, Richter A. The caffeine test of isolated human muscle<br />

in relation to malignant hyperthermia. Can Anaesth Soc J. 1977;24:678–694.<br />

64. Ellis FR, Harriman DG. A new screening test for susceptibility to<br />

malignant hyperpyrexia. Br J Anaesth. 1973;45:638.<br />

65. Ellis FR. European Malignant Hyperpyrexia Group. Br J Anaesth. 1984;<br />

56:11<strong>81</strong>–1182.<br />

66. Larach MG. Standardization of the caffeine halothane muscle contracture<br />

test. North American Malignant Hyperthermia Group. Anesth Analg.<br />

1989;69:511–515.<br />

67. Ording H. The European Malignant hyperthermia Group in vitro<br />

contracture test for diagnosis of malignant hyperthermia following the<br />

protocol of the European MH Group: results of testing patients surviving<br />

fulminant MH and un-related low-risk subjects. Acta Anaesthesiol Scand.<br />

1997;41:955–966.<br />

68. Maccani RM, Wedel DJ, Melton A, et al. Femoral and lateral femoral<br />

cutaneous nerve block for muscle biopsies in children. Paediatr Anaesth.<br />

1995;5:223–227.<br />

69. Ummenhofer W, Roesslein R, Sutter PM, et al. Muscle biopsy for<br />

malignant hyperthermia screening in children. Eur J Pediatr Surg. 1997;<br />

7:259–262.<br />

70. Isaacs H, Badenhorst M. False-negative results with muscle caffeine<br />

halothane contracture testing for malignant hyperthermia. Anesthesiology.<br />

1993;79:5–9.<br />

71. Monnier N, Romero NB, Lerale J, et al. An autosomal dominant con -<br />

genital myopathy with cores and rods is associated with a neomutation in<br />

the RYR1 gene encoding the skeletal muscle ryanodine receptor. Hum<br />

Mol Genet. 2000;9:2599–2608.<br />

72. Lynch PJ, Krivosic-Horber R, Reyford H, et al. Identification of<br />

heterozygous and homozygous individuals with the novel RYR1 mutation<br />

Cys35Arg in a large kindred. Anesthesiology. 1997;86:620–626.<br />

73. Islander G, Bendixen D, Ranklev-Twetman E, et al. Results of in vitro<br />

contracture testing of both parents of malignant hyperthermia susceptible<br />

probands. Acta Anaesthesiol Scand. 1996;40:579–584.<br />

74. McCarthy TV, Healy JM, Heffron JJ, et al. Localisation of the malignant<br />

hyperthermia susceptibility locus to human chromosome 19q 12–132.<br />

Nature. 1990;343:562–564.<br />

75. Monnier N, Kozak-Ribbens G, Krivosic-Horber R, et al. Correlations<br />

between genotype and pharmacological, histological, functional, and<br />

clinical phenotypes in malignant hyperthermia susceptibility. Hum Mutat.<br />

2005;26:413–425.<br />

76. Monnier N, Romero NB, Lerale J, et al. Familial and sporadic forms of<br />

central core disease are associated with mutations in the C-terminal<br />

domain of the skeletal muscle ryanodine receptor. Hum Mol Genet.<br />

2001;10:25<strong>81</strong>–2592.<br />

77. Girard T, Litman RS. Molecular genetic testing to diagnose malignant<br />

hyperthermia susceptibility. J Clin Anesth. 2008;20:161–163.


1378 PART 4 ■ Special Monitoring and Resuscitation Techniques<br />

78. Urwyler A, Deufel T, McCarthy T, et al. European Malignant Hyper -<br />

thermia Group. Guidelines for molecular genetic detection of suscep -<br />

tibility to malignant hyperthermia. Br J Anaesth. 2001;86:283–287.<br />

79. Girard T, Johr M, Schaefer C, et al. Perinatal diagnosis of malignant<br />

hyperthermia susceptibility. Anesthesiology. 2006;104:1353–1354.<br />

80. Krivosic-Horber R, Krivosic I. Central core disease associated with<br />

malignant hyperthermia sensitivity. Presse Med. 1989;18:828–831.<br />

<strong>81</strong>. Quinlivan RM, Muller CR, Davis M, et al. Central core disease: clinical,<br />

pathological, and genetic features. Arch Dis Child. 2003;88:1051–1055.<br />

82. Stamm SD, AylsworthAS, Stajich JM, et al. Native American myopathy:<br />

congenital myopathy with cleft palate, skeletal anomalies, and sus -<br />

ceptibility to malignant hyperthermia. Am J Med Genet. 2008;146A:1832–<br />

1841.<br />

83. King JO, Denborough MA. Anesthetic-induced malignant hyperpyrexia<br />

in children. J Pediatr. 1973;83:37–40.<br />

84. Habib AS, Millar S, Deballi P, et al. Anesthetic management of a<br />

ventilator-dependent parturient with the King-Denborough syndrome.<br />

Can J Anaesth. 2003;50:589–592.<br />

85. D’Arcy CE, Bjorksten A, Yiu EM, et al. King-denborough syndrome<br />

caused by a novel mutation in the ryanodine receptor gene. Neurology.<br />

2008;71:776–777.<br />

86. Duchenne and Becker muscular dystrophy. Available at: www.orphanet.<br />

net Accessed September 14, 2010.<br />

87. Larach MG, Rosenberg H, Gronert GA, et al. Hyperkalemic cardiac arrest<br />

during anesthesia in infants and children with occult myopathies. Clin<br />

Pediatr (Phila). 1997;36:9–16.<br />

88. Pedrozzi NE, Ramelli GP, Tomasetti R, et al. Rhabdomyolysis and<br />

anesthesia: a report of two cases and review of the literature. Pediatr<br />

Neurol. 1996;15:254–257.<br />

89. Breucking E, Reimnitz P, Schara U, et al. Anesthetic complications.<br />

The incidence of severe anesthetic complications in patients and families<br />

with progressive muscular dystrophy of the Duchenne and Becker types.<br />

Anaesthesist. 2000;49:187–195.<br />

90. Schulte-Sasse U, Eberlein HJ, Schmücker I, et al. Should the use of<br />

succinylcholine in pediatric anesthesia be re-evaluated? Anaesthesiol<br />

Reanim. 1996;18:13–19.<br />

91. Goudsouzian NG. Recent changes in the package insert for succinyl -<br />

choline chloride: should this drug be contraindicated for routine use in<br />

children and adolescents? (Summary of the Discussions of the<br />

Anesthetic and Life Support Drug Advisory Meeting of the Food and<br />

Drug Administration, FDA Building, Rockville, MD, June 9, 1994.)<br />

Anesth Analg. 1995;80:204–212.<br />

92. Hayes J, Veyckemans F, Bissonnette B. Duchenne muscular dystrophy:<br />

an old anesthesia problem revisited. Paediatr Anaesth. 2008;18:<br />

100–106.<br />

93. Flick RP, Gleich SJ, Herr MM, et al. The risk of malignant hyperthermia<br />

in children undergoing muscle biopsy for suspected neuromuscular<br />

disorder. Paediatr Anaesth. 2007;17:22–27.<br />

94. Bollig G, Mohr S, Raeder J. McArdle’s disease and anaesthesia: case<br />

reports. Review of potential problems and association with malignant<br />

hyperthermia. Acta Anaesthesiol Scand. 2005;49:1077–1083.<br />

95. Ellis FR, Halsall PJ, Harriman DG. Malignant hyperpyrexia and sudden<br />

infant death syndrome. Br J Anaesth. 1988;60:28–30.<br />

96. Lee CK, Chang BS, Hong YM, et al. Spinal deformities in Noonan syn -<br />

drome: a clinical review of sixty cases. J Bone Joint Surg Am. 2001;83:<br />

1495–1502.<br />

97. Porsborg P, Astrup G, Bendixen D, et al. Osteogenesis imperfecta and<br />

malignant hyperthermia. Is there a relationship? Anaesthesia. 1996;51:<br />

863–865.<br />

98. Hollander AS, Olney RC, Blackett PR, et al. Fatal malignant<br />

hyper thermia–like syndrome with rhabdomyolysis complicating the<br />

presen tation of diabetes mellitus in adolescent males. Pediatrics. 2003;111:<br />

1447–1452.<br />

99. Kilbane BJ, Mehta S, Backeljauw PF, et al. Approach to management of<br />

malignant hyperthermia–like syndrome in pediatric diabetes mellitus.<br />

Pediatr Crit Care Med. 2006;7:169–173.<br />

100. Yentis SM, Levine MF, Hartley EJ. Should all children with suspected<br />

or confirmed malignant hyperthermia susceptibility be admitted after<br />

surgery? A 10-year review. Anesth Analg. 1992;75:345–350.<br />

101. Crawford MW, Prinzhausen H, Petroz GC. Accelerating the washout of<br />

inhalational anesthetics from the Dräger Primus anesthetic workstation:<br />

effect of exchangeable internal components. Anesthesiology. 2007;2:<br />

289–294.<br />

102. Gunter JB, Ball J, Than-Win S. Preparation of the Dräger Fabius<br />

anesthesia machine for the malignant-hyperthermia susceptible patient.<br />

Anesth Analg. 2008;107:1936–1945.


1380 PART 4 ■ Special Monitoring and Resuscitation Techniques<br />

Figure 82-1. Immune response.<br />

ANESTHETIC AGENTS AND<br />

IMMUNE FUNCTION<br />

Numerous studies using clinical and in vitro techniques have<br />

examined the effects of inhaled and intravenous anesthetic agents<br />

on the function of immune cells (Table 82–1). 8–16 The resulting<br />

data reveal multiple and often conflicting effects on immune cells;<br />

however, the preponderance of findings suggest that, in most cir -<br />

cumstances, volatile and intravenous anesthetics depress immune<br />

cell functions.<br />

In attempt to understand the immune system effects of<br />

anesthesia-related drugs during surgery in children, it is important<br />

to consider the context of the evidence. Procopio and coworkers<br />

exposed healthy volunteers to mask thiopental isoflurane–nitrous<br />

oxide general anesthesia and then to lidocaine lumbar epidural<br />

anesthesia in the absence of surgery; the immune function of<br />

sampled ex vivo cells from these volunteers was unaffected. 17<br />

Clearly, the interaction among the stressors of surgery, the per -<br />

ianesthesia environment, and drugs of anesthesia is necessary to<br />

account for postoperative immune modulation. In considering the<br />

extremely complex milieu of a surgical patient under anesthesia,<br />

the net immune effect of these drugs may not be deleterious,<br />

because it will be determined not just by the drug’s propensity to<br />

directly depress immune cell function but also by its efficacy in<br />

TABLE 82-1. Effect of Anesthetic Drugs and Opioids on Immune Cell Function<br />

Immune Effects<br />

Macrophage exposure decreases mediators molecule release.<br />

Transcription factors blocked in T lymphocytes, cell death (apoptosis) is induced.<br />

Lymphocyte functions are suppressed.<br />

Less suppressive in two studies; other investigations found immune cell impairment<br />

on exposure.<br />

Inflammation reduced; may favor adaptive immunity<br />

Direct, M opioid receptor–mediated depression of immune cell function; indirect support<br />

of adaptive immune function through decreasing stress hormone release; potential antiinflammatory<br />

effects.<br />

Anesthesia<br />

All inhaled anesthetics<br />

Sevoflurane, isoflurane<br />

Thiopental, etomidate, ketamine<br />

Propofol<br />

Amide local anesthetics<br />

Opioids


CHAPTER 82 ■ Influence of Anesthesia on the Immune System in Children 13<strong>81</strong><br />

suppressing various aspects of the stress response to the surgical<br />

intervention.<br />

Anesthesia and Adaptive Immune Response<br />

Previous reports had demonstrated that anesthetics can influence<br />

various aspects of lymphocyte function in vitro and in vivo, but<br />

the results of these were often contradictory. 18–21 Stevenson and<br />

colleagues observed that anesthesia and surgery depress both<br />

T-cell and B-cell responsiveness as well as nonspecific host resist -<br />

ance mechanisms, including phagocytosis. 22 Barbiturates, such<br />

as thiopental, have already been shown to reduce the growth of<br />

phytohemagglutinin (PHA)-stimulated peripheral blood mono -<br />

nuclear cells (PBMCs) or isolated T lymphocytes. 12,23,24 The effects<br />

are more pronounced in neonates than in older children.<br />

The depressed proliferative T-cell response makes patients<br />

susceptible to postoperative infections, 25 and barbiturates adminis -<br />

tered over long periods may cause iatrogenic immunosuppression.<br />

Indeed, a higher incidence of infections has been described in<br />

head-injured patients receiving prolonged infusions of thiopental<br />

to control increased intracranial pressure. 26,27<br />

Thiopental has an elimination half-life of approximately<br />

11 hours and may accumulate during long-term administration.<br />

Thus, the in vivo effects of thiopental on the proliferative capacity<br />

of lymphocytes may be of particular relevance. The reduction of<br />

mitogen-induced PBMC proliferation is comparable with that<br />

induced by 10 –7 mol/L methylprednisolone. 28<br />

Propofol has been shown to produce a mild inhibition of PHAinduced<br />

PBMC proliferation at high concentrations. 29 These data<br />

are in accordance with results published by Pirttikangas and<br />

associates, who observed a proliferation suppressing effect of<br />

propofol in vitro only in PBMCs obtained from critically ill<br />

patients, who were primarily immunosuppressed. 29 Devlin and<br />

coworkers investigated the effects of thiopental and propofol on<br />

lymphocyte proliferation after PHA stimulation. 12 In their studies,<br />

neither propofol nor its solvent intralipid caused T lymphocyte<br />

depression. 12,24 In this context, the authors concluded that propofol<br />

may be the safest drug for patients undergoing prolonged surgery<br />

or for sedation in the intensive care unit.<br />

The activation of T and B lymphocytes is characterized by<br />

the release of the autocrine growth factor interleukin-2 (IL-2).<br />

The up-regulation of the high-affinity IL-2 receptor trimmer and<br />

the simultaneous release of a soluble form of the IL-2 receptor<br />

α-subunit (shedding) are suggested. Dysregulation of IL-2-<br />

production and IL-2 receptor expression in the accessory signaling<br />

pathways have been shown to result in immune defects or autoimmune<br />

diseases. 30-31 This can explain the immune suppression<br />

effect of certain anesthetic drugs. Conversely, whereas the release<br />

of sIL-2R was depressed in the presence of higher propofol<br />

concentrations, IL-2 production by PBMC was found to increase<br />

with increasing propofol concentrations. Salo and colleagues did<br />

not find any effect of propofol on IL-2 production, but described<br />

an increased production of the Thl cytokine interferon-γ (IFN-γ)<br />

by isolated T cells with an increase in the IFN-γ/IL-4 ratio at<br />

propofol concentrations up to 10 pg/mL. 32 In addition, barbi -<br />

turates, but not propofol, suppressed the activation of transcrip -<br />

tion factor nuclear κB in human T cells. 33<br />

Loop and associates described that, with thiopental, there was<br />

a decrease in the production of the IL-2, IL-6, and IL-8 as well as<br />

IFN-γ by CD3+ lymphocytes. 11 This group provided data sug -<br />

gesting that thiopental interferes with activation of nuclear<br />

transcription factor κB in T-lymphocytes, which is probably<br />

mediated via the suppression of κB kinase.<br />

Furthermore, the immunosuppressive effects of thiopental are<br />

associated with a marked reduction in the density of IL-2 receptors<br />

on the cell membrane and the numbers of cells expressing<br />

IL-2 receptors. 12<br />

In summary, a decrease in lymphocytes, T- and B-cell counts,<br />

as well as a depression of lymphocyte reactivity and alteration in<br />

B-cell response to pokeweed mitogen (PWM) due to use of anes -<br />

thesia were all inversely related to age of children from 1 month to<br />

12 years.<br />

Opioid use is ubiquitous in anesthesia practice, because these<br />

drugs are most effective and essential in the treatment of acute<br />

surgical pain and are used as an important component of chronic<br />

pain management. For these reasons and because of their complex<br />

effects on perioperative immune function, extra space is devoted<br />

to presenting relevant evidence. Review of the relevant literature<br />

reveals multiple and often conflicting reports of opioid effects on<br />

immune system functions (see Table 82–1).<br />

Of great concern when using opioids to manage perioperative<br />

pain is evidence that opioid analgesics are immunosuppressive.<br />

Animal studies have shown the prototypical opioid morphine to<br />

suppress NK cell activity, mitogen-induced lymphocyte prolif -<br />

eration, and inflammatory cytokine production. 34 Other drugs<br />

with mu-opioid receptor agonist effects, including buprenorphine<br />

and fentanyl, also depress immune function in a naltrexonereversible<br />

manner. 35 In contrast with these findings, a preclinical<br />

study found immune suppression associated with fentanyl was<br />

limited to the first 72 postoperative hours and was nonexistent for<br />

buprenorphine. 36 Interestingly, in postoperative animals, the im -<br />

mune suppression associated with surgery was ameliorated with<br />

morphine administration, whereas buprenorphine restored im -<br />

mune parameters to normal levels. By using a well-demonstrated<br />

animal model of postoperative pain and tumor metastasis,<br />

researchers have shown that animals treated with fentanyl or<br />

intrathecal morphine/bupivacaine have a significantly lower<br />

tumor burden than animals without postoperative analgesia. 37<br />

It is important to note that the fentanyl-associated immune cell<br />

depression seen in control animals was absent in the postoperative<br />

group. In contrast with the effects of morphine and fentanyl,<br />

tramadol, a novel opioid analgesic with inhibitory effects on<br />

norepinephrine and serotonin uptake, has been found to have no<br />

depressant effects on immune parameters in a study of postop -<br />

erative pain treatment in patients with cancer. 38<br />

However, there are well-documented, dose-dependent, immu -<br />

nosuppressive effects of morphine that are known to impair mon -<br />

ocyte and neutrophil functions, NK cell–mediated cytotoxicity,<br />

lymphocyte proliferation, and cytokine release. Morphine pro -<br />

motes apoptosis in lymphocytes and macrophages by activating<br />

enzymes involved in apoptotic cell death. Furthermore, it affects<br />

nitric oxide release and inhibits cell adhesion. Opioids are known<br />

to exert their effects via specific opioid receptors expressed on<br />

immunocompetent cells. It is reported that morphine induces<br />

DNA damage through the action on the kappa opioid receptor,<br />

which leads to immune suppression by activation of P53-mediated<br />

signal transduction. Studies estimating the effects of synthetic<br />

opioids used in general anesthesia showed no more transient<br />

immunomodulatory changes. 37–39<br />

Another study by Yeager and coworkers reported enhanced<br />

NK-cell cytotoxicity and increased relative number of CD16+ and


1382 PART 4 ■ Special Monitoring and Resuscitation Techniques<br />

CD8+ after an intravenous bolus dose and subsequent infusion of<br />

fentanyl (1.2 pg/kg/h for 2 h) in healthy volunteers. 40<br />

In a similar study, fentanyl increased the NK-cell (CDl6+/<br />

CD56+) number, but superoxide production of polymorpho -<br />

nuclear cells and the number of circulating B and T lymphocytes<br />

remained unchanged. 41 These results suggest a centrally mediated<br />

rather than a direct effect of fentanyl on NK cells.<br />

Previous in vitro studies have provided evidence that volatile<br />

anesthetics might alter the immune response. In cultured human<br />

leucocytes, halothane inhibited mitogen-induced RNA and<br />

protein synthesis and depressed the secretion of IFN. 42<br />

Extended exposure suppressed the mitogen-induced lym -<br />

phocyte proliferation and the expression of the IL-2 receptor.<br />

Mitsuhata and colleagues investigated the effects of volatile anes -<br />

thetics (sevoflurane, isoflurane, enflurane) in clinically relevant<br />

concentrations on the cytokine release of human PBMC-stimulated<br />

NK-sensitive tumor cells. 43 None of the anesthetics reduced the<br />

levels of IL-2.<br />

ANESTHESIA AND PHAGOCYTOSIS<br />

An intact phagocytic function is essential for the host defense<br />

(Figure 82–2). Several human and animal studies of the anesthetic<br />

effects of phagocytic activity in the past have produced<br />

contradictory evidence. At the turn of the century, animal studies<br />

concluded that ether and chloroform inhibited phagocytosis in a<br />

dose-dependent manner.<br />

Human studies after either halothane or nitrous oxide–narcotic<br />

anesthesia without surgery revealed a decrease in phagocytosis of<br />

latex particles and nitroblue tetrazolium (NBT) reduction in one<br />

instance, but showed only a minimal inhibition in another after<br />

halothane 0.5 to 2.5% or nitrous oxide 80%. Reduced phagocytic<br />

activity by fixed macrophages of the reticuloendothelial system<br />

has also been reported during anesthesia in humans and in<br />

animals, although the reduction was only minimal. In addition to<br />

the effects of volatile inhalational anesthetic agents, a study of the<br />

effects of intravenous induction and local anesthetic agents on the<br />

human leukocyte phagocytic activity revealed a dose-dependent,<br />

statistically significant depression of phagocytic activity after in<br />

vitro exposure to varying concentrations of intravenous induction<br />

agents, narcotics, and local anesthetic agents. These observations<br />

were not the result of a nonspecific drug concentration effect,<br />

because equimolar concentrations of other nonanesthetic agents<br />

failed to produce any depression of phagocytosis. 44<br />

Thus, it would appear that some of the anesthetic agents may<br />

potentially enhance the risk of perioperative infection by reducing<br />

the phagocytic activity. This depression may be further compounded<br />

by the surgical intervention per se, because several other<br />

studies have shown impaired leukocyte function by the stress of<br />

surgery both in humans and in animals. 30<br />

In addition to a decrease in phagocytosis, anesthetic agents can<br />

also inhibit bactericidal activities. Earlier studies utilized NBT<br />

reduction as an index of bactericidal activity.<br />

More recently, utilizing the technique of chemiluminescence, in<br />

which the light emission by the highly reactive and excited oxygen<br />

radical is evaluated, it was shown that only enflurane and not<br />

isoflurane inhibited the bactericidal activity. Similar observations<br />

were also made after exposure to halothane. Using the superoxideinduced<br />

chemiluminescence, a dose-dependent depression of bac -<br />

tericidal activity was also reported with thiopentone and althesin;<br />

however, methohexitone, morphine, diazepam, and lidocaine<br />

failed to produce similar effects. These findings support the<br />

concept that different anesthetic agents may interfere with various<br />

aspects of the nonspecific immune response.<br />

A suppression of leukocyte function in particular could be<br />

relevant in the pathogenesis of postoperative infection. However,<br />

the true clinical significance of these observations remains to<br />

be ascertained. 45<br />

Figure 82-2. Phagocytosis steps in<br />

neutrophils.


CHAPTER 82 ■ Influence of Anesthesia on the Immune System in Children 1383<br />

Figure 82-3. Anaphylactic reaction.<br />

ANESTHESIA AND<br />

ALLERGIC REACTIONS<br />

Intraoperative allergic reactions occur once in every 5000 to<br />

25,000 anesthetics with 3.4% mortality. 46,47 More than 90% of<br />

allergic reactions evoked by intravenous drugs occur within<br />

3 minutes of administration; this is usually called an anaphylactic<br />

reaction (Figure 82–3). In the anesthetized patient, an anaphylactic<br />

reaction is the most common life-threatening manifestation of an<br />

allergic reaction and is associated with circulatory collapse,<br />

reflecting vasodilatation with resulting decreased venous return<br />

and cardiac output. Although the immune system functions to<br />

provide host defense, it can respond inappropriately to produce<br />

various hypersensitivity reactions. A spectrum of life-threatening<br />

allergic reactions to any drug can occur in the perioperative<br />

period. The enigma of these reactions lies in their unpredictable<br />

nature.<br />

However, a high index of suspicion and vigilance, prompt<br />

recognition, and appropriate and aggressive therapy can help to<br />

avoid a disastrous outcome.<br />

Recognition of Anaphylaxis<br />

The onset and severity of the reaction relate to the mediator’s<br />

specific end-organ effects. Antigenic challenge in a sensitized<br />

individual usually produces immediate clinical manifestations,<br />

allowing only a short period before appropriate therapy. Indi -<br />

viduals may vary greatly in their manifestation and course of<br />

anaphylaxis, ranging from minor clinical features to the full-blown<br />

syndrome leading to death. The enigma of anaphylaxis lies in the<br />

unpredictability of its occurrence, severity of attack, and lack of a<br />

previous allergic history. 46<br />

Diagnostic Tests<br />

Tests performed during the reaction include<br />

●<br />

Plasma histamine levels<br />

Plasma histamine levels when elevated, particularly to levels<br />

greater than 20 nmol, confirm that its involvement in the<br />

immune reaction is anaphylactic. The converse that no elevation<br />

in plasma histamine levels means a lack of its involvement is not<br />

true. Plasma histamine levels rise only transiently and sampling<br />

must occur within 10 minutes. 47<br />

Urinary or plasma methylhistamine<br />

Even though measurement of urinary or plasma methyl -<br />

histamine has the advantages over plasma histamine because it<br />

has a more prolonged rise and provides a simple assay, it is less<br />

sensitive.<br />

Immunoglobulin E levels<br />

Drugs specific immunoglobulin E (IgE) can be detected in<br />

blood taken during an immune reaction or before a reaction<br />

and postmortem and usually reflects the results of delayed<br />

sampling at the time of skin testing. 47<br />

Complement levels<br />

As a diagnostic tool in clinical anaphylaxis, complement level<br />

measurement has limited value. Changes in serum complement<br />

levels and activation of the classic and alternate pathways have<br />

been demonstrated after clinical anaphylaxis, particularly due<br />

to adhesion, protamine, and contrast media. 48,49 Activation<br />

should be measured rather than levels of complement fraction,<br />

because there are major dilutional effects during anaphylaxis. 50<br />

Mast cell tryptase<br />

Mast cell tryptase is an important advance in the diagnosis of<br />

anaphylactoid reactions during anesthesia. In anaphylactic reac -<br />

tions, the levels are elevated for 1 to 5 hours after the beginning<br />

of the reaction, enabling the delay of sampling until resuscitation<br />

is over, 51 and can be obtained in samples taken postmortem. 52<br />

Elevated mast cell tryptase levels are highly specific and sensitive<br />

of an immune reaction, with the exception of reactions to gelatin<br />

and contrast. All patients with elevated mast cell tryptase levels<br />

should be considered to have a severe immune reaction and<br />

investigated further.<br />

●<br />

●<br />

●<br />

●<br />

Tests After the Reaction<br />

●<br />

Skin testing<br />

Skin testing is performed 4 to 6 weeks after the reaction. 53 Skin<br />

tests have the advantages over other tests because of the high<br />

yield of positive results (reflecting the high incidence of IgE


1384 PART 4 ■ Special Monitoring and Resuscitation Techniques<br />

●<br />

involve ment in severe reactions) and ease in performing the<br />

tests. 54–56 Two forms of skin testing are used—intradermal tests<br />

and prick tests.<br />

Prick testing is less traumatic to the skin, easily done, and<br />

safe. Their disadvantage is that they tend to produce more false<br />

negatives, whereas intradermal tests tend to produce more false<br />

positives. The consequences of a false-negative test for<br />

subsequent anesthesia are obviously greater than those of a false<br />

positive. Skin tests are of little value in reactions to colloids,<br />

contrast media, and blood products.<br />

With local anesthetics, genuine anaphylactic reactions are<br />

rare, and the goal is to exclude allergy. Skin testing on its own is<br />

inadequate. If there was not a clear-cut history suggestive of<br />

anaphylaxis and a clear-cut positive wheal and flare reaction<br />

at 1:100 dilution of 0.5% local anesthetic, the dosage should be<br />

increased to 2 mL of undiluted local anesthetic solution. It is<br />

important to note that deaths have occurred in association to<br />

skin testing. Resuscitation facilities should always be available.<br />

Radioimmunoassay tests<br />

Radioimmunoassay (RIA) tests have been used to detect IgE<br />

drug-specific antibodies. RIA tests have sensitivity similar to<br />

skin tests. However, RIAs are available for propofol, thiopen -<br />

tone, succinylcholine, vecuronium, pancuronium, gallamine,<br />

D-tubocurarine, and alcuronium. Combination of RIA and skin<br />

tests detects a drug responsible for reaction better than either<br />

test alone.<br />

Cross-sensitivities are better determined by RIAs than by<br />

cutaneous tests. There is significant in vitro cross-sensitivity<br />

between thiopentone and non-depolarizing muscle relaxants<br />

(NDMR), which is not reflected clinically. However, in patients<br />

who are allergic to both, skin tests and RIA inhibition are<br />

necessary to distinguish. 57<br />

In practice, when the results of tests disagree, the patient<br />

should be warned off all positive drugs.<br />

In summary, an often-overlooked factor in the investigation<br />

of patients with anesthetic allergy is the need for accurate docu -<br />

mentation and communication. The patients should be encour -<br />

aged to wear a warning device (e.g., bracelet) stating the drugs that<br />

should be avoided. It should also be accompanied with detailed<br />

information about the potential reaction if the medication is given,<br />

which tests were performed, the results of those tests, and the<br />

conclusion. Anesthetic allergy has been shown to persist up to<br />

27 years, and few patients lose their sensitivity over this period<br />

of time. 58,59<br />

Preoperative Testing<br />

Recently, it has been argued that patients presenting for anesthesia<br />

should be preoperatively screened by RIA/skin tests to reduce the<br />

risk of anaphylaxis. The case for this testing is poor. The selection<br />

of at-risk group, such as women of child-bearing age with or<br />

without a history of allergy, will improve the cost-effectiveness<br />

of testing and lead to a major reduction in reactors detected.<br />

If screening is performed, prick testing would be the only feasible<br />

and sufficiently reliable test.<br />

The most important variable in the case of screening is the<br />

prevalence of anesthetic allergy, which is different from and<br />

unlikely to be greater than the incidence of reactions, because a<br />

number of allergic patients will undergo anesthesia without<br />

exposure to a drug to which they may be allergic. In spite of the<br />

limitations of diagnostic tests, when patients are investigated using<br />

skin testing and RIA where available, subsequent anesthesia is<br />

usually safe, irrespective of whether the reaction was diagnosed as<br />

anaphylactic or not or whether the drug responsible was detected.<br />

EFFECT OF ANESTHESIA ON<br />

VACCINATION OF CHILDREN<br />

Anesthesia and surgery exert immunomodulatory effects, and<br />

some authors argue that they may exert additive or synergistic<br />

influences on vaccine efficacy and safety. Alternatively, inflam -<br />

matory responses and fever elicited by vaccines may interfere with<br />

the postoperative course. There is a lack of consensus approach<br />

among anesthesiologists to the theoretical risk of anesthesia and<br />

vaccination. Few studies have assessed the influence of anesthesia<br />

and surgery on pediatric vaccine responses. 60<br />

Anesthetists are often faced with a child who has been recently<br />

immunized presenting for either emergency or elective surgery. The<br />

question is then raised as to whether anesthesia or surgery will<br />

affect response of child to vaccine in achieving seroconversion or,<br />

more seriously, whether the vaccine may cause more serious<br />

adverse reactions in these circumstances. A further question may be<br />

raised as to how soon after surgery it is safe to administer vaccine. 61<br />

There is no direct evidence of any major interaction between<br />

immunization and commonly used anesthetic agents and tech -<br />

niques in children, but it is possible that immunosuppression<br />

caused by anesthesia and surgery may lead to decreased vaccine<br />

effectiveness or an increased risk of complications. 62,63<br />

Based on the theoretical risk of interaction between anesthesia<br />

and immunization, it is believed that it could be eliminated by<br />

ensuring a 3-week delay between the two. An international survey<br />

was undertaken to discover whether there is a consensus among<br />

pediatric anesthetists on this issue. 62 Two hundred and ninetysix<br />

(52.1%) Association of Paediatric Anaesthetists of Great Britain<br />

and Ireland (APAGBI) and 86 (49.4%) Society of Paediatric<br />

Anaesthesia in New Zealand and Australia (SPANZA) responses<br />

were analyzed. There was no consensus of approach to this theore -<br />

tical risk among respondents. In total, 60% of respondents would<br />

anesthetize a child for elective surgery within 1 week of receiving<br />

a live, attenuated vaccine, but 40% would not. Few hospitals have<br />

formal policies on this issue, and government guidance is based on<br />

a lack of evidence for adverse events rather than positive evidence<br />

of safety. An international postal survey failed to find a consensus<br />

to this risk among pediatric anesthetists. From a risk management<br />

perspective, a review of the available evidence suggests that it<br />

would be prudent to adopt a cautious approach in which the<br />

timing of elective surgery is discretionary.<br />

In children, few studies have assessed the influence of anes -<br />

thesia on immune responses. Hence, anesthetists and surgeons are<br />

confronted with unanswered questions 60 :<br />

1. Are there interferences between general anesthesia and routine<br />

vaccination?<br />

2. Should elective surgery under general anesthesia be postponed<br />

in recently vaccinated children?<br />

3. Should elective vaccination be postponed in children scheduled<br />

for elective surgery?<br />

4. What are the risks of a general anesthesia in recently vaccinated<br />

children, particularly when anesthesia must be performed in<br />

case of emergency?<br />

Children are routinely immunized from a very early age to<br />

protect them against the devastating morbidity and mortality


CHAPTER 82 ■ Influence of Anesthesia on the Immune System in Children 1385<br />

previously associated with many infectious diseases. Such immu -<br />

nization relies on the activation of the immune system in response<br />

to modified antigen, the formation of immunologic memory, and<br />

an enhanced immune response to subsequent exposure to the<br />

disease. Effective immunization thus requires an intact, func -<br />

tioning immune system. Exposure to modified antigen, as well as<br />

triggering the production of protective antibodies, may cause side<br />

effects ranging from mild fever to development of a noninfectious<br />

form of the disease. 64<br />

The first risk the authors identify is that the efficacy of<br />

vaccination may be reduced by the immunosuppressive effect of<br />

anesthesia and surgery. The duration of any immunosuppression<br />

because of anesthesia is short, and there is no evidence from<br />

humans that any of the routine childhood vaccines have failed<br />

because of anesthesia or surgery. 65<br />

If anesthetics or surgery was significantly immunosuppressive,<br />

then infections such as endogenous bacteremias would be regular<br />

sequelae, but this is not observed. The example that Short and<br />

associates cite is a single case report of the failure of postexposure<br />

rabies vaccination of a child bitten on the face. 62 Such failures are<br />

recognized irrespective of anesthesia or surgery and are most<br />

frequent in facial bites and when, as in this case, administration of<br />

human rabies immunoglobulin is delayed.<br />

Comparing this scenario with the routine immunization<br />

program in which children receive multiple doses of all the recom -<br />

mended vaccines. It is unlikely that anesthesia after any one dose<br />

of vaccine would have any significant impact on the efficacy of the<br />

whole series. For example, antibody levels are proxies for immu -<br />

nity and a poor measure of immunologic priming. Furthermore,<br />

if there were a concern about the efficacy of a vaccine being<br />

reduced, the most logical way to ensure that the child is sufficiently<br />

protected would be to give him or her an additional dose of<br />

vaccine, not to delay vaccination. 65<br />

The second risk is of increased complications after vaccination<br />

at the same time as a child receives an anesthetic and surgery.<br />

This possibility would again be more important in relation to the<br />

live, attenuated vaccines such as polio, and measles, mumps, and<br />

rubella (MMR). 62<br />

Healthy children receiving immunizations with killed vaccine<br />

commonly suffer mild reactions with local symptoms of redness,<br />

swelling at the injection site, mild fever, and irritability. 61 This<br />

occurs within a limited time of a few days and would be unlikely<br />

to complicate postoperative assessment. Fever, which would be<br />

more difficult to distinguish from a postoperative complication,<br />

occurs less frequently. 66 There has also been a switch to using<br />

acellular pertussis vaccine rather than whole cell pertussis vaccine,<br />

because it causes fewer localized reactions. 62 The period of risk<br />

is again a few days shorter than the 1-week delay in surgery<br />

suggested by the authors. 65<br />

After administration of live vaccine, the reaction may be<br />

delayed and present as a mild form of the illness. The reaction will,<br />

therefore, depend on the incubation period of the illness itself<br />

so that mild measles may occur 5 to 10 days after MMR and<br />

mild mumps 14 to 21 days after MMR. If mild rubella occurs, it is<br />

hard to distinguish from measles; severe reactions are rare, for<br />

example 61 :<br />

1. Polio<br />

One in 2 or 3 million doses of vaccine can revert toward the<br />

characteristics of the wild type and could then cause polio in<br />

nonimmunized persons or close contacts such as other children<br />

on the ward. 67 The vaccine is genetically unstable and may<br />

change its structure. 68<br />

The U.K. Immunization “Green Book” states that the<br />

possibility of a very small risk of poliomyelitis induced by oral<br />

vaccine cannot be ignored but is insufficient to warrant a<br />

change in immunization policy. 64 Since the book was published<br />

in 1996, however, there has been just such a change in policy,<br />

and the use of inactivated polio vaccine became routine in the<br />

United Kingdom at the end of 2004, eliminating the small risk<br />

of vaccine-associated polio. 62<br />

2. MMR<br />

Complications of these illnesses can occur, but more rarely with<br />

the vaccine than with the illness itself. For example, mumps<br />

meningitis occurs in about 1 in 300,000 immunizations but in<br />

approximately 1 in 1000 cases of mumps. Idiopathic thrombo -<br />

cytopenic purpura occurs as a complication of MMR, but it is<br />

extremely rare, affecting fewer than 1 in 32,000 children from<br />

MMR. 69 This reaction occurs up to 6 weeks after vaccination,<br />

so the 3-week period proposed in papers would not exclude it.<br />

Similarly, vaccine-derived mumps occurs 3 to 4 weeks after<br />

vaccination. As a result of the fact that the MMR vaccine con -<br />

tains live, attenuated viruses, it is also considered biologically<br />

plausible that it could cause encephalitis, although the risk is<br />

believed to be small. 62<br />

It may be policy in some units that anesthesia be delayed for<br />

up to 6 weeks after an immunization. Parents may also be<br />

advised not to have their children immunized for 6 weeks after<br />

surgery. This would make the planning of routine surgery<br />

difficult in the early months of life and may interfere with the<br />

immunization schedule. 61<br />

It is interesting that a recent survey of health professionals<br />

found a poor knowledge of these rare but serious side effects<br />

among the general practitioners, health visitors, and practice<br />

nurses who administer the vaccines. 62<br />

3. Haemophilus influenzae<br />

The conjugate Haemophilus influenzae B vaccine may be a risk<br />

factor for the development of two autoantibodies frequently<br />

associated with type 1 diabetes. 70<br />

Given these incubation periods, and the rarity of severe<br />

reactions, postponing anesthesia for 3 weeks after immunization,<br />

as recommended by Short and associates, 62 would seem<br />

appro priate. For inactivated vaccines, delaying anesthesia for<br />

7 days would allow resolution of the symptoms and should<br />

eliminate any effect of modulation of the immune system on<br />

the desired response to the vaccine. 61<br />

The final risk is that complications of vaccination may confuse<br />

postoperative assessment (Table 82–2). 65<br />

There are few definitive studies on human patients that directly<br />

assess the effect of surgery and anesthesia on the effectiveness of<br />

immunization. In humans, various studies demonstrated that<br />

antitetanus toxoid antibody response was reduced after surgery. 71,72<br />

Celiker and coworkers advised postponing elective tonsillec -<br />

tomy in a child until postexposure immunization was complete. 73<br />

Vaccination and anesthesia separately might be harmless but<br />

in combination could cause problems. However, Salo 74 pointed out<br />

that no harmful effects have been reported in Finland where<br />

recent vaccination is not regarded as a contraindication to surgery<br />

in children. Salo also commented that elective operations should<br />

not be carried out immediately after vaccination with live or<br />

weakened viruses (e.g., oral polio vaccine). In theory, the patient


1386 PART 4 ■ Special Monitoring and Resuscitation Techniques<br />

TABLE 82-2. Side Effects of Anesthesia and Surgery<br />

Local<br />

Systemic<br />

Immunization<br />

Inflammation<br />

Pain<br />

Granuloma and necrosis<br />

(uncommon)<br />

Lymphadenopathy and<br />

abscess (uncommon)<br />

Fever<br />

Irritability<br />

Exanthema<br />

Prolonged inconsolable<br />

crying (≥3 h)<br />

Neurodeficiency<br />

Thrombocytopenic purpura<br />

Anaphylaxis with shocklike<br />

state<br />

Surgery<br />

Inflammation<br />

(wound infection)<br />

Postoperative pain<br />

Fever (sepsis)<br />

Irritability<br />

Anesthesia-induced<br />

rash<br />

Crying<br />

Postanesthesia<br />

agitation and<br />

confusion<br />

Septic petechiae<br />

Septic shock<br />

or other patients on the ward may be at risk of infection because<br />

of operation-induced immunosuppression. There is no such risk<br />

with toxoid or purified component vaccines.<br />

It is highly likely that children who have recently been immu -<br />

nized will present for both elective and emergency surgery.<br />

In cases of surgical emergency, the immunization history will<br />

assume very little importance. However, the situation may be<br />

different when elective surgery is planned. The systemic side<br />

effects from vaccination may be a relative contraindication to<br />

anesthesia (e.g., fever, coryza, myalgia, and malaise) and may lead<br />

to postponement of surgery on the day of admission if present.<br />

Postoperative confusion may arise between complications of<br />

the surgical procedure and effects of immunization (e.g., crying,<br />

pain, fever, vomiting), and significant diagnostic and clinical man -<br />

agement difficulties may be encountered if the more serious side<br />

effects present in postoperative children, such as arthritis, throm -<br />

bocytopenia, hypotonia, or/and encephalopathy. The risk of these<br />

complications is rare, but their coincidence with the postoperative<br />

period can be avoided by delaying elective surgery until after the<br />

period in which symptoms occur. 62<br />

The immunosuppression caused by anesthesia and surgery lasts<br />

for approximately 2 days. It would, therefore, seem reasonable<br />

to advise that a small risk associated with anesthesia in recently<br />

immunized children can be minimized or eliminated by ensuring<br />

a delay of 1 week after inactive vaccination and 3 weeks after live,<br />

attenuated vaccination and by avoiding routine immunization<br />

until 1 week after surgery has been performed. Because children<br />

are immunized three times in the first 6 months of life, it would<br />

seem reasonable to delay the surgery rather than the immu -<br />

nization in that age group. In older children, immunizations could<br />

be more easily planned around dates for elective surgery. 60<br />

The World Health Organization states: “No child should be de -<br />

nied any indicated immunization without serious thought as to the<br />

consequences for the individual child or to the larger community.” 61<br />

Different centers give different recommendations for anesthetic<br />

procedures in recently vaccinated infants and children. For<br />

example; the New Zealand Immunization Handbook recommends<br />

postponing vaccination when elective surgery is planned. 75 Other<br />

guidelines, such as in the Australian Immunization Handbook<br />

or the Green Book edited by the U.K. Department of Health, 61<br />

describe surgery as compatible with vaccination. The U.S. Centers<br />

for Disease Control and Prevention (CDC), as well as most<br />

European National Guidelines, do not mention the issue of anes -<br />

thesia or surgery in relation to immunizations.<br />

Based on this review, it appears that, although anesthesia exerts<br />

immunomodulatory effects, these are transient and without major<br />

impact on recently vaccinated adults and children. According<br />

to the current evidence and to our understanding of the level<br />

of immunosuppression that is associated with impaired vaccine<br />

efficacy, there is no immunologic contraindication to vaccinate<br />

healthy children scheduled for elective surgery with either live or<br />

inactivated vaccines. Nevertheless, physicians should be aware<br />

that vaccine-driven adverse events may arise and should not be<br />

misinterpreted as a postoperative complication.<br />

Because children remain at risk of contracting vaccine-pre -<br />

ventable diseases, including during their stay in hospital, we<br />

recommend not postponing immunization in children scheduled<br />

for elective surgery. This consideration is especially important<br />

during the first year of life, when many vaccines are scheduled.<br />

Elective surgery should also be postponed only in recently<br />

vaccinated children if this delay creates no danger. 60<br />

Overall, the impact of either delaying surgery or delaying<br />

vaccination is likely to do more harm than good for the individual<br />

children concerned. The safest outcome for the overwhelming<br />

majority of children is, when in doubt, to vaccinate. 65<br />

In summary, the immunomodulatory influence of anesthesia<br />

during elective surgery is both minor and transient (~48 h) and<br />

the current evidence does not provide any contraindication to the<br />

immunization of healthy children scheduled for elective surgery.<br />

However, Siebert and colleagues suggested the following as a<br />

guideline 60 (Table 82–3):<br />

1. Postpone all elective procedures requiring anesthesia rather<br />

than immunization, especially in infants.<br />

2. Opportunistic immunization during general anesthesia is<br />

inadvisable.<br />

TABLE 82-3. Answers and Recommendations of Effect of<br />

Vaccination on Anesthesia<br />

Are there interactions<br />

between general<br />

anesthesia and the<br />

immune system?<br />

Should anesthesia be<br />

postponed in recently<br />

vaccinated children?<br />

Should vaccination be<br />

postponed in children<br />

scheduled for surgery?<br />

What are the risks of<br />

general anesthesia in<br />

recently vaccinated<br />

children?<br />

Yes, but minor and transient.<br />

No, although a delay before<br />

elective surgery may be useful<br />

to avoid vaccine related adverse<br />

events to be misinterpreted as<br />

postoperative complications: 7 d<br />

after a non-live vaccine; 21 d<br />

after a live viral vaccine.<br />

Not to postpone immunization in<br />

children scheduled for elective<br />

surgery.<br />

Not different from the risk in<br />

unvaccinated children.


CHAPTER 82 ■ Influence of Anesthesia on the Immune System in Children 1387<br />

3. Postpone anesthesia for 1 week after vaccination with inactive<br />

vaccines: diphtheria, tetanus, pertussis, inactive polio.<br />

4. Postpone anesthesia for 3 weeks after vaccination with live,<br />

attenuated vaccines: measles, mumps, rubella, oral polio vaccine,<br />

and bacille Calmette-Guérin (BCG; against tuberculosis).<br />

5. Delay immunization for 1 week after surgery has been<br />

performed.<br />

ANESTHESIA IN HIV-<br />

INFECTED CHILDREN<br />

It has been estimated that 20 to 25% of HIV-positive patients<br />

will require surgery during their illness. Therefore, most anes -<br />

thesiologists, also pediatric anesthesiologists, will care for HIVpositive<br />

patients. These anesthesiologists face a complex disease,<br />

which can vary from asymptomatic patients to multiple organ<br />

disease with opportunistic infections. 76<br />

The rapid and early diagnosis of HIV infection in exposed<br />

infants is complicated by transplacental passage of maternal<br />

IgG antibodies to the virus. Virtually all these children are HIV<br />

antibody–positive at birth, although only 15 to 30% is actually<br />

infected. This antibody usually becomes undetectable by 9 months<br />

of age but occasionally remains detectable until 18 months of age.<br />

Therefore, standard anti-HIV IgG antibody tests indicate HIV<br />

infection in children older than 12 to 18 months. In young infants<br />

younger than 18 months, the diagnosis of HIV infection now relies<br />

on virologic assays: HIV DNA polymerase chain reaction (PCR)<br />

and HIV peripheral blood lymphocyte culture. 77<br />

Pediatric HIV infection can be classified according to immu -<br />

nologic status (Table 82–4) and clinical status. 78 Once classified,<br />

an HIV-infected child cannot be reclassified in a less severe<br />

category even if the clinical or immunologic status improves.<br />

CD4+ T lymphocyte depletion is a major consequence of HIV<br />

infection and is responsible for many severe manifestations of<br />

HIV infection. However, normal CD4+ counts in infants and<br />

young children are higher than in adults and decline in the first<br />

years of life. They vary depending on the age of the child.<br />

Moreover, children may develop opportunistic infections at higher<br />

CD4+ levels than adults. Hence, age-specific CD4+ T lymphocyte<br />

counts and CD4+ percentage of total lymphocytes are used to<br />

classify the severity of immunosuppression attributable to HIV<br />

infection in children. Three immunologic categories are described<br />

based on the signs, symptoms, or diagnoses related to HIV<br />

infection. 77<br />

Anesthesia Considerations in<br />

Pediatric HIV Infection<br />

Anesthesia in a child with HIV infection is challenging. The<br />

spectrum of disease varies from asymptomatic to severe multiple<br />

organ involvement. There is no major anesthetic problem in<br />

an asymptomatic patient with seropositivity alone, because<br />

this group of patients is not treated with antiretroviral agents.<br />

Conversely, careful anesthesia management is needed in patients<br />

with multiorgan involvement. Anesthesia begins with preoperative<br />

assessment, which consists of history, physical examination, and<br />

laboratory studies. History and physical examination focus on the<br />

current status of HIV infection, coexisting organ involvement, and<br />

concurrent medications. Children with a lower CD4+ count reveal<br />

more advanced disease and need particular attention. In patients<br />

who received multiple medications, adverse drug effects and<br />

interactions with drug used in anesthesia should be taken into<br />

consideration. 77<br />

There is no specific recommended anesthesia technique or<br />

agent for HIV-infected children. However, general anesthesia is the<br />

most often used technique in pediatric anesthesia practice. Anes -<br />

thesia is based on the extent of organ involvement. For example,<br />

high-inspired oxygen concentration is necessary in children with<br />

pulmonary complications and impaired gas exchange. In patients<br />

with cardiovascular involvement, titration of anesthetics with<br />

careful hemodynamic monitoring is advised. Narcotics and muscle<br />

relaxants are chosen and dosed according to alterations in renal<br />

and hepatic function. The use of succinylcholine is avoided in<br />

progressive neuropathy, myopathy, and muscle weakness because<br />

of the potential risk of hyperkalemia. Immunologic suppression by<br />

anesthetic agents may be of minor importance for subjects with<br />

a normal immune system, but it is important in patients with<br />

immune dysfunction. 79<br />

Central neural blockade, especially caudal anesthesia, is a<br />

common regional anesthesia technique in children. Because there<br />

is the possibility of neurologic involvement in HIV infection,<br />

performing central neural blockade in HIV-infected patients is an<br />

issue for discussion. It is suggested that, before performing central<br />

neural blockade, the patient should be carefully evaluated for<br />

central nervous system involvement as well as any contraindication<br />

to central neural blockade such as thrombocytopenia, which is a<br />

side effect of antiretroviral agents. 80<br />

Patients with HIV infection are administered several medi -<br />

cations concomitantly to combat HIV-related conditions and<br />

to prevent or treat opportunistic infections. Some of these<br />

drugs interact with those used in anesthesia. Prolonged neuro -<br />

muscular blockade after vecuronium in patients with HIV<br />

infection is described. Protease inhibitors are inhibitors of cyto -<br />

chrome P450, which is involved in the metabolism of many<br />

anesthetic and anal gesic agents. Ritonavir, a protease inhibitor,<br />

can reduce clearance of fentanyl and prolong fentanyl half-life. <strong>81</strong><br />

If only small bolus doses of fentanyl are administered, a dose<br />

adjustment of fentanyl is probably not needed, but the dosage<br />

of fentanyl should be reduced if administered by infusion.<br />

Saquinavir can inhibit midazolam metabolism and prolong<br />

midazolam’s effect. 82<br />

TABLE 82-4. Immunologic Classification of HIV in Children Based on Age-Specific CD4+ T<br />

Lymphocyte Counts and Percentage of Total Lymphocytes<br />

Immunologic Categories ≤12 mo, µL (%) 1–5 y, µl (%) 6–12 y, µl (%)<br />

No evidence of suppression ≥1500 (≥25) ≥1000 (≥25) ≥500 (≥25)<br />

Evidence of moderate suppression 750–1499 (15-24) 500–999 (15–24) 200–499 (15–24)<br />

Evidence of severe suppression


1388 PART 4 ■ Special Monitoring and Resuscitation Techniques<br />

ANESTHESIA IN IMMUNOSUPPRESSED<br />

CHILDREN WITH GRAFT-<br />

VERSUS-HOST DISEASE<br />

Preanesthetic evaluation should focus on the extent of systemic<br />

involvement secondary to graft-versus-host disease (GVHD) and<br />

the status of immunosuppression. Interaction of anesthetics with<br />

immunosuppressive drugs is an important consideration. When<br />

hepatic and renal functions are normal, there are no contrain -<br />

dications to the use of any anesthetic agent. Inhalational agents<br />

such as halothane and sevoflurane can be used for induction, and<br />

agents such as isoflurane, sevoflurane, and desflurane can be used<br />

for maintenance of anesthesia. Intravenous agents and local anes -<br />

thetics may also be used. There is no evidence of any increased<br />

risk associated with central neuraxial blockade in children on<br />

long-term steroids. 83<br />

Cyclosporine enhances the effects of muscle relaxants. The<br />

dosage of nondepolarizing muscle relaxants including atracurium<br />

should be reduced in patients receiving cyclosporine, and the<br />

recovery time of nondepolarizing muscle relaxants may be pro -<br />

longed. Azathioprine has little interaction with nondepolarizing<br />

muscle relaxants in humans. Propofol infusion does not modify<br />

cyclosporine blood levels in humans. Atracurium or cisatracurium<br />

is specifically indicated in children with renal or hepatic im -<br />

pairment. 83<br />

The effects of nitrous oxide on the bone marrow cells may be a<br />

concern. Bone marrow suppression is more likely to occur after<br />

prolonged exposure (>6 h) at high concentrations. Methotrexate<br />

impairs folate metabolism and can cause bone marrow depression.<br />

Nitrous oxide may reduce the therapeutic benefit and increase<br />

the side effects of methotrexate therapy. Hence, nitrous oxide<br />

should be used with caution in patients on methotrexate therapy<br />

(as GVHD prophylaxis). In the presence of pancytopenia and<br />

refractory thrombocytopenia, it may be better to avoid this agent<br />

for long procedures. 84<br />

Because hepatic dysfunction is common in GVHD and renal<br />

dysfunction, it is imperative to avoid anesthesia agents with poten -<br />

tial hepatorenal toxicity such as halothane (for maintenance), longacting<br />

opioids such as morphine and pethidine, benzodiazepines<br />

undergoing oxidative metabolism (e.g., diazepam), long-acting<br />

muscle relaxants such as pancuronium, pipecuronium, doxa -<br />

curium, and non-steroidal anti-inflammatory drugs (NSAIDs).<br />

NSAIDs may augment the nephrotoxicity of cyclosporine or<br />

tacrolimus. 83<br />

REGIONAL ANESTHESIA IN<br />

IMMUNOCOMPROMISED PATIENTS<br />

Regional (neuraxial) anesthesia and analgesia provide several<br />

advantages over systemic opioids, including superior analgesia,<br />

reduced pulmonary complications, and improved joint mobility<br />

after major orthopedic surgery. 85<br />

In addition, neuraxial analgesia may decrease the risk of infec -<br />

tion through attenuation of the stress response and preservation of<br />

immune function. Despite these benefits, patients with altered<br />

immune status because of diabetes, neoplasm, immunosuppres -<br />

sion after solid organ transplantation, and chronic infection with<br />

HIV or herpes simplex virus (HSV) are often not considered<br />

candidates for neuraxial techniques because of the risk of infection<br />

around the spinal cord or within the spinal canal. These patients<br />

are more susceptible to infection with opportunistic pathogens<br />

compared with patients with normal immune function. Thus, a<br />

depressed immune state increases both frequency and severity<br />

of infection. 86<br />

In addition, in terminally ill patients, the risk of infection with<br />

long-term epidural catheterization is acceptable, but careful moni -<br />

toring is recommended to avoid serious neurologic sequelae. 87<br />

Immunocompromised patients often present with pre-existing<br />

neurologic dysfunction caused by their primary disease process<br />

(spinal cord compression from vertebral column neoplasm,<br />

peripheral neuropathy associated with HIV, or diabetes) or<br />

because of treatment. Based on the “double-crush” theory, which<br />

hypothesizes that axons injured at primary site may be particularly<br />

susceptible to damage, these patients may be at increased risk for<br />

neurologic complications of regional anesthesia. Furthermore, the<br />

damage of the dual injury exceeds the expected additive damage<br />

caused by each isolated injury. Thus, the effects of needle trauma,<br />

ischemia, and local anesthetic toxicity are exaggerated and a<br />

relatively minor injury applied to a previously dysfunctional nerve<br />

may result in new (or exacerbation of existing) symptoms. 88<br />

Recommendations for Safe<br />

Use of Regional Anesthesia in<br />

Immunocompromised Patients 86<br />

1. Clinical series and epidemiologic data provide guidance in the<br />

administration of spinal or epidural anesthesia in the febrile<br />

patient. However, as with all clinical judgments, the decision<br />

to perform a regional anesthetic technique must be made on<br />

an individual basis considering the anesthetic alternatives, the<br />

benefits of regional anesthesia, and the risk of central nervous<br />

system infection (which theoretically are more likely to occur<br />

in the immunocompromised patient), as well as the risk of<br />

hemorrhagic or neurologic complications.<br />

2. Consultation with an infectious disease specialist is advised to<br />

facilitate initiation of early and effective therapy, because there<br />

is attenuated inflammatory response within the immuno -<br />

compromised patient that may diminish the clinical signs and<br />

symptoms often associated with infection. Likewise, the range<br />

of microorganisms causing invasive infection in the immuno -<br />

compromised host is much broader than that affecting the<br />

general population and includes atypical and opportunistic<br />

pathogen.<br />

3. The diagnosis and treatment of central nervous system<br />

infections should not be delayed because it worsens neurologic<br />

outcome and increases mortality.<br />

4. Prolonged duration of epidural catheterization increases the<br />

risk of epidural abscess.<br />

5. Central neuronal block has been showing safety in patients<br />

with recurrent HSV infections, although exacerbations of<br />

HSV-1 have been reported in association with intrathecal and<br />

epidural opioids.<br />

AUTOIMMUNE HEPATITIS CAUSED<br />

BY VOLATILE ANESTHETICS IN<br />

PEDIATRIC ANESTHESIOLOGISTS<br />

Pediatric anesthesiologists are exposed to significant levels of<br />

volatile anesthetics during frequent facemask inductions of general<br />

anesthesia and from the practice of using uncuffed endotracheal


CHAPTER 82 ■ Influence of Anesthesia on the Immune System in Children 1389<br />

tubes. Previous investigations have demonstrated that several<br />

specific hepatic proteins become covalently trifluoroacetylated<br />

(TFA) by the reactive metabolites of halothane and isoflurane.<br />

Similar effect may also be formed at very small levels after exposure<br />

to desflurane. These TFA neoantigens are important because it is<br />

thought that they induce immune responses against either the TFA<br />

neoantigens, the native protein components (autoantigens) of the<br />

TFA neoantigens, or both of these classes of antigens in individuals<br />

who are susceptible to inhaled anesthetic-induced hepatitis. 89<br />

It has been established that cytochrome P450 2E1 (CYP 2E1),<br />

the primary enzyme responsible for the oxidative metabolism<br />

of most volatile anesthetics, also becomes TFA-altered when it<br />

metabolizes halothane. In addition, autoantibodies reacting with<br />

CYP 2E1 are significantly elevated in the sera of 45 to 70% of<br />

patients diagnosed with halothane hepatitis, whereas control sub -<br />

jects did not demonstrate increased levels of these autoantibodies.<br />

These findings suggest that pathogenic antibodies directed against<br />

ERp58, CYP 2E1, or both may have a role in the etiology of volatile<br />

anesthetic hepatitis. 90<br />

Njoku and associates demonstrated that pediatric anes -<br />

thesiologists had higher levels of both ERp58 and CYP 2E1 auto -<br />

antibodies than did general anesthesiologists and control patients,<br />

whereas general anesthesiologists have increased serum levels<br />

of CYP 2E1 autoantibodies only when compared with the control<br />

patients. 91 However, because most anesthesiologists do not develop<br />

volatile anesthetic–induced liver injury, the results suggest that<br />

pathogenic ERp58 and CYP 2E1 autoantibodies may not cause<br />

volatile anesthetic hepatitis.<br />

These results suggest that chronic occupational exposure to<br />

volatile anesthetics can lead to continuous hepatic metabolism of<br />

volatile anesthetics and formation of immunogenic TFA protein<br />

adducts, including TFA-ERp58 and TFA-CYP 2E1, which can<br />

induce formation of the ERp58 and CYP 2E1 autoantibodies<br />

associated with volatile anesthetic hepatitis. 92<br />

The results have also shown that female anesthesiologists have<br />

higher levels of ERp58 autoantibodies than male anesthesiologists.<br />

Also, female pediatric anesthesiologists have higher levels of<br />

CYP 2E1 autoantibodies than all other anesthesiologists. Only one<br />

female pediatric anesthesiologist developed liver injury, even when<br />

all of the pediatric anesthesiologists as a group had increased<br />

serum levels of anesthetic hepatitis–associated ERp58 and CYP<br />

2E1 autoantibodies that were not significantly different from<br />

those of halothane hepatitis patients. These finding suggest that<br />

ERp58 and CYP 2E1 autoantibodies may not have a role in the<br />

development of inhaled anesthetic hepatitis. Alternatively, it is<br />

possible that antigen-specific cytotoxic T cells, instead of autoan -<br />

tibodies, which are directed against peptides derived from ERp58<br />

and CYP 2E1, may cause volatile anesthetic hepatitis. By contrast,<br />

humoral reactions, cellular immune reactions, or both against<br />

native ERp58 and CYP 2E1 may not have a role in volatile<br />

anesthetic–induced hepatitis. Perhaps only TFA-altered forms of<br />

these autoantigens can be targets of pathogenic antibodies or<br />

cytotoxic T cells. 92<br />

In this regard, previous studies have demonstrated that only<br />

halothane hepatitis patients, but not patients exposed to halothane<br />

who did not develop hepatitis or patients with other forms of<br />

liver disease, have serum antibodies that react with TFA liver<br />

microsomal antigens. Furthermore, other cellular targets of the<br />

reactive acyl halide metabolites of volatile anesthetics that also<br />

become TFA modified, such as a carboxylesterase, protein disul -<br />

ide isomerase, ERp72, and glucose-related proteins 78 and 94,<br />

could potentially become the immunogens that lead to volatile<br />

anesthetic–induced hepatitis. 92<br />

In summary, a significantly higher level of ERp58 and CYP 2E1<br />

serum autoantibodies was found in pediatric anesthesiologists<br />

than in general anesthesiologists. Female anesthesiologists, both<br />

pediatric and general, have higher levels of autoantibodies to<br />

ERp58 than male anesthesiologists, and female pediatric anes -<br />

thesiologists tend to have higher levels of CYP 2E1 autoantibodies<br />

than male pediatric anesthesiologists. Still, only one of the female<br />

pediatric anesthesiologists developed symptoms of anesthetic<br />

hepatitis, even though these autoantibodies have been associated<br />

with volatile anesthetic–induced hepatitis. These findings suggest<br />

that ERp58 and CYP 2E1 serum autoantibodies may not have a<br />

significant role in volatile anesthetic–induced hepatitis and that<br />

other immune mechanisms may determine whether halogenated<br />

volatile anesthetic–induced hepatitis occurs. 92<br />

CONCLUSION<br />

Intraoperative anaphylaxis is the most severe complication of<br />

pediatric anesthesia and it is prevented by careful identification of<br />

risk groups, identification of causative agent, and serologic and<br />

skin tests. Postoperative immunosuppressive effects are transient<br />

for approximately 48 hours irrespective of the mechanism of<br />

drug actions.<br />

Immunocompromised patients should be carefully observed<br />

for fear of postoperative infections and neurologic complications,<br />

which occur as a result of regional anesthesia technique. Anes -<br />

thesia should be postponed for 1 week for killed vaccines and for<br />

3 weeks for live vaccines for fear of increased complications of<br />

vaccines or a decrease in the effectiveness of vaccines.<br />

REFERENCES<br />

1. Sao M. Effects of anaesthesia and surgery on the immune response. Acta<br />

Anaesthesiol Scand. 1992;36:201–220.<br />

2. McBride WT, Armstrong MA, McBride SJ. Immunomodulation: an<br />

important concept in modern anaesthesia. Anaesthesia. 1996;51:465–73.<br />

3. Delves PJ, Roitt FS. The immune system. First of two parts. N Engl J Med.<br />

2000;343:37–49.<br />

4. Goldsby, R. A., Kindt TJ, Osborne BA. Immunoglobulins: structure and<br />

function. In: Kindt TJ, R. A. Goldsby RA, Osborne BA, (eds.), Kuby<br />

immunology, 4th ed. W. H. Freeman and Company, New York, N.Y. 2000;<br />

p. 83–113.<br />

5. Ni Choileain N, Redmond HP. Cell response to surgery. Arch Surg.<br />

2006;16:1132–1140.<br />

6. McMahon SB, Bennett DL. Inflammatory mediators and modulators of<br />

pain. In: McMahon SB, Koitzenburg M, editors. Wall and Mehack’s<br />

Textbook of Pain. 5th ed. Philadelphia: Elsevier Churchill Livingstone;<br />

2006. pp. 49–72.<br />

7. Moraska A, Campisi J, Nguyen K, et al. Elevated IL-lb contributes to anti -<br />

body suppression produced by stress. J Appl Physiol. 2002;93:207–215.<br />

8. Sacerdote P. Opioids and the immune system. Palliat Med. 2006;20(Suppl<br />

I):s9–s15.<br />

9. Molina PE. Opioids and opiates: analgesia with cardiovascular, haemo -<br />

dynamic and immune implications in critical illness. J Intern Med. 2006;<br />

259:138–154.<br />

10. Homburger JA, Meiler SE. Anesthesia drugs, immunity and long-term<br />

outcome. Curr Opin Anaesthesiol. 2006;19:423–428.<br />

11. Loop T, Dovi-Akue D, Frick M, et al. Volatile anesthetics induce caspasedependent,<br />

mitochondria-mediated apoptosis in human T-lymphocytes<br />

in vitro. Anesthesiology. 2005;102:147–1157.<br />

12. Devlin EG, Clarke RS, Mirakhur RK, et al. Effect of four IV induction<br />

agents on T-lymphocyte proliferations to PHA in vitro. Br J Anaesth.<br />

1994;73:315–317.


1390 PART 4 ■ Special Monitoring and Resuscitation Techniques<br />

13. Chang Y, Chen T. Sheu J, et al. Suppressive effects of ketamine on<br />

macrophage functions. Toxicol Appl Pharmacol. 2005;204:27–35.<br />

14. Inada T, Yamanouchi Y, Jomura S, et al. Effect of propofol and isoflurane<br />

anaesthesia on the immune response to surgery. Anaesthesia. 2004;59:<br />

954–959.<br />

15. Kelbel I, Weiss M. Anaesthetics and immune function. Curr Opin<br />

Anaesthesiol. 2001;14:685–691.<br />

16. Schneemilch CE, Hachenberg T, Ansorge S, et al. Effects of different<br />

anaesthetic agents on immune cell function in vitro. Eur J Anaesthesiol.<br />

2005;22:616–623.<br />

17. Procopio M, Rassias AJ, DeLeo JA, et al. The in vivo effects of general and<br />

epidural anesthesia on human immune function. Anesth Analg. 2000;<br />

93:460–465.<br />

18. Correa-Sak’S C, Tosta CE, Rizzo LV. The effects of anesthesia with<br />

thiopental on T lymphocyte responses to antigen and mitogens in vivo<br />

and in vitro. Int J Immunopharmacol. 1997;19:117–128.<br />

19. Hamra JG, Yaksh TL. Halothane inhibits T cell proliferation and<br />

interleukin-2 receptor expression in rats. Immunopharmacol Immuno -<br />

toxicol. 1996;18:323–336.<br />

20. Mikawa K, Akamatsu H, Nishina K, et al. Propofol inhibits human<br />

neutrophil functions. Anesth Analg. 1998;87:695–700.<br />

21. Galley HF, Webster NR. Effects of propofol and thiopentone on the<br />

immune response. Anaesthesia. 1994;52:921–923.<br />

22. Stevenson GW, Hall SC, Rudnik S, et al. The effects of anesthetic agents<br />

on the human immune response. Anesthesiology. 1990;72:144.<br />

23. Spiers EM, Potts RC, Simpson JR, et al. Mechanisms by which barbiturates<br />

suppress lymphocyte responses to phytohaemagglutinin stimulation.<br />

Int J Immunopharmacol. 1987;9:505–512.<br />

24. Devlin EG, Clarke RSJ, Mirakhur RK, et al. The effects of thiopentone<br />

and propofol on delayed hypersensitivity reactions. Anaesthesia. 1995;50:<br />

496–498.<br />

25. Hensler T, Hecker H, Heeg K, et al. Distinct mechanisms of immuno -<br />

suppression as a consequence of major surgery. Infect Immunol. 1997;65:<br />

2283–2291.<br />

26. Stover JF, Stocker R. Barbiturate coma may promote reversible bone<br />

marrow suppression in patients with severe isolated traumatic brain<br />

injury. Eur J Clin Pharmacol. 1998;54:529–534.<br />

27. Eberhardt KE, Thimm BM, Spring A, et al. Dose dependent rate of<br />

nosocomial pulmonary infection in mechanically ventilated patients with<br />

brain oedema receiving barbiturates: a prospective case study. Infection.<br />

1992;20:12–18.<br />

28. Briggs WA, Eustace J, Gimenez LF, et al. Lymphocyte suppression<br />

by glucocorticoids with cyclosporine, tacrolimus, pentoxifylline, and<br />

mycophenoiic acid. J Clin Pharmacol. 1999;39:125–130.<br />

29. Pirttikangas CO, Perttila J, Salo M. Propofol emulsion reduces prolif -<br />

erative responses of lymphocytes from intensive care patients. Intensive<br />

Care Med. 1993;19:299–302.<br />

30. Angele MK, Faist E. Clinical review: immunodepression in the surgical<br />

patient and increased susceptibility to infections. Crit Care. 2002;6:298–305<br />

31. Hanisch UK, Quirion R. Interleukin-2 as a neuroregulatory cytokine.<br />

1996;21:246–284.<br />

32. Salo M, Pirttikangas CO, Pulkki K. Effects of propofol emulsion and<br />

thiopentone on T helper cell type-l/rype-2 balance in vitro. Anaesthesia.<br />

1997;52:341–344.<br />

33. Loop T, Liu Z, Humar M, et al. Thiopental inhibits the activation of<br />

nuclear factor kappaB. Anesthesiology. 2002;96:1202–1213.<br />

34. Ben-Eliyahu S. The promotion of tumor metastasis surgery and stress:<br />

immunological basis and indications for psychoneuroimmunology. Brain<br />

Behav Immun. 2003;17:S27–S36.<br />

35. Carrigan KA, Saurer TB, James SG, et al. Buprenorphine produces<br />

naltrexone reversible alterations of immune status. Int Immunopharmacol.<br />

2004;4:419–428.<br />

36. Martucci C, Panerai AE, Sacerdote R. Chronic fentanyl or buprenorphine<br />

infusion in the mouse: similar analgesic profile but different effects on<br />

immune responses. Pain. 2004;110:385–392.<br />

37. Tsujikawa H, Shoda T, Mizota T, Fukuda K. Morphine induces DNA<br />

damage and P53 activation in CD3+ T cells. Biochem Biophys Acta.<br />

2009;1790:793–793.<br />

38. Sacerdote P, Bianchi M, Gaspani L, et al. The effects of tramadol and<br />

morphine on immune responses and pain after surgery in cancer patients.<br />

Anesth Analg. 2000;90:1411–1414.<br />

39. Stoelting RK, Hillier SC. Pharmacology and Physiology in Anesthesia<br />

Practice. 4th ed. Philadelphia: Lippincott Williams & Wilkins, 2005,<br />

p. 850.<br />

40. Yeager MP, Procopio MA, DeLeo JA, et al. Intravenous fentanyl increases<br />

natural killer cell cytotoxicity and circulating CD16(+) lymphocytes in<br />

humans. Anesth Analg. 2002;94:94–99.<br />

41. Jacobs R, Karst M, Scheinichen D, et al. Effects of fentanyl on cellular<br />

immune functions in man. Int J Immunopharmacol. 1999;21:445.<br />

42. Bruce DL. Halothane inhibition of RNA and protein synthesis of PHAinduced<br />

human lymphocytes. Anesthesiology. 1975;42:11–14.<br />

43. Mitsuhata H, Shimizu R, Yokoyama MM. Suppressive effects of volatile<br />

anaesthetics on cytokine release in human peripheral blood mononuclear<br />

cells. Int J Immunopharmacol. 1995;17:529–534.<br />

44. Delves PJ, Roitt IM. Advances in immunology: the immune system.<br />

N Engl J Med. 2000;343:30–36.<br />

45. Moudgil GC. Update on anesthesia and the immune response. Can J<br />

Anaesth. 1986:33:354–360.<br />

46. Fisher MM, More DG. The epidemiology and clinical features of<br />

anaphylactic reactions in anaesthesia. Anaesth Intensive Care. 19<strong>81</strong>;9:<br />

226.<br />

47. Weiss ME, Adkinson NF, Hirshman CA. Evaluation of allergic reactions<br />

in the perioperative period. Anaesthesia. 1989,71:43.<br />

48. Laroche D, Lefrancois C, Gerard JL, et al. Early diagnosis of anaphylactic<br />

reactions to neuromuscular blocking drugs. Br J Anaesth. 1992;69:<br />

611–614.<br />

49. Watkins J, Appleyard TN, Thornton JA, Padfield A. Immune mediated<br />

reactions to anaesthesia (alphaxalone). Br J Anaesth. 1976,48:8<strong>81</strong>–886.<br />

50. Best N, Teisner B, Grudzinskas JG, et al. Classic pathway activation during<br />

an adverse response to protamine sulphate. Br J Anaesth. 1983;55:1149–<br />

1153.<br />

51. Cogen FC, Norman ME, Dunsky E, et al. Histamine release and<br />

complement changes following injection of contrast media in humans.<br />

J Allergy Clin Immunol. 1979;64:299–303.<br />

52. Fisher MM. Tiessner B, Charlesworth JC. Significance of sequential<br />

changes in serum complement levels during anaphylactoid reactions. Crit<br />

Care Med. 1984;12:351–354.<br />

53. Schwartz LB, Yunginger JW, Miller J, et al. Time course of appearance and<br />

disappearance of human mast cell tryptase in the circulation after<br />

anaphylaxis. J Clin Invest. 1989;83:1551–1555.<br />

54. Yunginger JW, Nelson DR, Squilace DL, et al. Laboratory investigations<br />

of death due to anaphylaxis. J Forensic Sci. 1991;36:857–865.<br />

55. Fisher MM. Intradermal testing after anaphylactoid reaction to<br />

anaesthetic drugs: practical aspects of performance and interpretation.<br />

Anaesth Intensive Care. 1984;12:115–120.<br />

56. Sage D. Intradermal drug testing following anaphylactoid reactions<br />

during anaesthesia. Anaesth Intensive Care. 19<strong>81</strong>;6:3<strong>81</strong>–390.<br />

57. Moscicki RA, Sockin SM, Corsello BF, et al. Anaphylaxis during induction<br />

of general anaesthesia: subsequent evaluation and management. J Allergy<br />

Clin Immunol. 1990;86:325–332.<br />

58. Harle DG, Baldo BA, Smal MA, et al. Detection of thiopentone-reactive<br />

IgE antibodies following anaphylactoid reactions during anaesthesia. Clin<br />

Allergy. 1986;16:493–498.<br />

59. Fisher MM, Baldo BA. Persistence of allergy to anaesthetic drugs. Anaesth<br />

Intensive Care. 1992;20:143–146.<br />

60. Siebert J, Posfay-Barbe K, Habre W, et al. Influence of anesthesia on<br />

immune responses and its effect on vaccination in children: review of<br />

evidence. Paediatr Anaesth. 2007;17:410–420.<br />

61. Currie J. Vaccination: is it a real problem for anesthesia and surgery?<br />

Paediatr Anaesth. 2006;16:501–503.<br />

62. Short J, Van der Walt J, Zoanetti D. Immunization and anesthesia—an<br />

international survey. Paediatr Anaesth. 2006;16:514–522.<br />

63. Van der Walt J, Roberton D. Anaesthesia and recently vaccinated children.<br />

Paediatr Anaesth. 1996;6:135–141.<br />

64. Salisbury D, Begg, N, Noakes K, (eds). Immunization against Infectious<br />

Disease – The Green Book. 4th edn. London: The Stationery Office, 2006,<br />

p. 468.<br />

65. Crowcroft N, Elliman D. Vaccination and anesthesia: the precautionary<br />

principle is to vaccinate. Paediatr Anaesth. 2007;17:1215–1227.<br />

66. Kitchin N, Southern J, Morris R. A randomised controlled study of the<br />

reactogenicity of an acellular pertussis-containing pentavalent infant<br />

vaccine compared to a quadrivalent whole cell pertussis-containing<br />

vaccine and oral poliomyelitis vaccine, when given concurrently with<br />

meningococcal group C conjugate vaccine to healthy UK infants at 2, 3<br />

and 4 months of age. Vaccine. 2006;24:3964–3970.<br />

67. Centers for Disease Control and Prevention (CDC). Imported vaccineassociated<br />

paralytic poliomyelitis. MMWR Morb Mortal Wkly Rep. 2006;<br />

55:97–99.


CHAPTER 82 ■ Influence of Anesthesia on the Immune System in Children 1391<br />

68. Karakasiliotis J, Paximadi E, Markoulatos P. Evolution of a rare vaccinederived<br />

multi recombinant poliovirus. J Gen Virol. 2005;86:3137–3142.<br />

69. Miller E, Waight P, Farrington C. Idiopathic thrombocytopenic purpura<br />

and MMR vaccine. Arch Dis Child. 2001;84:227–229.<br />

70. Song B, Katial R. Update on side effects from common vaccines. Curr<br />

Allergy Asthma Rep. 2004;4:447–453.<br />

71. Nielsen H, Hammer J, Moesgaard F. Ranitidine improves postoperative<br />

suppression of antibody response to preoperative vaccination. Surgery.<br />

1992;111:69–73.<br />

72. Clayer M, Drew P, Jamieson G. Antibody responses following splenectomy:<br />

implications for the timing of prophylactic vaccination. Aust N Z J Surg.<br />

1992;62:142–146.<br />

73. Celiker V, Basgu E, Peker L. Rabies vaccine and anesthesia. Paediatr<br />

Anaesth. 2004;14:365–366.<br />

74. Salo M. Effects of anesthesia and surgery on the immune response. Acta<br />

Anaesthesiol Scand. 1992;36:201–220.<br />

75. Immunization Handbook Working Group (ITWG). New Zealand<br />

Immunisation Schedule Wellington, New Zealand, New Zealand Ministry<br />

of Health, April 2006. pp. 23–27.<br />

76. Eichler A, Eiden U, Kessler P. AIDS and anesthesia. Anaesthesist. 2000;49:<br />

1006–1017.<br />

77. Leelaukrom R, Pancharoen C. Anesthesia in HIV-infected children.<br />

Paediatr Anaesth. 2007;17:509–519.<br />

78. Centers for Disease Control and Prevention (CDC). Revised classification<br />

system for human immunodeficiency virus infection in children less than<br />

13 years of age. MMWR Morb Mortal Wkly Rep. 1994;43:1–10.<br />

79. Schneemilch C, Ittenson A, Ansorge S. Effect of 2 anesthetic techniques<br />

on the postoperative proinflammatory and anti-inflammatory cytokine<br />

response and cellular immune function to minor surgery. J Clin Anesth.<br />

2005;17:517–527.<br />

80. Tom D, Gulevich S, Shapiro H. Epidural blood patch in the HIV-positive<br />

patient. Review of clinical experience. Anesthesiology. 1992;76:943–947.<br />

<strong>81</strong>. Olkkola K, Palkama V, Neuvonen P. Ritonavir’s role in reducing fentanyl<br />

clearance and prolonging its half-life. Anesthesiology. 1999;91:6<strong>81</strong>–685.<br />

82. Palkama V, Ahonen J, Neuvonen P. Effect of sanquinavir on the pharma -<br />

cokinetics and pharmacodynamics of oral and intravenous midazolam.<br />

Clin Pharmacol Ther. 1999;66:33–39.<br />

83. Venkatesan T, Jacob R. Anesthesia and graft-vs-host disease after<br />

hematopoietic stem cell transplantation. Paediatr Anaesth. 2007;17:7–15.<br />

84. Wyatt S, Gill RS. An absolute contraindication to nitrous oxide. Anaes -<br />

thesia. 1999;54:307–308.<br />

85. Liu S, Carpenter R, Neal J. Epidural anesthesia and analgesia. Their role<br />

in postoperative outcome. Anesthesiology. 1995;82:1474–1506.<br />

86. Capdevila X, Barthelet Y, Biboulet P, et al. Effects of perioperative analgesic<br />

technique on the surgical outcome and duration of rehabilitation after<br />

major knee surgery. Anesthesiology. 1999;91:8–15.<br />

87. Terese T, Horlocker M, Denise J, et al. Regional anesthesia in the<br />

immunocompromised patient. Reg Anesth Pain Med. 2006;31:334–345.<br />

88. Moen V, Dahlgren N, Irestedt L. Severe neurological complications<br />

after central neuraxial blockades in Sweden 1990–1999. Anesthesiology.<br />

2004;101:950–959.<br />

89. Njoku D, Laster M, Gong D. Biotransformation of halothane, enflurane,<br />

isoflurane, and desflurane to trifluoroacetylated liver proteins: association<br />

between protein acylation and hepatic injury. Anesth Analg. 1997;84:<br />

173–178.<br />

90. Bourdi M, Chen W, Peter R. Human cytochrome P450–2E1 is a major<br />

autoantigen associated with halothane hepatitis. Chem Res Toxicol. 1996;<br />

9:1159–1166.<br />

91. Njoku D, Greenberg R, Bourdi M, et al. Autoantibodies associated with<br />

volatile anesthetic hepatitis found in the sera of a large cohort of paediatric<br />

anaesthesiologists. Paediatr Anaesth. 2002;94:243–249.<br />

92. Pohl L, Pumford N, Martin J. Mechanism chemical structures and drug<br />

metabolism. Eur J Haematol. 1996;60:98–100.


83<br />

CHAPTER<br />

Specific Problems and Anesthesia<br />

Management of Extremely Low<br />

Birthweight Infants<br />

Richard J. Ing and Allison Kinder Ross<br />

INTRODUCTION AND DEFINITIONS<br />

Despite tremendous advances in the care of the extremely low<br />

birthweight (ELBW) infant since the early 2000s, complication<br />

rates still remain high. A recent study on morbidity and mortality<br />

(1998–2003) in 572 live-born infants born at less than 26 weeks’<br />

gestation and admitted to a neonatal intensive care unit (NICU)<br />

showed that 62.3% survived until day 28 and 59.6% were dis -<br />

charged home. 1 Among infants discharged home, bronchopul -<br />

monary dysplasia (BPD) was diagnosed in 52.2%, intraventricular<br />

hemorrhage (IVH) of grade III or greater and/or periventricular<br />

leukomalacia was present in 15%, and severe retinopathy of<br />

prematurity (ROP) was seen in 29.8%. 1 Another recent 5-year<br />

longitudinal cohort neurodevelopmental disability study has<br />

shown that long-term significant disability was highest (49%) in<br />

children born at 24 to 28 weeks’ gestation. 2 These unique longterm<br />

morbidity challenges of the ELBW infant patient group are<br />

significant and must, therefore, be taken into consideration by the<br />

pediatric anesthesiologist when caring for these children.<br />

ELBW infants are usually born at 24 to 30 weeks’ gestation<br />

and weigh between 401 and 1000 g. The current definitions of<br />

newborn infants are presented in Table 83–1. The ELBW infant<br />

accounts for approximately 1% of all live-born infants. Premature<br />

delivery occurs before intrauterine physiologic transitional<br />

development and major organ maturation have been completed.<br />

As a result, the immature ELBW infant is initially ill-prepared to<br />

regulate body temperature, energy requirements, or electrolyte<br />

balance. In addition, the infant’s immature cardiovascular and<br />

respiratory systems in the extrauterine environment predispose<br />

them to bradycardia, hypotension, apnea, and cyanosis. Transient<br />

hypoxia, the need for mechanical ventilation, cardiovascular<br />

resuscitation, inotropes, and surfactant administration at the time<br />

of birth all increase the risks of iatrogenic morbidity and mortality.<br />

TABLE 83-1. Definitions<br />

Neonate<br />

Premature<br />

Preterm<br />

LBW<br />

VLBW<br />

ELBW<br />

First 44 wk postconceptional age<br />


CHAPTER 83 ■ Specific Problems and Anesthesia Management of Extremely Low Birthweight Infants 1393<br />

with complete anatomic closure usually achieved by 3 weeks of<br />

age. In ELBW infants specifically, this process may take longer to<br />

complete, and commonly, a reversal or an incomplete transitional<br />

physiologic state remains. In this situation, the ELBW infant’s<br />

circulation may revert to the fetal state under times of hypoxemia,<br />

hypercarbia, acidosis, hypothermia, or pain and infection. Apnea<br />

may be associated with sinus and nonsinus bradycardia, partic -<br />

ularly in ELBW infants. The etiology may be related to a matu -<br />

rational immaturity in the ionic channels of the heart associated<br />

with increased vagal tone. 3,4 Goals during initial resuscitation and<br />

for the first 72 hours or so of life are directed not only at the basic<br />

ventilatory and circulatory requirements but also to avoid the<br />

insults that cause the infant to shunt blood from right-to-left<br />

through a PDA and right-to-left across a foramen ovale owing to<br />

persistent pulmonary hypertension of the newborn (PPHN). This<br />

is particularly challenging in the operating room under conditions<br />

of surgical stress and fluid shifts.<br />

RESUSCITATION OF THE<br />

ELBW INFANT AT BIRTH<br />

The anesthesiologist may be consulted in the delivery room<br />

to assist resuscitation in the ELBW who is particularly prone<br />

to asphyxia and metabolic stress at the time of birth. Early<br />

resuscitation is important. Intracranial hemorrhages in the very<br />

preterm infant during a complicated delivery are often anticipated,<br />

and urgent cesarean section is sometimes advocated to protect<br />

the ELBW infant’s brain from trauma during vaginal delivery.<br />

Metabolic stress predisposes the ELBW infant to anaerobic meta -<br />

bolism and a metabolic acidosis. Complicated protracted labor<br />

and delivery decrease the ELBW infant’s cardiac output further,<br />

thus exacerbating the metabolic acidosis. Resuscitation of the<br />

ELBW infant should begin immediately after delivery by intu -<br />

bation of the trachea and then gentle ventilation of the lungs to<br />

increase the PaO 2<br />

while preventing any lung volume trauma.<br />

The cardiovascular system resuscitation often responds without<br />

inotropic support to correction of the metabolic acidosis with<br />

dilute 4.25% NaHCO 3<br />

at 1 mEq/kg/min to a pH of 7.3. Ensure the<br />

blood glucose concentration is in the range 47 to 90 mg/dL. These<br />

precautions will prevent any further central nervous system injury<br />

from too rapid an intravascular volume expansion with a hyper -<br />

tonic solution (8.5% NaHCO 3<br />

) or hypo- or hyperglycemia. The<br />

infant should be warmed immediately and kept in a thermoneutral<br />

zone aiming for a body temperature of 36°C. A prospective,<br />

randomized trial comparing 55 very low birth weight (VLBW)<br />

infants to 28 ELBW infants requiring intubation and resuscitation<br />

in the delivery room found that goal-directed therapy targeting<br />

arterial oxygen saturation, administration of surfactant, correction<br />

of acidosis, and intravascular volume resuscitation resulted in a<br />

matched study control reduction in mortality from 46% to 18%. 5<br />

The persistence of high lactate levels and a severe metabolic<br />

acidosis despite resuscitative efforts, together with intrauterine<br />

growth retardation, was found to be bad prognostic criteria for<br />

outcome. 5<br />

EARLY MANAGEMENT<br />

Tracheal Intubation<br />

Endotracheal intubation is typically required immediately after<br />

birth in the ELBW infant for respiratory support, and the provider<br />

should take several considerations into account. The concern<br />

during awake intubation in the ELBW infant is that struggling and<br />

crying increase intrathoracic pressure and decrease cerebral<br />

venous return. Increased cerebral venous hypertension occurs<br />

and may lead to increased intracranial pressure and a risk of<br />

intracranial hemorrhage. 6 Further, complications such as laryn -<br />

gospasm, oxygen desaturation, breath-holding, bradycardia, in -<br />

creased systolic blood pressure, or trauma to the airway can<br />

occur. 7–9 The long-term neurocognitive outcomes of performing<br />

awake intubation in ELBW infants are not known. Since 2000,<br />

there has been an increased trend to sedate and administer<br />

analgesia before intubation in neonates of all ages. 9 The issue<br />

of whether it is necessary to give analgesic or anesthetic drugs<br />

before neonatal intubation should now be replaced by a different<br />

question: Is there a reason not to give analgesic or anesthetic<br />

drugs before neonatal intubation? 8,9 Current recommendations<br />

suggest that endotracheal intubation without the use of sedation<br />

or analgesia should be performed only when required emergently<br />

during resuscitation in the delivery room or in other lifethreatening<br />

situations when intravenous access is unavailable. 9–11<br />

The approach to securing the ELBW infant airway via intu -<br />

bation requires meticulous attention, patience, and planning. First,<br />

agents that are used to secure the airway such as succinylcholine,<br />

pancuronium, midazolam, and fentanyl should be prepared<br />

undiluted in 1-mL syringes. Resuscitation medications such<br />

as epinephrine and atropine should also be drawn up in 1-mL<br />

syringes. Second, preparation of the intubating environment<br />

is essential. This involves ensuring a selection of appropriate<br />

laryngoscope blades along with back-up blades. The Miller Zero<br />

is a blade commonly used for the ELBW infant because of its<br />

small size and the light positioned near the tip of the blade for<br />

good visibility. A neonatal McGill forceps and neonatal intubating<br />

stylet should also be on hand. Suction must be available, with a<br />

neonatal catheter tip for clearing the pharynx if needed. There<br />

should be a selection of appropriately small neonatal facemasks,<br />

oral airways, connectors, and an Ambu bag with free-flowing<br />

oxygen attached either from the wall mount in the intensive care<br />

unit or from anesthetic machine if present in the operating room.<br />

The choice of outer diameter (OD) endotracheal tube (ETT)<br />

size is very important if airway injury in the VLBW infant is to be<br />

avoided. A recent study highlights that the VLBW infant airway is<br />

at risk for injury predominantly in the posterior region of the<br />

glottis owing to increased elasticity of these structures. 12 This<br />

elasticity disappears around 37 weeks’ gestational age, and thus,<br />

the slightly older premature infant then becomes at risk for<br />

subglottic injury. Although the normal tracheal mucosa capillary<br />

pressure in preterm infants has not been reported, for cuffed or<br />

uncuffed ETTs, most clinicians accept a leak around the ETT of<br />

less than 20 cmH 2<br />

O. Currently, the recommended safe OD of an<br />

ETT for a 500-g infant is 3 mm and for an 800-g infant 3.5 mm.<br />

This corresponds to an internal diameter of 2.0 mm with the<br />

Mallinckrodt contour ETT. 12 It becomes imperative for the<br />

pediatric anesthesiologist to know the OD of the ETT that is<br />

planned to be placed in the ELBW infant.<br />

Once the practitioner is ready to intubate the VLBW infant,<br />

brief preoxygenation is performed. The use of 100% oxygenation<br />

during resuscitation in term asphyxiated infants may have a longer<br />

deleterious effect as measured by the presence of intraerythrocyte<br />

oxidized glutathione concentration. 13 This oxidative stress<br />

may impair the infant’s ability to mount an antioxidant response<br />

and create a protracted pro-oxidant status in the asphyxiated


1394 PART 4 ■ Special Monitoring and Resuscitation Techniques<br />

newborn infant. 13 More work is needed to determine oxygen’s<br />

role in this setting, but it is reasonable to allow a brief period of<br />

denitrogenation with oxygen before laryngoscopy. A sedation<br />

protocol for intubating ELBW infants can be guided by a recent<br />

double-blind, randomized study comparing the intubating<br />

conditions between two dosing regimen. In preterm infants<br />

weighing less than 1000 g, a 1-minute bolus of midazolam 200 µg/<br />

kg plus morphine 150 µg/kg was compared with a 1-minute<br />

bolus of midazolam 200 µg/kg plus remifentanil 1 µg/kg. 14<br />

The overall intubating conditions were found to be significantly<br />

better (P = .0034) in the remifentanil group than in the morphine<br />

group. 14 However, as discussed later in this chapter in the<br />

“Intraoperative Management” under “Anesthetic Agents,” there are<br />

concerns with the use of midazolam and possible accelerated<br />

neuroapoptosis. The reader is directed to that section and also<br />

<strong>Chapter</strong> 3 for further information on this topic.<br />

Temperature Regulation<br />

Preterm ELBW infants have poor temperature regulation because<br />

they have very little insulation with almost no subcutaneous fat<br />

(see <strong>Chapter</strong> 14). They have translucent skin through which the<br />

blood vessels can be seen coursing very close to the surface; as a<br />

result, exposure to a cold environment will cause them to lose heat<br />

to the environment and increase their oxygen requirements, which<br />

are already high at 4.3 to 5.4 mL/kg/min at birth. The thermoge -<br />

nesis brown fat cells are very sparse in the ELBW infant. They start<br />

to differentiate at 26 to 30 weeks postconception, and continue<br />

differentiating up to 46 weeks. Nonshivering thermogenesis is<br />

immature in the ELBW infant, but does begin to occur at approxi -<br />

mately 30 weeks. Cold perception causes release of norepine -<br />

phrine, which leads to adrenergic stimulation and release of fatty<br />

acids that are combusted exclusively in mito chondria by a unique<br />

brown fat protein called uncoupling protein (UCP) or thermo -<br />

genin. Activation of thermogenin depends on a modulator that<br />

has not yet been identified. 15 The combination of these inabilities<br />

to adapt to the environment requires the use of body wrap and/<br />

or warming devices that preserve heat without drying. Covering<br />

the head in VLBW infants may preserve heat loss by as much as<br />

63 to 73%. 16<br />

Hyperbilirubinemia<br />

In the ELBW infant, liver conjugation and bilirubin elimination<br />

is immature, bowel motility immaturity is present, faster red blood<br />

cell turnover occurs; as a result, these infants are at greater risk for<br />

kernicterus than neonates born at term. Kernicterus involves<br />

unconjugated bilirubin deposition in the pons, cerebellum, and<br />

basal ganglia. Hypoproteinemia and metabolic acidosis increase<br />

the free bilirubin that could potentially cross the immature<br />

blood-brain barrier and worsen kernicterus. Some NICUs begin<br />

phototherapy in ELBW infants at birth; others wait for bilirubin<br />

to increase by 50% of the level at birth. Exchange transfusion<br />

is strongly considered in ELBW infants if bilirubin approaches<br />

10 mg/dL/kg.<br />

Transfusion Practices<br />

The blood volume of ELBW infants is 100 mL/kg. Immature<br />

erythropoiesis in the ELBW infant, together with frequent blood<br />

sampling in the NICU, creates the need for multiple blood trans -<br />

fusions if apnea spells and cerebral bleeds are to be minimized in<br />

these patients. Current management is influenced by two recent<br />

trials. One was directed at maintaining a more liberal hemoglobin<br />

level closer to 12 to 14 g/dL because of the potential benefit of less<br />

intracranial bleeds in ELBW infants. 17 The second study found<br />

that a restrictive transfusion policy resulted in fewer ELBW infants<br />

receiving a transfusion. 18 The balance probably lies somewhere<br />

between the two current views. Clinically controlling the trans -<br />

fusion practice to the degree of respiratory support required by<br />

the infant (restrictive) when they are on room air spontaneously<br />

breathing or (liberal) when intubated and ventilated guides<br />

practice. 18,19 At least one study has addressed the concern of a<br />

decrease in hemoglobin oxygen affinity after a VLBW infant<br />

receives an adult red blood cell transfusion. 20 In this study,<br />

11 infants weighing 736 g (±125 g) received 26.9 mL/kg of packed<br />

red cells. The mean fetal hemoglobin (HbF) fell from 92.9 ± 1.1%<br />

to 42.6 ± 5.7%. Lowering the inspired oxygen concentration and<br />

targeting an arterial saturation of 85% might be an excellent<br />

protective strategy against potential hyperoxygenation and free<br />

radical damage after a blood transfusion in VLBW infants. 20<br />

Leukoreduction before transfusing blood to ELBW infants has<br />

been associated with a decreased incidence of BPD, ROP, NEC,<br />

and IVH in a retrospective study of 515 infants weighing less<br />

than 1250 g. 21 Further studies are required to assess whether<br />

leukoreduction has any effect on mortality. Single-donor exposure<br />

for each ELBW infant is also advocated by many units and blood<br />

banks. 22 An alternative approach is administration of erythro -<br />

poietin 200 IU/kg/dose subcutaneously twice a week with 4 to<br />

6 mg/kg/d iron supplement. This strategy, however, did not<br />

prevent 2 to 3 blood transfusions/patient/NICU stay in a group of<br />

ELBW patients. 23 A recent Cochrane review found similar results<br />

in which fewer blood transfusions are given if erythropoietin is<br />

used, but no clinically significant evidence is noted in current<br />

clinical trials. 24 Other factors determining the need for transfusion<br />

include birthweight; initial hemoglobin value; 5-min Apgar score;<br />

phlebotomy loss; duration of mechanical ventilation; duration of<br />

oxygen supplement; and incidence of IVH, surgery, and chronic<br />

lung disease. 24 Other studies have shown a decrease in the need<br />

for red blood cell transfusion in ELBW infants when a more<br />

restrictive transfusion guideline policy is adopted. The incidence<br />

of complications and chronic disease in these NICU graduates was<br />

no different in this study. 25 The rational intraoperative strategy<br />

from these studies informs the anesthesiologist to consider that a<br />

600-g ELBW infant only has 60 mL of circulating blood volume.<br />

Any significant surgical losses should be replaced promptly, ideally<br />

with irradiated leukoreduced red blood cells during anesthesia.<br />

The clinical signs of acute anemia in VLBW infants are often not<br />

easily seen until cardiovascular collapse occurs. It may be inferred<br />

from loss of the pulse oximetry trace or a sudden drop in<br />

cardiac output associated with a fall in end-tidal carbon dioxide<br />

measurement. At all times during anesthesia, blood loss must be<br />

looked for clinically and corrected promptly. Regular checks of<br />

hemoglobin should be made during protracted surgery.<br />

RESPIRATORY CONSIDERATIONS<br />

Bronchopulmonary Dysplasia<br />

Northway and colleagues published two seminal papers on lung<br />

disease following mechanical ventilation and oxygen supplemen -<br />

tation in premature infants. 26-27 They showed that changes in the


CHAPTER 83 ■ Specific Problems and Anesthesia Management of Extremely Low Birthweight Infants 1395<br />

lungs that occur at birth can have effects well into adolescence and<br />

adulthood. BPD is defined as a respiratory disease requiring<br />

supplemental oxygen for more than 28 days after birth. The grade<br />

of severity is determined at 36 postgestational weeks for infants<br />

born less than 32 weeks of age and at 56 days of life for infants<br />

born at greater than 32 weeks. Mild disease requires no additional<br />

fractional concentration of oxygen in inspired gas (FI O2<br />

) over 21%,<br />

moderate disease requires a supplemental FI O2 of 0.22 to 0.29, and<br />

severe disease requires an FIO2 of greater than 0.30, continuous<br />

positive airway pressure, or mechanical ventilation. 28 BPD occurs<br />

in about 20% of infants less than 1500 g born at less than 30 weeks<br />

postconception. 29<br />

The important implications for the anesthesiologist are that<br />

these babies exhibit bronchospasm more frequently than controls,<br />

and as many as 50% will require a hospital admission within the<br />

first year of life. 28,30 In addition, 50 to 60% of adolescents who had<br />

BPD as neonates will have hyperreactive airways. 31 Under<br />

anesthesia, the ex-BPD patient may not respond in quite the same<br />

positive therapeutic way as asthmatics do when given a β 2<br />

agonist<br />

bronchodilator or steroidal anti-inflammatory inhalational<br />

therapy. 28 This puts the anesthesiologist in a difficult position to<br />

manage any intraoperative bronchospastic events that result in<br />

desaturations.<br />

These infants may have a higher baseline arterial carbon<br />

dioxide pressure (PaCO 2<br />

), and it is important not to hyperventilate<br />

them during anesthesia or expose them to too high an FI O2<br />

concentration despite the natural tendency to do so. In view of<br />

increased airway reactivity, it would be prudent to plan an<br />

anesthetic without having to instrument the patient’s airway if at<br />

all possible in those infants who have a history of BPD but are<br />

otherwise stable from a respiratory standpoint. Although there are<br />

no randomized, prospective studies on this topic, it is common<br />

practice in some centers to try to avoid general anesthesia for these<br />

children. Regional anesthetic techniques avoid instrumentation<br />

of the airway.<br />

Persistent Pulmonary Hypertension<br />

PPHN is a disease that is found usually secondary to hypoxic<br />

respiratory failure that was induced by significant lung pathology<br />

soon after birth. Such insults that can lead to PPHN include<br />

meconium aspiration, pneumonia, BPD, and infant respiratory<br />

distress syndrome. Other causes include intrauterine maternal<br />

factors that induce significant pulmonary hypoplasia such as<br />

premature rupture of membranes, oligohydramnios, and sepsis.<br />

Congenital diaphragmatic hernia is another cause of pulmonary<br />

hypoplasia. Intrauterine renal disorders have also been implicated<br />

in oligohydramnios and pulmonary hypoplasia. 32 The use of<br />

inhaled nitric oxide (iNO), a potent pulmonary vasodilator, is<br />

clearly beneficial in improving arterial oxygenation and reducing<br />

the rate of extracorporeal membrane oxygenation use in term<br />

or near-term infants with PPHN. 32–34 This has also been shown in<br />

animal studies. This same benefit, however, is not seen in the<br />

ELBW infant or in premature animal studies, and this is probably<br />

because of the physiologic immaturity of the nitric oxide signaling<br />

pathways and the anatomic immaturity of the vascular smooth<br />

muscle in this premature infant population group. 32 A Cochrane<br />

review 33 and a consensus paper 34 have confirmed that there is<br />

currently no support for the use of iNO in ELBW and preterm<br />

infants with respiratory failure.<br />

High-frequency oscillatory ventilation of the ELBW infant is<br />

sometimes necessary to prevent lung injury via conventional<br />

mechanical ventilation. This often prevents an ELBW infant from<br />

safely being transported to the operating room. If surgery is<br />

emergent, it should then be instituted at the bedside in the NICU.<br />

Mechanical Ventilation<br />

and Weaning Strategies<br />

The need to intubate the trachea, mechanically ventilate and<br />

support the respiratory system, in ELBW infants is one of the<br />

therapeutic cornerstones of NICU care. The challenge is to allow<br />

transitional physiologic and anatomic growth and development of<br />

the pulmonary vasculature, bronchial tree, and alveoli to occur<br />

while avoiding the development of chronic lung disease changes<br />

of pulmonary hypertension or BPD. A goal of therapy may be to<br />

allow the ELBW infant to be discharged from the NICU without<br />

the need for supplemental inspired oxygen at 36 weeks post -<br />

conceptional age.<br />

Despite the potential life-saving benefits that are obtained by<br />

intubating and ventilating critically ill infants, it is still evident that<br />

mechanical ventilation can cause lung injury. 35,36 Modern NICU<br />

mechanical ventilation strategies aim at gentle ventilatory support<br />

with small tidal volumes and low mean airway pressures to avoid<br />

volume trauma and barotrauma. Experimental evidence indicates<br />

that low tidal volumes and higher peak pressures are responsible<br />

for the least amount of lung injury with decreased incidence of<br />

pulmonary edema, epithelial injury, and hyaline membrane<br />

disease. 36,37 Experimental evidence also indicates that high endinspiratory<br />

volume may be the determining factor in ventilatorinduced<br />

acute lung injury rather than functional residual capacity<br />

or tidal volume. 36,38 To further protect the neonatal lung, applying<br />

high positive end-expiratory pressure (PEEP) to each breath cycle<br />

will decrease damaging repeated opening and collapse of alveoli<br />

throughout each cycle. 36,39 To confirm the benefits of these venti -<br />

latory approaches, markers of lung injury including filtration<br />

coefficient and lymphatic flow remain normal when a low–tidal<br />

volume, high-pressure strategy of ventilation is used. 36,40<br />

These ventilator strategies often result in a rise in the patient’s<br />

PaCO 2<br />

and a decrease in PaO 2<br />

and may be considered by the<br />

terms permissive hypercapnia and permissive hypoxia. Along with<br />

the administration of aerosolized surfactant, these ventilatory<br />

strategies that allow permissive hypercapnia and a lower PaO 2<br />

are<br />

arguably some of the greatest medical advances in neonatology<br />

since the early 2000s. In a premature lamb model, high tidal<br />

volumes reduced lung compliance by at least 50% within 4 hours<br />

after birth. 41 In addition, in the lamb with surfactant-deficient<br />

lungs, even six breaths with tidal volumes of 35 to 40 cm 3 /kg will<br />

reduce lung compliance. 36,41<br />

It is up to the pediatric anesthesia providers to continue these<br />

strategies of protective ventilation when the ELBW infant is in the<br />

operating room under their care. Anesthesia machines and<br />

operating room ventilators have developed significantly since the<br />

early 2000s; however, it is not necessarily common practice to have<br />

a neonatal intensive care ventilator in the operating room. The<br />

typical operating room ventilator has fewer options than those in<br />

the NICU in terms of mode of ventilation. In addition, there is<br />

more deadspace and seldom does an operating room ventilator<br />

have a decelerating gas insufflation waveform pattern that would<br />

mimic the physiologic ELBW lung gas flow. It is often beneficial


1396 PART 4 ■ Special Monitoring and Resuscitation Techniques<br />

to bring the infant’s personal intensive care ventilators to the<br />

operating room for those infants on specialized settings and to<br />

communicate with the neonatology team as to the ventilatory<br />

strategy that has worked best for that particular infant. Although<br />

the goal would be to use the exact respiratory parameters used in<br />

the NICU, it may be necessary to adjust the operating room<br />

ventilators to mimic that used in the NICU without being exact<br />

owing to the differences in ventilators. For example, in the infant<br />

who is on intermittent positive-pressure ventilation (IPPV) at a<br />

peak pressure of 16 cmH 2<br />

O and PEEP of 5 cmH 2<br />

O in the NICU at<br />

a rate of 24, these same parameters should be set for the operating<br />

room. However, once the infant is attached to the operating room<br />

ventilator, one must watch the chest movement and CO 2<br />

waveform<br />

to ensure that there is no hypoinflation or overinflation. Ideally,<br />

a ventilator with flow volume loops in the operating room<br />

would be invaluable, but often not practical in many settings. Once<br />

the infant has her or his parameters adjusted for normal CO 2<br />

waveform and bilateral breath sounds with normal excursion<br />

for an ELBW infant, the FIO 2<br />

should be decreased to a value that<br />

provides saturations in the low to mid 90s as discussed in Mecha -<br />

nical Ventilation. An arterial blood gas should be drawn when<br />

possible to determine adequacy of ventilation and oxy genation at<br />

steady state. If this is not possible, close monitoring of the CO 2<br />

by<br />

end-tidal measurement with particular attention to the plateau of<br />

the waveform (goal of flat waveform vs sloping that is suggestive<br />

of obstructive pattern) should be sufficient.<br />

When an infant is brought to the operating room not mechani -<br />

cally ventilated, once the trachea of the infant is intubated, settings<br />

of peak inspiratory pressure of 16 to 18 cmH 2<br />

O minimum along<br />

with PEEP of 5 cmH 2<br />

O and rate of 20 may be considered a safe<br />

starting point. Modern anesthesia machines are currently able to<br />

deliver IPPV and PEEP suitable for ELBW infants as part of the<br />

upgrades for anesthesia delivery systems. However, they remain<br />

unable to deliver high-frequency oscillation or high-frequency jet<br />

ventilation, and those infants still require that their ventilator be<br />

transported to the operating room with them.<br />

As the ventilated ELBW infant’s lungs mature, an extubation<br />

strategy needs to be planned. A recent study of weaned ELBW<br />

infants in the NICU compared delayed extubation (36 h) to imme -<br />

diate extubation once weaning criteria were met. 42 Re-intubation<br />

within 7 days of extubation was the primary endpoint. Delayed<br />

extubation did not prevent re-intubation statistically in this study<br />

of 86 ELBW infants aged younger than 28 weeks’ gestation. In the<br />

event that an ELBW infant has been successfully weaned off the<br />

ventilator, but now must come to the operating room for a surgical<br />

procedure, the anesthesia team must likewise determine an<br />

extubation strategy. Although it is difficult to extrapolate NICU<br />

data regarding weaning to the operating room, the principles<br />

remain the same and continued vigilance is required when the<br />

anesthesiologist deems an ELBW infant ready for extubation. The<br />

risk of apnea and the need for re-intubation are ever present in<br />

the ELBW infant. Very often, the safest option for these small<br />

babies is to return them intubated to the NICU after an anesthetic<br />

is administered in the operating room.<br />

In the infant who is extubated postoperatively but continues to<br />

have periods of apnea, therapy is usually supportive with 4 to 6 cm<br />

of continuous positive airway pressure (CPAP) and/or caffeine at<br />

a dose of up to 10 mg/kg. 43 Caffeine, a xanthine oxidase inhibitor,<br />

is thought to act as an adenosine receptor antagonist. 44 Activation<br />

of adenosine 2A receptors appears to excite GABAminergic inter -<br />

neurons, and it is hypothesized that released gamma-aminobutyric<br />

acid (GABA) may contribute to the respiratory inhibition. 44 The<br />

use of caffeine in ex–premature infants has been well described in<br />

the literature for use in avoiding apnea during the perioperative<br />

period. 43 This is primarily recommended for ex–premature infants<br />

for hernia repair but can be applicable to the rare particularly<br />

robust ELBW infant. A recent study measured serum caffeine<br />

concentrations approximately 7 days after starting therapy with a<br />

20- or 25-mg/kg loading dose and a 6-mg/kg/d maintenance dose<br />

in 154 infants with a mean gestational age of 29 weeks. 45 The 25th<br />

to 75th percentile range for the serum caffeine concentrations<br />

with the two dosing regimens was equivalent, approximately 18<br />

to 23 mg/L. 45 The authors conclude that routine measurement<br />

of steady-state serum caffeine concentrations in infants of 24 to<br />

35 weeks gestational age is not required in the absence of ongoing<br />

apnea/hypopnea or signs compatible with toxicity. 45<br />

Chronic Lung Disease<br />

Preventing chronic lung disease from the complication of severe<br />

unilateral pulmonary interstitial emphysema is a challenge in<br />

ELBW infants. It is often unilateral and caused by barotrauma after<br />

mechanical ventilation. 46 A variety of options exist for therapy<br />

including selective left or right lung ventilation for 1 to 10 days<br />

to allow resolution of the emphysema. 47 A recent case report<br />

highlighting the benefit of a successful single-lung ventilation<br />

technique in the ELBW infant indicates that single-lung venti -<br />

lation in these children is often therapeutic. 48<br />

CARDIOVASCULAR CONSIDERATIONS<br />

The most commonly used clinical “rule of thumb” is that the<br />

minimal mean blood pressure in millimeters of mercury in the<br />

first days after birth should be equal to the gestational age in<br />

weeks. 49–52 This has been widely recommended since 1992 and is<br />

often used as the criterion for hypotension; however, there does<br />

not appear to be any published literature to support this widely<br />

held recommendation. 51,52 After a few days of birth, the blood<br />

pressure steadily increases in well ELBW infants.<br />

The problem for clinicians is what blood pressure value is<br />

deemed hypotensive? What should it be treated with? What are<br />

the risks of hypotension in the ELBW infant?<br />

The initial treatment for hypotension among 96% of Canadian<br />

neonatologists is the administration of an intravenous fluid bolus. 53<br />

Despite this, it has been shown repeatedly over many years that<br />

there is no relationship between blood pressure and intravascular<br />

volume in ELBW infants. 52,54 Currently, it is not clinically advised<br />

to treat a low blood pressure number in ELBW infants with an<br />

intravascular fluid bolus. We now know that administration of<br />

excessive intravascular fluids is deleterious to the ELBW infant.<br />

It is, therefore, recommended to look for additional signs of<br />

organ hypoperfusion such as poor capillary refill time, oliguria, or<br />

metabolic acidosis or cerebral hypoperfusion. 55,56 The major<br />

concern for the ELBW infant relates to whether or not cerebral<br />

blood flow (CBF) will remain adequate preventing an intracerebral<br />

hemorrhage during a period of systemic hypotension. The concern<br />

for neonatologists is that CBF may not be autoregulated adequately<br />

in the presence of systemic hypotension in ELBW infants.<br />

Neonatologists try to keep the mean blood pressure in ELBW<br />

infants at 30 mmHg to prevent cerebral white matter injury and<br />

cerebral hemorrhage. 57 At least one study in 1.5- to 40-hour-old


CHAPTER 83 ■ Specific Problems and Anesthesia Management of Extremely Low Birthweight Infants 1397<br />

ELBW infants, utilizing near-infrared spectroscopy to monitor<br />

cerebral tissue oxygenation has addressed this problem. It was<br />

found that, in this ELBW patient study group, cerebral autoregu -<br />

lation is functional in normotensive but not hypotensive infants. 56<br />

A threshold below which CBF autoregulation fails to adequately<br />

protect the ELBW infant brain was found to be at 29 to 30 mmHg.<br />

In this study, the administration of dopamine improved both mean<br />

arterial pressure (MAP) and CBF. 56 The authors stress that there is<br />

a ceiling to higher blood pressure, above which the risk of cerebral<br />

autoregulation would fail and CBF would remain pressure passive<br />

flow-related. It is believed that this hypertension would increase<br />

the risk of intracranial hemorrhage. What that blood pressure<br />

would be is hard to quantify. A recent study in 30 patients looking<br />

at CBF velocities in ELBW infants found that there was no<br />

difference in mean CBF velocity (P = .934) in infants with hypo -<br />

tension (MAP 23 mmHg [range 20–24.9]) compared with infants<br />

with normal blood pressure (MAP 32.6 mmHg [range 27.5–35.7]).<br />

Important from this study is that hypotension may not always<br />

be an indicator of decreased CBF. Therefore, before treating all<br />

hypotension, one should be observant of other signs of other organ<br />

hypoperfusion such as lactic acidosis and decreased urine output. 58<br />

When an ELBW infant receives an inotrope, blood pressure<br />

must be monitored, ideally utilizing invasive blood pressure<br />

monitoring. Until randomized, prospective, controlled trials are<br />

completed comparing long-term outcomes in ELBW infants with<br />

or without hypotension, it is still recommended to aim for an MAP<br />

close to the age of the ELBW infant in postconceptional weeks.<br />

From these data, it can be seen that the pediatric anesthesiologist<br />

needs to be vigilant in controlling blood pressure in ELBW infants<br />

during anesthesia, ideally aiming for an MAP of 30 mmHg and<br />

not being too hasty in administering intravascular volume unless<br />

it is clear that hypovolemia exists. An inotrope such as dopamine<br />

at low-dose 3 to 5 µg/kg/min might be a more appropriate choice<br />

aiming to keep the MAP between 30 and 40 mmHg. Caution<br />

needs to be exercised with the addition of inotropes because<br />

raising blood pressure in ELBW infants may be associated with<br />

intracerebral bleeds. 56,58<br />

The one area of concern when providing anesthesia during the<br />

first week of life to ELBW infants is that 30% of these patients may<br />

have delayed refractory hypotension nonresponsive to fluid,<br />

inotrope, or hydrocortisone therapy. 59 A differential diagnosis must<br />

be considered: including PDA, adrenal insufficiency, high mean<br />

airway pressure during mechanical ventilation, pulmonary air<br />

leaks, cardiac arrhythmias, electrolyte imbalance (hypocalcemia,<br />

hyperkalemia, hypomagnesemia), myocardial depression from<br />

TABLE 83-2. Causes of Severe Hypotension in the<br />

Extremely Low Birthweight Infant<br />

PDA<br />

Adrenal insufficiency<br />

High mean airway pressure, pulmonary air leaks<br />

Cardiac arrhythmia<br />

Myocardial depression from hypoglycemia<br />

Sepsis, necrotizing enterocolitis<br />

Metabolic acidosis—inborn errors of metabolism<br />

Electrolyte imbalance (hypocalcemia, hyperkalemia,<br />

hypomagnesemia)<br />

PDA = patent ductus arteriosus.<br />

From Sakar 2007 reference 117.<br />

hypoglycemia, sepsis, NEC, or severe metabolic acidosis from<br />

inborn errors of metabolism. 59 The PDA is often the cause and<br />

usually follows administration of surfactant. 59 If detected, it should<br />

be closed medically or surgically as soon as feasible.<br />

INTRAOPERATIVE MANAGEMENT<br />

Fluid, Energy, and Acid-Base Considerations<br />

Perhaps one of the more controversial topics in the management<br />

of the preterm infant, particularly one who is ELBW, is the use of<br />

intraoperative fluids. Traditionally, neonatologists manage hypo -<br />

tension with vasopressors whereas anesthesiologists administer<br />

volume expanders. One must remember that causes of hypo -<br />

tension are different in both situations. For instance, the use of<br />

fluid intraoperatively is supported by the fact that vasoactive<br />

anesthetics needed for surgical conditions cause a drop in systemic<br />

vascular resistance (SVR) and hypovolemia that needs to be<br />

first treated intravascularly with the administration of liberal<br />

intravenous fluids to make up for any preoperative hypovolemia.<br />

The current data would support that the neonatologist’s<br />

viewpoint outside the scope of anesthesia is more appropriate.<br />

A recent Cochrane review on fluid management of preterm infants<br />

was designed specifically to look at the effects of water intake on<br />

postnatal weight loss and the risks of dehydration, PDA, NEC,<br />

BPD, intracranial hemorrhage, and death in premature infants. 60<br />

Analysis of five studies that fulfilled the strict criteria for review<br />

when taken together indicate that restricted water intake signi -<br />

ficantly increases postnatal weight loss and significantly reduces<br />

the risks of PDA and NEC. With restricted water intake, there<br />

are trends toward increased risk of dehydration and reduced<br />

risks of BPD, intracranial hemorrhage, and death, but these<br />

trends are not statistically significant. Care should be exercised<br />

not to overhydrate the ELBW infant perioperatively. 60 However,<br />

these precautions must be weighed against the risks that occur<br />

with ongoing surgical losses of fluid and blood during surgical<br />

procedures.<br />

Essentially, the three major considerations in the adminis -<br />

tration of fluids to ELBW infant perioperatively are<br />

1. Strict control of the volume and rate of infusion of intravenous<br />

fluids.<br />

2. Meticulous attention to the infused sodium concentration.<br />

3. Prevention of hyper- or hypoglycemia.<br />

Intravascular hypovolemia should be corrected promptly with<br />

20 to 40 mL/kg of normal saline. 61,62 Currently, particularly in the<br />

postoperative period, there is a strong trend to limit the average<br />

fluid maintenance volume to half or two thirds of Holliday and<br />

Segar’s recommended 4/2/1 rule of maintenance need for water<br />

in parenteral fluid therapy. 61–63 In ELBW infants, if fluid volumes<br />

greater than 130 mL/kg are administered daily, the ductus arte -<br />

riosus is more likely to remain open. This increases the chance of<br />

congestive heart failure and low cardiac output and may result in<br />

diminished gastrointestinal perfusion. Diminished gastrointestinal<br />

perfusion is a major contributor to the risk of developing NEC<br />

and one of the main reasons why fluids are restricted in ELBW<br />

infants who are at risk of maintaining a PDA. The other reason to<br />

restrict fluids in these infants is to try to avoid the development of<br />

BPD. Overhydration, particularly when associated with hypo -<br />

natremia less than 120 mEq/L, will predispose ELBW infants to<br />

seizure development. Should hyponatremia develop, therapy


1398 PART 4 ■ Special Monitoring and Resuscitation Techniques<br />

should be aimed at fluid restriction. It is important for the<br />

anesthesiologist to be aware that, in the first 12 to 48 hours of life,<br />

the ELBW infant undergoes a significant shift in potassium from<br />

the intracellular space to the extracellular fluid compartment.<br />

During this time, 32 to 50% of ELBW may have a hyperkalemia<br />

greater than 6.7 mmol/L. 64 Pathologic and significantly dangerous<br />

hyperkalemia greater than 6.7 mmol/L should be treated by<br />

insulin and dextrose infusion as the first line of therapy in the<br />

premature infant. Oral and rectal ion exchange resins should be<br />

avoided because they are ineffective and associated with signifi -<br />

cant and potentially life-threatening complications. Exchange<br />

transfusion may be used as a last-resort therapy. 64 Nonpathologic<br />

hyperkalemia can also be worrying and require similar therapy.<br />

It is often seen before ELBW infants have started a diuresis. 65,66<br />

During the subsequent diuresis stage as they begin to lose 10%<br />

of their initial birth weight due to maturation in their kidneys<br />

and glomerular filtration rate, a hypokalemia may be present.<br />

During this time, potassium supplementation becomes necessary.<br />

These considerations should be taken into account by the anes -<br />

thesiologist when planning intravascular blood and fluid replace -<br />

ment in the ELBW infant undergoing surgery.<br />

From 24 to 28 weeks’ gestation, the fetus grows at approxi -<br />

mately 15 g/kg/d, accumulating approximately 25 kcal/kg of new<br />

tissue per day. 67 In premature infants of approximately 32 to<br />

34 weeks’ gestational age, in order to continue this type of weight<br />

gain, an enteral intake of 120 to 130 kcal/kg/d would be required. 68<br />

The energy requirements of ELBW infants intraoperatively<br />

have not been fully elucidated. The ELBW infant’s basal glucose<br />

production is 6 to 10 mg/kg/min. Because of their catabolic state<br />

immediately after delivery, additional dextrose needs to be admin -<br />

istered intravenously. The amount administered is important in<br />

order to prevent any hyperosmolar hypotonic intravascular<br />

environment from developing. Targeting osmotic diuresis with the<br />

presence of greater than 1000 mg/dL or 56 mmol/L of urinary<br />

glucose may suggest a risk of osmolar changes and warrant strict<br />

glucose control in sick infants. 69,70<br />

Practice guidelines warn that to discontinue parenteral nutri -<br />

tion intraoperatively without providing a dextrose infusion could<br />

precipitously lower the blood glucose level. 71 This lack of energy<br />

source could potentially contribute to neurologic damage in<br />

the developing ELBW infant. There appears to be no consensus<br />

regarding the optimal dextrose content of infused fluids periop -<br />

eratively. The current trends are to administer only low-dose<br />

dextrose (0.95 or 1%), rather than a 5 to 10% dextrose-containing<br />

intravenous fluid solution when measured blood glucose is less<br />

than 2.8 mmol/L or 50 mg/dL. During long surgeries, frequent<br />

measurement of the blood glucose level is recommended. 62,72 The<br />

route of intravenous fluid administration is usually through a<br />

24- to 26-gauge peripherally placed central catheter. As an aside,<br />

these lines should not be flushed with syringes smaller than<br />

10 mL, thus preventing pressure-related catheter rupture. Mainte -<br />

nance fluids and total parenteral nutrition from the unit should<br />

be continued while a venous cannula should be inserted in order<br />

to cautiously administer lactated Ringer’s solution for replacement<br />

of surgical third space losses. Intravascular fluid requirements are<br />

titrated to blood pressure and heart rate. Surgical blood loss should<br />

promptly be replaced with an equal volume of packed red cells<br />

(see <strong>Chapter</strong> 53).<br />

Acid-base management pertains to first ensuring adequate<br />

cardiac output in the VLBW infant. If a metabolic acidosis occurs,<br />

the cause should immediately be sought and treated. Cardio -<br />

vascular resuscitation and support of the infant should be<br />

instituted before metabolic correction of the pH is attempted.<br />

Administration of dilute intravenous NaHCO 3<br />

must always be<br />

titrated carefully to avoid hypernatremia and the risk of IVH.<br />

Pain and Sedation Requirements<br />

Premature infants have a lower pain threshold than term babies,<br />

and pain activates cortical regions in the preterm infant brain. 73–75<br />

This finding has been observed by cortical spectroscopy. 74 In<br />

addition, repeated painful stimuli in preterm infants, particularly<br />

in the slower maturing inhibitory nerve fibers of the lower limb<br />

compared with the upper limb, result in wind-up, temporal sum -<br />

mation, and hyperalgesia. 75 These effects may last for as long as<br />

30 to 60 minutes poststimulation and may lead to a maturation of<br />

the cortical response to pain in preterm infants. 73,75<br />

A recent study concluded that the average NICU infant receives<br />

as many as 12 to 15 moderate to severely painful procedures per<br />

day. Despite this, less than 35% of these procedures are preceded<br />

by any pre-emptive analgesia. 75 Pre-emptive morphine infusions<br />

have not been found to decrease the frequency of severe IVH,<br />

periventricular leukoplakia, or death in ventilated preterm<br />

neonates. However, a loading dose of 100 µg/kg in addition to an<br />

infusion of morphine 3 to 5 µg/kg/h may provide effective<br />

sedation and analgesia in the ELBW infant only if they are already<br />

intubated and ventilated but not tolerating their ventilatory<br />

support. 75 Because of a more favorable withdrawal profile, metha -<br />

done is being investigated for use in the ELBW infant. 75 Fentanyl,<br />

with an onset time of 2 to 3 minutes, short duration of action of<br />

60 minutes, and stable hemodynamic profile, is a analgesic com -<br />

monly used by anesthesiologists for infants in the perioperative<br />

period. Although there is ongoing investigation on the effects<br />

of fentanyl in the developing brain and an association with peri -<br />

ventricular leukomalacia, the benefits of using appropriate<br />

opioids such as fentanyl outweigh the risks of the stress of invasive<br />

procedures in infants who do not receive opioids. These risks and<br />

benefits must be considered with all agents used in this fragile<br />

population. 76<br />

Adequate analgesia needs to be provided by the anesthe -<br />

siologist caring for the ELBW infant in the perioperative period.<br />

However, excessive administration of opioids may be associated<br />

with hypotension and lead to undesirable secondary effects.<br />

This is especially true in infants under 750 grams when cortisol<br />

production can be inhibited by fentanyl in doses of greater than<br />

20 µg/kg. This decrease in cortisol production can result in<br />

refractory hypotension. 77 As with many therapeutic modalities in<br />

the ELBW infant, the administration of analgesia becomes a<br />

balancing act and the choices must be made when providing nar -<br />

cotics for analgesia, because the lack of analgesia itself may also<br />

have deleterious effects secondary to activation of the neuro -<br />

endocrine stress response.<br />

The use of sedatives also deserves significant consideration in<br />

this population of tiny patients. Administration of 0.2 mg/kg of<br />

midazolam followed by 0.2 mg/kg/h in ELBW infants has been<br />

shown to be associated with a transient 14.7% decrease in CBF<br />

velocity as measured by near-infrared spectroscopy. 78 In this same<br />

study, when 0.05 mg/kg of morphine was followed by 0.01 mg/<br />

kg/h, an increase in cerebral blood volume of 11% was seen<br />

without any significant change in CBF velocity. 78 The conclusion<br />

for the anesthesia provider from this recent study is that medi -<br />

cations used in the operating room and NICU can significantly


CHAPTER 83 ■ Specific Problems and Anesthesia Management of Extremely Low Birthweight Infants 1399<br />

affect CBF and blood volume. Therefore, attention needs to be<br />

directed to a judicious and gradually titrated analgesic or sedative<br />

effect. In addition, as is discussed the following section, there are<br />

concerns with the use of midazolam and possible accelerated<br />

neuroapoptosis.<br />

Anesthetic Agents<br />

The potential for accelerated neural cell death or apoptosis in<br />

premature human infant brains exposed to multiple anesthetic<br />

agents has been suggested and has prompted international interest<br />

in the use of anesthetic agents in infants with developing brains 79,80<br />

(see <strong>Chapter</strong>s 3). This speculative frenzy has been fueled by neural<br />

degeneration evidence from postnatal rodent and primate in vivo<br />

and in vitro animal models. In these models, control of apoptotic<br />

variables such as hemodynamics, nutrition, oxygenation, and CO 2<br />

elimination was achieved. <strong>81</strong>–83 For iso flurane, for example, an agedependent<br />

and duration-dependent relationship has been shown<br />

between the isoflurane exposure and rodent perinatal neuronal<br />

death. 79,80,82 A variety of general anes thetics, which act primarily as<br />

GABA receptor modulators and N-methyl-D-aspartic acid gluta -<br />

mate receptor antagonists, has been shown to cause this rodent<br />

neuron degeneration. <strong>81</strong>,83,84 At this time, the major question as to<br />

how these neonatal animal studies may be translated to human<br />

clinical significance remains unanswered. It is unclear whether<br />

high-dose anesthetic agent cocktail exposure over several hours<br />

in immature mice or rat brains and the resultant apoptosis can be<br />

comparable with anesthetic agent exposure in the premature<br />

human infant brain that takes many months to develop. <strong>81</strong>,82,84 Until<br />

anesthetic agent–induced neuronal degeneration in humans is<br />

identified, the applicability of this animal data to premature<br />

human infants remains speculative. Current analgesic and anes -<br />

thetic practice in ELBW infants should continue, particularly<br />

based on past evidence that shows lack of anesthesia or analgesia<br />

results in significant morbidity or mortality in the fetus and<br />

infant, 84–87 although it is important to note that our NICU tries to<br />

avoid the use of intravenous benzodiazepines for sedation in the<br />

ELBW infant.<br />

Because of the surrounding issues on the use of anesthetic<br />

agents in premature infants, it is imperative that the anesthe -<br />

siologist be aware of the dosing and potential hazards before<br />

delivering these agents to this fragile population. With years of<br />

experience and data on safety, inhalational anesthetic agents<br />

currently continue to be commonly used in premature infants<br />

undergoing general anesthesia. In a study involving 10 premature<br />

infants, it was shown that the blood-gas partition coefficients of<br />

isoflurane, halothane, and sevoflurane in preterm neonates are<br />

similar to those in full-term neonates and that gestational age<br />

does not significantly affect the blood-gas solubility. 88 There are,<br />

however, no reports specifically randomizing ELBW infants to<br />

different inhalational type anesthetics. In one study, 30 infants less<br />

than 37 weeks’ gestation and less than 47 weeks’ postconceptional<br />

age undergoing inguinal herniotomy had an inhalational induc -<br />

tion with sevoflurane and were randomly allocated to sevoflurane<br />

or desflurane for maintenance. Median times to first movement,<br />

tracheal extubation, eye opening, and first cry were all faster when<br />

anesthesia was maintained with desflurane as compared with<br />

sevoflurane. No difference in postoperative respiratory apnea<br />

events was demonstrated between the groups. 89 However, des -<br />

flurane is not commonly used in young children, particularly for<br />

induction, owing to its risk of airway reactivity, and there are no<br />

data to support its use in the ELBW infant. 90<br />

A recent NICU randomized trial comparing intubation under<br />

2 to 5% sevoflurane compared with 100% oxygen in 30 premature<br />

and small neonates found that intubation and glottic visualization<br />

was easier in the sevoflurane group: no movements: 95.5%<br />

versus 28% (P < .005); good glottis visualization: 73% versus 33%<br />

(P = .013) compared with the oxygen-alone group. Untoward<br />

effects of hypertension in the sevoflurane group 25% versus<br />

56.3% (P = .04) in the oxygen-only group and the incidence<br />

of bradycardias 8.3% versus 44.4% (P < .01) were greater in the<br />

oxygen control group. This important prospective, randomized<br />

trial confirms the safety of sevoflurane inhalational induction<br />

of anesthesia for intubation in small infants. 91 Regarding other<br />

medications used in ELBW infants and children, recently a study<br />

noted 45% of parenteral medications used in neonates were used<br />

for off-label purposes. The parenteral medications most frequently<br />

used were analgesics, vasopressors, and hematologic agents. 92<br />

The extent of this off-label usage of medications problem warrants<br />

further drug studies, particularly in small children.<br />

ANESTHESIA FOR<br />

SPECIFIC PROCEDURES<br />

The ELBW infant may present for a variety of procedures with<br />

some of the more common ones being presented here. Regardless<br />

of the type of procedures, certain standards must apply to all of<br />

these young patients. First, the procedure room must be equipped<br />

with the small supplies appropriate for the size of these patients.<br />

The room should be warmed during setup so that heat loss is at a<br />

minimum from any conductive surfaces or instruments. Fluids<br />

must be setup on pumps to avoid unintentional volume overload,<br />

and drugs must be put in appropriately sized syringes at concen -<br />

trations that allow accurate delivery.<br />

All infants in this age require the usual American Society<br />

of Anesthesiologists (ASA) monitors, but additional helpful<br />

information may be obtained by a second pulse oximeter to allow<br />

preductal and postductal saturation monitoring.<br />

Intravenous access is essential. Percutaneous intravenous<br />

central catheter (PICC) lines are often used in small infants, but<br />

their small diameter and long length do not allow for rapid<br />

infusion of medications or fluids that are often required in an<br />

operative setting. In fact, a 24-gauge PICC line requires a 10-mL<br />

syringe to be used for infusion because smaller syringes allow<br />

greater pressures that may rupture the catheter. Whether to<br />

perform procedures in the NICU versus the operating room is<br />

often institution-dependent. An operating room environment<br />

typically provides more room, better light, and the ability to use<br />

the anesthesia team and operating room nurses effectively. The<br />

primary advantage to operating in the NICU is that transport<br />

becomes unnecessary, and it is often life-threatening in a severely<br />

ill premature infant who is on significant respiratory and/or<br />

hemodynamic support. Each of these cases should be considered<br />

on an individual basis with an open discussion between the<br />

anesthesia, the surgical, and the neonatal teams.<br />

Ligation of PDA<br />

Owing to the advances in neonatology and improved outcomes<br />

for the ELBW infant, PDA is the most common congenital heart<br />

defect (CHD) in the neonate. It is present in 0.05% of term infants,


1400 PART 4 ■ Special Monitoring and Resuscitation Techniques<br />

20% of 32-week-old infants, and as common as 60% in ELBW<br />

infants less than 28 weeks; postconceptional age. The ductus<br />

arteriosus is derived from the sixth aortic arch embryologically<br />

and connects the main pulmonary artery to the descending aorta.<br />

In fetal life, the high pulmonary vascular resistance (PVR) directs<br />

90% of right ventricular output through the ductus arteriosus to<br />

the aorta. Intrauterine ductus arteriosus patency is under control<br />

of chemical, neural, and hemodynamic blood flow stimuli. Locally<br />

produced prostaglandin E 2<br />

(PGE 2<br />

) maintains ductal patency with<br />

the production of PGE 2<br />

decreasing with increasing gestational age.<br />

Postnatal ductus arteriosus closure usually starts within<br />

72 hours of life in healthy term neonates. Ductal closure after birth<br />

is initiated by local and circulating neurohumoral factors as well<br />

as higher partial oxygen content, both of which initiate ductal<br />

smooth muscle constriction. This sensitivity to oxygen is maximal<br />

in term infants and within 24 hours of birth. Ductal closure takes<br />

longer to initiate in the ELBW infant and often is initiated medi -<br />

cally by the administration of prostaglandin inhibitors such<br />

as indomethocin. Delayed closure can be a result of respiratory<br />

prematurity with a high PVR or respiratory disease in the ELBW<br />

infant. Another cause of delayed closure of the ductus arteriosus<br />

is excessive intravenous fluid administration greater than 170 mL/<br />

kg/d, particularly on day 3 postnatally in ELBW infants. 93 Other<br />

factors include hypoxia, hypercarbia, and acidosis, which all<br />

increase PVR. These causes of a sustained high PVR owing to<br />

PPHN are important to avoid so that continued shunting at the<br />

PDA or patent foramen ovale level does not occur. A physiologic<br />

and anatomic cardiac right-to-left shunt then remains, with<br />

desaturated blood being shunted into the systemic circulation,<br />

causing severe systemic hypoxemia. Conversely, if the PDA<br />

remains open without PPHN, and the PVR continues to drop as<br />

it should do with increasing postconceptional age, the PDA will<br />

shunt cardiac blood in a left-to-right manner, and actually cause<br />

a systemic cardiac output “steal” phenomenon. Less blood will<br />

reach the systemic circulation (Qs) and more will circulate<br />

through the lungs (Qp). An increased Qp:Qs often leads to con -<br />

gested lungs, apnea, cardiac failure, and a low systemic blood<br />

pressure. Owing to the low-pressure pulmonary circulation acting<br />

as a “sink,” a low diastolic blood pressure often heralds a PDA in<br />

the ELBW infant. As a result, this low blood pressure and low sys -<br />

temic blood flow may be associated with diminished gastro -<br />

intestinal blood and renal blood flow. This pathophysiology often<br />

occurs at about the time of introduction of oral feeding in growing<br />

ELBW infants. This combination of increased requirements<br />

of gastrointestinal blood flow at a time of decreased supply is<br />

implicated in the increased incidence of NEC in the ELBW<br />

infant. 93 To avoid these complications, it is necessary to attempt<br />

to close the PDA with non-steroidal anti-inflammatory drugs<br />

(NSAIDs). A recent Cochrane review comparing indomethacin<br />

with ibuprofen found no statistical difference in ductal closure rate<br />

with either medication. 94 There are reports of greater pulmonary<br />

hypertension with ibuprofen; however, studies with outcome<br />

measurements to school-age children will be needed to make any<br />

conclusions regarding survival without impairment. 94 If the PDA<br />

remains open, despite medical intervention, and the cardio -<br />

vascular system remains compromised, it becomes mandatory to<br />

close the PDA surgically.<br />

Repair of PDA in the NICU<br />

These infants are commonly operated on in the NICU in order<br />

to avoid a transportation of a critically ill small infant to the oper -<br />

ating room, where the hazards of accidental tracheal extubation,<br />

hypothermia, and intravenous line disconnection can be pre -<br />

vented. If not already on a ventilator, the infant should be<br />

intubated orally with the appropriately sized ETT. This is usually<br />

a 2- to 2.5-mm internal diameter ETT secured at 5 to 6 cm at the<br />

mouth in the ELBW infant. Although the black line on the tube is<br />

used to ensure that the tip of the ETT is above the carina, it is<br />

recommended in ELBW infant to confirm the final position of<br />

the ETT by preoperative x-ray. Preoperative x-ray also confirms<br />

whether the aorta is left- or right-sided. If the infant is not<br />

intubated, this will often be completed by the NICU team before<br />

the surgical team arrives in the NICU. An x-ray will confirm<br />

correct ETT placement, and once the hemodynamics are stable,<br />

anesthesia and surgery will begin. Usually, surgical antibiotic<br />

prophylaxis is not administered in the NICU.<br />

A left thoracotomy is usually performed with a left-sided<br />

thoracic aorta. The infant is placed in the right lateral position, a<br />

small roll is placed under the right shoulder, the head and legs are<br />

padded for any pressure points, and an infant diathermy dispersive<br />

electrode pad is placed on the infant’s back. The lowest possible<br />

setting should always be set on the diathermy machine before<br />

surgery. The head of the patient should be directed toward the foot<br />

of the bed or open side of the NICU, and the ventilator is placed<br />

directly at the foot of the bed. A neonatal resuscitation ventilation<br />

or Ambu bag with an independent oxygen supply is close at hand<br />

but not under the drapes, and the oxygen should not be left<br />

running through the Ambu bag during surgery. The reason for<br />

this is to avoid an increase in oxygen under the surgical drapes.<br />

High concentrations of oxygen under surgical drapes may allow<br />

cautery sparks to be combustible, causing fires. For the same<br />

reason, an alcohol skin preparation should not be used. A warmed<br />

iodine skin preparation is preferred. It is imperative to ensure that<br />

a free flowing 24-gauge intravenous cannula has been sited before<br />

surgery. This line is attached to a length of extension tubing and a<br />

three-way stopcock. This allows administration of anesthetic<br />

drugs and fluids through the cannula insertion site, which will be<br />

under the surgical drapes during the procedure. It is very difficult<br />

to administer blood with a syringe through a PICC line. These<br />

lines are used in the NICU to administer inotropes and total<br />

parenteral nutrition, and it is safer to leave well enough alone and<br />

not contaminate by adding an anesthetic infusion line to this<br />

system. The second reason is that these lines can rupture when<br />

fluids are administered under pressure and only 10-mL syringes<br />

should be used. Cross-match one leukoreduced gamma-irradiated<br />

adult unit of packed red blood cells before surgery and have this<br />

in a cooler on ice in the unit before surgery. The advantage of this<br />

is that when the blood is not used (which is the majority of the<br />

time), it can be returned to the blood bank and re-issued for<br />

another patient without wastage. Ensure an intravenous blood<br />

administration set is also at the bedside because these are com -<br />

monly not present because blood ordered in the NICU is usually<br />

prefiltered by the blood bank. A blood pressure cuff and pulse<br />

oximeters should be placed on one of the arms and one of the legs.<br />

This is important to confirm correct surgical clip placement on<br />

the ductus during surgery. If end-tidal CO 2<br />

monitoring is present,<br />

it may be very useful during these surgical procedures. Sometimes,<br />

the anatomy of the aorta can be difficult and this ensures that the<br />

aorta is not inadvertently obstructed at the time of surgery.<br />

Lung ventilation of these infants during thoracotomy is chal -<br />

lenging. Usually, a slight increase of 2 to 3 mmHg in the inspired<br />

ventilatory pressure together with a 10 to 25% increase in the FI O2


CHAPTER 83 ■ Specific Problems and Anesthesia Management of Extremely Low Birthweight Infants 1401<br />

together with a 5% increase in respiratory rate is all that is required<br />

to ensure adequate oxygenation and ventilation during the<br />

thoracotomy. Pulse oximetry levels of 89 to 94% are aimed for<br />

during anesthesia. This is essential during thoracotomy and lung<br />

collapse with surgical swabs and retraction. Should a pulmonary<br />

hypertensive crisis occur during surgery, it should be treated by<br />

control of acid-base status and lowering PaCO 2<br />

by ventilation.<br />

If right ventricular failure occurs, it may be augmented with an<br />

epinephrine infusion of 0.03 µg/kg/min titrated to effect.<br />

Anesthetic drugs used are aimed at providing the correct<br />

amount to ablate the neuroendocrine stress response to surgery,<br />

but at the same time, excessive administration of narcotics, benzo -<br />

diazepines, and muscle relaxants must be avoided. Otherwise,<br />

depression of the infant’s autonomic nervous system will result in<br />

a greater requirement for inotrope administration. It is, therefore,<br />

a balance, and the medication doses recommended are fentanyl 2<br />

to 5 µg/kg, midazolam 0.1 mg/kg, and pancuronium 0.08 mg/kg.<br />

It is possible to use ketamine as the anesthetic agent, although this<br />

is not common practice in the ELBW infant. The administration<br />

of fluids at this time should not be overzealous. These infants are<br />

critically balanced fluidwise, and once the duct is clipped and the<br />

left ventricle is now expected to eject blood in the presence of a<br />

higher SVR, these infants can progress to cardiac failure requiring<br />

greater inotropic support. At the end of the surgery, once the<br />

surgical swabs have been removed from the chest, anesthesia is<br />

asked to re-expand the collapsed lung. Complications are usually<br />

infrequent with this surgical procedure. However, a recent report<br />

has shown that there may be an incidence as high as 67% unilateral<br />

vocal cord paralysis with respiratory and swallowing difficulties<br />

after ligation or ductal clipping of the PDA in ELBW patients. 95<br />

The ELBW infants in this series had significantly longer tube<br />

feeding (relative risk 8.25; 95% confidence interval 1.93–46.98<br />

[P = .003]; supplemental oxygen use [P = .004]; and ventilatory<br />

support [P = .001]) and had a longer hospital stay. 95<br />

Is there a role for prophylactic surgical closure of the PDA in<br />

ELBW infants? The current answer is based on a recent Cochrane<br />

database report indicating that surgical PDA closure prophylaxis<br />

in ELBW infants at this time is not warranted. 96 Only 84 patients<br />

have been randomized to any prospective study on this topic. In<br />

this study, prophylactic PDA surgical closure did not decrease<br />

mortality or BPD in ELBW infants. Postoperatively, a morphine or<br />

fentanyl infusion is commonly used to continue ablating the<br />

deleterious neuroendocrine stress response.<br />

Eye Surgery<br />

The retina usually does not fully mature before 42 to 44 weeks’<br />

gestation, and most elective surgery should ideally be delayed until<br />

after 44 weeks’ gestation if at all possible and pulse oximetry<br />

should be targeted at all times to maintain arterial saturations of<br />

89 to 95% in the ELBW infant. ROP occurs in infants usually<br />

weighing less than 1000 g and these infants are not usually<br />

screened until 31.5 weeks’ gestational age. There is still a high<br />

incidence of ROP in graduates of the NICU and the reason might<br />

be that the more aggressive resuscitation and care offered to the<br />

sicker, more premature infants might be increasing the incidence<br />

of ROP. 97 It is believed to be caused by multiple etiologies.<br />

Hypoxia, hyperoxia, hypercarbia, sepsis, and multiple blood<br />

transfusions have all been implicated. Interestingly, however, ROP<br />

has also occurred in infants who have received no supplemental<br />

oxygen. Laser surgery is used to ablate abnormal vessels before<br />

retinal detachment and progression to blindness. This surgery is<br />

traditionally done in infants with their trachea intubated and lungs<br />

mechanically ventilated under general anesthesia. 98 One problem<br />

with providing anesthesia for this procedure is that many infants<br />

who are weaned and extubated in the NICU must then return to<br />

the NICU after surgery still intubated, requiring mechanical<br />

ventilation and reweaning over a number of days. Overcoming<br />

this problem is important, and a recent alternative anesthetic<br />

approach deserves consideration. In a series of 54 infants for laser<br />

ablation for ROP, 73% of infants had been successfully weaned<br />

from mechanical ventilation before surgery. 99 If intubation and<br />

mechanical ventilation were instituted for the surgery alone<br />

(group 1), 77% of the infants remained intubated and mechani -<br />

cally ventilated on postoperative day 1. Fifty percent of infants<br />

intubated for surgery were still intubated and ventilated on<br />

postoperative day 3. This adds to the cost as well as to the risks of<br />

the respiratory support in the NICU. An important and novel<br />

nasopharyngeal prong anesthetic administration technique aimed<br />

at avoiding intubation for laser surgery in (group 2, N = 24 infants)<br />

found significantly(P < .001) that only 1 infant of those not intu -<br />

bated for surgery required a period of postoperative mechanical<br />

ventilation. 99 Management with nasopharyngeal prongs did<br />

not result in higher perioperative PaCO 2<br />

or lower pH values. This<br />

study is noteworthy because avoiding postoperative intubation<br />

would be a great benefit to the ELBW infant. A larger series needs<br />

to be performed to confirm the advantage of this novel anesthetic<br />

technique. Many NICUs have a procedure room, and with gentle<br />

sedation, these patients are screened by ophthalmology in the unit<br />

and laser can then be done after appropriate “laser in use” signs<br />

and precautions are instituted. These patients may need to be<br />

followed up by ophthalmology even after discharge from the<br />

NICU to ensure their eyesight is preserved.<br />

Gastrointestinal Procedures<br />

The classic paper on staging and management of NEC published<br />

by Bell and coworkers in 1978 has guided physicians caring<br />

for small infants with acquired intestinal disease since then. 100<br />

The current era of lung maturation steroid administration ante -<br />

natally to mothers and newborn ELBW infants, together with<br />

concomitant surfactant and NSAIDs administration soon<br />

after birth, has seen the emergence of newly acquired neonatal<br />

intestinal disease syndromes called sudden intestinal perforation<br />

(SIP) 101,102 and feeding intolerance of prematurity (FIP). 102 SIP is a<br />

new distinct entity and no longer fits into the classic definitions<br />

previously described. 100 Feeding intolerance has arisen because of<br />

the need to feed smaller and smaller infants orally at a time when<br />

they should still be in utero. 102 The etiology of gastrointestinal tract<br />

disruption and NEC in ELBW infants is still poorly understood<br />

and multifactorial. Certainly, neonatal asphyxia, oral feeding,<br />

overzealous intravenous fluid administration resulting in a PDA,<br />

and diminished blood flow to the gastrointestinal tract can cause<br />

bowel ischemia and perforation. Mucosal integrity break-down<br />

and gastrointestinal bacterial colonization play a role. Unfortu -<br />

nately, because an increasing number of ELBW infants currently<br />

survive and must be admitted to a NICU, the in-hospital mortality<br />

rate postsurgical intervention for NEC or SIP in this age group is<br />

still approximately 50%. 103 Gastrointestinal disruption in ELBW<br />

infants tends to follow one of three clinical pathways: SIP with<br />

sudden free air, metabolic derangement (MD) complicated by


1402 PART 4 ■ Special Monitoring and Resuscitation Techniques<br />

the appearance of free air, or progressive MD without evidence of<br />

free air. 104 MD is more like classic NEC and includes thrombo -<br />

cytopenia, metabolic acidosis, neutropenia, left shift of segmented<br />

neutrophils, hyponatremia, bacteremia, and hypotension. Surgical<br />

management remains controversial. Absence of MD warrants<br />

consideration for peritoneal drainage rather than a laparotomy,<br />

especially for the appearance of sudden free air. 104 Surgery should<br />

be planned based on degree of MD and organ dysfunction rather<br />

than the age of the patient. 105 When performing these procedures<br />

in the NICU, the same presurgery preparation and added<br />

precautions need to be taken as if preparing an operating room.<br />

Airway equipment, monitoring, suction, and a modern intensive<br />

care ventilator should be appropriately set for an ELBW infant<br />

before beginning anesthesia. The operating room or intensive care<br />

operating area should be warm. Often, 20 to 80 mL/kg of warmed<br />

colloid, blood, or fresh frozen plasma fluids may need to be<br />

administered in severe NEC depending on the extent of third<br />

space losses and blood loss with surgery. An intravenous catheter<br />

for this fluid administration should be present independent of<br />

the intravenous maintenance fluids being administered by the<br />

NICU. An infant who is diagnosed with NEC will often be on an<br />

antibiotic regimen that includes coverage for coagulase-positive,<br />

coagulase-negative, and anaerobic organisms, and this should be<br />

continued during the procedure.<br />

Prospective, randomized trials analyzing therapy for more<br />

major surgery in the ELBW infant being practiced at the bedside<br />

in the NICU soon after delivery needs to be done; although this is<br />

widely practiced (e.g., PDA repair, NEC surgery, gastroschesis<br />

reduction, congenital diaphragmatic hernia), no data currently<br />

exist to refute or prove the benefit of NICU bedside surgery. 106<br />

What is clearly emerging is that enterally administered probiotics<br />

are beneficial in the prevention of NEC in infants only if greater<br />

than 1000 g. This may become part of standard practice in neo -<br />

natal intensive care therapy; however, in ELBW infants weighing<br />

less than 1000 g, further research is needed. 107<br />

Neurosurgical Procedures<br />

Currently 60,000 infants in the United States are born with<br />

a birthweight less than 1500 g. 108 At least 50% of these NICU<br />

graduates will have some magnetic resonance imaging (MRI)<br />

evidence of injury to the cerebral white matter. 108–110 Common to<br />

most of this complex white matter injury is termed encephalopathy<br />

of prematurity. This is defined by the combination of predo -<br />

minantly noncystic periventricular leukomalacia and neuronal<br />

deficits. 110 Neuronal injury leads later in life to degrees of cerebral<br />

palsy, seizures, and hydrochepalus. These neurologic quality-oflife<br />

outcomes are currently the most challenging clinical problems<br />

to solve in neonatology. There are multiple etiologies why pre -<br />

mature and ELBW infants present for neurosurgery. They include<br />

posthemorrhagic hydrocephalus (PPH) requiring a ventricular<br />

shunt after IVH. These bleeds may be caused by birth delivery<br />

trauma, asphyxia and resuscitation at birth, NSAID medications to<br />

close a PDA, hypoxia and the need for ventilatory support while<br />

the lungs mature, periods of hypotension and decreased CBF, or<br />

hypertension and increased CBF. Periventricular leukomalacia<br />

is the predominant injury, and it may have multiple etiologies<br />

including those just listed for brain damage. All of these causes of<br />

brain damage may lead to cerebral palsy. Recent evidence in a<br />

rodent model indicates that high oxygen exposure in maturationdependent<br />

brain tissue causes accelerated apoptosis. 111 Clinical<br />

studies need to be performed to determine whether this may<br />

be an additional cause of periventricular leukomalacia in the<br />

ELBW infant.<br />

Anesthesia for neurosurgical procedure in ELBW infants<br />

mostly involves external ventricular drains and Omaya reservoir<br />

placement. These procedures are often staged to prevent infected<br />

central nervous system shunts in at least 11% in one series 112 and<br />

result in multiple neurosurgical procedures in ELBW infants.<br />

Positioning of the large hydrocephalic occiput in relation to the<br />

body on the operating table is often eased by placing layers under<br />

the infant’s body to create a higher horizontal plane with the neck<br />

in the neutral position. This facilitates the view of the larynx at<br />

the time of intubation. Infants must be kept warm. Hyperventi -<br />

lation should be avoided because a drop in PaCO 2<br />

may adversely<br />

affect CBF and normocapnea should be the goal. After surgery,<br />

extubation of the trachea is often not performed in the operating<br />

room in the ELBW infant for fear of apnea and hypoventilation<br />

during transport back to the NICU.<br />

REGIONAL ANESTHESIA<br />

CONSIDERATIONS<br />

There are no randomized studies on regional anesthesia in ELBW<br />

infants, but a recent report of a successful caudal in a 740-g infant<br />

indicates that further study is warranted. 113 A 22-gauge epidural<br />

catheter was inserted caudally to a midthoracic level in a 740-g<br />

ELBW infant undergoing laparotomy for intestinal perforation.<br />

Analgesia was continued epidurally with ropivicaine 2 mg/mL at<br />

0.2 mg/h for 48 hours postoperatively. Bowel sounds returned after<br />

3 hours and stool was passed on the same postoperative day.<br />

Extubation was achieved on postoperative day 1. The epidural<br />

catheter was removed after the platelet count was greater than<br />

80 × 10 9 /L. 113 The pharmacokinetics of local anesthetics in ELBW<br />

infants have not been fully elucidated. 114 One might expect that<br />

the clearance of ropivicaine is less in the premature infant than<br />

in the neonate at term. 114 It is for this reason that regional catheter<br />

use in ELBW infants for longer than 48 hours is currently not<br />

recommended. 113 It is imperative that the same vigilance and<br />

precautions one would take while providing regional anesthesia<br />

to a larger child should equally be considered when administering<br />

regional anesthesia to the ELBW infant.<br />

CHALLENGES OF THE ELBW<br />

INFANT LATER IN LIFE<br />

Nearly 50% of ELBW infants discharged home are readmitted<br />

to hospital mostly for respiratory problems, and this risk persists<br />

up until school age. 115 The risks of BPD leading to chronic lung<br />

disease are a major consideration of the anesthesiologist taking<br />

care of the ELBW infant graduate. There is a greater incidence<br />

of higher systolic blood pressure in adolescents and adults who<br />

were ELBW infants and is recorded as a 3- to 8-mmHg increase. 116<br />

The importance of this hypertension is that a 2-mmHg reduction<br />

in the diastolic blood pressure would result in a 17% decrease in<br />

the prevalence of hypertension, a 6% reduction in the risk of CHD,<br />

and a 15% reduction in the risk of stroke or transient ischemic<br />

attack later in life. 116 Many of these ELBW children will require<br />

on-going surgeries and medical care well into adulthood.


CHAPTER 83 ■ Specific Problems and Anesthesia Management of Extremely Low Birthweight Infants 1403<br />

CONCLUSION<br />

The anesthetic care of the ELBW infant is both challenging and<br />

rewarding. This chapter has highlighted the specific transitional<br />

physiology and pathophysiology commonly seen in the ELBW<br />

infant. Keeping these children warm with minimal handling and<br />

ablating the neuroendocrine stress response with adequate<br />

analgesia during anesthesia are essential. Careful airway control<br />

and a thoughtful lung protective ventilation strategy will be re -<br />

quired when caring for the ELBW infant. In addition, maintaining<br />

a stable cardiovascular system and delivering appropriate intra -<br />

venous fluids and red blood cells will ensure continued excellent<br />

anesthetic outcomes in these patients.<br />

REFERENCES<br />

1. Landmann E, Misselwitz B, Steiss JO, et al. Mortality and morbidity of<br />

neonates born at


1404 PART 4 ■ Special Monitoring and Resuscitation Techniques<br />

44. Wilson CG, Martin RJ, Jaber M, et al. Adenosine A 2A receptors interact<br />

with GABAergic pathways to modulate respiration in neonatal piglets.<br />

Respir Physiol Neurobiol. 2004;141:201–211.<br />

45. Leon AE, Michienzi K, Ma CX, et al. Serum caffeine concentrations in<br />

preterm neonates. Am J Perinatol. 2007;1:39–47.<br />

46. Swingle HM, Eggert LD, Bucciarelli RL. New approach to management of<br />

unilateral tension pulmonary interstitial emphysema in premature<br />

infants. Pediatrics. 1984;74:354–357.<br />

47. Meyer MT, Rice TB, Glaspey JC. Selective fiberoptic left mainstem<br />

intubation for severe unilateral barotrauma in a 24-week premature<br />

infant. Pediatr Pulmonol. 2002;33:227–231.<br />

48. Chalak LF, Kaiser JR, Arrington RW. Resolution of pulmonary interstitial<br />

emphysema following selective left main stem intubation in a<br />

premature newborn: an old procedure revisited. Paediatr Anaesth. 2007;<br />

17:183–186.<br />

49. Versmold HT, Kitterman JA, Phibbs RH, et al. Aortic blood pressure<br />

during the first 12 hours of life in infants with birth weight 610 to 4,220<br />

grams. Pediatrics. 19<strong>81</strong>;67:607–613.<br />

50. Levene M, Chiswick M, Field D, et al. Joint Working Group of the British<br />

Association of Perinatal Medicine and the Research Unit of the Royal<br />

College of Physicians. Development of audit measures and guidelines for<br />

good practice in the management of neonatal respiratory distress<br />

syndrome. Arch Dis Child. 1992;67:1221–1227.<br />

51. Evans JR, Lou Short B, Van Meurs K, et al. Cardiovascular support in<br />

preterm infants. Clin Ther. 2006;28:1366–1384.<br />

52. Dempsey EM, Barrington KJ. Treating hypotension in the preterm infant;<br />

when and with what: a critical and systematic review. J Perinatol.<br />

2007;27:469–478.<br />

53. Dempsey EM, Barrington KJ. Diagnostic criteria and therapeutic<br />

interventions for the hypotensive very low birth weight infant. J Perinatol.<br />

2006;26:677–6<strong>81</strong>.<br />

54. Aladangady N, Aitchison TC, Beckett C, et al. Is it possible to predict the<br />

blood volume of a sick preterm infant? Arch Dis Child Fetal Neonatal Ed.<br />

2004;89:344–347.<br />

55. Lee J, Rajadurai VS, Tan KW. Blood pressure standards for very low<br />

birthweight infants during the first day of life. Arch Dis Child Fetal<br />

Neonatal Ed. 1999;<strong>81</strong>:168–170.<br />

56. Munro MJ, Walker AM, Barfield CP. Hypotensive extremely low birth<br />

weight infants have reduced cerebral blood flow. Pediatrics. 2004;114:<br />

1591–1596.<br />

57. Miall-Allen VM, de Vries LS, Whitelaw AG. Mean arterial blood pressure<br />

and neonatal cerebral lesions. Arch Dis Child. 1987;62:1068–1069.<br />

58. Lightburn MH, Gauss CH, Williams DK, et al. Cerebral blood flow<br />

velocities in extremely low birth weight infants with hypotension and<br />

infants with normal blood pressure. J Pediatr. 2009;6:824–828.<br />

59. Sarkar S, Dechert R, Schumacher RE, et al. Is refractory hypotension in<br />

preterm infants a manifestation of early ductal shunting? J Perinatol.<br />

2007:27:353–358.<br />

60. Bell E, Acarregui M. Restricted versus liberal water intake for preventing<br />

morbidity and mortality in preterm infants. Cochrane Database Syst Rev.<br />

2008;1:CD000503.<br />

61. Holliday M, Segar W. Reducing errors in fluid therapy management.<br />

Pediatrics. 2003;111:424–425.<br />

62. Paut O, Lacroix F. Recent developments in the perioperative fluid<br />

management for the paediatric patient. Curr Opin Anaesthesiol. 2006;19:<br />

268–277.<br />

63. Holliday M, Segar W. The maintenance need for water in parenteral fluid<br />

therapy. Pediatrics. 1957;19:823–832.<br />

64. O’Hare FM, Molloy EJ. What is the best treatment for hyperkalaemia in<br />

a preterm infant? Arch Dis Child. 2008;2:174–176.<br />

65. Stefano JL, Norman ME. Nitrogen balance in extremely low birth weight<br />

infants with nonoliguric hyperkalemia. J Pediatr. 1993;123:632–635.<br />

66. Lorenz JM, Kleinman LI, Markarian K. Potassium metabolism in<br />

extremely low birth weight infants in the first week of life. J Pediatr. 1997;<br />

131:<strong>81</strong>–86.<br />

67. Ziegler E, O’Donnell A, Nelson S. Body composition of the reference<br />

fetus. Growth. 1976;40:329.<br />

68. Hay W. Nutritional requirements of extremely low birthweight infants.<br />

Acta Paediatr Suppl. 1994;402:94.<br />

69. Coulthard MG, Hey EN. Renal processing of glucose in well and sick<br />

neonates. Arch Dis Child Fetal Neonatal Ed, 1999;2:F92–F98.<br />

70. Kairamkonda VR, Khashu M. Controversies in the management of<br />

hyperglycemia in the ELBW infant. Indian Pediatr. 2008;1:29–38.<br />

71. Siker D. Pediatric fluids, electrolytes, and nutrition. In: Gregory GA,<br />

editor. Pediatric Anesthesia. 3rd ed. New York: Churchill Livingstone;<br />

1994. pp. 83–117.<br />

72. Ayers J, Graves SA. Perioperative management of total parenteral<br />

nutrition, glucose containing solutions, and intraoperative glucose<br />

monitoring in paediatric patients: a survey of clinical practice. Paediatr<br />

Anaesth. 2001;11:41–44.<br />

73. Fitzgerald M, Millard C, MacIntosh N. Hyperalgesia in premature infants.<br />

Lancet. 1988;6:8580.<br />

74. Bartocci M, Bergqvist LL, Lagercrantz H, et al. Pain activates cortical areas<br />

in the preterm newborn brain. Pain. 2006;122:109–117.<br />

75. Anand KJS. Pharmacological approaches to the management of pain in<br />

the neonatal intensive care unit. J Perinatol. 2007;27:S4–S11.<br />

76. Laudenbach V, Calo G, Guerrini R, et al. Nociceptin/orphanin FQ<br />

exacerbates excitotoxic white-matter lesions in the murine neonatal brain.<br />

J Clin Invest. 2001;4:457–466.<br />

77. Setzer N. Anesthesia, premature infants, and hypotension. Pediatrics.<br />

1994;5:870.<br />

78. van Alfen-van der Velden AA, Hopman JC, Klaessens JH, et al. Effects of<br />

midazolam and morphine on cerebral oxygenation and hemodynamics in<br />

ventilated premature infants. Biol Neonat. 2006;90:197–202.<br />

79. Nikizad H, Yon JH, Carter LB, et al. Early Exposure to general anesthesia<br />

causes significant neuronal deletion in the developing rat brain. Ann N Y<br />

Acad Sci. 2007;1122:69–82.<br />

80. Johnson SA, Young C, Olney JW. Isoflurane-induced neuroapoptosis in<br />

the developing brain of nonhypoglycemic mice. Neurosurg Anesthesiol.<br />

2008;20:21–28.<br />

<strong>81</strong>. Jevtovic-Todorovic V, Beals J, Benshoff N, et al. Prolonged exposure to<br />

inhalational anesthetic nitrous oxide kills neurons in adult rat brain.<br />

Neuroscience. 2003;122:609–616.<br />

82. Wise-Faberowski L, Zhang H, Ing R, et al. Isoflurane-induced neuronal<br />

degeneration: an evaluation in organotypic hippocampal slice cultures.<br />

Anesth Analg. 2005;101:651–657.<br />

83. Zou X, Patterson TA, Divine RL, et al. Prolonged exposure to ketamine<br />

increases neurodegeneration in the developing monkey brain. Int J Dev<br />

Neurosci. 2009;277:727–731.<br />

84. Culley DJ, Xie Z, Crosby G. General anesthetic-induced neurotoxicity:<br />

an emerging problem for the young and old? Curr Opin Anaesthesiol.<br />

2007;5:408–413.<br />

85. Anand KJ, Sippell WG, Aynsley-Green A. Randomised trial of fentanyl<br />

anaesthesia in preterm babies undergoing surgery: effects on the stress<br />

response. Lancet. 1987;10:62–66.<br />

86. Anand KJ, Hickey PR. Halothane-morphine compared with high-dose<br />

sufentanil for anesthesia and postoperative analgesia in neonatal cardiac<br />

surgery. N Engl J Med. 1992;1:1–9.<br />

87. Soriano SG, Anand KJ. Anesthetics and brain toxicity. Curr Opin<br />

Anaesthesiol. 2005;18:293–297.<br />

88. Malviya S, Lerman J. The blood/gas solubilities of sevoflurane, isoflurane,<br />

halothane, and serum constituent concentrations in neonates and adults.<br />

Anesthesiology. 1990;72:793–796.<br />

89. Sale SM, Read JA, Stoddart PA, et al. Prospective comparison of<br />

sevoflurane and desflurane in formerly premature infants undergoing<br />

inguinal herniotomy. Br J Anaesth. 2006;96:774–778.<br />

90. von Ungern-Sternberg BS, Saudan S, Petak F, et al. Desflurane but not<br />

sevoflurane impairs airway and respiratory tissue mechanics in children<br />

with susceptible airways. Anesthesiology. 2008;2:216–224.<br />

91. Hassid S, Nicaise C, Michel F, et al. Randomized controlled trial of sevo -<br />

flurane for intubation in neonates. Paediatr Anaesth. 2007;17:1053–1058.<br />

92. Kumar P, Walker JK, Hurt KM, et al. Medication use in the neonatal<br />

intensive care unit: current patterns and off-label use of parenteral<br />

medications. J Pediatr. 2008;3:412–415.<br />

93. Stephens BE, Gargus RA, Walden RV, et al. Fluid regimens in the first<br />

week of life may increase risk of patent ductus arteriosus in extremely low<br />

birth weight infants. J Perinatol. 2008;28:123–128.<br />

94. Ohlsson A, Walia R, Shah S. Ibuprofen for the treatment of patent ductus<br />

arteriosus in preterm and/or low birth weight infants. Cochrane Database<br />

Syst Rev. 2008;1:CD0034<strong>81</strong>.<br />

95. Clement WA, El-Hakim H, Phillipos EZ, et al. Unilateral vocal cord<br />

paralysis following patent ductus arteriosus ligation in extremely lowbirth-weight<br />

infants. Arch Otolaryngol Head Neck Surg. 2008;134:28–33.<br />

96. Mosalli R, Alfaleh K. Prophylactic surgical ligation of patent ductus<br />

arteriosus for prevention of mortality and morbidity in extremely low<br />

birth weight infants. Cochrane Database Syst Rev. 2008;1:CD0061<strong>81</strong>.


CHAPTER 83 ■ Specific Problems and Anesthesia Management of Extremely Low Birthweight Infants 1405<br />

97. Vyas J, Field D, Draper ES, et al. Severe retinopathy of prematurity and<br />

its association with different rates of survival in infants of less than 1251<br />

g birthweight. Arch Dis Child Fetal Neonatal Ed. 2000;82:145–149.<br />

98. Sullivan TJ, Clarke MP, Tuli R, et al. General anesthesia with endotra -<br />

cheal intubation for cryotherapy for retinopathy of prematurity. Eur J<br />

Ophthalmol. 1995;5:187–191.<br />

99. Woodhead DD, Lamber DK, Molloy DA, et al. Avoiding endotracheal<br />

intubation of neonates undergoing laser surgery for retinopathy of<br />

prematurity. J Perinatol. 2007;27:209–213.<br />

100. Bell MJ, Ternberg JL, Feigin RD, et al. Neonatal necrotizing enterocolitis;<br />

therapeutic decisions based upon clinical staging. Ann Surg. 1978;187:1–7.<br />

101. Aschner JL, Deluga KS, Metlay LA, et al. Spontaneous focal gastro intes -<br />

tinal perforation in very low birth weight infants. J Pediatr. 1988;113:<br />

364–367.<br />

102. Gordon PV, Swanson JR, Attridge JT, et al. Emerging trends in acquired<br />

neonatal intestinal disease: is it time to abandon Bell’s criteria?<br />

J Perinatol. 2007;27:661–671.<br />

103. Blakely ML, Lally KP, McDonald S, et al. NEC Subcommittee of the<br />

NICHD Neonatal Research Network. Postoperative outcomes of<br />

extremely low birth-weight infants with necrotizing enterocolitis or<br />

isolated intestinal perforation; a prospective cohort study by NICHD<br />

Neonatal Research Network. Ann Surg. 2005;241:984–989.<br />

104. Tepas JJ 3rd, Sharma R, Hudak ML, et al. Coming full circle: an evidencebased<br />

definition of the timing and type of surgical management of very<br />

low-birth-weight


84<br />

CHAPTER<br />

Miscellaneous Techniques<br />

Pedro Paulo Vanzillotta<br />

INTRODUCTION<br />

The field of anesthesia is expanding considerably as anesthe -<br />

siologists become more involved in specialized monitoring, in<br />

invasive and therapeutic procedures that, at first sight, might not<br />

appear directly related to anesthesia. Some acute illnesses and<br />

particularly trauma (e.g., fractures, traumatic and septic arthritis,<br />

different kinds of wounds) may present in emergency pediatric<br />

patients, requiring medical and surgical care. Pediatric anesthe -<br />

siologists, therefore, require basic knowledge about traumatic<br />

and postoperative compartment syndromes, principles of wound<br />

management, arthrocentesis, and immobilization techniques.<br />

Continuous infusion techniques are also frequently indicated<br />

when managing patients on parenteral drug and fluid therapy,<br />

during intravenous anesthesia, and for postoperative pain control.<br />

Intraosseous Infusion<br />

With the renewed interest in the use of intraosseous infusions,<br />

cases of iatrogenic compartment syndromes after fluid adminis -<br />

tration through this route are being reported. 1 Symptoms may<br />

develop within 20 minutes of the infusion. The outcomes vary<br />

from no residual deficits to through-the-knee amputations. 6<br />

The compartment presumably becomes congested secondary to<br />

COMPARTMENT SYNDROMES AND<br />

MUSCLE PRESSURE MEASUREMENT<br />

Definition and Anatomic Considerations<br />

Compartment syndromes occur when the tissues pressure within<br />

an osteofascial compartment compromises blood flow to that<br />

compartment resulting in nerve and muscle damage. 1–3 The syn -<br />

drome may be seen in the forearm and all parts of the leg, the<br />

lower leg being affected more commonly than the buttocks, thigh,<br />

or foot. The forearm is divided into dorsal and superficial volar<br />

and deep volar compartments. The lower leg is divided into four<br />

compartments: superficial posterior, deep posterior, anterior,<br />

and anterolateral (Figure 84–1). The thigh is divided into three<br />

compartments and the foot has four. The back muscles and but -<br />

tocks also have compartments. 2<br />

Etiologies<br />

Trauma<br />

Compartment syndromes are frequently associated with crush<br />

injuries, fractures, and blunt trauma. They can also occur with<br />

penetrating arterial trauma and after reperfusion of an acutely<br />

ischemic limb. Tissue fluid pressure elevation exceeding 30 mmHg,<br />

after trauma, ischemia, and reperfusion injury, leads to lymphatic<br />

vessel collapse, impairing drainage of the edematous muscle. 3,4<br />

Ultimately, arteriolar spasm occurs, decreasing oxygen delivery<br />

to the tissue. The adverse effects of the ischemia are time- and<br />

pressure-related. Elevated pressures might be tolerated for short<br />

periods, but when abnormal pressures persist, neuronal and then<br />

muscle damage occurs. 5<br />

Figure 84-1. Upper view: Mid lower leg sagittal plane. Lower<br />

view: Fascial compartments of the lower leg: (1) anterior;<br />

(2) lateral; (3) superficial posterior; and (4) deep posterior<br />

compartments.


CHAPTER 84 ■ Miscellaneous Techniques 1407<br />

Figure 84-2. A simple method of<br />

measuring compartment pressure.<br />

extravasation of fluid at a puncture site or through the foramina of<br />

the nutrient vessel. Extravasation may be caused by previous<br />

infusions, incomplete penetration of a needle through the cortical<br />

bone, or extension of the cannula through the bone into the<br />

posterior compartment. Risk of compartment syndrome increases<br />

with pressure infusion, infusion of irritating drugs, use of inappro -<br />

priate needles, and extended duration of intraosseous infusion. 7–9<br />

Other Iatrogenic Causes<br />

Compartment syndromes may also develop after surgical pro -<br />

cedures in the limbs (e.g., plastic and orthopedic procedures). 10<br />

In the postoperative period, early clinical diagnosis may be dif -<br />

ficult because of residual regional anesthesia/analgesia 11 and the<br />

immobilization devices usually applied. Other described iatro -<br />

genic causes of particular interest to anesthetists include arterial<br />

cannula extravasation, 12 hyperosmolar Bier block, 13 intra-arterial<br />

barbiturates, 14 and extravasation to pressurized intravenous<br />

infused fluids. 12,15–17 Prompt recognition of an iatrogenic compart -<br />

ment syndrome is essential for preservation of limb function. 1<br />

Clinical Diagnosis<br />

Although the diagnosis may initially be subtle, failure to recognize<br />

and aggressively treat this entity can be devastating. The clinical<br />

findings in patients with incipient compartment syndrome are<br />

essentially similar, regardless of etiology. The four Ps—pain, pallor,<br />

paralysis, and pulselessness—are considered diagnostic, but all<br />

may be absent in early or late compartment syndrome. A high<br />

index of suspicion and repetitive examination are necessary for<br />

early diagnosis. Patients often present with pain that seems to<br />

be out of proportion to the injury they have suffered. A careful<br />

examination will help in localizing the compartment involved.<br />

Pain increases on passive stretching of the involved muscles. 1–3<br />

Measurement of Muscle<br />

Compartment Pressure<br />

Measurement of compartment pressure is the most reliable<br />

method to diagnose compartment syndrome because, in all cases,<br />

there is an increase in compartment pressure. Compartment<br />

pressures can facilitate the diagnosis of compartment syndrome<br />

in patients with peripheral nerve injuries, altered levels of con -<br />

sciousness, residual effects of anesthesia/analgesia, or equivocal<br />

clinical findings. The most appropriate time for fasciotomy can be<br />

identified by repeatedly monitoring the changes in compartment<br />

pressure. 3 Several available techniques for measuring compart -<br />

ment pressures have been described. 17–19 One of the simplest and<br />

easiest methods is to use an 18-gauge needle attached to a mercury<br />

manometer. More accurate results are obtained if a saline infusion<br />

is used to prevent the needle from becoming blocked 20 (Figure<br />

84–2). Solid-state, commercial devices are now readily available,<br />

such as the solid-state transducer intracompartmental catheter<br />

(STIC, Stryker Surgical, Kalamazoo, Mich) or the pressure sense<br />

intracompartmental pressure monitor (Horizon Medical, Inc,<br />

Santa Ana, Calif). The latter is the most compact of these devices. 3<br />

It is important to remember that only the pressure in the com -<br />

partment in which the needle is placed is being measured. Normal<br />

pressure in one compartment does not mean that there is normal<br />

pressure in all the compartments of the limb. 2 In humans, com -<br />

partment syndrome and muscle necrosis do not occur at pressures<br />

below 45 mmHg but always occur at pressures above 60 mmHg. 5<br />

The exact pressure at which tissue damage occurs varies in patients<br />

and is still controversial. 19–21 There is no precise pressure at which<br />

fasciotomy is invariably indicated, but when one is concerned<br />

about an impending compartment syndrome, it is best to err on<br />

the side of early fasciotomy. 3,5 The pressure in each compartment<br />

should be checked after decompression. 22 Full recovery is possible<br />

if treatment is initiated within 4 hours. Clinical experience and<br />

judgment are necessary in selecting the appropriate time for<br />

surgical intervention. Lack of recognition and delay in instituting<br />

treatment will lead to an increased incidence of amputation or loss<br />

of limb function.<br />

PRINCIPLES OF<br />

WOUND MANAGEMENT<br />

One of the most common procedures in the practice of emergency<br />

medicine is acute traumatic wound management. Pediatric<br />

patients frequently present for these procedures. The primary<br />

objectives in wound care are preserving viable tissue, restoring<br />

tissue continuity and function, optimizing conditions for the<br />

development of wound strength, preventing excessive or pro -<br />

longed inflammation, avoiding infection and other delays to<br />

healing, and minimizing scar formation. 23


1408 PART 4 ■ Special Monitoring and Resuscitation Techniques<br />

Normal Physiology of Wound Healing<br />

A basic understanding of the process of wound healing in essential<br />

to achieve these objectives. Wound healing is a nonspecific repair<br />

process that involves five stages. 24–26<br />

1. Inflammation: Inflammation is a beneficial response that<br />

serves to remove bacteria, foreign debris, and devitalized tissue.<br />

Significant contamination with bacteria of foreign material<br />

may prolong the inflammatory process, interfering with epi -<br />

thelialization and fibroplasia and, consequently, impairing<br />

wound healing.<br />

2. Epithelialization: An epithelial covering, developed by epi -<br />

thelial cells migration, makes the surface of sutured wounds<br />

impermeable to water within 24 to 48 hours. This process is<br />

impaired by surface debris and eschar.<br />

3. Fibroplasia refers to collagen and protein polysaccharides<br />

synthesis by transformed fibroblasts that initiates this stage of<br />

scar formation. It is influenced by physical forces acting across<br />

the wound, mainly the stress imposed by movement.<br />

4. Contraction: The movement of skin edges toward the center<br />

of the wound, primarily in the direction of underlying muscle,<br />

is more important during wound healing by secondary<br />

intention.<br />

5. Scar maturation: tensile strength improvement begins after the<br />

fifth day following injury. It increases rapidly for 6 to 17 days,<br />

more slowly in the next 10 to 14 days, and lasts almost imper -<br />

ceptibly for as long as 2 years.<br />

Although the whole process can be divided into these stages,<br />

they are not independent. Different events during each stage<br />

are interrelated and influence the final result. Phagocytosis and<br />

leukocyte death, occurring during the inflammatory process, are<br />

responsible for the purulent exudates that may accumulate in<br />

the wound, impairing epithelial cell migration and fibroblastic<br />

synthesis of new collagen (fibroplasia) as opposed to the lysis of<br />

old collagen (fibrinolysis). When deciding the best time for suture<br />

removal, a persistent or prolonged inflammatory response and<br />

the physical forces exerted on the wound edges should be con -<br />

sidered. The cosmetic and functional results depend on this<br />

reparative process and the medical conditions provided during<br />

wound management.<br />

Initial Evaluation of the Wound<br />

Surgical incisions (e.g., those for thoracic drainage, vascular<br />

cutdown, tunneled catheters, subcutaneous injection device place -<br />

ment) are the most suitable for management and have the best<br />

healing scar results. These therapeutic procedures are carried<br />

out under ideal conditions such as aseptic skin surfaces, lack of<br />

particulate matter and tissue debris, incisions parallel to flexion<br />

lines, and minimal tension sutures. Accidental wounds should be<br />

carefully evaluated and need previous preparation. The “Clinical<br />

Policy for the Initial Approach to Patients Presenting with Pene -<br />

trating Extremity Trauma” of the American College of Emergency<br />

Physicians provides a useful approach to the evaluation of all the<br />

wounds. 27 Extrinsic and intrinsic factors that impair healing and<br />

promote infection should be identified before treatment. Higher<br />

infection rates are reported for patients at extremes of age (e.g.,<br />

infants ≤ 6 mo) and with medical illness. 28 Other factors include<br />

current medication, allergies, tetanus immunization status, poten -<br />

tial exposure to rabies, presence of foreign bodies embedded<br />

in the wound, associated injuries (fracture joint penetration),<br />

availability for follow-up, and parents’ understanding of wound<br />

care. 29 Nonaccompanied pediatric patients impose an additional<br />

difficulty in identifying these historical factors. The same problem<br />

should be expected when trying to examine a wounded and awake<br />

infant or child. These situations may contribute to underestimat -<br />

ing the amount of tissue destruction, the degree of contamination,<br />

the damage to underlying structures, and the failure to detect the<br />

presence of foreign bodies.<br />

Cleaning Procedures<br />

All wounds must be cleaned immediately after evaluation.<br />

Removing bacteria, particulate matter, and tissue debris will reduce<br />

infection rates and shorten the inflammatory response. 30 All<br />

procedures should be explained to parents and patients, in an<br />

appropriate manner, to alleviate fears, stimulate cooperation, and<br />

provide assurance. Despite that, pediatric patients may need some<br />

intravenous or inhalation sedation associated with local or regional<br />

anesthesia. 23 The patient’s level of understanding and cooperation<br />

will determine which anesthetic technique should be used to<br />

provide the best conditions for evaluating, cleaning, scrubbing,<br />

irrigating, and suturing wounds. The physician must keep in mind<br />

that a good surgical result has to be emotionally atraumatic as well.<br />

The goals of wound cleaning may be accomplished by extensive<br />

scrubbing with a 1% povidone-iodine solution. Povidone-iodine<br />

as a 10% solution and other antiseptics (e.g., hexachlorophene and<br />

chlorhexidine) are harmful to tissues and increase infection rates,<br />

thus prohibiting their application to intact skin surfaces (Table<br />

84–1). 30–34 After a through cleansing, surgical débridement, exci -<br />

sion, and hemostasis, the wound is ready for closure.<br />

Techniques of Wound Closure<br />

Overview<br />

When the wound is considered clean or becomes clean after<br />

scrubbing, irrigation, and débridement, it must be closed. This<br />

procedure minimizes inflammation, fibroplasias, contracture,<br />

scar width, and contamination. 35 Wounds that close by secondary<br />

intention result in more deformity and loss of function. 36–38<br />

Despite this, certain wounds should almost always be left open<br />

or closure delayed. Included in this category are those already<br />

infected or heavily contaminated by soil, organic matter, or feces;<br />

those associated with extensive tissue damage (e.g., high-velocity<br />

missile injuries, explosion injuries of the hand, or complex crush<br />

injuries); and most bite wounds. 38–40 One of the available closure<br />

techniques must be selected according to location and config -<br />

uration of the wound (Table 84–2).<br />

Hair Tying of Scalp Wounds<br />

Closure of scalp wounds by hair tying offers an attractive alter -<br />

native for closure of small superficial scalp wounds (1–2 cm in<br />

length) in children. 41 It is a particularly humane method and<br />

relatively painless because a local anesthetic injection is not<br />

needed. In a series of 25 children younger than 8 years whose scalp<br />

wounds were closed by hair tying, a 48-hour follow-up showed no<br />

evidence of wound infection and only 2 cases of mild (2–4 mm)<br />

wound separation. 42


TABLE 84-1. Cleaning Solutions for Wound Care<br />

CHAPTER 84 ■ Miscellaneous Techniques 1409<br />

Antiseptic Solution Biologic Activity Indications Comments<br />

Povidone-iodine detergent<br />

solution (10%) a<br />

Povidone-iodine aqueous<br />

solution (10%) a<br />

Povidone-iodine diluted<br />

aqueous solution (l%) a<br />

Povidone-iodine alcoholic<br />

solution (10%) a<br />

Chlorhexidine gluconate<br />

detergent solution (4%)<br />

Hexachlorophene detergent<br />

solution (pHisoHex)<br />

Hydrogen peroxide<br />

Strong germicidal<br />

Strong germicidal<br />

Same as full-strength solution<br />

Strong germicidal<br />

Strongly gram-positive<br />

bactericidal<br />

Less strong against gramnegative<br />

bacteria<br />

Gram-positive bacteriostatic<br />

Poor activity against gramnegative<br />

bacteria<br />

Very weak antibacterial agent<br />

Hand and intact skin cleanser<br />

Wound periphery cleanser<br />

Mucosal antiseptic<br />

Open wound cleanser<br />

Wound irrigation<br />

Surgical field antiseptic<br />

Invasive procedures<br />

antiseptic<br />

Hand cleanser<br />

Alternative hand cleanser<br />

Clean skin encrusted with<br />

blood and coagulum<br />

Soak off adherent bloodsoaked<br />

dressings<br />

a<br />

Iodine and chlorhexidine solutions should be applied for at least 2 min to obtain maximal residual bactericidal activity, >60 min.<br />

Toxic to wound tissues<br />

Painful to open wounds<br />

Minimally toxic to wound tissues<br />

No significant tissue toxicity<br />

Safest and most effective product<br />

for open wound cleaning<br />

Irritating and toxic to open<br />

wounds and mucosa<br />

Toxic to cellular components of<br />

blood (in vitro studies)<br />

Avoid use in open wounds and<br />

eye/ear cleaning<br />

Toxic to wound tissues<br />

Neurotoxic and teratogenic<br />

through skin absorption<br />

No more effective than ordinary<br />

soap and water<br />

Toxic to tissue/red cells<br />

Foaming activity useful to remove<br />

debris/coagulated blood<br />

Wound Taping<br />

The primary indication for tape closure is a superficial straight<br />

laceration with no significant tension (Figure 84–3). Tape closure<br />

can be used with deep sutures to reduce tension. Some wounds in<br />

anxious children, when sutures are not clearly indicated, may be<br />

closed with tape. The most suitable areas are the hairless surfaces<br />

of the forehead, chin, malar eminence, thorax, and nonjoint areas<br />

of the extremities. It should be considered for wounds with<br />

potential for infection, under plaster casts, and is mandatory when<br />

reducing the chance of a permanent suture mark scar. 43–45 Several<br />

brands of tapes are available with specific properties: air and water<br />

porosity, flexibility, strength, and adhesiveness. 46,47<br />

Application of Tissue Adhesive<br />

The tissue adhesive N-2-butylcyanoacrylate (Histoacryl glue) is a<br />

bonding agent that can be used instead of tape strips on tensionless<br />

superficial wounds. Its advantage is that there is no limitation to<br />

hairless surfaces. Some studies found no difference between the<br />

TABLE 84-2. Techniques of Wound Closure<br />

Technique Indications Comments<br />

Hair tying<br />

Wound tape a<br />

Tissue adhesive a<br />

Wound staples a<br />

Sutures<br />

Small superficial scalp wounds<br />

Superficial straight lacerations on hairless<br />

surfaces, forehead, chin, malar eminence,<br />

thorax, and nonjoint areas of the extremities<br />

Tensionless wounds or secondary support to<br />

skin suture<br />

Same as wound tape<br />

Linear lacerations with straight, sharp edges on<br />

an extremity, the trunk, or the scalp<br />

Wounds with complex configurations<br />

Wounds extending into deeper layers<br />

Wounds in mobile areas<br />

a<br />

Must be associated with deep, absorbable sutures, whenever necessary to reduce tension.<br />

Painless method<br />

No inflammatory response to suture material<br />

Painless method<br />

Can be associated with deep sutures to reduce tension<br />

No limitation to hairless surfaces<br />

Three to four times faster than suturing<br />

Especially useful for agitated, uncooperative pediatric<br />

patients; probably same cost as that of suturing<br />

Most commonly used method<br />

Different materials for two categories to be used<br />

(absorbable and nonahsorbable sutures)<br />

Suture materials impair host defenses and infection<br />

resistance and provoke inflammation


1410 PART 4 ■ Special Monitoring and Resuscitation Techniques<br />

Figure 84-3. Tape closure of a<br />

superficial wound.<br />

tensile strength and overall degree of inflammation in wounds<br />

closed with adhesive and with sutures. 48,49 Cosmetic results were<br />

also similar. 50,51<br />

Use of Wound Staples<br />

Nowadays, the indications for stapling are limited to relatively<br />

linear lacerations with straight, sharp edges located on an extrem -<br />

ity, the trunk, or the scalp. The most significant advantage of<br />

wound stapling over suturing is speed of closure. Although it must<br />

be performed under local anesthesia, and deep, absorbable sutures<br />

whenever necessary to reduce tension, it is a procedure three to<br />

four times faster than suturing and especially useful for agitated,<br />

uncooperative emergency pediatric patients. 52–55 Many stapling<br />

devices are commercially available. With the introduction of new<br />

devices, the cost of wound stapling is comparable with that of<br />

suturing. 54 Because of the increased availability and versatility<br />

of stapling instruments, they are being used more frequently in<br />

traumatic wound management. Four studies have demonstrated<br />

their application in both adult and pediatric patients in the<br />

emergency department setting. 52–55<br />

Sutures<br />

The traditional and most commonly used method of closure is<br />

suturing. Suture repair is the most appropriate method for wounds<br />

with complex configurations, those that extend into deeper layers,<br />

and those in mobile areas. 35 All sutures will damage host defenses,<br />

provoke inflammation, and impair the ability of the wound to<br />

resist infection. 56 For most wounds with more than one layer of<br />

tissue to be closed, suture materials must be chosen from two<br />

general categories 57 : (1) an absorbable suture for the subcutaneous<br />

or deeper layers (e.g., plain gut, chromic gut, polyglactin, poly -<br />

dioxanone) and (2) a nonabsorbable suture for skin closure (e.g.,<br />

nylon, polypropylene, cotton, silk). Multifilament synthetic ab -<br />

sorbable sutures (e.g., Vicryl, coated Vicryl) have the best handling<br />

characteristics, although they provoke more inflammation and<br />

infection than monofilament sutures. 58,59 Polydioxanone sutures<br />

provide the advantages of a monofilament synthetic suture in<br />

an absorbable form, making it a good choice as a subcuticular<br />

stitch. 60,61 Absorbable gut sutures have many disadvantages—<br />

including relatively low and variable strength, a tendency to fray<br />

when handled, stiffness despite being packaged in a softening<br />

fluid—and are the most reactive. 62,63 Natural suture materials have<br />

been made obsolete by the newer, synthetic products. They have<br />

improved handling characteristics, knot security and tensile<br />

strength, and predictable absorption rates and induce minimal<br />

tissue reactivity. 56,64,65 In the emergency department, the most<br />

useful suture materials for wound closure are Dexon or coated<br />

Vicryl for subcutaneous or deeper layers and nylon or polypro -<br />

pylene for skin closure. In most situations, 3-0 or 4-0 sutures are<br />

used to the repair of fascia, 4-0 or 5-0 absorbable sutures in<br />

subcutaneous closure, and 4-0 or 5-0 nonabsorbable sutures<br />

in skin closure. 35<br />

The most important principle applied to suturing is relieving<br />

tension exerted on edges by tissue plane undermining and layered<br />

closure (Figure 84–4A). Separate approximation of fascia and<br />

subcutaneous layers hastens the healing and return of muscle<br />

function, preventing deformation of the surface of the wound. 66–68


CHAPTER 84 ■ Miscellaneous Techniques 1411<br />

Figure 84-4. Surgical closure of a<br />

wound. A: Technique of tissue plane<br />

undermining. B: Interrupted stitches.<br />

C: continuous stitch. D: Surgical tape<br />

as support to skin movements.<br />

Skin closure consists of matching the epidermis and the superficial<br />

layer of dermis on each side without excessive tension stitches.<br />

Three general methods should be used:<br />

1. Interrupted stitch: It is the most frequently used technique for<br />

skin closure. Although time-consuming, the advantage is that<br />

each stitch holds the wound together independently from the<br />

other (see Figure 84–4B).<br />

2. Continuous stitch: In a continuous stitch, the loops are the<br />

exposed portions of a helical coil that is tied at each end of the<br />

wound. It is more rapidly performed and has the advantage of<br />

strength, fewer knots, and more effective hemostasis. It is<br />

indicated for closing relatively clean wounds that are under<br />

little or no tension and are on flat, immobile skin surfaces<br />

(see Figure 84–4C).<br />

3. Continuous subcuticular stitch: This stitch is the ideal tech -<br />

nique for pediatric patients who are likely to be as frightened<br />

and uncooperative for suture removal as for suture placement.<br />

An absorbable subcuticular suture (e.g., polydioxanone) may<br />

be used, obviating the need for later removal. It can be left in<br />

place for a longer period than a percutaneous suture because<br />

stitch marks are avoided.<br />

Repair of Special Structures<br />

Repair of special structures requires specific procedures. 35,68<br />

FACIAL WOUNDS: Subcutaneous fat should be preserved, if<br />

possible, to prevent eventual depression of the scar and preserve<br />

normal facial contours. Surgical tape is useful as a secondary<br />

support, protecting the epithelial stitch from the stress produced<br />

by normal skin movements (see Figure 84–4D).<br />

EYEBROW AND EYELID LACERATIONS: Minimal, if any, débridement<br />

should be done. A vertical excision may produce a linear<br />

alopecia in the eyebrow, whereas with simple closure, the scar<br />

remains hidden under the hair. The eyebrow must not be shaved<br />

because it often results in more deformity than the injury itself.<br />

Eyelid wounds should be carefully evaluated for concomitant<br />

lesions to other structures (e.g., tarsal plate, levator palpebrae<br />

muscle, lacrimal duct), and an ophthalmologist’s opinion may be<br />

required.<br />

EAR LACERATIONS: The primary goals are expedient coverage of<br />

exposed cartilage and minimization of wound hematoma.<br />

LIP AND INTRAORAL LACERATIONS: The vermilion-cutaneous<br />

junction of the lip is a critical landmark that, if divided, must be<br />

repositioned with precision. Small flaps of oral mucosa may<br />

be excised, whereas extensive lesions should be closed with<br />

interrupted 4-0 Vicryl suturing.<br />

TONGUE LACERATIONS: Even large (simple or linear) lacerations,<br />

especially those in the central portion of the tongue, heal quickly<br />

with minimal risk of infection. Only those lacerations that involve<br />

the edge or pass completely through the tongue, flap lacerations,<br />

and excessively bleeding lacerations need to be sutured. Large flaps<br />

require repair, but small ones on the edge may be excised. Most<br />

lacerations that need to be sutured in uncooperative children must<br />

be performed under general anesthesia. Interrupted stitches with<br />

4-0 absorbable sutures are used including all thicknesses of the<br />

tongue. In small children, a surface suture is likely to be interfered<br />

with, and closure of only the muscle layer with a deep absorbable<br />

suture may be preferred (mucosal healing is rapid).<br />

SCALP LACERATIONS: The rich vascular network found in the<br />

scalp fascia results in abundant bleeding from scalp wounds.<br />

Profuse bleeding may have serious consequences in small infants<br />

without prompt treatment. 69,70 It is best controlled by expeditious<br />

suturing with 3-0 nylon or polypropylene, incorporating skin,<br />

subcutaneous tissue, and galea in a single stitch.<br />

NAIL LACERATIONS: Injuries to the nail or nailbed are common<br />

problems in pediatric emergency patients. The finger extremity<br />

has to be cleaned, simple nailbed lacerations repaired (6-0 or 7-0<br />

absorbable sutures), and the avulsed nail reapplied to cover the<br />

sensitive area and maintain the fold for new nail growth. When<br />

the nailbed has been extensively lacerated or partially avulsed, a<br />

hand surgeon should be consulted. 71<br />

Instructions to the Patient Before Discharge<br />

Wound healing and its result depend on the care given to the<br />

wound after treatment. Surgical wounds usually need no special<br />

care before dressing removal. Nurses are advised to inquire about


1412 PART 4 ■ Special Monitoring and Resuscitation Techniques<br />

pain and look for inflammatory signs in hospitalized patients.<br />

The same instructions should be given to parents in a thorough<br />

and understandable way before hospital discharge. The dressing<br />

must be kept clean and dry for the first 24 to 48 hours; after that,<br />

the wound is essentially impermeable to water and bacteria. 23<br />

Complicated or infection-prone wounds have to be reviewed in<br />

2 days. If there is no sign of infection, parents may provide a daily<br />

gentle washing with mild soap and water until it is time for the<br />

removal of the sutures. Vigorous scrubbing should be discouraged.<br />

Rabies and tetanus postexposure prophylaxis schedule must be<br />

explained as well. 72,73<br />

Suture Removal<br />

There is no standard time for suture removal because wounds do<br />

not heal at a standard rate. As a general rule, sutures are removed<br />

within 7 days, and if necessary, wound repair can be maintained<br />

with strips of surgical skin tape. Sutures left in place for longer<br />

than 7 to 10 days are associated with extensive inflammatory<br />

response and stitch abscesses that determine the severity of stitch<br />

marks. 25,74 Whenever possible, absorbable subcuticular sutures<br />

should be used to avoid this stressful and traumatic procedure for<br />

infants and children.<br />

ARTHROCENTESIS<br />

Arthrocentesis, the puncture and aspiration of a joint, is an<br />

acknowledged useful diagnostic and therapeutic procedure that is<br />

easily performed in the emergency department and indicated for<br />

a variety of clinical situations. 75–77<br />

Indications and Contraindications<br />

Because an acutely swollen joint may be indicative of a number<br />

of disease entities, thorough history and physical examination are<br />

the cornerstones of evaluation, followed by arthrocentesis. 75,76<br />

Indications<br />

The indications for arthrocentesis are 77–80<br />

●<br />

●<br />

●<br />

●<br />

●<br />

●<br />

●<br />

Diagnosis of nontraumatic joint disease by synovial fluid analysis<br />

(e.g., septic arthritis).<br />

Differential diagnosis between a traumatic and an inflammatory<br />

joint effusion confirmed by hemarthrosis.<br />

Differential diagnosis between articular and periarticular<br />

disease (e.g., tendinitis, bursitis, contusion, cellulites, or phle -<br />

bitis).<br />

Diagnosis of an intra-articular fracture by the presence of blood<br />

with fat globules in the aspiration.<br />

Pain relief of an acute hemarthrosis of a tense effusion.<br />

Local injection of medications in acute and chronic inflammatory<br />

arthritides.<br />

Obtaining fluid for culture, Gram staining, immunologic studies,<br />

and cell count in cases of suspected joint infection.<br />

SEPTIC ARTHRITIS: Acute monoarticular arthritis is a common<br />

problem in emergency medicine. Although there are many causes<br />

of acute monoarticular arthritis, the one most requiring urgent<br />

diagnosis and treatment is septic arthritis. Confirmation requires<br />

arthrocentesis and synovial fluid culture. <strong>81</strong> Infants aged 6 months<br />

to 2 years show a higher incidence of Haemophilus influenzae<br />

cultures, whereas in neonates, staphylococci and Escherichia coli<br />

predominate this infection. 82 Neisseria gonorrhoeae is the most<br />

common organism causing septic arthritis among adolescents and<br />

young adults. 83 Patients with other medical illnesses are more<br />

likely to have staphylococcal joint infections.<br />

HEMARTHROSIS: Hemarthrosis after trauma is a frequent occur -<br />

rence and often denotes significant internal damage (e.g., a tear in<br />

a ligamentous structure, knee capsule, or a fracture). If the history<br />

of trauma is vague, arthrocentesis may be required to differentiate<br />

hemorrhage from other causes of joint effusion. Isolated nontrau -<br />

matic hemarthrosis may occasionally be seen by the emergency<br />

physician, especially in patients with bleeding diatheses (e.g.,<br />

hemophilia or oral anticoagulant therapy). 84,85 Distention of the<br />

joint by effusion or hemorrhage causes considerable pain and<br />

disability. 86 Therapeutic arthrocentesis to drain a symptomatic<br />

traumatic effusion is a well-accepted practice. 87<br />

INTRA-ARTICULAR CORTICOSTEROID INJECTION: The use of<br />

steroids has proved to be a dependable method for providing<br />

rapid relief (within 6–12 h after the injection) from pain and<br />

swelling of inflamed joints, although it is strictly local, usually<br />

temporary, and rarely curative. 88–90 Corticosteroid injections<br />

are most helpful when only a few of a patient’s joints are<br />

actively inflamed. 91 The most serious complication of this practice<br />

is intra-articular infection. Steroids are contraindicated when an<br />

infection is suspected.<br />

Contraindications<br />

The most important contraindication to arthrocentesis is the<br />

presence of infection in the tissue overlying the site to be punctured<br />

(e.g., an abscess or frank cellulitis). However, inflammation with<br />

warmth, swelling, and tenderness may overlie an acutely arthritic<br />

joint, and this condition may mimic a soft tissue infection. Once<br />

convinced that cellulitis does not exist, the clinician should not<br />

hesitate to obtain the necessary diagnostic joint fluid. 75,76 A relative<br />

contraindication to joint puncture is the presence of a bacteremia.<br />

Bleeding diatheses may at times be a relative contraindication, but<br />

arthrocentesis to relieve a tense hemarthrosis in bleeding disorders,<br />

such as hemophilia, is an accepted practice after infusion of<br />

the appropriate clotting factors. Arthrocentesis is also relatively<br />

contraindicated for a patient receiving anticoagulants or in the<br />

presence of a joint prosthesis, unless the procedure is being<br />

performed to exclude infection. 75,79<br />

General Arthrocentesis Technique<br />

Although one may successfully aspirate where the joint bulges<br />

maximally, certain landmarks are important (see Specific Arthro -<br />

centesis Techniques). 75,76,79 The most crucial part of arthrocentesis<br />

is spending adequate time in defining the joint anatomy by<br />

palpating the bony landmarks as a guide. An aseptic technique is<br />

essential to avoid introducing infection. It is important to have the<br />

patient relax during the procedure. Even with appropriate local<br />

anesthesia, arthrocentesis is a relatively distressing procedure to<br />

pediatric patients. Tense muscles narrow the joint space and make<br />

the procedure more difficult, requiring repeated attempts at<br />

aspiration, and which results in inadequate drainage, bleeding, or<br />

unwarranted cartilage damage. As a general rule, the procedure<br />

should be performed under general anesthesia associated with a<br />

regional block.


CHAPTER 84 ■ Miscellaneous Techniques 1413<br />

Figure 84-6. Elbow arthrocentesis<br />

Figure 84-5. Wrist arthrocentesis.<br />

Specific Arthrocentesis Techniques<br />

The following are arthrocentesis techniques for specific parts of<br />

the body. 75,76<br />

Wrist<br />

The dorsal radial tubercle (Lister’s tubercle) is an elevation found<br />

in the center of the dorsal aspect of the distal end of the radius.<br />

The extensor pollicis longus tendon runs in a groove on the radial<br />

side of the tubercle. The tendon can be palpated by active<br />

extension of the wrist and thumb. The wrist should be positioned<br />

in approximately 20 to 30 degrees of flexion and accompanying<br />

ulnar deviation (Figure 84–5). Traction is applied to the hand.<br />

A 22- to 24-gauge needle is inserted dorsally, just distal to the<br />

dorsal tubercle on the ulnar side of the extensor pollicis longus<br />

tendon. The anatomic snuffbox, located more radially, should<br />

be avoided.<br />

to the coracoid process and directed posteriorly toward the<br />

glenoid rim (Figure 84–7). Arthrocentesis of this joint is of mod -<br />

erate difficulty. Other approaches have been suggested but are less<br />

well accepted.<br />

Ankle<br />

The lateral malleolus and the distal tibia are the main landmarks<br />

of the ankle. With dorsiflexion of the foot, a 20- to 22-gauge needle<br />

is inserted 1 cm medial and 1 cm cephalad to the lateral malleolus,<br />

at the lateral sulcus between the articular surface of the talus and<br />

the distal fibula, in an anteroposterior direction (Figure 84–8).<br />

Knee<br />

The middle or superior portion of the lateral surface of the<br />

patella is the landmark for the knee joint (Figure 84–9). The knee<br />

Elbow<br />

The lateral epicondyle of the humerus and the head of the radius<br />

are the landmarks for arthrocentesis of the radiohumeral joint<br />

(Figure 84–6). The depression between the radial head and the<br />

lateral epicondyle of the humerus is palpated during elbow<br />

extension. With the elbow flexed 90 degrees, the forearm pronated,<br />

and the palm placed down flat on a table, a 22-gauge needle is<br />

inserted from the lateral aspect just below the lateral epicondyle<br />

and directed parallel to the shaft of the radius. A medial approach<br />

should not be used, because the ulnar nerve and the superior ulnar<br />

collateral artery may be damaged.<br />

Shoulder<br />

The coracoid process medially and the proximal humerus laterally<br />

are palpated anteriorly while the shoulder is externally rotated.<br />

A 20- to 22-gauge needle is inserted at a point inferior and lateral<br />

Figure 84-7. Shoulder arthrocentesis.


1414 PART 4 ■ Special Monitoring and Resuscitation Techniques<br />

Figure 84-8. Ankle arthrocentesis.<br />

is fully extended as far as possible. Relaxation of the quadriceps<br />

muscle greatly facilitates needle placement. The foot is kept<br />

perpendicular to the floor. An 18-gauge needle is inserted at the<br />

midpoint or superior portion of the patella approximately 1 cm<br />

lateral to the anterolateral patellar edge. The needle is directed<br />

between the posterior surface of the patella and the intercondylar<br />

femoral notch.<br />

Hip<br />

The anterior superior iliac spine, the femoral artery, the inguinal<br />

ligament, and the greater trochanter should be identified (Figure<br />

84–10). For the anterior approach, a mark should be made 2 cm<br />

Figure 84-10. Hip arthrocentesis. A: landmarks. B: anterior<br />

approach.<br />

below the inguinal ligament and 2 cm lateral to the femoral artery.<br />

A 20-gauge needle is inserted at a 60-degree angle to the skin and<br />

directed posteriorly and medially until synovial fluid is obtained.<br />

In the lateral approach, the needle is inserted just anterior to the<br />

greater trochanter and directed medially and slightly cephalad<br />

toward a point below the middle of the inguinal ligament. This is<br />

a difficult joint for arthrocentesis, and ultrasound guidance in<br />

children is very helpful. 92,93<br />

Complications<br />

Infection, the most serious complication, is extremely rare.<br />

Various studies report the incidence of infection after routine<br />

arthrocentesis to be in the range of 0.001 to 0.005%. 94,95 The most<br />

common adverse reaction is related to corticosteroid injection and<br />

consists of corticosteroid crystal-induced synovitis, with a 1 to 2%<br />

occurrence. 95 Repeated corticosteroid injections into one joint<br />

carry the risk of necrosis of the juxta-articular bone with subse -<br />

quent joint destruction and instability. Other rare complications<br />

include local soft tissue atrophy and calcification, tendon rupture,<br />

intra-articular bleeding, and transient nerve palsy. 90<br />

Figure 84-9. Knee arthrocentesis.<br />

IMMOBILIZATION TECHNIQUES<br />

Initial Evaluation<br />

The initial evaluation of a patient with a fracture or another injury<br />

needing some form of immobilization involves at least the<br />

following objectives: (1) to relieve pain, (2) to obtain and maintain<br />

a satisfactory position of the fracture fragments, and (3) to avoid<br />

secondary damage to soft tissues. 96 The rescuer must not let<br />

obvious injuries to the extremities be a distraction to the treatment<br />

of more life-threatening lesions. Special attention to airway,<br />

breathing, and circulation, as established for routine emergency

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!