Physical Address
304 North Cardinal St.
Dorchester Center, MA 02124
The field of pediatric anesthesia has mirrored that of pediatric surgery—developing as a result of the need for specialized treatment of uncommon and complex congenital anomalies and childhood surgical diseases ( ; ). Despite the complexity, general surgery of the neonate is becoming increasingly common and safe. Twenty-five percent of the most common pediatric general surgery operations occur in neonates ( ). Improved safety within the neonatal population has been linked to improvements in surgical technique, anesthetic care, and development of the modern-day neonatal intensive care unit (NICU) ( ).
Newborn babies who need intensive medical care are admitted to the NICU. The NICU specializes in the unique factors associated with neonates, some of which include prematurity (gestational age <37 weeks), low birth weight, or congenital anomalies. Extreme prematurity and associated complications remain the leading causes of neonatal mortality and a major source of morbidity requiring surgery ( ). The introduction of specialized intraoperative care by pediatric surgery and anesthesia are key contributing factors to the overall reduction in mortality since 1960 ( ). The framework for the specialized anesthetic care provided to neonates requires an understanding of neonatal pharmacology and physiology.
This chapter addresses anesthetic pharmacology for the neonate, approaches to preoperative assessment, and intraoperative monitoring of the neonate, as well as the more common neonatal general surgical procedures with respect to perioperative management, embryology, and long-term outcome.
The landmark studies of Anand and Hickey in the 1980s demonstrated that general anesthesia in neonates prevented the acute surgical stress response and decreased morbidity and mortality ( ; ). Increasing evidence confirms that the neonate has a physiologic response to painful stimuli similar to that of adults, whereby hormonal, metabolic, and cardiorespiratory changes can be observed (see Chapter 23 , “Acute Pain Management” and Chapter 25 , “Chronic Pain Management”). Anesthesia for the neonate has the same goals as caring for older patients: avoid pain and its adverse cardiovascular and neurologic consequences. A significant challenge in neonates is the variable response to medications, including opioids and inhalation agents. Understanding the pharmacokinetics and pharmacodynamics of the neonate is imperative for careful titration of these agents when undergoing surgery. See Chapter 27 , “Neonatology for Anesthesiologists” and Part II, “Pharmacology.”
Pharmacokinetics and pharmacodynamics of drugs are affected by anatomic factors relating to body composition and distribution of water, as well as physiologic factors such as metabolism (i.e., hepatic biotransformation), protein binding, and pathologic factors (e.g., disease, anesthesia, and surgery) (see Part II, “Pharmacology”). Compared with older children and adults, neonates have significant body composition and physiologic differences. In early fetal development, water constitutes approximately 94% of body weight. As gestation continues, the total body water decreases so that at 32 weeks, 80% to 90% of body weight is water, and at term, total body water is approximately 70% to 75% of body weight. Adult proportions of fluid to body weight (55%) are reached between 9 months and 2 years of age. The distribution of water between the extracellular and intracellular compartments also changes during fetal growth. Extracellular water (interstitial fluid plus plasma volume) decreases from 60% of body weight at the fifth month of fetal life to approximately 45% at term. Intracellular water increases from 25% in the fifth month of fetal life to 33% at birth; therefore the extracellular fluid compartment of the newborn is greater than or equal to the intracellular fluid space. In adults, the intracellular and extracellular fluid compartments are approximately 40% and 20% of body weight, respectively. Because the plasma component of the extracellular fluid compartment remains at approximately 5% of body weight throughout life, it is the interstitial water that is greater in infancy (40%) and declines to 10% to 15% in the adult ( ).
Age-dependent changes in body composition also occur. At term, fat constitutes 11% of body weight. Fat content doubles by 6 months of age and is approximately 30% at 1 year. Teenage girls remain with approximately 20% to 30% fat, whereas in teenage boys it decreases to 10% to 15% fat. Moreover, the composition of fat tissue changes with age. Fat in the newborn may contain as much as 57% water and 35% lipids; adults have 26% water and 71% lipids ( ). Skeletal muscle comprises 25% of total body mass in a full-term newborn compared with 43% in an adult.
The binding of drugs to serum proteins depends on several factors, including the concentration of protein, the number of binding sites on these proteins, and the affinity of the binding sites. The concentration of total serum protein, albumin, and α 1 -acid glycoprotein is lower in early infancy and reaches adult levels by approximately 1 year of age ( ). Albumin primarily binds acidic drugs; α 1 -acid glycoprotein binds basic drugs. The concentration of these two proteins and their binding affinities are deficient in the newborn ( ).
The primary organ for drug biotransformation is the liver, but the kidneys, intestines, lungs, and skin also have minor roles. Hepatic oxidation, reduction, and hydrolysis (nonsynthetic, phase I reactions) mature rapidly, achieving adult rates by 6 months of age ( ). Drugs metabolized via this cytochrome P450–dependent monooxygenase system include phenobarbital and phenytoin. Conjugation reactions (synthetic, phase II reactions) convert drugs into more polar compounds to facilitate renal excretion. These systems also mature postnatally.
The renal excretion of drugs is a function of glomerular filtration rate (GFR), active secretion, and passive reabsorption. GFR and secretion increase in an age-dependent manner. Renal blood flow and GFR increase dramatically during the first postnatal week and more gradually during the next several months, and adult performance is achieved at approximately 6 to 12 months of age ( ; ). Tubular secretory and reabsorptive capacity also mature postnatally ( ). (See Chapter 6 , “Regulation of Fluids and Electrolytes.”)
Cardiac output and its distribution to various organs contribute to drug elimination. The perinatal adaptation to extrauterine life demands rapid changes in the circulation. This process may be inhibited as a result of congenital heart disease or acid-base problems. Drug metabolism and elimination may be drastically affected when cardiovascular function is abnormal.
Infants have a higher incidence of cardiovascular instability and cardiac arrest during induction of inhalational anesthesia than do older persons ( ; ; ; ; ) (see Chapter 10 , “Inhaled Anesthetic Agents”). This untoward effect of potent inhalational agents can be attributed to several factors, including faster equilibration, rapid myocardial uptake in infants, increased anesthetic requirement, and sensitivity of the neonatal myocardium. Infants attain a higher concentration of inhaled anesthetic agents in the heart and brain than do adults at the same inspired concentration. (See Chapter 10 , “Inhaled Anesthetic Agents,” Fig. 10.6 ). Moreover, the neonatal myocardium has a smaller fraction of contractile mass, and the magnitude and velocity of fiber shortening are less than in the adult myocardium. These factors and the increased anesthetic requirement, which is inversely related to age, all produce a higher incidence of adverse cardiovascular effects in infants.
The rate of rise of the alveolar concentration of an inhaled anesthetic depends on several factors: the inspired concentration, alveolar ventilation, and uptake. The greater the alveolar ventilation, the faster the rate of rise of the alveolar concentration ( Box 28.1 ). This effect of alveolar ventilation is affected by the size of the functional residual capacity (FRC). Infants and children have an FRC similar to that of the adult, 30 mL/kg per minute. In contrast, alveolar ventilation is much higher in the infant (100 to 150 mL/kg) compared with the adult (60 mL/kg per minute). This difference parallels the greater oxygen consumption of the infant. Thus in the normal-term newborn who weighs 3.0 kg, the ratio of alveolar ventilation to FRC is approximately 5:1, compared with the adult, in whom the same ratio is 1.5:1. As a result of this difference, the time constant of the inhaled anesthetic equilibrium for infants is much shorter than that for the adult. Consequently, changes in concentrations of inspired gas are reflected rapidly in alveolar levels. In fact, it has been demonstrated that alveolar levels of inhalational anesthetic agents reach equilibrium faster in infants than in adults (see Chapter 10 , “Inhaled Anesthetic Agents”).
V a /FRC 5:1 compared with adult 1.5:1
Increase cardiac output to vessel-rich group
Decreased tissue solubility
The rise in alveolar concentration of an inhaled anesthetic is opposed by uptake of the agent into lung tissue and, more importantly, blood. Three factors determine inhaled anesthetic uptake: cardiac output, the alveolar–to–mixed venous anesthetic partial pressure difference, and solubility. Each of these factors has unique aspects in the infant compared with in the adult and consequently affects the pharmacology of the uptake of inhaled agents.
The greater the cardiac output, the greater the anesthetic uptake. The cardiac output of the newborn is 250 to 300 mL/kg per minute. By 8 weeks of age, the cardiac output has decreased to 150 mL/kg per minute. The cardiac output of young infants is approximately three to six times that of the normal adult (70 mL/kg per minute). By itself, this high cardiac output should significantly decrease the rate of rise of the alveolar concentration of the soluble anesthetic agents. However, the newborn distributes a greater proportion of this cardiac output to the vessel-rich group of organs and a smaller proportion to the muscle and fat group. The equilibrium between the inspired and alveolar concentrations of inhaled agent occurs more rapidly because uptake decreases faster.
Because cardiac output is predominantly distributed to the vessel-rich group and because the muscle group is small, the arterial-venous partial pressure difference narrows quickly in the young and thereby decreases uptake.
Both left-to-right and right-to-left shunting occurs in infants. A left-to-right shunt results in an increase in total cardiac output. However, the shunted blood does not lose anesthetic to tissue; instead it returns to the lung with the same anesthetic partial pressure. This recycled blood cannot accept more anesthetic agent unless the alveolar partial pressure has risen. Thus a left-to-right shunt has no effect on anesthetic uptake. A right-to-left shunt slows the rate of rise of the alveolar concentration of an inhaled anesthetic. The anesthetic-deficient shunted blood dilutes the concentration of the anesthetic in the blood, decreasing the partial pressure of anesthetic in the arterial circulation. This slows the rate of rise of the anesthetic by slowing tissue uptake and equilibration.
Blood-gas partition coefficients of isoflurane, halothane, and sevoflurane did not differ in preterm infants compared with full-term infants but were lower than in adults. Only serum cholesterol correlated with the blood-gas partition coefficients ( ). The blood-gas partition coefficient is an important determinant of solubility and therefore the rate of rise of the alveolar concentration of an inhaled agent ( ).
The effect of age on the solubility of the inhaled agents in tissue is also important in determining the rate of rise of the alveolar concentration of the agent—the rate of anesthetic induction. Data by Lerman and colleagues are consistent with earlier work documenting that anesthetic solubility in brain, heart, liver, and muscle increases with age ( ). An increase in solubility may prolong uptake, delay equilibration of the tissue partial pressure of anesthetic, and prolong the time of induction. found that the rate of increase in tissue anesthetic partial pressure, and therefore alveolar anesthetic partial pressure, is approximately 30% more rapid in newborns than in adults.
Minimal alveolar concentration (MAC) is an estimate of anesthetic requirement and changes with age ( Fig. 28.1 ) ( ; ; ). In the original study, Gregory and coworkers reported that infants in the first 6 months of life had the highest MAC ( ). In a later study, newborns were noted to require approximately 25% less halothane at MAC compared with infants who were between 1 and 6 months of age ( ) (see Part II, “Pharmacology”).
Several studies have shown an increased sensitivity to and more prolonged effects of barbiturates and morphine in the neonate and young infant ( ; ). (See Chapter 12 , “Opioids.”) These features have been attributed in part to the immaturity of the blood-brain barrier, allowing faster and greater penetration and therefore higher concentration of these drugs in the brain. (See Chapter 12 , “Opioids,” Fig. 12.2 ).
In 1981, Robinson and Gregory reported that after a 10 mL/kg bolus of lactated Ringer’s solution, 30 to 50 mcg/kg of fentanyl was a safe anesthetic for premature infants undergoing ligation of a patent ductus arteriosus (PDA). Several years later, evidence was presented that infants who received fentanyl in combination with d -tubocurarine and nitrous oxide in oxygen had an improved perioperative course compared with those infants who did not receive analgesics ( ).
Plasma levels of fentanyl are lower in infants versus children versus adults (newborns were not studied) after similar intravenous doses ( ). Gauntlett and colleagues noted that clearance of fentanyl in newborns increased during the first few weeks of life ( ). Elimination half-life and volume of distribution did not change. In a study of newborns who were administered continuous fentanyl infusions, Saarenmaa and associates noted that plasma clearance correlated with maturity (gestational age) and weight ( ), whereas Santeiro and coworkers noted a correlation of clearance with postnatal age ( ) ( Fig. 28.2 A and B). Koehntop and colleagues showed a highly variable disposition and elimination of fentanyl in neonates ( ). In addition, infants with increased intraabdominal pressure (e.g., omphalocele, gastroschisis, or septic ileus) appeared to have a further increase in the elimination half-life compared with infants undergoing repair of a PDA or myelomeningocele. Davis and associates noted that the clearance of alfentanil in newborn premature infants was markedly reduced compared with older children ( ) ( Fig. 28.3 ).
Koren and coworkers showed that the elimination half-life for morphine (13.9 hours) in human neonates is markedly prolonged compared with that in older children and adults (2 hours) ( ). They also showed reduced clearance and higher serum concentration in neonates compared with those seen in older children after morphine infusion. Because of the large variability in clearance among neonates of different ages, the dose of opioids needs to be carefully titrated for each patient and each clinical setting.
Remifentanil is an ultra-short-acting opioid. Remifentanil is metabolized by plasma and tissue esterases, and its metabolism is independent of organ elimination. As a result, in neonates its pharmacokinetic profile is different from any other opioid. The remifentanil clearance rate in neonates is greater than in any other age group. Although its volume of distribution is large, its terminal elimination half-life does not change with age ( ). Because remifentanil is metabolized in tissues and plasma, it does not accumulate, and consequently its context-sensitive half-time is flat (i.e., the time for 50% reduction at the effect site is independent of drug duration). Thus remifentanil may be an ideal drug for anesthetic use in neonates.
Ketamine requirements are greater (mg per kg of body weight) in infants than in older children ( ). Ketamine has been shown to produce apnea in infants with increased intracranial pressure ( ). Ketamine produces hypertension and tachycardia, which some anesthesiologists have taken advantage of in caring for infants and children with congenital heart disease, cardiovascular instability, or both.
Propofol is commonly administered to infants. Propofol clearance is dependent on hepatic blood flow (high extraction coefficient) with subsequent metabolism and glucuronidation. In a population pharmacokinetic analysis, Allegaert and colleagues noted that postmenstrual age and postnatal age were predictive covariants for clearance and that developmental maturation occurs within the first 2 weeks of life ( ) ( Fig. 28.4 ).
Midazolam is an extensively used drug in both full-term and preterm infants and is metabolized by the P450 3A subfamily. Midazolam clearance in preterm infants is less than in children and older infants, and it may reflect the pattern of CYP3A4 ontogeny ( Fig. 28.5 A and B). In a study of 24 preterm infants De Wildt and associates found no relationship between age (postconceptual, gestational, or postnatal) and midazolam clearance ( ). Of note was that in infants exposed postnatally to indomethacin, plasma clearance was higher and the volume of distribution was larger than in those infants not exposed to indomethacin. Because CYP3A4 expression is actuated during the first week after birth regardless of gestational age at birth, the decrease in midazolam clearance probably represents a decrease in CYP3A activity.
Developmental pharmacologic changes influence the requirements for muscle relaxants in infants and older children. Synaptic transmission is slow at birth, the rate at which acetylcholine is released during repetitive stimulation is limited, and neuromuscular reserve is reduced ( Fig. 28.6 ). In addition, the reported sensitivity of infants to the effects of neuromuscular blocking agents has differed depending on whether drug administration was indexed to body weight or to body surface area. Because most neuromuscular blocking agents are distributed in the extracellular space and the extracellular space is related to the body surface area, dosage requirements for neuromuscular blocking agents often correlate with surface area rather than with body weight. Neonates and infants have a higher sensitivity (lower ED 50 and ED 95 ) for most nondepolarizing blocking agents.
Fisher and coworkers studied infants’ sensitivity to nondepolarizing muscle relaxants using the pharmacodynamic and pharmacokinetic properties of d-tubocurarine. These investigators determined the steady-state plasma concentration associated with 50% neuromuscular blockade (CPss50) and noted that infants had a lower CPss50 than older children ( ). Because the volume of distribution of d-tubocurarine in infants is significantly larger than that in older children, the dose (mg per kg of body weight) required to achieve the same degree of neuromuscular blockade appeared the same for infants and older children. Although the pharmacokinetic data reveal similar clearance values for infants and older children, the infant’s larger volume of distribution and consequently longer elimination half-life suggest that infants need less frequent and smaller supplemental doses for continued neuromuscular relaxation. Although these data are specific for d -tubocurarine, the general principles can be extrapolated to other hydrophilic compounds that are primarily distributed to the “central compartment” (i.e., small volume of distribution).
Studies of rocuronium in infants and children have shown that onset time in small infants is faster, and the occurrence of 100% block at lower doses compared with older children suggests a greater potency in infants. In addition, rocuronium has an age-dependent difference in duration of action and in recovery after 0.45 mg/kg and 0.6 mg/kg doses ( Fig. 28.7 ) ( ; ). Historically, medications that inhibit acetylcholinesterase, such as neostigmine or edrophonium, were administered to competitively reverse residual neuromuscular blockade ( ).
In 2015 the U.S. Food and Drug Administration (FDA) approved sugammadex as the first noncompetitive antagonist for the reversal of neuromuscular blockade for clinical use in adults ( ). Sugammadex has a completely different mechanism of action compared with acetylcholinesterase inhibitors; it encapsulates rocuronium or vecuronium, providing rapid and complete recovery of neuromuscular blockade. The reported adverse effect profile of sugammadex has included minor and self-limited issues (nausea, vomiting, hypotension, etc). During a preclinical trial, severe adverse effects such as bradycardia and anaphylactoid reactions were noted ( ). Sugammadex has not received FDA approval for use in children. Published neonatal data on sugammadex use are relatively sparse with three case reports and one open-label trial. However, these studies reflect the overall safety profile of sugammadex in the pediatric population ( ).
Safe and effective intraoperative management of the newborn depends on understanding basic principles of physiology and pharmacology, as well as understanding the technical aspects of monitoring and the anesthesia equipment.
Perioperative hypothermia is a challenge when caring for neonates outside the NICU. In these patients, hypothermia is associated with increased morbidity ( ). Measures must be taken to protect against heat loss during surgery ( Box 28.2 ).
Transport neonate in a heated isolette.
Warm the operating room to more than 27° C (80° F).
Use a warming mattress (water temperature of 40° C [104° F]).
Heat and humidify gases to 36° C (96.8° F) (at the trachea).
Use a radiant heat warmer with a servocontrol mechanism.
Wrap noninvolved areas with plastic.
Warm intravenous fluids and blood.
Warm scrubbing and irrigation solutions.
Monitor the temperature in the operating room.
Attention must be paid to fine, seemingly insignificant details in the care of a sick neonate because the margin of safety is narrow. Although advanced electronic monitoring has contributed significantly to the safety of these babies, the anesthesiologists’ clinical skills, judgment, and evaluation remain indispensable.
Color (e.g., cyanosis and pallor), chest mobility (e.g., bilateral expansion, respiratory pattern, and chest compliance), and palpation (e.g., warmth, pulses, and peripheral perfusion) are often difficult to assess because of the patient’s position and draping on the operating room bed. The use of specific monitors depends on the planned surgical intervention and the underlying disease state.
A precordial stethoscope is a simple and effective means to assess the quality of heart sounds, rate, and rhythm, as well as breath sounds. A change in the intensity of heart sounds indicates a decrease in blood pressure and possibly cardiac output. Depending on the surgical procedure and the method of airway management (mask vs. laryngeal mask airway vs. endotracheal tube), an esophageal stethoscope is an alternative monitor for noninvasive beat-to-beat cardiovascular monitoring. Although the sensitivity of the stethoscope has been discussed, the simplicity and accuracy of the precordial and esophageal devices to monitor heart tones and breath sounds during surgery involving pediatric patients cannot be denied ( ).
The primary role for continuous electrocardiography during anesthesia for the newborn is to detect arrhythmias, especially in the setting of electrolyte disturbance or as evidence of adverse effects of various drugs. Simple monitoring of heart rate is available via the pulse oximeter, now a routine monitor for all patients in the operating room.
In most cases, the blood pressure can be accurately monitored with an automated device based on oscillometry (e.g., Dinamap) or ultrasonic flow (e.g., Arteriosonde) if the appropriate-sized cuff is available. Cuff inflation of the automatic devices should cycle no more frequently than every 3 to 4 minutes to avoid ischemia to the arm ( ). Systolic blood pressure measurements correlate with the circulating blood volume and therefore are essential to monitor and guide fluid and blood replacement. Normal values of blood pressure change with age, gestational age, and anesthetic conditions. See Fig. 5.17 in Chapter 5 , “Cardiovascular Physiology,” and Fig. 21.7 in Chapter 21 , “Induction, Maintenance, and Recovery.” Another alternative system is a Doppler ultrasonic transducer, which has a characteristic sound that decreases in intensity with a decrease in blood pressure.
An indwelling arterial cannula allows repeated blood sampling for cardiopulmonary and biochemical evaluation. A 24-gauge cannula can be inserted percutaneously or by cutdown into a variety of sites, including the radial, dorsalis pedis, or posterior tibial arteries. Percutaneous insertion aided by two-dimensional ultrasound has shown higher first-attempt success rate, fewer overall needle passes, and faster time to cannulation ( ). The axillary artery is rarely cannulated. In general, the umbilical artery can be cannulated in the first 4 to 7 days of life, but this is usually avoided after the first 2 days of life because of the risk for infection and vascular emboli. The umbilical catheter tip can be placed high (T10 to T12) or low, above the bifurcation of the aorta and below the level of the renal arteries (L4, L5). If an umbilical vein catheter is placed for long-term access, the line should be advanced into the inferior vena cava just beneath the right atrium. This is usually 10 to 12 cm in a full-term neonate. The placement of the catheter must be confirmed by a radiograph ( Fig. 28.8 ).
The risk for retinopathy of prematurity (ROP) necessitates meticulous monitoring of oxygen saturation in the neonate, especially the very low-birth-weight (VLBW) infant. Most neonatologists recommend adjusting the inspired oxygen to maintain oxygen saturation between 90% and 95%, depending on the underlying medical status, gestational age, hemoglobin, and postnatal age (i.e., quantity of fetal hemoglobin HgbF) (see Chapter 27 , “Neonatology for Anesthesiologists”). Of importance, if blood is shunting right to left through a PDA, the oxygen saturation measured in the lower extremities or umbilical artery (postductal site) does not reflect the oxygen saturation in the retinal vessels (preductal site). To enable simultaneous monitoring of both preductal and postductal oxygen saturation, two pulse oximeters are placed—one on the right hand (preductal) and one on a lower extremity (postductal). During right-to-left shunting through the PDA, the preductal oxygen saturation is higher than the postductal value, and the difference depends on the amount of shunting. If blood is shunting right to left only via the foramen ovale or other intracardiac sites (e.g., ventriculoseptal defect), the preductal and postductal oxygen saturation are equal. (See Chapter 3 , “Respiratory Physiology,” and Chapter 5 , “Cardiovascular Physiology.”)
An arterial catheter must be connected to a pressure transducer and slowly and continuously infused with a small volume of diluted heparin solution (0.1 to 1 units/mL) at the rate of 0.5 to 1 mL/hr. In the extremely low-birth-weight (ELBW) neonate, the flush volume should be measured and included in calculating the total daily fluid intake. In addition, extreme caution is critical while flushing an arterial cannula, because retrograde embolization into the cerebral circulation is possible in the small neonate, especially with a patent foramen ovale or PDA.
A central venous catheter may be indicated to administer blood, fluid, total parenteral nutrition (TPN), and medications and to monitor central venous pressure (CVP). Using the Seldinger technique, a catheter can be inserted percutaneously into the subclavian, internal jugular, external jugular, or femoral veins. In emergencies or situations of difficult venous access, the umbilical vein catheter can be inserted and passed into the right atrium (see Fig. 28.8 ). Its location must be verified by radiograph or by a CVP tracing. Umbilical vein catheters have been associated with portal vein thrombosis. All central venous catheters are associated with significant morbidity, including thrombosis, emboli, and infection. Central venous catheters in the low-birth-weight (LBW) neonate have additional risks, from malpositioning and from the disruption of venous flow (ratio of the size of the vessel to the size of the catheter is low).
The combination of immature and fragile central nervous and cardiorespiratory systems coupled with unstable chest wall mechanics and variable responses to anesthetic agents are often indications for mechanical ventilatory support both during and after surgery for the newborn. For each patient and for each procedure, the anesthesiologist must evaluate the needs and the requirements for mechanical ventilation. For some neonates, the ventilatory status might be so precarious that using a more advanced ventilator from the NICU intraoperatively might offer additional options for responding to intraoperative events affecting ventilatory status. Of note, if a neonate requires a specific mode of ventilatory support, such as high-frequency oscillation, the operating room ventilatory strategy should be coordinated with the critical care team.
Manual ventilation has been proposed as a technique to allow the anesthesiologist to continuously sense changes in compliance of the chest and airways. However, Spears and colleagues noted that manual ventilation can be extremely unreliable for sensing changes in airway compliance ( ). In addition to monitoring heart sounds, the precordial or esophageal stethoscope is a simple system to monitor ventilation and quality of breath sounds. Peak airway and end-expiratory pressures should also be measured. End-tidal carbon dioxide devices (mass spectrometers or infrared analyzers) are now the “standard of care” to continuously monitor the adequacy of respiratory exchange. These devices provide a breath-to-breath level of carbon dioxide tension, and the waveform of this measurement can provide information about rebreathing, ventilator disconnection, suspected air embolism, and hypermetabolic states (see Chapter 17 , “Equipment”).
The pulse oximeter provides a precise, continuous readout of the hemoglobin oxygen saturation. During the first 1 to 2 weeks of life and without transfusion of autologous blood, the oxygen dissociation curve of HgF is shifted to the left of the adult curve so that hemoglobin saturation of 95% to 97% corresponds to an arterial oxygen tension (Pao 2 ) of 52 to 77 mm Hg, assuming a Pao 2 at 50% hemoglobin saturation (P50) of 19 mm Hg. The hemoglobin saturation should be correlated with an arterial partial pressure of oxygen (Po 2 ) measurement to ensure valid interpretation of oxygen saturation data in the operating room (see Chapter 3 , “Respiratory Physiology”).
Neuromuscular blockade can be monitored with a battery-operated nerve stimulator. The simple twitch and train-of-four are elicited by stimulating the ulnar nerve at the wrist or the posterior tibial nerve at the ankle. The neonate’s neuromuscular response to nerve stimulation allows the anesthesiologist to titrate further doses of a muscle relaxant and avoid excessive neuromuscular blockade. However, neuromuscular monitoring is technically challenging for VLBW neonates because of the small size of their muscles. Accurate data from the transcutaneous devices are often impossible to obtain. Inserting needles into a neonate’s extremity should be justified because such trauma may cause bleeding or infection. Furthermore, even with needles in place, obtaining a reliable response using standard battery-operated devices is unpredictable. Acceleromyography has been described in neonates, and although it may provide more accurate information, it can be technically challenging because of size ( ).
Of significance, most of these neonates require mechanical ventilation postoperatively, so that documenting full recovery to neuromuscular blockade is often unnecessary immediately after surgery.
Devices that collect urine during surgery (specifically, Foley catheters or modifications) are helpful, because in the absence of glycosuria, urine output is a good indicator of hydration, circulating volume, and renal function. The desirable range of urine output in the neonate under anesthesia is 0.5 to 2.0 mL/kg per hour. Note that for a 1-kg neonate, 0.5 to 2 mL of urine per hour is difficult to reliably collect in the setting of surgical drapes, lack of direct access to the patient, and easy kinking of drainage tubing secondary to pressure and positioning. Thus in actual practice, accurately assessing urine output is difficult.
Minimal-access surgery (MAS) refers to a collection of surgical techniques that aim to minimize the morbidity of “conventional” open surgery ( ). In the neonate, MAS typically refers to either laparoscopy or thoracoscopy. Despite recent technologic advancements in MAS, the core components remain unchanged—one must have a scope/video equipment, access to the surgical site (typically via trocar), suitable instruments, and insufflation (using carbon dioxide). Most complex neonatal procedures, including congenital diaphragmatic hernia (CDH) and tracheoesophageal fistula (TEF), are amenable to minimal-access surgical repair because of advancements in endoscopic tools and techniques ( ; ; ). In patients with TEF, the thoracoscopic approach improves the surgeon’s view because the fistula is seen perpendicular to its connection to the membranous trachea; consequently, the surgeon can more easily identify the exact site for ligation ( Fig. 28.9 ) ( ; ). In patients with abdominal surgery, MAS techniques reduce the incidence of postoperative adhesions after laparoscopy ( ).
Even in the neonatal population, MAS has been shown to decrease postoperative pain, length of stay, and surgical site infections ( ; ; ). This is partly because of the smaller incisions. An open incision can experience a relative fivefold greater wound closing tension than incisions used in a MAS approach. The morbidities associated with incision such as pain, scar, infection risk, and dehiscence depend directly on a wound’s closing tension ( ; ; ). In some specific procedures the MAS approach simply avoids potential morbidities associated with the open approach. In TEF repair, avoiding a posterior-lateral thoracotomy may prevent thoracic nerve damage, winged scapula, chest wall asymmetry, scoliosis, and breast deformities ( ; ; ). In spite of these advantages MAS has specific dilemmas and limitations.
For endoscopic procedures, the small working space secondary to the size of the newborn’s chest, coupled with the unique aspects of neonatal physiology, creates specific challenges during single-lung ventilation. The learning curve to efficiently perform minimally invasive surgery in the newborn is striking. In the early learning phase, surgical time may be markedly prolonged compared with those for the open procedures ( ; ; ; ). In addition to longer surgical times, operative morbidity and conversion rates to open procedures vary among institutions and surgeons ( ). Thus efficient and safe endoscopic surgery requires both surgical and biotechnical expertise, as well as highly skilled operating room and anesthesia teams ( ).
Challenges associated with MAS in any size patient are compounded in the neonate. MAS creates a unique set of operative conditions that may impose significant cardiorespiratory effects: positioning, insufflation, prolonged pneumoperitoneum or pneumothorax, and hypercarbia. The insufflation of any substance into the peritoneum or the thorax must be controlled to avoid excessive pressure in the cavity. The simple increase in pressure can dramatically affect hemodynamic and respiratory function. The development of low-flow devices and valved trocars has allowed surgeons to adopt specific approaches for the newborn or small neonate (intrathoracic pressure less than 10 mm Hg and intraabdominal pressure less than 15 mm Hg). CO 2 has become the most commonly used agent for insufflation because of its high solubility, low expense, and noncombustible nature. On the other hand, CO 2 is readily absorbed from both the peritoneum and the thoracic cavity. The cardiorespiratory effects of insufflation with CO 2 during laparoscopy and thoracoscopy have been well documented in children, infants, and neonates.
The physiologic changes associated with pneumoperitoneum can have a significant impact on the neonate. Bannister and colleagues reported on the pulmonary effects of pneumoperitoneum in infants (younger than 10 months of age) who were undergoing a variety of procedures that required at least 30 minutes of laparoscopy but no change in position ( ). Patients were divided into two groups: those weighing less than 5 kg (intraabdominal pressure <12 mm Hg) and those weighing more than 5 kg (intraabdominal pressure <15 mm Hg). Again, peak inspiratory pressure increased (18%) and correlated with insufflation pressure, and dynamic compliance decreased (48%). In at least 40% of patients, tidal volume decreased approximately 33%, and oxygen saturation decreased 2% to 11%. To maintain Petco 2 within 10% of baseline, ventilation was adjusted approximately 20 times. Neither heart rate nor blood pressure changed with insufflation or ventilatory adjustments at pressures of 5 mm Hg, but significant changes from baseline status were noted at insufflation pressures of 10 mm Hg and greater. Similar minimal hemodynamic effects of low-pressure pneumoperitoneum have been documented in young children elsewhere ( ).
Because of unique cerebrovascular physiology and impaired autoregulation of cerebral blood flow, newborns may experience more profound cerebral vascular effects from increased intraabdominal pressure (see Chapter 27 , “Neonatology for Anesthesiologists”). This absence of predictable autoregulation combined with the significant risk for injury secondary to oxygen and positive pressure ventilation imply unique factors for anesthetic management of the newborn during either laparoscopy or thoracoscopy. Although indirect monitoring of cerebral blood flow is available (e.g., near-infrared spectroscopy [NIRS]), a direct, reliable, continuous method to monitor cerebral perfusion pressure would be of benefit in this setting ( ). In fact, during thoracoscopic repair of either a congenital diaphragmatic hernia or a tracheoesophageal fistula, significant absorption of CO 2 was associated with severe hypercapnia ( ) and acidosis. Of importance, at the same time, cerebral hemoglobin oxygen saturation (measured via NIRS) decreased during insufflation ( ).
Although the range of procedures managed by minimally invasive surgery has expanded dramatically in the newborn over the last 2 decades, morbidity in this age group continues to be considerable because of the neonate’s unique cardiorespiratory physiology. Specifically, the transitional circulation may predispose the newborn to exaggerated risks secondary to insufflation pressure and enhanced absorption of carbon dioxide during laparoscopy and thoracoscopy. Kalfa and colleagues noted that hemodynamic, respiratory, and/or thermal stability was disrupted in 12% of newborns during either laparoscopy or thoracoscopy ( ).
Increasing intrathoracic or intraabdominal pressure (e.g., >8 mm Hg) may decrease venous return and therefore cardiac output and blood pressure. In most cases, temporarily interrupting insufflation, limiting the peak pressure, and/or delivering volume expansion promptly alleviates hypotension, with a low incidence of converting to an open technique ( ). In fact, only 20% of patients encounter significant hypotension ( ). Although exceedingly rare, the newborn maintains a higher risk for cardiovascular collapse secondary to paradoxical emboli via the PDA/foramen ovale during laparoscopy ( ; ). Gas entry probably occurs through a transversed umbilical vein by the trocar.
Interfering with ventilation of the lungs during insufflation may induce hypoxia and/or hypercarbia, which can profoundly influence the transitional circulation ( ; ). As expected, these effects are more common during thoracoscopy ( ) and are proportional to the pressure, flow rates, and duration of insufflation. The low quantity of peritoneal fat serves a buffer, since CO 2 is soluble in fat, and the higher permeability of the relatively thin surface allows the CO 2 to be absorbed more efficiently from the peritoneum as a function of decreasing age ( ), exacerbating the direct and indirect effects of insufflation on respiratory function. Beyond the newborn period in hemodynamically stable patients, hypercarbia is not associated with major clinical side effects, and the Pco 2 is usually controlled to acceptable levels (e.g., Pco 2 <45 to 50 torr) by increasing minute ventilation. However, in the setting of major developmental anomalies (e.g., tracheoesophageal atresia or CDH), hypercarbia may cause more hemodynamic changes, especially on the central nervous system. Bishay and associates noted that cerebral oxygenation decreased during insufflation, and it was associated with hypercarbia and acidosis ( ). These authors suggest that this finding should prompt reassessing the risks associated with thoracoscopic repair of these lesions. In addition, increasing peak airway pressure/minute ventilation and fraction of inspired oxygen (Fio 2 ) are associated with a higher risk for long-term morbidity in the immature lung (see Chapter 27 , “Neonatology for Anesthesiologists”).
In some cases, in spite of increased minute ventilation, hypercarbia persists and only returns to baseline levels approximately 15 minutes after desufflation ( ). For unclear reasons, younger patients often demonstrate a period of increased CO 2 elimination after desufflation, which may be related to increased venous return and/or minute ventilation after the decrease in intraabdominal pressure. On the other hand, this postinsufflation phenomenon is not universal, implying a persistent risk for hypercarbia in some patients into the postoperative period.
Of concern, end-tidal CO 2 may less accurately reflect arterial levels during laparoscopy ( ), especially in the presence of cyanotic heart disease ( ; ). That is, the alveolar-arterial difference may be greater in the newborn during insufflation with CO 2 .
Delivering gas into body cavities can decrease core body temperature, but the incidence of hypothermia varies. For example, in one study, 50% of newborns developed a postoperative core temperature less than 36° C, especially in the setting of a prolonged operative time (insufflation time longer than 100 minutes) ( ), but severe hypothermia (<34.5° C) is uncommon (12%). With improved warming techniques and shorter times for insufflation, postoperative hypothermia (<34.9° C) was noted in less than 2% of newborns ( ).
Because many of the strategies to affect single-lung ventilation (e.g., double-lumen endotracheal tubes, endobronchial blockers) are not available for the newborn, adequate surgical exposure for thoracoscopy relies heavily on insufflation to collapse the lung. Thus single-lung ventilation is challenging to achieve and at times is not tolerated by a newborn. Many pediatric surgeons suggest that insufflation of the chest with a low flow of CO 2 (1 to 2 L/min, peak pressure of 4 to 6 mm Hg) in the 30- to 45-degree prone position induces adequate lung collapse ( ; ). Lungs with abnormal distribution of ventilation impede uniform collapse with insufflation, so the newborn with abnormal lung function may not be eligible for thoracoscopic surgery.
In spite of the myriad of critical physiologic challenges, endoscopic procedures have been conducted successfully in high-risk infants with conditions such as hypoplastic left heart syndrome, complex cyanotic heart disease, and patients with single-ventricle physiology ( ; ; ). With increasing expertise among surgeons and anesthesiologists, risk factors are successfully managed with close collaboration and preoperative planning so that MAS has become the standard for complex surgery in the newborn.
Gastroschisis and omphalocele are the most common abdominal wall defects, but they are still rare. The National Birth Defects Prevention Network (NBDPN) examined a 5-year birth cohort (birth years 2012–2016) using data from 30 population-based birth defect surveillance programs in the United States and found the overall prevalence estimates (per 10,000 live births) were 4.3 (95% confidence interval [CI]: 4.1–4.4) for gastroschisis and 2.1 (95% CI: 2.0–2.2) for omphalocele ( ). An ultrasound easily confirms the presence of these lesions in approximately 95% to 100% of cases at the time of the nuchal scan at 10 to 13 weeks of gestation ( ; ). In some cases, an elevated level of α-fetoprotein may initially suggest an in utero abnormality; this marker is elevated more often in gastroschisis than in omphalocele ( Box 28.3 ). According to some reports, the increasing prevalence of gastroschisis has been allocated to younger women ( ; ; ), but others suggest that the phenomenon is not restricted to those younger than 20 years of age ( ). In the cohort of births studied by the NBDPN, mothers of neonates with gastroschisis were more likely to be underweight/normal weight prior to pregnancy. Little evidence points to a significant genetic link. However, in utero exposure to acetaminophen, aspirin, and pseudoephedrine has been associated with an increased incidence of gastroschisis ( ; ). Furthermore, a recent ecologic analysis found a higher prevalence of gastroschisis in areas where opioid prescription rates were high, supporting epidemiologic data suggesting an association between opioid use during pregnancy and gastroschisis ( ).
In contrast to gastroschisis, the prevalence of omphalocele has been stable. Genetic factors have a major role ( ). In fact, the in utero diagnosis should prompt further noninvasive prenatal testing, because this lesion is commonly associated (50% to 80%) with chromosomal abnormalities or other complex syndromes (e.g., cloacal exstrophy, Donnai-Barrow syndrome, pentalogy of Cantrell, Beckwith-Wiedemann syndrome); 30% to 40% of omphaloceles are associated with trisomy 21, 18, or 13. Multiple associated anomalies are identified in 65% to 88% of cases ( ; ; ; ). These anomalies are more common with nongiant omphaloceles (<4 cm) than with giant omphaloceles (55% vs. 36%) ( ). If the omphalocele contains only bowel and if oligohydramnios or polyhydramnios are present, the likelihood of an associated chromosomal abnormality increases. Mann and colleagues noted that omphaloceles that contain liver and other viscera are more likely to have cardiac, renal, or limb anomalies, whereas the smaller omphaloceles are more likely to have gastrointestinal or central nervous system malformations ( ). Syndromes of midline defects often include an omphalocele, and this lesion also has been included in the nonsyndromic multiple congenital anomalies (MCA) complex.
The concept of an “isolated omphalocele” must be considered from the perspective of recent data suggesting that 30% of the fetuses allocated to the category of “isolated cases” are then found to have multiple defects. Only 14% of omphaloceles were truly isolated lesions ( ). Similarly, Porter and colleagues noted that 26% of cases classified prenatally as “isolated” had significant anomalies (e.g., cardiac) identified postnatally. In addition, one-third of these infants were premature, and the mortality rate was 50% ( ). In contrast to omphalocele, only 15% to 20% of neonates with gastroschisis have associated malformations, and only rarely is a complex pattern of malformation identified ( ).
Whether isolated or part of a more complex disorder, an in utero diagnosis of an abdominal wall defect implies delivery at a medical center with resources for high-risk obstetric, surgical, anesthetic, and neonatal care. In general, it is the associated lesions (both extraintestinal and intraintestinal) as opposed to the abdominal defect itself that contributes to short-term and long-term outcomes of patients with gastroschisis or omphalocele. Because prognosis correlates with the severity of associated anomalies, identifying a structural or karyotypic abnormality is critical in predicting outcome and counseling parents.
Although similar in gross physical appearance, omphalocele and gastroschisis are distinct lesions. An omphalocele is a central defect of the umbilical ring; the abdominal contents herniate into the intact umbilical sac, and the bowel remains protected throughout gestation unless the sac ruptures in utero ( Fig. 28.10 ). The umbilical cord inserts into the sac, which contains an internal peritoneal membrane, Wharton jelly, and an external amniotic membrane. By definition, the lesion has a fascial defect of more than 4 cm (<4 cm is considered an umbilical hernia) and often as large as 10 to 12 cm. The sac often contains the stomach, loops of the small and large intestines, and in about 30% to 50% of cases, the liver.
A gastroschisis is a full-thickness abdominal wall defect usually 2 to 5 cm in diameter, almost always to the right of the umbilical cord, with evisceration of bowel (usually only small and large bowel, rarely liver) through the defect, and without a covering membrane ( Fig. 28.11 ). The bowel is exposed to the intrauterine environment with no sac, predisposing the loops to become matted, thickened, and often covered with an inflammatory coating or peel. Whether this exudative peel is secondary to a specific inflammatory pathway or simply an effect of amniotic fluid is unclear. The umbilical cord is normal and separate from the defect. Cryptorchidism coexists when the testes exit along with the bowel ( ; ). Although “simple gastroschisis” is not commonly associated with other anomalies, the presence of a complex intestinal disorder such as an atresia (5% to 15%) or volvulus increases morbidity. In the setting of atresia, Kronfli and colleagues noted that prolonged feeding intolerance, recurrent sepsis, short bowel syndrome/intestinal failure, and mortality were higher ( ).
With either omphalocele or gastroschisis, the rotation of the gut is incomplete in utero. This results in various “malrotation” phenotypes.
Although many theories have been proposed, the embryologic etiology of these lesions is not completely defined ( ). For example, Sadler suggested that the embryologic events of ectopia cordis (5.5 to 7.9/1 million live births), bladder exstrophy (1/40,000 live births), and gastroschisis are closely related ( ). That is, he proposes that abnormal closure of the ventral body wall by the lateral folds during the fourth week of gestation defines the fundamental disorder in the three lesions. This school of thought also hypothesizes that an omphalocele simply represents a failure of the loops of bowel, which normally herniate into the umbilical cord between weeks 6 and 10 of gestation, to reenter the abdominal cavity ( ).
Others have associated ectopia cordis, exstrophy of the bladder, and omphalocele with a common embryologic event. In this model, a “classic omphalocele” results when closure of the lateral folds is interrupted, ectopia cordis results from abnormal closure of the cephalic folds, and bladder exstrophy results from failure of the caudal folds to close ( ). This model is also consistent with syndromes such as pentalogy of Cantrell (cleft sternum, diaphragmatic and pericardial defects, cardiac anomalies, and omphalocele) or omphalocele, exstrophy, imperforate anus, and spinal defects (OEIS).
Single-gene mutations have been identified in patients with omphalocele and multiple anomalies, such as the filamin A, alpha (FLNA) gene in otopalatodigital syndrome. Otopalatodigital syndrome is characterized by abnormalities in the axial and appendicular skeleton along with extraskeletal anomalies such as omphalocele ( ). Pentalogy of Cantrell (omphalocele, diaphragmatic hernia, sternal abnormalities, and ectopic and anomalous heart) includes gene abnormalities at Xq25 to Xq26.1. Thus identifying an omphalocele in utero demands an intense evaluation for associated anomalies that may be part of a recognizable malformation syndrome or chromosomally based disorder.
The mode of delivery for neonates with abdominal wall defects remains controversial, but studies have shown that in gastroschisis and nongiant omphaloceles (i.e., liver extracorporeal), outcome after vaginal delivery is no different than with cesarean section ( ). The controversy centers on delivery with gastroschisis because of the unprotected viscera, but no advantages of cesarean section are consistently apparent when survival and incidence of complications are evaluated ( ; ). Thus intervening with a cesarean section should be based on the obstetric indications.
Preoperative management of abdominal wall defects primarily focuses on fluid resuscitation, minimizing heat loss, treating sepsis, and avoiding direct trauma to the herniated organs. Rather than “rushing” to close the defect, careful assessment of associated defects, establishing a smooth cardiorespiratory transition, and ensuring adequate intravascular volume comprise the critical aspects of preoperative management.
Normothermia should be maintained or achieved by preventing heat loss from the exposed viscera. A bowel bag may be used for this purpose ( ). Of importance, although loss of fluid and heat is greater with gastroschisis, either lesion should be covered with a nonadherent dressing and a plastic cover. Although moist (saline-soaked) material is often recommended, moisture tends to exacerbate heat loss ( ), so this type of dressing should be covered in plastic. Distorting or twisting the sac at the base should be meticulously avoided; lateral positioning should be considered. Decompressing the stomach with an orogastric or nasogastric tube minimizes regurgitation, aspiration pneumonia, and further bowel distension. Broad-spectrum antibiotics and intravenous fluid therapy (e.g., normal saline or lactated Ringer’s solution) three to four times (150 to 300 mL/kg per day) the usual maintenance rate (80 to 100 mL/kg per day) may be needed to provide adequate hydration and to compensate for a combination of peritonitis, edema, ischemia, protein loss, and significant third-space loss. Although urine output may be less on day 1 of life, in general, a urine output of 1 to 2 mL/kg per hour suggests adequate hydration, but should also be guided by clinical and acid-base status. Because large volumes of fluid are often required, acid-base status and electrolyte levels should be monitored serially. Rarely, if severe metabolic acidosis develops in spite of aggressive fluid delivery (e.g., coexisting sepsis), colloids or vasopressors may be required. In general, delivery of crystalloid and colloid provides adequate support.
Surgical management aims to repair the abdominal wall defect and reduce the protruded viscera. If primary closure is not possible, a staged repair is planned, including the use of a silo chimney or a silastic silo prosthesis ( ). The “silo” consists of a silastic or Teflon mesh that is sutured to the fascia of the defect. The synthetic material used to cover the lesion and the specific mechanism for placing the organs into the abdomen (e.g., umbilical tapes or umbilical cord clamps) vary from center to center. After the silo is in place, the extraabdominal organs are gradually returned to the peritoneal cavity over 3 to 10 days. Improved outcome using the delayed-repair approach after nonoperative placement of a spring-loaded silo has gained increased attention ( ). For example, inserting a spring-loaded device is accomplished in the NICU or delivery room without general anesthesia. After the herniated bowel is reduced, the prosthesis is removed, the lesion is reduced under general anesthesia, and eventually the defect is closed. However, complications have been associated with this “simple” device so that advantages over traditional methods may be less than initially hypothesized. In some cases, ischemic injury developed that resulted in a complex hospital course ( ). Although some authors claim that the time to both first and full feedings was shorter in neonates who underwent delayed closure, this outcome seems to be more closely correlated with the ability to achieve primary closure without an increase in intraabdominal pressure, the presence of other intraabdominal lesions (e.g., atresia), and requirement for ventilator support ( ; ).
Forcing the viscera into an underdeveloped abdominal cavity that cannot accommodate the herniated bowel and tight closure of the defect can restrict diaphragmatic excursion, possibly compress the lungs, and may produce abdominal compartment syndrome (ACS). Although gastroschisis may lead to these events, large omphaloceles are more likely to precipitate significantly increased intraabdominal pressure. ACS not only impairs respiratory function, but the high intraabdominal pressure can diminish blood flow to the kidneys, liver, and other viscera; impair venous return; and even increase intracranial pressure via effects on intrathoracic pressure ( ; ). Thus during abdominal closure, the anesthesiologist must monitor airway pressures to identify decreased pulmonary compliance, as well as observe for evidence of decreased perfusion to the lower extremities (evidence of ACS). In some cases, if primary closure is questionable, intraabdominal pressure might be measured directly (intravesical or intragastric). The surgeon and the anesthesiologist should cooperate to assess the feasibility of a primary closure. Yaster and colleagues noted that an increase in intragastric pressure of greater than 20 mm Hg and CVP of more than 4 mm Hg above baseline were often associated with reductions in venous return and cardiac index, requiring surgical decompression of the abdomen ( ). The impact of ACS on hemodynamic function and its treatment with percutaneous drainage of the peritoneal cavity has been described in adults and older children, especially in the setting of trauma, and recently in an LBW neonate with sepsis and an intestinal perforation ( ; ; ; ).
Intraoperatively, an arterial catheter facilitates blood sampling and continuous monitoring of blood pressure. However, a CVP catheter might be equally valuable for evaluating changes in blood volume and the degree of visceral compression during abdominal closure and to allow metabolic monitoring (e.g., serial intraoperative glucose levels in infants with Beckwith-Wiedemann syndrome) ( ). Intravenous fluids often consist of 5% to 10% dextrose in 0.2% saline to deliver the maintenance therapy and normal saline or lactated Ringer’s solution (8 to 15 mL/kg, or more, per hour) for third-space loss. Vigilant efforts at preventing heat loss should be sustained (see Chapter 7 , “Thermoregulation: Physiology and Perioperative Disturbances”).
After decompression of the stomach, anesthesia may be induced with inhalation or intravenous agents ( Box 28.4 ). Because it distends the bowel, nitrous oxide is avoided. In most cases, neuromuscular blockade facilitates abdominal decompression or closure. Ventilation with an air and oxygen mixture with low concentrations of an inhalation anesthetic plus intravenous opioids are titrated in response to the hemodynamic status. In newborns who have received transfusions of adult blood, the inspired oxygen concentration must be adjusted to maintain oxygen saturation between 90% and 95% (see Chapter 27 , “Neonatology for Anesthesiologists”). In neonates who have not received adult hemoglobin transfusions, a P50 of 19 mm Hg and an O 2 saturation between 90% and 97% result in a Pao 2 of 40 to 64 mm Hg. In the preterm neonate, an O 2 saturation of 90% to 95% results in a Pao 2 of 40 to 52 mm Hg (see Chapter 3 , “Respiratory Physiology”).
Adequate hydration
Maintenance of temperature, prevention of heat loss
Evaluation for congenital heart disease and other associated abnormalities (omphalocele > gastroschisis)
Broad-spectrum antibiotics
Measure electrolytes
Rapid-sequence induction
Avoid mask ventilation and abdominal distension
Muscle relaxation (nondepolarizing muscle relaxants)
Inhalational agents as hemodynamically tolerable
Opioids for analgesic
Consider regional anesthesia for intraoperative and postoperative care
Monitors: standard monitors, arterial catheter if hemodynamically unstable, glucometer
Consider central venous catheter for both intraoperative monitoring and postoperative nutrition
Monitor inspiratory pressures during defect closing
Right upper-extremity pulse oximeter (preductal)
Pulse oximetry on lower extremity; used for postductal saturation and perfusion of lower extremity during abdominal closure
Prevent hypothermia; forced air warmer
Mechanical ventilation unless defect small
Regional anesthesia encouraged unless prolonged intubation expected
Escharotic therapy offers an alternative staged closure of an omphalocele that is relevant to neonates with cardiorespiratory dysfunction that prevents surgical intervention. Although other agents have been applied in the past (mercurochrome, povidone-iodine), silver sulfadiazine provides a safer treatment ( ). This intervention may require months to effectively epithelialize the sac. The resulting ventral hernia is electively repaired under general anesthesia when the infant is stable.
Although seldom used, Vane and colleagues demonstrated that spinal anesthesia is an effective modality for the repair of gastroschisis in selected patients ( ).
Unless the lesion is small, postoperatively after an uncomplicated primary closure of an abdominal wall defect, mechanical ventilation is often required for 24 to 48 hours or longer; thereafter, respiratory compliance usually improves dramatically ( ). Clearly, neonates undergoing a gradual reduction after placement of a silo ( Fig. 28.12 ) or similar device usually require mechanical ventilatory support during this process. Neonates with a small defect can sometimes be extubated at the conclusion of surgery. All patients must be carefully monitored for respiratory and infectious complications in an intensive care unit after closure of an abdominal wall defect. Inferior vena caval compression (evident by blueish lower limbs) or bowel ischemia (necrotizing enterocolitis [NE]) can occur as a result of increased abdominal pressure and may require surgical decompression.
The onset of peristalsis after repair of omphalocele or gastroschisis is usually delayed, and the resulting ileus may be prolonged so that TPN is generally required for days to weeks postoperatively ( ). Bowel hypomotility is most common in neonates with gastroschisis, especially those with an atresia or volvulus. In anticipation of this, most neonates with large lesions and/or gastroschisis should have appropriate intravenous access established in the operating room to facilitate early postoperative nutritional support, which is essential for healing and recovery. Prolonged feeding intolerance should precipitate an evaluation for an associated intestinal atresia or other intestinal lesion.
Survival of neonates born with anterior abdominal wall defects has improved dramatically as a result of prenatal diagnosis; improved surgical, anesthetic, and perioperative intensive care; and nutritional support. When the defect exists in isolation from other anomalies or malformations, a 95% to 97% survival rate is expected. Mortality and long-term outcome are related to associated anomalies (e.g., cardiac malfunction or intestinal atresia), complications of treatment (e.g., bowel perforation, NE, sepsis, or short bowel syndrome), and the side effects of intravenous alimentation (e.g., liver failure or sepsis). For example, if only 14% of omphaloceles are “isolated” ( ), the long-term outcome of this group is predominantly a function of the morbidity of associated lesions. On the other hand, for gastroschisis, outcomes tend to be excellent, even in the setting of atresia, if the bowel escapes major injury ( ).
In a single institutional report by Phillips and colleagues, approximately one-third of the patients with gastroschisis and intestinal atresia had significant intestinal dysmotility without short bowel syndrome or obstruction ( ). Although data reflecting outcomes of patients with abdominal wall defects vary, a consistent observation is that infants with a simple defect (i.e., no atresias, no extraintestinal anomalies, and defect smaller than 5 or 6 cm) and no postoperative complications (e.g., obstruction, NE, or short gut syndrome) have better outcomes compared with patients with complex lesions or difficult postoperative courses ( ; ). In patients with isolated lesions (either an omphalocele or gastroschisis), “catch-up” growth occurs over the first 2 years followed by a normal trajectory for subsequent growth and development. Outcomes for patients with a giant omphalocele (i.e., a liver-containing lesion through an abdominal defect wider than 5 cm) are affected by the incidence of long-term respiratory insufficiency that may be related to abnormal chest wall development ( ; ). Although exercise tolerance may be compromised in these patients, many with large abdominal wall defects eventually establish normal lung volumes ( ).
In a follow-up study of 23 neonates born with gastroschisis between 1972 and 1984 (older than 16 years of age at the time of the study), Davies and Stringer reported that 22 of these 23 neonates were in good health and that overall growth was normal ( ). About one-third of the patients with gastroschisis had undergone additional surgery for adhesions, bowel obstruction, or scar revision related to their defect. In a 30-year review of morbidity related to adhesions after neonatal repair of an abdominal wall defect, van Eijck and colleagues reported a 25% incidence of small bowel obstruction in the gastroschisis group and 13% in the patients with an omphalocele ( ). Examining the outcome of all patients, 88% required a laparotomy and 85% occurred in the first year of life. A history of sepsis or wound dehiscence was a factor for predicting the development of a small bowel obstruction. The authors suggest that pediatric surgeons should evaluate the relevance of adhesion prevention techniques and materials at the time of the initial surgical treatment of abdominal wall defects (e.g., component separation technique or hyaluronate-based barrier) ( ). Finally, patients with complicated neonatal courses (e.g., extensive bowel necrosis) resulting in short bowel syndrome may require parenteral nutrition (TPN) for months, years, or a lifetime. Those who are eventually weaned from TPN may experience malabsorption and nutritional challenges. Some are candidates for bowel transplantation.
Of note, in 25% to 60% of patients with anterior wall defects, the absence of an umbilicus was a distressing physical sign, especially during adolescence ( ; ). Studies involving shorter follow-up periods have noted patients with chronic abdominal pain and gastroesophageal reflux ( ). School-aged children may have some issues with cognition, but generally have reassuring outcomes ( ). Lastly, economic analysis of care of infants with gastroschisis emphasized that the cost of care is high and that on average 47 days of hospitalization were needed in order to establish full feedings ( ; ).
With an incidence between 1:2500 and 1:3000 live births, CDH ( ) results when intraabdominal organs extrude into the thoracic cavity secondary to failure of development of the diaphragm early in gestation ( Fig. 28.13 ). With the thorax containing variable quantities of intestine/stomach/liver, this disorder represents much more than an anatomic abnormality of a hole in the diaphragm. That is, in utero, CDH evolves into a disorder of lung development, including pulmonary hypoplasia and abnormal vasculature.
The normal patterns of postnatal transition from intrauterine to extrauterine physiology (the “transitional circulation”) seem to occur in the newborn with CDH, but the process may be protracted ( ; ). This constitutes the primary physiologic basis for delaying surgery for several days. Some have suggested that failure of improvement in the postnatal cardiorespiratory status in the setting of CDH correlates with a degree of pulmonary hypoplasia that is incompatible with life ( ; ). Finally, surfactant deficiency and left ventricular function may contribute to the postnatal clinical dysfunction in the newborn with CDH ( ; ).
CDH may be an isolated lesion, but approximately 40% of cases are associated with other anomalies, including 20% with congenital heart disease ( ). CDH imparts morbidity to a variety of syndromes, such as Beckwith-Wiedemann, CHARGE (coloboma, heart, atresia choanae, retardation, genital, and ear anomalies), Cornelia de Lange, and Denys-Drash syndromes. Trisomies 13, 18, 21, and 45X are the most common aneuploidies associated with CDH ( ). Chromosomal microarray analysis has become a “first-tier diagnostic test” in some institutions ( ). In addition to karyotype abnormalities, copy number variants (e.g., microdeletions, microduplications) have been diagnosed in 3.5% to 13% of isolated CDH patients ( ). In spite of recognizing a genetic basis for a greater number of CDHs, genetic evaluation in the setting of this lesion is usually normal. If the family history is negative, the risk for recurrence is minimal ( ).
The most common defect is posterolateral (Bochdalek hernia), accounting for more than 95% of cases, of which 80% to 87% are left-sided ( ; ). In approximately 2%, CDH is bilateral. Morgagni (anteromedial) and paraesophageal hernias and eventrations define the rarer versions ( Fig. 28.14 ). Of interest, trisomy 21 is the most frequent aneuploidy identified in Morgagni hernia ( ). Of major clinical relevance, CDH is associated with the following:
Varying degrees of bilateral lung hypoplasia
Pulmonary hypertension and arteriolar reactivity
Congenital anomalies (e.g., cardiac, gastrointestinal, genitourinary, skeletal, neural, and trisomic)
Since the 1980s, the overall survival of neonates with CDH has improved dramatically from 40% to 60% in the 1980s to 70% to 80% in subsequent years (1990 to 2000) ( ), with survival of patients who required extracorporeal membrane oxygenation (ECMO) of about 50%. Although some have reported no change in survival rates ( ; ), others have suggested that overall survival is as high as 85% to 95% ( ; ) and 55% to 85% when ECMO is required ( ; ). In summary, rates for mortality continue to vary widely, depending on how the cohort is analyzed.
Institution-based reports that only include data about live-born infants cared for in an NICU may underestimate the total mortality of CDH and the incidence and severity of associated anomalies. Two decades ago, survival rates associated with an isolated CDH approached 80% in those who did not require ECMO ( ; ). In the 1970s, Harrison and colleagues first noted that mortality may be “hidden” when in utero and that early postnatal deaths were not included in calculating mortality associated with CDH ( ). In fact, combining improved prenatal diagnosis with deliberate delivery at medical institutions equipped to provide the appropriate level of obstetric and neonatal care has eliminated some of the “hidden mortality” associated with CDH.
However, the impact of “hidden mortality” persists and should be analyzed when examining reports of increased survival. The Congenital Diaphragmatic Hernia Study Group reviewed outcomes from their international CDH registry (January 1995 to January 2010, 4390 patients). One notable result of this report was that if only surgical patients were included in the analysis (i.e., eliminate preoperative mortality, exclude patients who were not considered surgical candidates, stillbirths, spontaneous/therapeutic abortions), survival was high for those undergoing a minimally invasive approach (98.7%), as well as for those who had an open repair (82.9%). The higher survival among those undergoing minimally invasive surgery implies selection bias for patients with the requirement for less severely deranged cardiorespiratory status for this approach ( ; ).
Stillborn babies with CDH have a 95% incidence of other anomalies. A severe cardiac malformation combined with CDH implies a lethal outcome ( ; ). Various series rank the incidence of associated malformations differently. Stege and colleagues reported that cardiovascular lesions were most common, followed by chromosomal, skeletal, facial dysmorphism (cleft palate, ear deformity), gastrointestinal, and syndromal ( ). CDH is sometimes part of a complex set of anomalies: pentalogy of Cantrell (omphalocele, sternal cleft, ectopia cordis, and an intracardiac defect [ventricular septal defect or diverticulum of the left ventricle]) ( ). CDH has also been associated with Fryns syndrome, Goldenhar syndrome, Brachmann–de Lange syndrome, and Beckwith-Weidemann syndrome ( ; ; ; ).
The improved operative survival rate of CDH stems from the strategy of delaying surgery to ensure stabilization of the transitional circulation, adopting a lung-protective, or “gentle ventilatory,” strategy using small tidal volumes and as low as possible positive end-expiratory pressure (PEEP), and accepting higher Pco 2 (permissive hypercapnia) ( ; ; ). The lung-protective approach aims to avoid barotrauma and volutrauma ( ; ) (see the “Timing of Surgery” section later in the chapter).
CDH outcomes are a function of the underlying pulmonary hypoplasia and pulmonary hypertension. Dillon and colleagues correlated pulmonary artery pressure (measured via echocardiogram) with survival in a cohort of 47 full-term neonates treated with delayed surgery and gentle ventilatory approaches such as high-frequency oscillatory ventilation (HFOV), permissive hypercarbia, nitric oxide (NO), or ECMO. All 23 infants with estimated normal (i.e., less than 50% of systemic) pulmonary artery pressure within the first 3 weeks of life (49% of the study) survived. In contrast, none of the eight patients with persistent systemic (or higher) pulmonary artery pressure survived. Of the remaining 16 patients with pulmonary artery pressure between these two extremes, 12 (75%) of the 16 survived ( ). Similarly, proposed that preoperative pulmonary pressure greater than 90% of systemic pressure predicted an overwhelming risk for mortality. Clearly, the severity of pulmonary hypertension contributes significantly to mortality. Optimal in utero and postnatal strategies for infants with CDH, especially those with pulmonary hypertension, remain incompletely defined ( ).
The overall mortality (23.6%) among those recruited for the DHREAMS (Diaphragmatic Hernia Research and Exploration, Advancing Molecular Science) study ( ) correlates with other data. Morbidity and mortality strongly correlated with degree of pulmonary hypertension. In turn, other markers (e.g., prenatal diagnosis, need for a patch for repair, support with ECMO, nonisolated CDH) that track severity of pulmonary hypertension also predicted survival.
The diaphragm, lungs, and gastrointestinal tract develop simultaneously. The lungs begin as a ventral bud of the foregut. Airway development and branching begin between the fourth and fifth weeks of gestation and progress until the terminal bronchioles are formed by the seventeenth week. The ventral (membranous) component of the diaphragm is formed between the third and fourth weeks of gestation. At about the eighth week of gestation, this portion envelops the esophagus, inferior vena cava, and aorta and fuses with the foregut mesentery to form the posterior and medial (membranous) portions of the diaphragm. The lateral margins of the diaphragm seem to be derived from the muscular components of the body. The pleuroperitoneal canals close when all of the membranous portions of the diaphragm fuse together, and by the ninth week of gestation, diaphragmatic closure is usually complete ( ). If the closure (obliteration) of the pleuroperitoneal canals is delayed beyond the ninth to tenth weeks of development, or if the normal rotation and settling of the midgut occur before the tenth week or before the obliteration of the pleuroperitoneal canals, the midgut (abdominal viscera) herniates into the thoracic (pleural) cavity.
CDH seems to be linked to a disordered formation of the pleuroperitoneal fold—a much earlier event (fourth week of gestation). An abnormal formation of the framework of mesenchyme, which inhibits muscular development of the diaphragm, has been linked to abnormalities in the retinoid signaling pathway. This suggests that vitamin A may play a role in the pathophysiology of CDH ( ; ; ).
Commonly, the herniated abdominal viscera (which includes the midgut but may also include the stomach, parts of the descending colon, the left kidney, and the left lobe of the liver) occupies the left thoracic cavity and interferes with the development of the lungs. In most cases, depending on the severity of the hernia, some degree of pulmonary hypoplasia exists, the severity of which depends (at least in part) on the timing of the herniation and degree of compression during fetal development. The herniation of abdominal contents shifts the mediastinum to the right, compresses the contralateral lung, and, in part, contributes to abnormal development of that lung. Simple compression of the ipsilateral and contralateral lungs fails to explain the severe morphologic derangements in lung development, such as fewer alveoli with thickened walls, smaller alveolar gas-exchange surface area, decreased vasculature with medial hyperplasia, and extension of the muscle layer into intraacinar arterioles ( ).
The structural abnormalities of the pulmonary vasculature correlate with the pathophysiology, mortality, and morbidity of CDH. The low number of airways, the simple arterial branching pattern, the increase in smooth muscle mass at the level of the resistance vessels, and left ventricular abnormalities add to the severity of cardiorespiratory dysfunction produced by the hernia itself ( ). Alveolar development does occur postnatally, but growth at the preacinar level is limited, in that the number of airway generations remains constant after midgestation ( ). Postnatal vascular remodeling provides larger and less muscular arteries, so the pathology present at birth in the setting of CDH has been documented to reverse to some degree ( ). Nonetheless, the degree of pulmonary hypoplasia and hypertension is predictive not only of mortality but also the long-term sequelae in the survivor.
In summary, the anatomic abnormalities in the pulmonary vasculature of CDH imply that persistent pulmonary hypertension will commonly accompany this anomaly. Although small arteries predominantly determine pulmonary vascular resistance (PVR), other factors also influence the pulmonary circulation of the newborn.
The decrease in PVR (approximately 80%, which normally occurs in the first 24 hours of life) and rise in pulmonary blood flow that are essential for the transition from placental circulation to the postnatal pattern are dependent on adequate function of the endothelial cell (see Chapter 3 , “Respiratory Physiology”; Chapter 5 , “Cardiovascular Physiology”; and Chapter 27 , “Neonatology for Anesthesiologists”). Imbalance in the production, release, and circulating levels of vasoconstrictors (leukotrienes C4 and D4, thromboxane A2, platelet-activating factor) and vasodilators [NO, prostacyclin]) seems to be central to the right-to-left shunting observed with pulmonary hypertension associated with CDH ( ; ). Postnatally, various endothelially derived vasoconstrictive peptides are produced in response to inflammation, ischemia, and other stimuli. Multiple receptors, both in the vascular smooth muscle and in the vascular endothelial cell, mediate response to these molecules. In part, the release of these vasoactive agents is also elicited in response to ventilator-induced epithelial and endothelial damage from hyperinflation of the hypoplastic lungs. Active remodeling of the pulmonary vascular bed continues gradually over the first 2 to 4 weeks of life.
Accurately defining prenatal prognostic factors for this lesion deserves continued effort to ensure accurate prenatal counseling and to allow families to prepare for termination of a pregnancy in the setting of a poor prognosis or to anticipate a complicated neonatal course if less severe outcomes are predicted.
The prenatal diagnosis of CDH contributes to survival, because birth at a center with high-level neonatal intensive care and pediatric surgery improves outcome. In fact, prenatal diagnosis has increased from approximately 10% in 1985 ( ) to between 50% and 80% ( ; ). On routine ultrasound, the most common findings include displacement of the heart and fluid-filled bowel in the thorax and, in some cases, herniation of the liver ( ). Ultrasound diagnosis of a large defect (e.g., dilated intrathoracic stomach, herniated left lobe of the liver, or polyhydramnios) suggests a severe lesion.
In addition to establishing a diagnosis and evaluating for associated anomalies, ultrasound and magnetic resonance imaging (MRI) are relevant to predicting severity of the CDH. Lung size and the position of the liver can be evaluated via two-dimensional ultrasound. Both lung size and liver position reflect the risk for pulmonary hypoplasia. Over the last 2 decades, lung-to-head ratio (LHR) has been revised and validated ( ; ; ; ; ; ; ; ). However, techniques for generating these indices vary among institutions ( ).
The basic approach for generating the LHR consists of measuring the lung size contralateral to the CDH via one of several specified techniques (e.g., longest axis, level of four-chamber view) and dividing the value by the circumference of the fetal head. Because head circumference increases fourfold while lung area increases sixteenfold between 12 and 32 weeks’ gestation ( ; ), LHR has been standardized for gestational age by expressing the value as a percentage of the normal (i.e., observed/expected: O/E). Another predictive marker of mortality and morbidity in CDH is the presence or absence of the liver in the thorax. Ruano and colleagues estimated survival at 50% if the liver was herniated into the thorax on ultrasound examination (nonquantitative evaluation) ( ). Applying these algorithms to hundreds of affected fetuses, the antenatal CDH registry noted that a low O/E LHR with herniation of the liver reliably predicted a high risk for mortality and morbidity (e.g., need for ECMO, incidence of chronic lung disease) ( ; ; ) ( Box 28.5 ).
LHR is calculated by dividing fetal lung area (mm 2 ) by fetal head circumference (mm).
LHR >1.35 associated with 100% survival
LHR 1.35–0.6 associated with 61% survival
LHR <0.6 – no survival
Observed to expected LHR (O/E LHR) is calculated by dividing the observed LHR by the expected ratio for gestational age.
The fetal lung area increases sixteenfold compared with fourfold increase in the head circumference between 12 and 32 weeks’ gestation
O/E LHR <25% is considered severe CDH (survival 10% with liver up and 25% with liver down)
O/E LHR <15% with liver up – 100% mortality
Position of liver (or presence of liver herniation)
Liver herniation with LHR <1.0%–60% mortality
Liver in the thorax – 56% survival
LHR, Lung-to-head ratio.
Evaluating lung size as an O/E ratio has expanded to include three-dimensional ultrasound and MRI. Three-dimensional ultrasound measurements generate indices for total fetal or contralateral lung volume and position of the liver and stomach. In fact, MRI is preferred to estimate lung volume and is likely to evolve as the most highly reliable assessment tool for prenatal assessment of CDH ( ). MRI provided improved sensitivity and specificity for predicting survival ( ), in addition to intraobserver reliability ( ). Also, MRI allows a scaled method to specifically calculate the amount of liver herniated into the chest, rather than simply noting that the liver is “in the chest” or “in the abdomen” ( ).
Indices for right-sided diaphragmatic hernia differ from those for left-sided hernia because the liver is herniated into the chest in most cases of right-sided hernias. Moreover, right-sided CDH is more often missed at prenatal ultrasound ( ). The severity and outcome of right-sided CDH remain controversial. Some suggest that outcome is similar to that for the left-sided lesions ( ; ), whereas others note higher mortality in spite of similar lung volume before birth ( ). A graded system to include degree of herniation of the liver in right-sided CDH has not been developed.
Because the fundamental pathophysiology of CDH is pulmonary hypoplasia, various fetal surgical techniques to improve the growth of hypoplastic lungs in utero have been developed ( ). Starting with animal models in the 1980s, Harrison and colleagues gradually perfected several open fetal surgical procedures and introduced the ex utero intrapartum treatment (EXIT) procedure. The procedure was eventually abandoned in favor of a minimally invasive technique using fetoscopy to surgically occlude the trachea (fetal endoluminal tracheal occlusion [FETO]). Currently, FETO remains the only clinically relevant in utero intervention. Fetal lungs secrete approximately 100 mL/kg/day of fluid that normally exits the trachea and mouth to enter the amniotic fluid cavity. By preventing lung fluid from exiting the lung, tracheal occlusion aims to manipulate the normal physiology of fetal lung development by stretching the lung to accelerate growth. At the same time, the increased intrathoracic pressure tends to move the viscera out of the thorax. In a group of fetuses with moderate hypoplasia enrolled in a trial sponsored by the National Institutes of Health (NIH), the tracheal occlusion technique marginally improved survival of the affected fetus. However, the procedure was associated with significant complications, including premature labor and delivery. At the same time, survival of infants with CDH with conventional postnatal management improved ( ; ; ). Similarly, another report found that FETO was associated with an infant survival of 47% in cases with isolated unilateral severe CDH ( ).
In 2009, enrollment for the multicenter randomized Tracheal Occlusion to Accelerate Lung growth trial (TOTAL) was initiated ( ; www.totaltrial.eu ). The protocol targeted fetuses with moderate lung hypoplasia (O/E 25% to 35%, liver either in chest or abdomen; O/E 35% to 45%, liver in chest) and balloon occlusion of the trachea at 30 to 32 weeks’ gestation. The initial data demonstrated improved survival (49.1%) compared with historical controls from the antenatal CDH registry (24.1%). However, there were significant side effects (membrane rupture and preterm delivery rate) ( ). The TOTAL trial closed recruitment of the moderate lung hypoplasia arm in May 2019; however, enrollment continues for subjects with severe lung hypoplasia. In an effort to address the consequences of pulmonary hypertension in CDH, the role of antenatal administration of sildenafil (a selective phosphodiesterase inhibitor type 5) is being addressed in both animal models and human trials ( ; ; ).
Become a Clinical Tree membership for Full access and enjoy Unlimited articles
If you are a member. Log in here