Anesthesia for Obstetrics





Key Points





  • The normal physiologic changes of pregnancy begin in the first trimester, affect all organ systems, and alter pharmacokinetic and pharmacodynamic responses to many drugs commonly used in anesthesia.



  • Maternal-fetal exchange of most drugs and other substances occurs primarily by diffusion. The rate of diffusion and peak levels in the fetus depend on maternal-to-fetal concentration gradients, maternal protein binding, molecular weight of the substance, lipid solubility, and the degree of ionization of that substance.



  • All women in labor are considered to have a full stomach and an increased risk for pulmonary aspiration of gastric contents during induction of anesthesia and aspiration prophylaxis should be considered before all surgical procedures during pregnancy.



  • Uterine blood flow increases progressively during pregnancy from approximately 100 mL/min in the nonpregnant state to between 700 and 900 mL/min (∼10% of cardiac output) at term gestation. Consequently, hemorrhage during pregnancy carries significant morbidity and is a leading cause of maternal death worldwide. Early recognition with timely intervention, optimal team performance, and appropriate blood product transfusions are essential to patient outcomes.



  • Uterine and placental blood flow depend on maternal cardiac output and are directly related to uterine perfusion pressure and inversely related to uterine vascular resistance. Decreased perfusion pressure can result from maternal hypotension secondary to hypovolemia, aortocaval compression, sympathetic blockade, and decreased systemic resistance from either general or neuraxial anesthesia. Prophylactic or therapeutic phenylephrine in boluses or as an infusion reduces the incidence and severity of hypotension from spinal anesthesia for cesarean delivery. In comparison to ephedrine, phenylephrine results in less fetal acidosis.



  • During pregnancy, the maternal oxyhemoglobin dissociation curve shifts to the right with pregnancy while the fetal oxyhemoglobin dissociation curve lies to the left. This facilitates oxygen transfer from maternal to fetal hemoglobin. Fetal O 2 saturation does not exceed 60% even with 100% O 2 delivery to the mother. Maternal Pa CO 2 decreases from 40 mm Hg to approximately 30 mm Hg during the first trimester. This reduction facilitates carbon dioxide transfer across the placenta, which is primarily limited by blood flow and not diffusion.



  • Labor is a continuous process separated into first, second, and third stages. The first stage of labor includes the change of the uterine cervix from a thick closed tube to an opening of approximately 10 cm through which the fetus can be expelled. This stage is further divided into latent and active phases.



  • Neuraxial analgesia is the most reliable and effective method of reducing pain during labor. Adequate analgesia is achieved with blockade of T10 to L1 during the first stage of labor and requires extension to include S2 to S4 during the second stage of labor.



  • Neuraxial analgesia in comparison to unmedicated birth or intravenous opioid analgesia may prolong the second stage of labor but does not increase the risk for cesarean delivery. Epidural analgesia inserted early compared to late in labor does not increase the risk for cesarean delivery or prolong the first stage of labor.



  • Remifentanil patient-controlled analgesia (PCA) may offer superior pain relief and less fetal effects than other intravenous opioid analgesics but its analgesic effects are inferior to epidural labor analgesia and it requires careful maternal oxygenation and ventilation monitoring.



  • Hypertensive disorders of pregnancy complicate 5% to 10% of worldwide pregnancies and can cause maternal and fetal mortality. Patients with preeclampsia are at increased risk for cerebral hemorrhage, pulmonary edema, and coagulopathy. Systolic and diastolic blood pressure higher than 160/110 mm Hg should be treated to prevent intracerebral hemorrhage.



  • Sepsis is a leading cause of maternal morbidity and mortality in the UK and United States. Early identification and treatment has been shown to improve outcomes.





Acknowledgment


The editors and the publisher would like to thank Drs. Pamela Flood and Mark D. Rollins for contributing a chapter on this topic in the prior edition of this book. It has served as the foundation for the current chapter.




Physiologic Changes During Pregnancy and Delivery


During pregnancy and the peripartum period, substantial changes in maternal anatomy and physiology occur secondary to (1) changes in hormone activity, (2) mechanical effects of an enlarging uterus, and (3) increased maternal metabolic demands and biochemical alterations induced by the fetoplacental unit. These changes have a significant impact on anesthetic pharmacology and physiology resulting in unique anesthesia management requirements during pregnancy. Pregnant women with comorbid conditions require even greater anesthetic modifications.


Cardiovascular Changes


Changes in the cardiovascular system occur throughout gestation and include (1) anatomic changes, (2) an increase in intravascular volumes, (3) an increase in cardiac output, (4) a decrease in vascular resistance, and (5) the presence of supine hypotension. Table 62.1 and the following sections detail these changes.



Table 62.1

Changes in the Cardiovascular System During Pregnancy

Data from references Bucklin BA, Fuller AJ. Physiologic Changes of Pregnancy. In: Sures MS, Segal BS, Preston RL, Fernando R, Mason CL, eds. Shnider and Levinson’s Anesthesia for Obstetrics . 5th ed. Philadelphia: Lippincott Williams & Wilkins 2013; Kron J, Conti JB. Arrhythmias in the pregnant patient: current concepts in evaluation and management. J Interv Card Electrophysiol . 2007;19:95–107 ; and Conklin KA. Maternal physiologic adaptations during gestation, labor, and puerperium. Semin Anesth . 1991;10:221–234.



















































Cardiovascular Parameter Value at Term Compared With Nonpregnant Value
Intravascular fluid volume Increased 35%-45%
Plasma volume Increased 45%-55%
Erythrocyte volume Increased 20%-30%
Cardiac output Increased 40%-50%
Stroke volume Increased 25%-30%
Heart rate Increased 15%-25%
Vascular Pressures and Resistances
Systemic vascular resistance Decreased 20%
Pulmonary vascular resistance Decreased 35%
Central venous pressure No change
Pulmonary capillary wedge pressure No change
Femoral venous pressure Increased 15%
Clinical Studies
Electrocardiography Heart rate dependent decrease in PR and QT intervals
Small QRS axis shift to right (first TM) or left (third TM)
ST depression (1 mm) in left precordial and limb leads
Isoelectric T-waves in left precordial and limb leads
Small Q-wave and inverted T-wave in lead III
Echocardiography Heart is displaced anteriorly and leftward
Right-sided chambers increase in size by 20%
Left-sided chambers increase in size by 10%-12%
Left ventricular eccentric hypertrophy
Ejection fraction increases
Mitral, tricuspid, and pulmonic valve annuli increase
Aortic annulus not dilated
Tricuspid and pulmonic valve regurgitation common
Occasional mitral regurgitation (27%)
Small insignificant pericardial effusions may be present

TM, Trimester.


Physical Examination and Cardiac Studies


The cardiovascular changes of a normal pregnancy are significant. In cardiac auscultation an accentuated first heart sound (S 1 ) can be heard, with an increased splitting noted from dissociated closure of the tricuspid and mitral valves. A third heart sound (S 3 ) is often heard in the final trimester, and a fourth heart sound (S 4 ) can also be heard in some pregnant patients as a result of increased volume and turbulent flow. Neither the S 3 nor S 4 heart sounds have clinical significance. In addition, a benign grade 2/6 systolic ejection murmur is typically heard over the left sternal border and is secondary to mild regurgitation at the tricuspid valve from the annular dilation associated with the increased cardiac volume. Table 62.1 details the effects of pregnancy on the electrocardiogram and echocardiography. The elevation of the diaphragm by the growing uterus shifts the heart anteriorly and to the left. Left axis deviation as well as left ventricular hypertrophy are common findings in normal pregnancy. Women who present with chest pain, syncope, high-grade flow murmurs, arrhythmias, or heart failure symptoms such as hypoxia or clinically significant shortness of breath should undergo appropriate diagnostic investigation and referral.


Intravascular Volume


Maternal intravascular fluid volume begins to increase in the first trimester secondary to changes in the renin-angiotensin-aldosterone system promoting sodium absorption and water retention. These changes are likely induced by rising progesterone from the gestational sac. Plasma protein concentrations accordingly decrease with a 25% decrease in albumin and 10% decrease in total protein at term compared with nonpregnant levels. Consequently, colloid osmotic pressure decreases from 27 to 22 mm Hg over the time of gestation. At term, the plasma volume is 50% to 55% above the nonpregnant level. It is thought that the increase in blood volume prepares the parturient for delivery blood loss. Blood volume returns to prepregnancy values approximately 6 to 9 weeks postpartum.


Cardiac Output


By the end of the first trimester, maternal cardiac output typically increases approximately 35% to 40% above prepregnancy values and continues to increase 40% to 50% by the end of the second trimester. Cardiac output remains stable throughout the third trimester. This increased cardiac output is secondary to increases in both stroke volume (25%-30%) and heart rate (15%-25%). Labor further increases cardiac output, which fluctuates with each uterine contraction. Increases above prelabor values of 10% to 25% occur during the first stage and 40% in the second stage. The largest increase in cardiac output occurs immediately after delivery, when cardiac output can increase by 80% to 100% more than prelabor values. This abrupt increase is secondary to the autotransfusion of uteroplacental blood as the evacuated uterus contracts, reduced maternal vascular capacitance from loss of the intervillous space, and diminished lower extremity venous pressure from release of the aortocaval compression. This large fluctuation in cardiac output presents a unique postpartum risk for patients with cardiac disease, especially those whose heart cannot accommodate an increase in cardiac output such as those with fixed valvular stenosis or pulmonary vascular hypertension. Cardiac output returns toward prelabor values within 24 hours postpartum depending on the mode of delivery and degree of blood loss. Cardiac output decreases substantially toward prepregnant values by 2 weeks postpartum, with complete return to nonpregnant levels between 12 and 24 weeks after delivery.


Systemic Vascular Resistance


Although cardiac output and plasma volume increase, systemic blood pressure decreases in an uncomplicated pregnancy secondary to a reduction in systemic vascular resistance. Systemic vascular resistance decreases as a result of the vasodilatory effects of progesterone and prostaglandins as well as the low resistance of the uteroplacental vascular bed. Although affected by positioning and parity, systolic, diastolic, and mean blood pressure may all decrease 5% to 20% by 20 weeks gestational age and then gradually increase toward nonpregnant values by term. Diastolic arterial blood pressure decreases more than systolic arterial blood pressure resulting in a slight increase in pulse pressure. Central venous and pulmonary capillary wedge pressures do not change during pregnancy, despite the increased plasma volume, because venous capacitance increases.


Aortocaval Compression


Aortocaval compression by the gravid uterus as a result of supine positioning is associated with a decrease in systemic blood pressure. Although the inferior vena cava is compressed in nearly all term parturients, supine hypotension syndrome (also known as aortocaval compression syndrome) is experienced by only 8% to 10% of women. Supine hypotension syndrome is defined as a decrease in mean arterial pressure of more than 15 mm Hg with an increase in heart rate of more than 20 beats/min and is often associated with diaphoresis, nausea, vomiting, and changes in mentation. At term, the inferior vena cava can be almost completely occluded in the supine position with the return of blood from the lower extremities through the epidural, azygos, and vertebral veins that become engorged ( Fig. 62.1 A ). Also, significant aortoiliac artery compression occurs in 15% to 20% of pregnant women. Inferior vena caval compression in the supine position causes a decrease in both stroke volume and cardiac output of 10% to 20% in comparison to the upright position (see Fig. 62.1 B ), and may exacerbate venous stasis in the legs and thereby result in ankle edema, varices, and increased risk for lower extremity deep venous thrombosis.




Fig. 62.1


Aortocaval compression.

(A) Cross-sectional views of aortocaval compression from the gravid uterus in the supine position with loss of compression in the lateral position. (B) Alterations in heart rate, stroke volume, and cardiac output for both supine and lateral positioning with increasing gestation of pregnancy. IVC, Inferior vena cava.

Reprinted with permission from Bonica JJ, ed. Obstetric Analgesia and Anesthesia . Amsterdam: World Federation of Societies of Anaesthesiologists; 1980.


Most pregnant women have compensatory adaptations that reduce supine hypotension symptoms despite aortocaval compression. One compensatory response is a reflexive increase in peripheral sympathetic nervous system activity. This increase in sympathetic activity results in increased systemic vascular resistance and permits arterial blood pressure to be maintained despite the reduced cardiac output. Consequently, the reduced sympathetic tone from neuraxial or general anesthetic techniques impairs the compensatory increase in vascular resistance and exacerbates the impact of hypotension from supine positioning.


Therefore in general, supine positioning is avoided during use of neuraxial techniques for labor analgesia and cesarean deliveries. Reducing the compression of the inferior vena cava and abdominal aorta with left tilt may mitigate the degree of hypotension and help maintain uterine and fetal blood flow. This is accomplished by positioning the patient laterally or by elevating the right hip 10 to 15 cm (with a historical goal of 15 degree left-tilt) with a blanket, wedge, or table tilt.


The practice of left uterine displacement has been challenged recently. In a magnetic resonance imaging (MRI) study of healthy pregnant volunteers, the volume of the inferior vena cava did not differ significantly between the supine position and the 15 degree left-tilt position but when the patients were tilted to the 30 degree left-tilt position, the inferior vena cava volume did increase. Additionally, healthy women undergoing elective cesarean delivery under spinal anesthesia and a phenylephrine infusion were randomized to supine or 15 degree left-tilt position and no difference was found on neonatal acid-base status; however, the supine patients had lower cardiac output and required more phenylephrine. Further studies are needed to investigate who benefits from left uterine displacement and the amount required to achieve the greatest benefit without hindering the surgical procedure. In the meantime, left uterine displacement should continue to be utilized during induction of neuraxial analgesia/anesthesia and during episodes of maternal hypotension or fetal compromise.


Respiratory System Changes


Pregnancy results in significant alterations in (1) the upper airway, (2) lung volumes and ventilation, and (3) O 2 consumption and metabolic rate ( Table 62.2 ).



Table 62.2

Changes in the Respiratory System at Term

Data from Conklin KA. Maternal physiologic adaptations during gestation, labor, and puerperium. Semin Anesth . 1991;10:221–234.































































Pulmonary Parameter Value Near Term Compared With Nonpregnant Value
Minute ventilation Increased 45%-50%
Respiratory rate
Tidal volume
Increased 0%-15%
Increased 40%-45%
Lung Volumes
Inspiratory reserve volume Increased 0%-5%
Tidal volume Increased 40%-45%
Expiratory reserve volume Decreased 20%-25%
Residual volume Decreased 15%-20%
Lung Capacities
Vital capacity No change
Inspiratory capacity Increased 5%-15%
Functional residual capacity Decreased 20%
Total lung capacity Decreased 0%-5%
Oxygen Consumption
Term Increased 20%-35%
Labor (first stage) Increased 40% above prelabor value
Labor (second stage) Increased 75% above prelabor value
Respiratory Measures
FEV 1 No change
FEV 1 /FVC No change
Closing capacity No change


The Upper Airway


Capillary engorgement with increased tissue friability and edema of the mucosal lining of the oropharynx, larynx, and trachea begins early in the first trimester. As a result, an increased risk for bleeding exists during manipulation of the upper airway, in addition to an increased risk of difficult mask ventilation and intubation of the trachea. Suctioning of the airway and placement of devices should be performed gently to prevent bleeding and nasal instrumentation should be avoided. Furthermore, there is increased risk for airway obstruction during mask ventilation and both laryngoscopy and tracheal intubation are more difficult. Also, after extubation, the airway may be compromised as a result of edema, with subsequent risk for airway obstruction in the immediate recovery period.


Consequently, attempts at laryngoscopy should be minimized and experts recommend a cuffed endotracheal tube with a smaller diameter (6.0-7.0 mm internal diameter) should be placed to minimize the chances of difficult placement secondary to airway edema. Airway edema can be more severe in patients with coexisting preeclampsia, in upper respiratory tract infections, and after active pushing as a result of associated increased venous pressure. In addition, pregnancy-associated weight gain and increase in breast tissue, particularly in women of short stature or with coexisting obesity, can make insertion of a laryngoscope difficult. A patient’s position should always be optimized and back-up airway instrumentation available before attempts are made at intubation of the trachea. The Obstetric Anaesthetists’ Association and Difficult Airway Society guidelines for the management of difficult and failed intubation in obstetrics recommends a videolaryngoscope should be immediately available for all obstetric general anesthetics.


Ventilation and Oxygenation


The increased O 2 demand and carbon dioxide production of the growing placenta and fetus cause minute ventilation to be elevated 45% to 50% more than nonpregnant values in the first trimester and for the remainder of the pregnancy. This larger minute ventilation is attained primarily as a result of a larger tidal volume and a slight increase in respiratory frequency. Maternal Pa CO 2 decreases from 40 mm Hg to approximately 30 mm Hg during the first trimester as a reflection of the increased minute ventilation. Arterial pH, however, remains only mildly alkalotic (typically 7.42-7.44) because of metabolic compensation with increased renal excretion of bicarbonate ions (HCO 3 is typically 20 or 21 mEq/L at term). Early in gestation, maternal room air PaO 2 is more than 100 mm Hg because of the presence of hyperventilation and the associated decrease in alveolar CO 2 . Later, PaO 2 becomes normal or even slightly decreased in the supine position, most likely reflecting small airway closure with normal tidal volume ventilation and intrapulmonary shunt. Arterial oxygenation can be significantly improved by moving the patient from the supine to the lateral position. With pregnancy, the maternal oxyhemoglobin dissociation curve shifts to the right, with the P50 (partial pressure of O 2 at which hemoglobin is 50% saturated with oxygen) increasing from 27 to approximately 30 mm Hg at term. The higher P50 in the mother and lower P50 in the fetus means that the fetal blood has higher affinity for O 2 and offloading of O 2 across the placenta is facilitated. A comparison of arterial blood gas measurements in pregnant versus nonpregnant patients is summarized in Table 62.3 .



Table 62.3

Arterial Blood Gas Measurements in Pregnancy




























Blood Gas Values Pregnant Nonpregnant
PaCO2 30 40
PaO2 103 100
HCO3 20 24
pH 7.44 7.4
P50 30 27


At term, O 2 consumption is increased by 20% to 35%. During the first stage of labor, O 2 consumption increases above prelabor values by 40% and during the second stage it is increased by 75%. The pain of labor can result in severe hyperventilation causing Pa CO 2 to occasionally decrease below 20 mm Hg.


Lung Volumes


During pregnancy, tidal volume increases 20% during the first trimester and increases up to 45% above nonpregnant values at term. The expanding uterus forces the diaphragm cephalad and creates a 20% decrease in functional residual capacity (FRC) by term (see Table 62.2 ). This reduction is comprised of nearly equal reductions in both the expiratory reserve volume (ERV) and residual volume (RV). However, closing capacity (CC) remains unchanged and creates a reduced FRC/CC ratio. This results in more rapid small airway closure with reduced lung volumes, and in the supine position FRC can be less than CC for many small airways, giving rise to atelectasis. Vital capacity does not change with pregnancy. The combination of increased minute ventilation and decreased FRC results in a more rapid rate at which changes in the alveolar concentration of inhaled anesthetics can be achieved. Spirometric measurements of bronchial flow are unchanged in pregnancy.


During induction of general anesthesia, desaturation and subsequent hypoxemia occur more rapidly than in a nonpregnant patient because of decreased O 2 reserve (secondary to decreased FRC) combined with increased O 2 uptake (resulting from increased metabolic rate). Preoxygenation before general anesthesia is critical for patient safety to mitigate these physiologic changes and increase apnea time. Preoxygenation with inhalation of 100% O 2 with a goal of end-tidal oxygen fraction greater than 0.9 is recommended (can usually be obtained with 2-3 minutes of preoxygenation before induction of anesthesia) (see Chapter 44 ). Although the use of high-flow humidified nasal oxygen has been shown to be as effective as conventional preoxygenation in nonpregnant patients, it has not been shown to achieve acceptable preoxygenation levels in term pregnant women. The increased airway edema makes both ventilation and tracheal intubation more difficult and further increases the potential for complications and morbidity of general anesthesia in pregnancy.


Gastrointestinal Changes


After midgestation, the induction of general anesthesia places pregnant women at increased risk for regurgitation, aspiration of gastric contents, and development of acid pneumonitis. The stomach and pylorus are moved cephalad by the gravid uterus, which repositions the intraabdominal portion of the esophagus intrathoracically and decreases the competence of the lower esophageal sphincter muscle. Higher progesterone and estrogen levels of pregnancy further reduce lower esophageal sphincter tone. Gastrin, secreted by the placenta, increases gastric hydrogen ion secretion and lowers the gastric pH in pregnant women. These changes in combination with the increased gastric pressure from the enlarged uterus increase the risk for acid reflux in pregnancy. Maternal gastric reflux with subsequent esophagitis (heartburn) is common in pregnant women and increases with increased gestational age. Gastric emptying is not prolonged in pregnancy. Conversely, gastric emptying is decreased with the onset of labor, pain, anxiety, or administration of opioids. Increased gastric contents can further increase the risk for aspiration. Epidural analgesia using local anesthetics alone does not further delay gastric emptying; however, epidural boluses of fentanyl can cause gastric emptying delay.


All women in labor are considered to have a full stomach and an increased risk for pulmonary aspiration of gastric contents during induction of anesthesia. To reduce this risk, a nonparticulate antacid, a rapid sequence induction of anesthesia technique including cricoid pressure, and endotracheal intubation are considered routine parts of general anesthesia in a pregnant woman past midgestation.


Hepatic and Biliary Changes


Blood flow to the liver does not change significantly with pregnancy. The markers of liver function, including aspartate aminotransferase (AST), alanine aminotransferase (ALT), and bilirubin, increase to the upper limits of normal with pregnancy. Alkaline phosphatase levels more than double secondary to placental production. Plasma protein concentrations are reduced during pregnancy, and the decreased serum albumin levels can result in elevated free blood levels of highly protein-bound drugs. Plasma cholinesterase (pseudocholinesterase) activity is decreased approximately 25% to 30% from the 10th week of gestation up to 6 weeks postpartum. Although neuromuscular transmission should be analyzed before extubation, the clinical consequence of the reduced cholinesterase activity is unlikely to be associated with marked prolongation of the neuromuscular block resulting from succinylcholine. The risk for gallbladder disease is increased during pregnancy with incomplete gallbladder emptying and changes in bile composition. Acute cholecystitis is the second most common cause of acute abdomen in pregnancy and occurs between 1 in 1600 to 1 in 10,000 pregnancies.


Renal Changes


Renal blood flow and the glomerular filtration rate (GFR) increase during pregnancy. Renal blood flow rises 60% to 80% by midpregnancy and in the third trimester is 50% greater than nonpregnant values. GFR is increased 50% above baseline by the third month of pregnancy and remains elevated until 3 months postpartum. Therefore the clearance of creatinine, urea, and uric acid are increased in pregnancy, and the upper laboratory limits for blood urea nitrogen and serum creatinine concentrations are decreased approximately 50% in pregnant women. Levels of urine protein and glucose are commonly increased as a result of decreased renal tubular resorption capacity. The upper limits of normal in pregnancy in a 24-hour urine collection are 300 mg protein.


Hematologic Changes


As previously discussed, blood volume increases during pregnancy. At term, the plasma volume has increased approximately 50% above prepregnancy values and the red cell volume has increased only approximately 25%. The greater increase in plasma volume creates a physiologic anemia of pregnancy with a hemoglobin value normally around 11.6 g/dL. Hemoglobin values less than this at any time during pregnancy are concerning for anemia. Overall oxygen delivery is not reduced by the normal physiologic anemia of pregnancy because of the subsequent increase in cardiac output. The additional intravascular fluid volume of approximately 1000 to 1500 mL at term helps compensate for the estimated blood loss of 300 to 500 mL typically associated with vaginal delivery and the estimated blood loss of 800 to 1000 mL that accompanies a standard cesarean delivery. After delivery, contraction of the evacuated uterus creates an autotransfusion of blood often in excess of 500 mL that offsets the blood loss from delivery.


Leukocytosis is common in pregnancy and is unrelated to infection. Leukocytosis is defined as a white blood cell (WBC) count greater than 10,000 WBCs/mm 3 of blood. In pregnancy, the normal range can extend to 13,000 WBCs/mm3. WBC count may rise in labor with the degree of increase related to the duration of elapsed labor. The WBC count may decrease over the first week postpartum but may take weeks or months to return to nonpregnant values.


Coagulation


Pregnancy is characterized by a hypercoagulable state with a marked increase in factor I (fibrinogen) and factor VII and lesser increases in other coagulation factors ( Table 62.4 ). Factors XI and XIII are decreased, and factors II and V typically remain unchanged. Antithrombin III and protein S are decreased during pregnancy and protein C levels remain unchanged. These changes result in an approximately 20% decrease in prothrombin time (PT) and partial thromboplastin time (PTT) in normal pregnancy. Platelet count may remain normal or slightly decreased (10%) at term as a result of dilution. However, 8% of otherwise healthy women have a platelet count less than 150,000/mm 3 . In the absence of other hematologic abnormalities, the cause is usually gestational thrombocytopenia, from which the platelet count does not usually decrease to less than 70,000/mm 3 . This syndrome is not associated with abnormal bleeding. Gestational thrombocytopenia is due to a combination of hemodilution and more rapid platelet turnover and is a diagnosis of exclusion. Other more consequential diagnoses such as idiopathic thrombocytopenic purpura and hemolysis, elevated liver enzyme, and low platelet count (HELLP) syndrome must be excluded (see section on maternal comorbidities, coagulopathies).



Table 62.4

Changes in Coagulation System at Term




























Pro-Coagulant Factors
Increased I, VII, VIII, IX, X, XII von Willebrand factor
Decreased XI, XIII
Unchanged II, V
Anti-Coagulant Factors
Increased None
Decreased Antithrombin III, Protein S
Unchanged Protein C
Platelets Decreased 0%-10%


Thromboelastography (TEG) is a hemostatic assay that measures the kinetics of clot formation and breakdown. It can provide information about clotting variables, including platelet function as well as the function of other coagulation factors (see also Chapter 50 ). At term gestation, TEG analysis reflects a hypercoagulable state with decreased time to start of clot formation (R), decreased time to specified clot strength (K), increased rate of clot formation (α), and increased clot strength (MA). Although the timing and degree of change in TEG analysis varies with each parameter, many of the changes begin to occur within the first trimester.


Neurologic Changes


Pregnant patients are considered more sensitive to both inhaled and local anesthetics. They have a reduced minimum alveolar concentration (MAC) for inhaled anesthetics. The MAC of a volatile anesthetic is reduced by 40% in animals and 28% in humans during the first trimester of pregnancy. However, it appears the decrease in MAC (i.e., immobility in response to volatile anesthetics among 50% of patients) occurs at the level of the spinal cord based on an electroencephalographic study suggesting that anesthetic effects of sevoflurane on the brain are similar in the pregnant and nonpregnant state. The underlying mechanism of reduced MAC in pregnancy remains unclear; it is likely multifactorial, and many postulate progesterone may have a role.


Pregnant women are more sensitive to local anesthetics and neuraxial anesthetic requirements are decreased by 40% by term. At term, the epidural veins are distended and the volume of epidural fat increases, which decreases the size of the epidural space and volume of cerebrospinal fluid (CSF) in the subarachnoid space. Although the decreased volume of these spaces facilitates the spread of local anesthetics, the local anesthetic dose requirement is decreased for neuraxial block as early as the first trimester, before significant aortocaval compression or other mechanical- or pressure-related changes occur. Consequently, the increased nerve sensitivity and decrease in local anesthetic dose requirements are likely biochemical in origin.




Uteroplacental Physiology


The placenta is a remarkable organ that undergoes vast changes from a fertilized ovum’s initial implantation into the uterine wall until birth. The placenta is composed of both maternal and fetal tissues and is the interface of maternal and fetal circulation systems. It provides a substrate for physiologic exchange between the two systems. The placenta is made up of a basal and a chorionic plate that are separated by the intervillous space. Maternal blood is delivered to the placenta by the uterine arteries and enters the intervillous space via the spiral arteries. It travels toward the chorionic plate, passing fetal villi where exchange takes place, and then drains back to veins in the basal plate and then ultimately away from the uterus via the uterine veins. The fetal blood arrives at the placenta via two umbilical arteries that form umbilical capillaries that cross the chorionic villi. After placental exchange, oxygen-rich, nutrient-rich, and waste-free blood is returned from the placenta to the fetus through a single umbilical vein.


Uterine Blood Flow


An understanding of uteroplacental blood flow is critical for appropriate clinical care. Uterine blood flow increases progressively during pregnancy from about 100 mL/min in the nonpregnant state to 700 to 900 mL/min (∼10% of cardiac output) at term gestation. Approximately 80% of the uterine blood flow perfuses the intervillous space (placenta) and the remainder perfuses the myometrium. Uterine blood flow has minimal autoregulation, and the vasculature remains essentially fully dilated during pregnancy. Uterine and placental blood flow depend upon maternal cardiac output and are directly related to uterine perfusion pressure and inversely related to uterine vascular resistance. Decreased perfusion pressure can result from maternal hypotension secondary to multiple causes including hypovolemia from blood loss or dehydration, decreased systemic resistance from general or neuraxial anesthesia, or aortocaval compression. Increased uterine venous pressure from aortocaval compression, frequent or prolonged uterine contractions, or prolonged abdominal musculature contraction with bearing down (Valsalva) during second-stage pushing can decrease uterine perfusion. Additionally, extreme hypocapnia (Pa CO 2 < 20 mm Hg) occasionally associated with hyperventilation secondary to severe labor pain can reduce uterine blood flow with resultant fetal hypoxemia and acidosis. Neuraxial blockade does not alter uterine blood flow as long as maternal hypotension is avoided but decreases in maternal blood pressure during neuraxial or general anesthesia should be immediately corrected.


Endogenous maternal catecholamines and exogenous vasopressors may cause increasing uterine arterial resistance and decreasing uterine blood flow depending on the class and amount given. In a pregnant ewe model, use of α-adrenergic vasopressors—methoxamine and metaraminol—increased uterine vascular resistance and decreased uterine blood flow, whereas administration of ephedrine did not reduce uterine blood flow despite drug-induced increases in maternal arterial blood pressure. As a result, ephedrine was previously considered the vasopressor of choice for the treatment of hypotension caused by the administration of neuraxial anesthesia to pregnant women. In complete contrast, more recent human trials demonstrate the use of phenylephrine (α-adrenergic agonist) for prophylaxis or treatment of neuraxial-induced hypotension is not only effective in preventing hypotension, but also is associated with less fetal acidosis and base deficit than the use of ephedrine. Other methods to reduce maternal hypotension with induction of regional or general anesthesia are discussed in the section on anesthesia for cesarean delivery.


Placental Exchange


Oxygen Transfer


The delivery of O 2 from the mother to the fetus depends on a variety of factors, including the ratio of maternal to fetal placental blood flow, the O 2 partial pressure gradient between the two circulations, the diffusion capacity of the placenta, the respective maternal and fetal hemoglobin concentrations and O 2 affinities, and the acid-base status of the fetal and maternal blood (Bohr effect). O 2 delivery to the fetus is facilitated primarily because the fetal oxyhemoglobin dissociation curve is to the left (greater O 2 affinity) of the maternal oxyhemoglobin dissociation curve (decreased O 2 affinity). Fetal hemoglobin has a higher O 2 affinity and lower partial pressure at which it is 50% saturated (P50: 18 mm Hg) compared to maternal hemoglobin (P50: 27 mm Hg). Fetal PaO 2 is normally 40 mm Hg and never more than 60 mm Hg, even if the mother is breathing 100% O 2 . Animal studies note that in the face of decreased O 2 delivery, fetal O 2 consumption can be maintained with increased O 2 extraction until the maternal O 2 delivery is approximately 50% of its normal state. CO 2 easily crosses the placenta and its transfer from the fetus to the mother is limited by blood flow and not diffusion.


Drug Transfer


Maternal-fetal exchange across the placenta occurs by one of four mechanisms: passive diffusion, facilitated diffusion, transporter-mediated mechanisms, and vesicular transport. Most drugs have molecular weights less than 1000 Daltons and, therefore, cross the placenta by diffusion if the drug is not ionized. The rate of diffusion and peak levels in the fetus depend on maternal-to-fetal concentration gradients, maternal protein binding, molecular weight, lipid solubility, and the degree of drug ionization. The maternal blood concentration of a drug is typically the primary determinant of how much drug will ultimately reach the fetus. Nondepolarizing neuromuscular blocking drugs are ionized, have a high molecular weight, and poor lipid solubility resulting in minimal placental transfer. Succinylcholine has a low molecular weight but is highly ionized and therefore does not readily cross the placenta unless given in large nonclinical doses. Thus during administration of a general anesthetic for cesarean delivery, the fetus or neonate is not paralyzed. Both heparin and glycopyrrolate have minimal placental transfer because they are highly charged. In contrast, placental transfer of volatile anesthetics, benzodiazepines, local anesthetics, and opioids is facilitated by the relatively low molecular weights of these drugs. Dexmedetomidine may cross the placental barrier but is stored within the placenta and transfer to the fetus is reduced. As a general consideration, drugs that readily cross the blood-brain barrier also readily cross the placenta. Therefore most centrally acting general anesthetics cross the placenta and affect the fetus. There is a paucity of evidence on the placental transfer of newer drugs such as liposomal bupivacaine and sugammadex at this time.


Fetal blood is more acidic than maternal blood, and the lower pH creates an environment in which weakly basic drugs, such as local anesthetics and opioids, cross the placenta as nonionized molecules and become ionized in the fetal circulation. Because this newly ionized molecule has more resistance to diffusion back across the placenta, the drug may accumulate in the fetal circulation and reach levels higher than the maternal blood. This process is referred to as “ion trapping.” During fetal distress (fetal acidemia), higher concentrations of these weakly basic drugs can be trapped. High concentrations of local anesthetics in the fetal circulation decrease neonatal neuromuscular tone. Extremely high levels, such as those associated with unintended maternal intravascular local anesthetic injection, result in a variety of fetal effects, including bradycardia, ventricular arrhythmias, acidosis, and severe cardiac depression. Placental transfer and fetal uptake of specific analgesic and anesthetic drugs are detailed later in the sections that discuss methods of labor analgesia and anesthesia for cesarean delivery.


Fetal Circulation and Physiology


Fetal blood volume increases throughout gestation. Approximately onethird of the fetal-placental blood volume is contained within the placenta. During the second and third trimester, the fetal blood volume is estimated to be approximately 120 to 160 mL/kg of fetal body weight. Thus the total blood volume of a term normal fetus is approximately 0.5 L. Although fetal liver function is not yet mature, coagulation factors are synthesized independent of the maternal circulation. The serum concentrations of these factors increase with gestational age and do not cross the placenta. However, fetal clot formation in response to tissue injury is decreased in comparison to that in adults.


The anatomy of the fetal circulation helps decrease fetal exposure to potentially high concentrations of drugs in umbilical venous blood. Approximately 75% of umbilical venous blood initially passes through the fetal liver, which may result in significant drug metabolism before the drug reaches the fetal heart and brain (first-pass metabolism). Fetal and neonatal enzymatic drug metabolism activities are lower than those of adults, but most drugs can be metabolized. In addition, drugs entering the fetal inferior vena cava via the ductus venosus, thus bypassing the portal circulation and the liver, are initially diluted by drug-free blood returning from the fetal lower extremities and pelvic viscera. These anatomic characteristics of the fetal circulation add to the complexity of maternal-fetal pharmacokinetics.




Labor Progress


Labor begins with the onset of repetitive uterine contractions that result in the dilation of the cervix, thus permitting passage of the fetus from the uterus through the birth canal. In reality, however, preparation for labor may begin several hours or days before active labor with an inflammatory process mediated by cellular infiltration and release of local cytokines that result in softening of the cervix. The signals that orchestrate the onset of spontaneous labor are not precisely known. However, “labor” is the onset of organized, regular uterine contractions that result in progressive cervical dilation and effacement. When spontaneous labor does not occur at an appropriate time, labor may be induced for fetal or maternal indications with various pharmacologic and physical methods.


Labor is a continuous process separated into first, second, and third stages. The first stage of labor begins with regular, painful uterine contractions and includes the change of the uterine cervix from a thick, closed tube to an opening of approximately 10 cm through which the fetus can be expelled. This stage is further divided into a latent phase and an active phase. The second stage of labor begins when the cervix is fully dilated and ends with the birth of the newborn. The third stage of labor is the delivery of the placenta. The time course of the first stage of labor was first studied by Emanuel Friedman who described a sigmoidal relationship between cervical dilation and time ( Fig. 62.2 A ). The sigmoidal nature of the relationship has since been challenged in that little evidence exists for a deceleration phase as the cervix approaches complete dilation (10 cm). However, the separation of the first stage of labor into an early slow phase termed latent labor and a more rapid phase of active labor has stood the test of time and advances in modeling techniques. To account for the contemporary obstetric population including an older maternal age and increased maternal and fetal body sizes, a new labor curve has been proposed after analysis of 62,415 parturients. The main difference of the newly proposed curve is when latent labor is considered to transition to active labor. Traditionally, this transition point was 4-cm dilation. However, the new curve proposes active labor beginning at 6-cm dilation in both multiparous and nulliparous parturients ( Fig. 62.2 B ) .




Fig. 62.2


Cervical dilation.

(A) Friedman’s original sigmoid model of the progress of cervical dilation over time based on an analysis of 500 nulliparas at term. (B) The modern labor curve, which eliminates the deceleration (Deceler) phase and changes the threshold for the active phase from 4 to 6 cm. Acceler, Acceleration; P0 , nulliparous women; P1, women of parity 1; P2+, women of parity 2 or higher. (B) (Legend from Zhang, Fig. 2)Average labor curves by parity in singleton term pregnancies with spontaneous onset of labor, vaginal delivery, and normal neonatal outcomes.

Reproduced with permission from [A] Friedman E. Primigravid labor: a graphicostatistical analysis. Obstet Gynecol . 1955;6:567–589; and [B] Zhang J, Landy HJ, Brand DW, et al. Contemporary patterns of spontaneous labor with normal neonatal outcomes. Obstet Gynecol . 2010;116:1281–1287. doi: 10.1097/AOG.0b013e318fdef6e.


Labor may be referred to as “abnormal” on the basis of having abnormally slow latent labor, arrest in the active phase, or arrest of descent (failure of stage 2). Dystocia, or abnormal labor, may be a result of inadequate uterine contractions, mismatch of fetal and pelvic size, or abnormal fetal position. The diagnosis of dystocia is based on deviation from normal values derived from populations; however, significant variability exists among individual laboring women. Various demographic and genetic factors contribute to the variability in labor progress. Multiparity is associated with faster labor. Greater maternal weight, older age, and larger fetal size have been associated with slower labor. Evidence also indicates a hereditary role in labor progress from epidemiologic studies. Specifically, β 2 -adrenergic and oxytocin receptor polymorphisms have been implicated in mediating variability in labor progress. An abnormally poor response to intrinsic or extrinsic oxytocin may result in abnormal contractility, as would an abnormally strong response to β 2 -adrenergic agonists (either exogenous or endogenous) which inhibit contractility.


Labor and Fetal Monitoring


Intrapartum fetal monitoring was created to evaluate fetal well-being and detect fetal distress earlier in labor to allow intervention prior to permanent fetal injury. Electronic fetal monitoring (EFM) combines interpretation of fetal heart rate (FHR) monitoring and uterine contraction monitoring. FHR monitoring was developed in the 1960s and its use has been increasing since. There is high interobserver and intraobserver variability of FHR tracing interpretation. A meta-analysis comparing EFM to intermittent FHR auscultation noted the use of EFM reduced the risk for neonatal seizures (relative risk [RR]: 0.50), but not the risks for perinatal mortality or cerebral palsy. The use of this monitoring has been shown to increase the rate of both operative and cesarean deliveries.


The nomenclature, interpretation, and management principles for FHR monitoring were updated in 2009 by the American Congress of Obstetricians and Gynecologists (ACOG). These current guidelines are detailed later, and related terminology is presented in Box 62.1 . An understanding of the specific uterine contraction and FHR monitoring terminology as well as the clinical implications is critical for optimal communication during emergent situations among anesthesiologists, obstetricians, midwives, and labor nurses.



Box 62.1

Fetal Heart Rate Monitoring Pattern Definitions

Data from Macones GA, Hankins GD, Spong CY, et al. The 2008 National Institute of Child Health and Human Development workshop report on electronic fetal monitoring: update on definitions, interpretation, and research guidelines. Obstet Gynecol . 2008;112:661–666.


Baseline





  • The mean FHR rounded to increments of 5 bpm during a 10-min segment, excluding:




    • Periodic or episodic changes



    • Periods of marked FHR variability



    • Segments of baseline that differ by more than 25 bpm




  • The baseline must be for a minimum of 2 min in any 10-min segment, or the baseline for that period is indeterminate. In this case, one may refer to the prior 10-min window for determination of baseline.



  • Normal FHR baseline: Rate is 110-160 bpm.



  • Tachycardia: FHR baseline is greater than 160 bpm.



  • Bradycardia: FHR baseline is less than 110 bpm.



Baseline Variability





  • Fluctuations occur in the baseline FHR that are irregular in amplitude and frequency.



  • Variability is visually quantitated as the amplitude of peak-to-trough in beats per minute.




    • Absent: Amplitude range is undetectable.



    • Minimal: Amplitude range is detectable but 5 bpm or fewer.



    • Moderate (normal): Amplitude range is 6-25 bpm.



    • Marked: Amplitude range is greater than 25 bpm.




Acceleration





  • A visually apparent abrupt increase (onset to peak in <30 s) occurs in the FHR.



  • At 32 weeks’ gestation and beyond, an acceleration has a peak of 15 or more bpm above baseline, with a duration of 15 s or more but less than 2 min from onset to return.



  • Before 32 weeks’ gestation, an acceleration has a peak of 10 or more bpm above baseline, with a duration of 10 s or more but less than 2 min from onset to return.



  • Prolonged acceleration lasts 2 min or more but less than 10 min.



  • If an acceleration lasts 10 min or longer, it is a baseline change.



Sinusoidal Pattern





  • Visually apparent, smooth, sine wave–like, undulating pattern occurring in FHR baseline, with a cycle frequency of 3-5 cycles/min that persists for 20 min or longer.



bpm, Beats per minute; FHR, fetal heart rate.



Contraction Monitoring


Uterine contractions can be monitored externally with a tocodynamometer or internally with an intrauterine pressure transducer. External monitors only allow determination of contraction frequency, whereas internal monitors also allow quantitative measurement of intrauterine pressure. The Montevideo unit is traditionally used by obstetricians to assess the adequacy of uterine contractions. The Montevideo unit is defined as the intensity of contractions (in millimeters of mercury, as measured with an intrauterine pressure catheter) multiplied by the number of contractions that occur in 10 minutes.


Uterine contractions are quantified over a 10-minute window that is averaged over a 30-minute window with guidelines provided by the ACOG. Normal contractions are defined as five or fewer contractions in 10 minutes, averaged over a 30-minute window. Tachysystole is defined as uterine activity greater than five contractions in 10 minutes, averaged over a 30-minute window. Tachysystole applies to both spontaneous and augmented labor and should always be qualified as to the presence or absence of associated FHR decelerations. Treatment of tachysystole during labor may differ depending on the clinical situation but may include sublingual or intravenous nitroglycerin to briefly relax the uterus, as well as the use of β 2 -adrenergic drugs such as terbutaline.


Fetal Heart Rate Tracing


FHR monitoring is most commonly accomplished with a surface Doppler ultrasound transducer (external monitoring), but it may be necessary to apply a fetal scalp electrode to obtain accurate continuous FHR monitoring (internal monitoring). For internal monitoring, a peak or threshold voltage of the fetal R wave from the scalp electrode is used to measure FHR. Of note, a fetal scalp electrode can be placed only if the cervix is minimally dilated and the membranes are ruptured. The FHR pattern changes in response to fetal asphyxia from activation of peripheral and central chemoreceptors and baroreceptors. It also shows changes as a result of various fetal brain metabolic changes that occur with asphyxia. These changes in the FHR produce specific patterns and characteristics that provide an evaluation of the fetal state.


The FHR tracing is used as a nonspecific reflection of fetal acidosis. It should be interpreted over a time course in relation to the clinical context and other known maternal and fetal comorbidities, because multiple factors other than fetal acidosis can influence the FHR tracing. Box 62.1 defines FHR baseline, variability, and accelerations. A normal baseline FHR ranges from 110 to 160 bpm. FHR variability are fluctuations in the baseline FHR that are irregular in frequency and amplitude. Normal FHR variability predicts early neonatal health and a fetal central nervous system that is normally interacting with the fetal heart. Accelerations are abrupt changes in the FHR above baseline and are defined by gestational age of the fetus.


Fig. 62.3 details FHR tracing deceleration characteristics. Late decelerations are a result of uteroplacental insufficiency causing relative fetal brain hypoxia during a contraction. The resulting sympathetic outflow elevates the fetal blood pressure and activates the fetal baroreceptors and an associated slowing in the FHR. A second type of late deceleration is from myocardial depression in the presence of increasing hypoxia. Therefore late decelerations are considered worrisome. On the other hand, early decelerations are considered benign and tend to mirror the uterine contraction and are believed to be in response to vagal stimuli, which are often the result of fetal head compression. Variable decelerations are associated with umbilical cord compression. A sinusoidal FHR pattern is associated with fetal anemia and is considered ominous. In general, minimal-to-undetectable FHR variability in the presence of variable or late decelerations is associated with fetal acidosis. Prolonged decelerations (<70 beats/min for >60 seconds) are associated with fetal acidemia and are extremely ominous, particularly with the absence of variability.




Fig. 62.3


Fetal heart rate deceleration.

(A) Early deceleration: visually apparent, usually symmetric gradual decrease and return of the fetal heart rate (FHR) with the nadir of the deceleration occurring at the same time as the peak of the contraction. (B) Variable decelerations: visually apparent, abrupt decrease in FHR ≥15 seconds and <2 minutes. The relationship of the onset of the deceleration compared to the contraction is variable as is the depth and duration. (C) Late FHR decelerations: visually apparent, usually symmetric gradual decrease and return of the FHR during which the nadir of the deceleration occurs after the peak of the contraction.


Fetal Heart Rate Categories


A three-tiered FHR category classification system is currently recommended for fetal assessment with the specific criteria for each category outlined in Box 62.2 . This system evaluates the fetus for the given moment of the assessment. The fetal condition may move back and forth among the categories over time. Specific terminology used for categorization is defined in Box 62.1 .



Box 62.2

Three-Tier Fetal Heart Rate Interpretation System

Data from Macones GA, Hankins GD, Spong CY, et al. The 2008 National Institute of Child Health and Human Development workshop report on electronic fetal monitoring: update on definitions, interpretation, and research guidelines. Obstet Gynecol. 2008;112:661–666.


Category I


Category I FHR tracings include all of the following:




  • Baseline rate of 110-160 beats/min



  • Moderate baseline FHR variability



  • Late or variable decelerations are absent



  • Accelerations and early decelerations may be present or absent



Category II


Category II FHR tracings include all FHR tracings not categorized as Category I or III. Category II FHR tracings may include any of the following:




  • Baseline rate:




    • Bradycardia not accompanied by absent baseline variability



    • Tachycardia




  • Baseline FHR variability




    • Minimal baseline variability



    • Absent baseline variability with no recurrent decelerations



    • Marked baseline variability




  • Accelerations




    • Absence of induced accelerations after fetal stimulation




  • Periodic or episodic decelerations




    • Recurrent variable decelerations accompanied by minimal or moderate baseline variability



    • Prolonged deceleration ≥2 min but <10 min



    • Recurrent late decelerations with moderate baseline variability



    • Variable decelerations with slow return to baseline, “overshoots,” or “shoulders”




Category III


Category III FHR tracings include either:




  • Absent baseline FHR variability plus any of the following:




    • Recurrent late decelerations



    • Recurrent variable decelerations



    • Bradycardia




  • Sinusoidal pattern



FHR, Fetal heart rate.



Category I FHR tracings are considered normal and are predictive of a normal fetal acid-base state at the time of observation, and no specific management is required.


Category II FHR tracings are considered indeterminate and include all tracings not in categories I or III. Category II tracings are not predictive of abnormal fetal acid-base status and require continued monitoring and reevaluation with consideration for the entire clinical picture. In some cases, additional tests may be obtained or intrauterine resuscitative measures taken to improve the fetal condition.


Category III FHR tracings are considered abnormal and are associated with an abnormal fetal acid-base state at the time of observation. These tracings require prompt patient evaluation and efforts to improve the fetal condition. These interventions may include intrauterine resuscitation with change in maternal position, discontinuation of labor augmentation, treatment of maternal hypotension with fluids and/or vasopressor administration, use of supplemental O 2 , and/or administration of a tocolytic agent such as terbutaline. If the FHR tracing does not improve, expeditious delivery should occur which may involve an assisted vaginal (forceps or vacuum) delivery or a cesarean delivery.




Labor Analgesia


Childbirth is a pinnacle event in a family’s life that is surrounded by many beliefs and traditions, some of which are founded in science and others more historical, cultural, personal, or even spiritual. In this context, several nonpharmacologic techniques have been used to relieve the pain of childbirth throughout history, including acupuncture, massage, and hypnosis. Drugs were not used in Western medicine to relieve pain in childbirth until the mid-1800s, most famously when the English Queen Victoria chose to inhale chloroform for analgesia during the birth of Prince Leopold.


For most women, labor is intensely painful. However, the time course of pain intensity is highly variable, dynamic, and unpredictable. Some women will experience severe pain only just before and during the second stage of labor, whereas others will report severe pain from their first contraction. Rarely do women experience a pain-free labor and give birth unexpectedly under inopportune conditions. The source of these differences in labor pain is not completely known but may be in part genetic. In one study, Asian women reported more pain in labor than women of other ethnic backgrounds. This association was also found with a single nucleotide polymorphism in the β 2 -adrenergic gene. Other factors may include parity; maternal pelvic size and shape; fetal size and presentation; maternal anxiety, pain tolerance, and other psychological variables; the presence of maternal social and psychological support during labor; induction of labor; and whether contractions are augmented.


Nonpharmacologic Labor Pain Management


Many patients prefer to use nonpharmacologic methods of pain management during all or part of labor. Acupuncture can be effective in treating postoperative pain after cesarean delivery, but it is not as effective for analgesia during labor. A systematic review and meta-analysis of acupuncture for pain relief in labor involving 10 randomized controlled trials ( n = 2038) found that acupuncture was not superior to sham acupuncture (superficial needling lateral to an actual acupuncture point) at 1 and 2 hours. Unfortunately, most of the trials were not properly blinded, increasing the likelihood of bias.


Several trials have found a reduction in pain and anxiety during the first stage of labor with the use of massage. A Cochrane review on massage in labor identified seven randomized trials of massage, six of which were judged to have low or unclear risk for bias. During the first stage of labor, pain was reduced in the massage group by −0.98 (confidence interval [CI], 1.17-0.47) on a 10-point pain scale. No difference was found in the use of pharmacologic pain relief between groups or in pain reported during the second and third stages of labor. Of note, one study of 60 women found that massage decreased anxiety.


Hypnosis has been used both as a relaxation technique and for management of pain during labor. When hypnosis was compared with standard care, no evidence was found that pain was less with the use of hypnosis, nor was evidence found for a difference in satisfaction with pain relief. A Cochrane review of 9 trials randomizing 2954 women found women in the hypnosis group were less likely to use pharmacologic pain relief compared to those in the control groups. There were no clear differences for satisfaction, spontaneous vaginal birth, and postpartum depression between the two groups.


Other nonpharmacologic techniques include the breathing techniques described by Lamaze, the LeBoyer technique, the Bradley method, transcutaneous nerve stimulation, hydrotherapy, presence of a support person, intradermal water injections, and biofeedback. A retrospective national survey of women’s childbearing experiences in the United States found that although neuraxial methods of pain relief were considered the most helpful and effective, nonpharmacologic methods of hydrotherapy and massage were rated more or equally helpful in relieving pain compared with the use of intravenous opioids. Although many nonpharmacologic techniques seem to reduce the perception of labor pain, most published studies lack the rigorous scientific methodology for useful comparison of these techniques to pharmacologic methods.


Considerations for Pharmacologic Treatment of Pain in Labor


Preprocedural assessment by an anesthesia provider should be performed for any candidate for neuraxial labor analgesia. Clinical assessment should be considered for all patients admitted to a labor and delivery floor not only to discuss labor analgesia options prior to excruciating pain, but also to assess the patient for comorbid conditions that could complicate labor, obstetric procedures, or anesthesia. The obstetric anesthesia team should be prepared to care for all admitted patients in the event of an obstetric emergency. In otherwise healthy women, laboratory testing is not required during a routine preprocedural obstetric assessment.


Although any laboring woman has the potential to require cesarean delivery, labor takes many hours and requires adequate nutrition and hydration. While balancing these two considerations, the ASA has recommended that moderate amounts of clear liquids be allowed during the administration of neuraxial analgesia and throughout labor. A period of abstention from solids before the placement of neuraxial analgesia is not required. However, the ASA does recommend the ingestion of solid foods be avoided in laboring patients.


Systemic Medications


Opioids can be used for labor analgesia. They are inexpensive, widely available, and can be administered intramuscularly without the need for intravenous access. Although there are differences among opioids, they all cross the placenta and can have fetal effects including dose-related respiratory depression and decreased FHR variability.


Although meperidine was once the most commonly used long-acting opioid in obstetric practice, it is the most likely to result in side effects. Meperidine is typically administered intravenously in doses of up to 50 mg or intramuscularly in doses ranging from 50 to 100 mg. Maternal half-life of meperidine is 2.5 to 3 hours whereas the half-life for its active metabolite normeperidine is 13 to 23 hours. The half-life of both is up to three times longer in the fetus and newborn. Normeperidine can accumulate with repeated doses and can be neurotoxic. With increased dosing and shorter intervals between doses and delivery, risk to the newborn is increased, including lower Apgar scores and prolonged time to sustained neonatal respiration.


Morphine is rarely used for labor pain. Like meperidine it has an active metabolite (morphine-6-glucuronide) with a half-life that is longer in neonates than in adults, and it produces significant maternal sedation. Obstetricians may use intramuscular morphine for analgesia, sedation, and rest. This produces analgesia with an onset of 10 to 20 minutes and is used most commonly in latent labor. Maternal side effects may include respiratory depression and histamine release resulting in pruritus and rash.


Mixed agonist-antagonist opioid analgesics such as nalbuphine and butorphanol are utilized to treat labor pain. Nalbuphine has similar analgesic potency as morphine. It is given either intravenously, intramuscularly, or by subcutaneous injection at doses of 10 to 20 mg every 4 to 6 hours. Butorphanol is five times as potent as morphine and 40 times more potent than meperidine. A dose of 1 to 2 mg intravenously or intramuscularly is commonly used for labor analgesia. Both medications are often well tolerated by the parturient.


Fentanyl and, more recently, remifentanil have become popular systemic opioid analgesics over the past two decades. Fentanyl is a synthetic opioid that is highly lipid-soluble and has a short duration and no active metabolites. When given in small IV doses of 50 to 100 μg/h, no significant differences are seen in neonatal Apgar scores and respiratory effort compared with those in newborns of mothers not receiving fentanyl. Fentanyl is also commonly used in patient-controlled analgesia (PCA) during labor. Common doses utilized for fentanyl PCA include a bolus dose of 10 to 25 μg with a lockout interval of 5 to 12 minutes. High doses of systemic fentanyl, especially immediately prior to birth, could result in neonatal depression.


Remifentanil PCA may offer superior pain relief and lesser fetal effects than other intravenous opioid analgesics but its analgesic effects are inferior to epidural labor analgesia and it requires careful maternal oxygenation and ventilation monitoring. The metabolism of remifentanil depends completely on tissue and plasma esterases, which are fully mature in the fetus. Further, it is more rapidly metabolized in the placenta (by placental esterases) than in the maternal plasma and thus the fetal-to-maternal ratio is small. In the pregnant ewe model, the maternal-to-fetal ratio of remifentanil is approximately tenfold, and the ratio was similar in human studies. Because of these characteristics, more remifentanil can be administered to the mother at times close to delivery than would be considered safe for longer-acting opioids that rely on slower metabolism by the liver. Remifentanil is more effective than long-acting opioids ; however, the improvement in pain relief may be because comparatively larger doses of remifentanil were given.


Although remifentanil may be superior to longer acting systemic opioids, it is inferior to epidural labor analgesia. A meta-analysis of randomized controlled trials comparing remifentanil PCA and epidural analgesia found that parturients with remifentanil PCA had higher pain scores at one hour than those who received epidural analgesia. The incidence of pruritus, nausea, and vomiting were not statistically different between the two groups; however, the confidence intervals were wide. A subsequent meta-analysis confirmed that women were less satisfied when receiving remifentanil PCA compared to women in the epidural group but more satisfied than women receiving other parenteral opioids. The major risk of remifentanil in labor is maternal respiratory depression. Careful surveillance is required to ensure adequate oxygenation and ventilation throughout treatment and for this reason, some practices choose not to offer remifentanil PCA labor analgesia.


Inhaled Analgesia


While volatile anesthetics are no longer used for labor analgesia, nitrous oxide (N 2 O) is commonly used worldwide. Typically, it is blended with O 2 in a 50:50 ratio for patient-inhaled self-administration just before and during contractions. A systematic review evaluating N 2 O for labor analgesia found that epidural analgesia provided more effective pain relief than N 2 O but most studies were of poor quality. Some studies have demonstrated moderate analgesia in response to inhaled N 2 O (pain scores of 8 in 10 reduced to 6 in 10), whereas others have shown no difference in visual analogue pain scores. Paradoxically, in this negative study, many women wished to continue using nitrous labor analgesia after the study period. Overall, although patients are less likely to report excellent pain control with N 2 O, they were as likely to express satisfaction with their anesthesia care. Therefore although it is less efficacious than neuraxial analgesia, it provides an alternative to patients who desire a less invasive analgesic approach as well as those who have a contraindication for neuraxial analgesia. Without the coadministration of opioids, the use of 50% N 2 O in O 2 is safe and does not result in hypoxia or unconsciousness.


Neuraxial Analgesia


Neuraxial analgesia is the most reliable and effective method of reducing pain during labor. A large meta-analysis found epidural analgesia offered better pain relief compared with placebo (median pain scale reduction 3.4 in 10, 95% CI, 1.3-5.4) and reduced the need for additional pain medication. Analgesia is given most commonly by epidural, spinal, combined spinal-epidural (CSE), or dural puncture epidural (DPE). These techniques provide superior analgesia to alternative approaches and are safe.


Neuraxial Analgesia and Progress of Labor


Considerable controversy has been generated regarding the effects of neuraxial analgesia on the progress of labor. Observational studies have suggested that epidural analgesia is associated with slower labor progress as well as higher cesarean delivery rates. Confounding variables are likely responsible for this association. For example, patients who have dysfunctional labor (which are at higher risk to proceed to cesarean delivery) would have more exposure to severe pain, would be more likely to request epidural analgesia, and more likely to request it earlier. Before-and-after studies and multiple prospective, randomized controlled trials have found no association between epidural labor analgesia and cesarean delivery. In fact, a 2011 Cochrane review of 38 studies involving 9658 women comparing epidural versus nonepidural analgesia in labor found no difference in the risk for cesarean delivery. Large prospective studies in which patients were randomly assigned to early or late neuraxial anesthesia have invariably concluded that earlier neuraxial labor analgesia does not affect the length of the first stage of labor or increase the risk for cesarean delivery. In contrast, several prospective trials and a meta-analysis suggest that neuraxial anesthesia may cause a modest prolongation of the second stage of labor by approximately 15 minutes. Increase in the duration of the second stage may occur because dense motor blockade could impede coordinated pushing. Local anesthetic dosing is sometimes reduced in the second stage if the blockade is too dense to allow for coordinated expulsion efforts. However, a recent double-blind trial randomized 400 women with an epidural to either saline or local anesthetic at the onset of the second stage of labor and found no difference in length.


Epidural labor analgesia may offer maternal benefits during the second stage. When labor is comfortable, tolerance for a longer second stage may allow uterine contractions to lower the fetal station before active pushing efforts begin, a technique sometimes called “laboring down.” A large randomized clinical trial of term nulliparous women receiving neuraxial analgesia found no difference in the rate of spontaneous vaginal delivery if they delayed pushing or immediately started pushing at the onset of the second stage. This technique may allow women a less strenuous delivery and can be employed for women with significant comorbid cardiac or vascular conditions to limit the hemodynamic effects of repetitive Valsalva maneuvers. Further, epidural labor analgesia may protect the perineum from injury during delivery by allowing a more controlled expulsion of the fetus that allows for stretching (instead of tearing) of tissues.


Timing of Placement


The optimal timing for the placement of a neuraxial anesthetic has been extensively studied. In 2011, a meta-analysis including prospective, randomized trials was conducted to test if the placement of neuraxial analgesia during the early first stage of labor was associated with a prolonged first stage of labor. Six studies of 15,399 parturients compared placement of neuraxial analgesia at 3-cm cervical dilation or less to placement in active labor. No increase in the incidence of cesarean delivery was found in the early epidural group nor was the first stage of labor prolonged. Thus if a parturient chooses neuraxial analgesia, there is no point during the first stage of labor that is “too early” to initiate epidural analgesia. Current ASA guidelines note that maternal request for labor pain relief is sufficient justification for epidural initiation and the timing should not depend on an arbitrary cervical dilation.


Epidural Analgesia


Lumbar epidural analgesia offers a safe and effective method of pain relief during labor and is the mainstay of labor analgesia. Epidural analgesia is most commonly initiated after placement of a catheter into the epidural space between L2-3 and L4-5 (see Chapter 45 ). The analgesia technique is versatile and the block may be made denser and prolonged if operative delivery is required. Typically, a combination of low-dose local anesthetic and opioid are administered to provide continuous sensory block during labor. Benefits of epidural analgesia include decreased maternal catecholamines, effective pain relief, increased patient satisfaction, and the ability to quickly achieve surgical anesthesia for an emergency cesarean delivery.


Prevention of accidental intravascular or intrathecal local anesthetic administration is paramount in the safety of epidural techniques. Initial dosing of local anesthetic through the needle within the epidural space is not recommended because of potential unintended intravascular or intrathecal placement that would result in local anesthetic systemic toxicity or total spinal. Further, most anesthesia providers “test dose” the epidural catheter after placement to assess for intravascular or intrathecal placement. A test dose may consist of a small dose of local anesthetic that would induce an altered sensorium (dizziness, buzzing in the ears, or numbness in the lips) if injected intravascularly but would not cause harm. Likewise, if this small dose were injected into the intrathecal space, it would cause numbness and motor block in the lower extremities but not a high spinal block. Some clinicians favor inclusion of a small dose of epinephrine in the test dose so that if the placement were intravascular, a slight tachycardic and/or hypertensive response would ensue. Overall, even with test dosing, unidentified intravascular or intrathecal catheter placement is possible. This risk can be mitigated by dosing slowly, aspirating the catheter intermittently throughout the injection process, watching for CSF or blood return in the aspirated catheter, and vigilance for rapid and unexpected changes in vital signs, central nervous system (CNS) symptoms, or motor block throughout the dosing process.


Spinal Analgesia


Intrathecal analgesia for labor can be administered as a single dose or as a continuous infusion. A single injection of opioid combined with a small dose of local anesthetic in the subarachnoid space is quick to perform, provides rapid analgesia, and dissipates when no longer needed. A single spinal injection for labor analgesia can be utilized in a parturient who is unable to hold still to facilitate placement of an epidural but is usually reserved for when the duration of labor can be reasonably estimated, such as in multiparous parturients with advanced dilation or in the second stage of labor. Continuous spinal analgesia with a spinal catheter can be considered in the case of accidental dural puncture or in the high-risk parturient. The high incidence of postdural puncture headache (PDPH) precludes the elective placement of spinal catheters through epidural needles in most patients. A catheter-over-needle system provides the option for a 23-gauge intrathecal catheter placed over a 27-gauge pencil-point spinal needle. Although theoretically this should decrease the rate of PDPH, a recent series of five cases reported two cases of PDPH. When utilized, continuous spinal analgesia provides excellent analgesia and can be quickly converted to surgical anesthesia if necessary.


Combined Spinal-Epidural Analgesia


The CSE has become increasingly popular in obstetric anesthesia practice. It provides effective, rapid onset of analgesia with minimal motor blockade. It is most commonly placed utilizing the “needle-through-needle” technique, which involves identification of the epidural space through a loss-of-resistance technique followed by insertion of a long, pencil-point spinal needle (25-27 gauge) into the intrathecal space. After free-flow of CSF is confirmed, an opioid, local anesthetic, or both, is injected. The spinal needle is removed and a catheter is threaded into the epidural space. A large systematic review of 27 trials involving 3275 women found CSE had a faster onset of analgesia and was less likely to need additional epidural boluses. There was no difference between CSE and traditional epidural in terms of incidence of PDPH or rate of cesarean delivery. In one large retrospective review, the incidences of overall failure, inadequate analgesia, and catheter placement were lower in parturients receiving CSE compared to epidural. Disadvantages of CSE include the inability to assess the effectiveness of the epidural catheter until the spinal medication has subsided; however, one study not only found that CSE had lower epidural catheter failures (6.6% vs. 1.6%; P = .001) but more failed catheters with CSE (48.4%) than with an epidural (30.6%) were recognized within 30 minutes of placement ( P = .009).


Dural Puncture Epidural


An emerging technique in labor analgesia is the DPE. After the epidural space is located with the epidural needle, a pencil-point spinal needle is inserted utilizing the “needle-through-needle” technique and the dura is punctured. A 25- or 26-gauge spinal needle is usually used because a DPE placed by a 27-gauge needle was shown to offer no benefit in a single study. No medication is directly introduced into the intrathecal space but the dural puncture may facilitate the intrathecal migration of medication administered into the epidural space. DPE has been associated with a decreased median time to adequate analgesia after block placement. One study found improved block quality (measured by the number of physician administered top-up boluses) and a lesser incidence of asymmetric block in the DPE compared to the epidural group, while another study did not observe this difference. More studies are needed to fully understand the role of the DPE in the neuraxial labor analgesia armamentarium.


Neuraxial Analgesic Medications


Any preservative-free local anesthetic can be used in an epidural catheter. A perfect labor analgesic recipe provides excellent analgesia without motor blockade or other maternal or fetal effects. Low concentrations of local anesthetics (alone or in combination with opioids) are used to maximize sensory blockade and minimize motor blockade and maternal hypotension from sympathetic blockade. Most commonly, bupivacaine (0.0625%-0.125%) and ropivacaine (0.0625%-0.2%) are used because the ratio of sensory to motor blockade is greater than that for lidocaine or 2-chloroprocaine. Ropivacaine and levobupivacaine were synthesized to reduce cardiotoxicity that occurs with inadvertent intravascular bolus doses of bupivacaine. However, with the dilute concentrations of local anesthetic currently used for labor analgesia, cardiotoxicity is uncommon.


The addition of fentanyl to bupivacaine has been shown to reduce local anesthetic requirements while still providing similar pain relief. Therefore lipid-soluble opioids, fentanyl 1 to 3 μg/mL, or sufentanil 0.1 to 0.5 μg/mL, are typically added to local anesthetic labor epidural mixtures to reduce total local anesthetic administration and thereby reduce motor blockade while preserving analgesia and enhancing maternal satisfaction. Of note, opioid-only epidural regimens do not provide adequate analgesia without unacceptable side effects. The most troublesome complication that limits the dose of epidural fentanyl and sufentanil is pruritus.


The search for the perfect labor epidural drug combination has led to the use of other adjuvant drugs that can reduce the dose of required local anesthetic. Most of these drugs act through activation of adrenergic receptors. Epinephrine is a nonselective adrenergic agonist activating α 1 -, α 2 -, β 1 -, and β 2 -adrenergic receptors. Activation of α 1 -receptors in the epidural vasculature causes vasoconstriction that delays the vascular uptake of local anesthetic and opioid. Additional analgesia is likely provided by epinephrine through activation of α 2 -adrenergic receptors. The dose of epidural epinephrine in labor epidural mixtures is typically dilute (1:400,000-1:800,000) secondary to concerns for uterine artery vasoconstriction by the systemic effects of higher doses. Intrathecal or epidural neostigmine produces analgesia by increasing acetylcholine stimulation of spinal muscarinic and nicotinic receptors. Intrathecal neostigmine, however, caused an unacceptable incidence of nausea and vomiting and continued clinical development was abandoned. However, epidural neostigmine has been shown to reduce local anesthetic requirements without nausea and vomiting. A randomized controlled trial compared bupivacaine use in laboring patients when neostigmine versus fentanyl was added and found no difference in bupivacaine requirements.


Clonidine is a relatively selective α 2 -adrenergic antagonist that when added to dilute local anesthetic solution provides adjuvant analgesia. Although clearly effective for labor analgesia, within the United States epidural clonidine carries a US Food and Drug Administration (FDA) warning that states it is “not recommended for obstetrical, postpartum, or perioperative pain management as the risk of hemodynamic instability (e.g., hypotension, bradycardia) may be unacceptable in this population.” Monitoring recommendations related to this statement declare that in “a rare obstetrical, postpartum, or perioperative patient, potential benefits may outweigh possible risks.” Dexmedetomidine is a highly selective α 2 -adrenergic receptor agonist that is not approved for neuraxial use in the United States. However, when used in combination with epidural bupivacaine or ropivacaine it has been found to be an efficacious adjunct for labor analgesia.


Administration Techniques


Epidural analgesia can be administered by continuous infusion, patient-controlled epidural analgesia (PCEA), or programmed intermittent epidural bolus (PIEB). The continuous infusion is commonly utilized because it allows the maintenance of a steady anesthesia level without frequent, time-consuming manual boluses by the anesthesia provider. PCEA allows the patient to self-deliver a dose through the epidural catheter with the use of a pump that limits the maximum drug dose per hour to prevent toxicity. PCEA can be used alone or in combination with a continuous infusion or PIEB. PCEA results in reduced local anesthetic use and decreased motor blockade. PIEB utilizes a pump that gives automated boluses at a fast rate at set intervals instead of a slow continuous infusion. Theoretically, the PIEB function allows for greater, more uniform spread of the epidural infusion thus reducing unilateral blocks, areas of block sparing, and total amounts of local anesthetic required for analgesia. Early studies indicate that this pump function may, in fact, offer such benefits.


Contraindications of Neuraxial Anesthesia


Contraindications to the placement of a neuraxial procedure include patient refusal, coagulopathy, infection at the site of needle insertion, uncorrected hypovolemic shock, increased intracranial pressure from mass effect, and inadequate resources or expertise. Relative contraindications may include systemic infection, preexisting neurologic disease, severe cardiac valvular stenosis, and pharmacologic anticoagulation. The decision to place neuraxial anesthesia should be individualized for the patient and the risks and benefits should be considered.


Other Regional Nerve Blocks


Local anesthetic nerve blocks have been used for many years to relieve labor pain, mostly by obstetricians. For a paracervical block, local anesthetic is injected lateral to the cervix at 4 o’clock and 10 o’clock, taking care to avoid vascular structures. It controls pain of the first stage of labor only and is more effective than placebo or intramuscular meperidine. No difference was found in pain relief comparing paracervical block to PCA with intravenous fentanyl. Paracervical block can be complicated by injection of local anesthetic into the presenting fetal head, which can have devastating consequences. More commonly, side effects of transient fetal bradycardia and maternal local anesthetic toxicity have been reported. Therefore for the delivery of a viable fetus, obstetricians in the United States avoid this procedure. The technique of paracervical block is still used in intrauterine fetal demise labor analgesia, dilation and curettage, and dilation and evacuation procedures. It has become safer with more superficial injection ensured by a needle guide and more dilute solutions of local anesthetic.


The pudendal nerve is derived from sacral nerve roots (S2-S4) and can be blocked with local anesthetic using a transvaginal or transperitoneal approach to treat pain during the second stage of labor and for episiotomy repair. Although a pudendal nerve block provides some relief during second stage, it is not as effective as a subarachnoid block with fentanyl and bupivacaine. A pudendal block can impede the urge to push during the second stage of labor. Other complications include a high rate of block failure; systemic local anesthetic toxicity; ischiorectal or vaginal hematoma; and, rarely, fetal injection of local anesthetic.




Anesthesia Considerations for Operative Delivery


Low-dose epidural analgesia can be inadequate for assisted vaginal delivery with forceps or vacuum. In this setting, a higher concentration local anesthetic can be administered through an indwelling epidural catheter. Supplementation of an indwelling epidural catheter with 5 to 10 mL of 1% to 2% lidocaine or 2% to 3% 2-chloroprocaine is usually adequate, depending on whether vacuum or forceps are being used. Pudendal nerve block also can be considered for operative delivery. Consideration could be given to a CSE approach instead of a single injection “second stage spinal” in the event that the operative vaginal delivery fails and cesarean delivery is subsequently required.




Anesthesia for Cesarean Delivery


Maternal Risks and Considerations


Cesarean delivery rates in the United States increased by 50% between 1998 and 2016, rising from 22% to 32% of all births. Common indications for cesarean delivery include fetal malpresentation, nonreassuring fetal status, labor dystocia, and prior cesarean delivery. Although maternal mortality substantially decreased during the first half of the twentieth century, the maternal mortality ratio has not declined in over 25 years and appears to have recently been increasing in the United States. Of interest, the incidence of maternal mortality is 10 times higher in women who underwent cesarean delivery versus vaginal delivery, based on a retrospective study of 1.5 million deliveries between 2000 and 2006.


Pregnant women who undergo general anesthesia for cesarean delivery are at increased risk of pulmonary aspiration of gastric contents, failed intubation of the trachea, or inadequate postoperative ventilation compared with those under neuraxial blockade, particularly in emergent situations. However, it appears that the risks associated with general anesthesia have decreased significantly over time to the point where it is difficult to say that avoiding general anesthesia prevents maternal mortality. Data in the United States from 1979 to 1990 noted a risk ratio of 16.7 for mortality with general anesthesia for cesarean delivery compared with neuraxial blockade, whereas between 1997 to 2002, the risk ratio was not significantly greater with general versus neuraxial anesthesia (RR 1.7; CI, 0.6-4.6, P = .2). This reduction in the risk ratio may be because of advanced airway techniques (e.g., supraglottic airways and video laryngoscopy) and safer airway practices (e.g., difficult airway algorithms).


Use of neuraxial anesthesia for cesarean delivery minimizes exposure of the neonate to maternal anesthetic medications, avoids airway manipulation, improves postoperative pain, and allows the mother to see the child almost immediately after birth. All pregnant women should undergo a preoperative evaluation, regardless of planned delivery mode or type of anesthetic technique, with appropriate risk and benefit counseling. The current status of the fetus and obstetric management plan also should be taken into consideration when formulating the anesthetic plan. In addition, appropriate equipment and medications should always remain readily available to safely provide general anesthesia for an emergent or unanticipated situation.


Although the rates of significant maternal aspiration of gastric contents with induction of general anesthesia are difficult to determine, the mortality from such an event is estimated at 5% to 15% based on retrospective data. ASA guidelines recommend aspiration prophylaxis with the administration of nonparticulate antacids, H 2 receptor antagonists, and/or metoclopramide before obstetric surgical procedures. The decision to use general anesthesia or neuraxial block for cesarean delivery is determined by a variety of factors that include the fetal condition and urgency of delivery, maternal comorbidities, presence of a previously placed epidural for labor analgesia, surgical considerations, and maternal wishes. At present, most cesarean deliveries in developed countries are performed with neuraxial techniques.


Spinal Anesthesia


If an epidural catheter is not already placed, spinal anesthesia is typically used for nonemergent cesarean deliveries. Compared with an epidural, a single injection spinal is often faster and technically easier to perform, allows adequate operating conditions in a shorter time, provides a denser block, is more cost effective, and is less likely to fail (failure rate <1%). Spinal anesthesia is most often administered through a small (24 gauge or smaller), pencil-point needle. On occasion, a continuous spinal catheter may be used for anesthesia for cesarean delivery. As previously discussed, a spinal catheter may be placed in the case of an inadvertent dural puncture but can be placed intentionally for cesarean delivery in high-risk obstetric patients.


The chance of significant maternal hypotension is greater with spinal anesthesia than with epidural anesthesia. Left uterine displacement with appropriate administration of fluids and use of vasopressor medications can minimize the associated hypotension. Intravenous administration of crystalloid or colloid can reduce the degree of hypotension after spinal anesthesia for cesarean delivery. A Cochrane review assessed 11 trials with 698 women comparing colloid versus crystalloid and found significantly fewer women became hypotensive after colloids (RR 0.68; 95% CI, 0.52-0.89). However, concern exists regarding the safety of synthetic colloids as part of intraoperative and critical care resuscitation. Of note, fluid co-loading is thought to have limited efficacy in consistently preventing postspinal hypotension and is typically utilized in combination with a vasopressor.


Historically, ephedrine was considered the vasopressor of choice to manage hypotension caused by neuraxial anesthesia in pregnancy; however, prophylactic or therapeutic phenylephrine in boluses or as an infusion is not only effective in reducing hypotension but also has less transfer to the fetus and results in less fetal acidosis than ephedrine. Phenylephrine is now considered the vasopressor of choice for the treatment of spinal hypotension, and there is accumulating evidence that administration by prophylactic infusion is most effective in preventing hypotension. A systematic review found prophylactic phenylephrine infusions compared to placebo significantly reduced the risk of hypotension (RR 0.36; 95% CI, 0.18-0.73) and nausea and vomiting (RR 0.39; 95% CI, 0.17-0.91). Phenylephrine is an α-adrenergic receptor agonist and is often associated with reflexive slowing of maternal heart rate and a decrease in cardiac output. There is increasing interest in norepinephrine as an alternative vasopressor for treating spinal hypotension. Compared to phenylephrine, norepinephrine had similar efficacy for maintaining arterial blood pressure during spinal anesthesia for cesarean delivery and was associated with a greater heart rate and cardiac output. Further work needs to be done to assess the safety and efficacy of norepinephrine as the vasopressor of choice to prevent and treat postspinal hypotension.


Although various local anesthetics can be used for spinal blockade, hyperbaric bupivacaine 10 to 12 mg is frequently used to achieve an adequate (T4) level block. Neither patient height nor weight affect block extension, although dosing may require adjustment at extremes of the height spectrum. Lipid soluble opioids (such as fentanyl or sufentanil) may be added to enhance neuraxial blockade by reducing local anesthetic dose and decreasing stimulation from surgical traction of the viscera. Epinephrine (0.1-0.2 mg), may be added to improve the quality and duration of the block. Clonidine can also prolong the duration of the block and improve intraoperative analgesia but can increase sedation and is considered off-label for this use. Preservative-free morphine 0.1 to 0.2 mg or hydromorphone 0.75 mg is frequently administered with the spinal to reduce postoperative pain for 18 to 24 hours after the anesthetic has dissipated.


Epidural Anesthesia


If an epidural catheter has already been inserted for labor analgesia, it provides an excellent method to provide surgical anesthesia for cesarean delivery. A catheter-based technique allows for the ability to titrate the local anesthetic to the proper block height and provide additional local anesthetic administration during the case. For patients who do not already have a catheter in place, this technique may be chosen if the procedure is anticipated to take additional time, or if maternal comorbidities would favor a more gradual, controlled onset of epidural anesthesia. Achieving surgical block conditions takes longer with an epidural than spinal technique but can be rapid enough for use in many urgent situations if already in place and used for maternal analgesia.


Although use of a rapid-onset local anesthetic such as 3% 2-chloroprocaine to attain a T4 level in a newly placed catheter may take 10 minutes, extension of a T10 analgesic level to a surgical anesthetic T4 level can be obtained in approximately 5 minutes using 3% 2-chloroprocaine or alkalinized 2% lidocaine. Bupivacaine 0.5% may be used if rapid onset is not needed, but is often avoided because of the increased risk of local anesthetic systemic toxicity. Typical epidural local anesthetic volumes required for cesarean delivery range between 10 and 20 mL, depending on whether the epidural is already in use. The administration of epidural local anesthetic should occur in divided doses to ensure that the catheter has not migrated into the intravascular or intrathecal space. Block quality can be improved with addition of epinephrine 1:200,000, fentanyl 50 to 100 μg, or sufentanil 10 to 20 μg. Epidural clonidine 50 to 100 μg can be useful in patients with preexisting chronic pain or severe hypertension if the benefit is judged to outweigh the risk for hypotension, bradycardia, and sedation. Epidural morphine 2 to 5 mg is frequently administered to improve postoperative pain.


Combined Spinal-Epidural Anesthesia


Use of a CSE for cesarean delivery may be optimal in some situations as it combines the benefits of the spinal and epidural techniques. This technique allows for the rapid onset of a dense reliable block while allowing the block time or height to be extended with use of the epidural catheter. A low-dose sequential CSE, a technique where a small intrathecal dose of local anesthetic is given followed by administration of epidural medications, can be used in patients with cardiac disease or patients with short stature. Possible disadvantages of a CSE for surgical anesthesia include the presence of an untested catheter and the possibility of a misplaced or nonfunctioning epidural. More details regarding the CSE neuraxial block are discussed in the previous section on labor analgesia.


General Anesthesia


Although neuraxial anesthesia is typically preferred, in certain emergent situations (e.g., fetal bradycardia, maternal hemorrhage or coagulopathy, uterine rupture, maternal trauma) general anesthesia may be needed for cesarean delivery because of its rapid onset. In addition, it allows for a controlled airway, controlled ventilation, and in some scenarios such as massive hemorrhage, improved hemodynamic control and perhaps decreased maternal psychological stress in comparison to neuraxial anesthesia.


Appropriate equipment preparation, knowledge of patient comorbidities, airway examination, and familiarity with the difficult airway algorithm are necessary preparation for delivering a safe general anesthetic. The ASA difficult airway algorithm has been modified slightly by some authors for use in the setting of cesarean delivery, and a previously published example is provided in Fig. 62.4 . Clear, concise communication among all members of the perioperative team is especially critical in urgent or emergent situations to maximize patient safety and minimize procedural complications. Open lines of communication are essential around the time of induction of anesthesia, airway management, and surgical incision.




Fig. 62.4


Algorithm for management of unanticipated difficult airway in obstetric patients.

BMV, Bag-mask ventilation; BP, blood pressure; CS, cesarean section; ETCO 2 , end-tidal carbon dioxide; HR, heart rate; LMA, laryngeal mask airway; SpO 2 , oxygen saturation.

Redrawn from Balki M, Cooke M, Dunington S, et al. Unanticipated difficult airway in obstetric patients: development of a new algorithm for formative assessment in high-fidelity simulation. Anesthesiology . 2012;117:883–897, with permission.


A rapid-sequence induction commences with preoxygenation, followed by the application of cricoid pressure and the administration of an intravenous induction drug (typically propofol) and a neuromuscular-blocking drug (typically succinylcholine or rocuronium). If endotracheal intubation fails, consider placement of a supraglottic airway device, such as a laryngeal mask airway (LMA), or ventilation with mask and cricoid pressure. The LMA has a high success rate for placement in obstetric patients; however, it does not protect against aspiration of gastric contents and should be used primarily for rescue of a failed intubation. Of note, the LMA has been used without observed aspiration or presence of hypoxia in a prospective study of over 1000 lean, low-risk patients undergoing elective cesarean delivery.


Complications as a result of general anesthesia for cesarean delivery can be more common than general anesthesia for other procedures. For example, although the overall risk of intraoperative awareness is estimated to be 1:19,000 general anesthetics, the awareness risk for cesarean delivery is estimated at 1:670 (1:380-1:1300). Anesthesia-related mortality from airway difficulties may occur during emergence and in the recovery period, as well as during induction. Improper monitoring, provider inexperience, emergent situations, and patient obesity all increase patient risk. Emergent cesarean delivery requiring general anesthesia is an uncommon but predictable emergency that can be practiced by teams using simulation to improve performance.


Induction of Anesthesia: Intravenously Administered Drugs


Premedication with lidocaine or fentanyl is typically avoided in cesarean delivery to limit fetal exposure. In scenarios in which hemodynamic stability is prioritized, such as preeclampsia or heart disease, remifentanil 1 to 2 μg/kg or fast-acting antihypertensives such as esmolol or labetalol can be used.


Propofol is most commonly used for induction of general anesthesia for cesarean delivery and is able to induce unconsciousness in approximately 45 seconds. Sodium thiopental 4 to 6 mg/kg intravenously is still used in many countries for induction of anesthesia. Propofol can result in significant hypotension and has an umbilical artery (UA) to umbilical vein (UV) ratio of 0.7. Propofol administration does not affect neonatal Apgar scores with typical intravenous induction doses (2-2.5 mg/kg), but repeated or larger cumulative doses (9 mg/kg) are associated with significant newborn depression.


Etomidate is quick acting and its rapid hydrolysis results in a relatively short duration of action. Unlike propofol and thiopental, etomidate has minimal direct effects on maternal hemodynamics and significant hypertension can occur when etomidate is used without adjuvant premedication in the healthy parturient. Etomidate has higher rates of nausea and vomiting and can increase risk for seizures in patients with decreased seizure threshold. At typical induction doses (0.3 mg/kg), decreased neonatal cortisol production for less than 6 hours was noted with unclear clinical significance.


Ketamine inhibits the N -methyl- d -aspartate receptor and has analgesic, amnestic, and hypnotic properties with minimal respiratory depression effects. At typical induction doses (1-1.5 mg/kg), ketamine causes central stimulation of the sympathetic nervous system and inhibits the reuptake of norepinephrine. This helps maintain arterial pressure, heart rate, and cardiac output but could result in hypertension in the preeclamptic patient. It is an ideal choice for a pregnant woman in hemodynamic compromise resulting from bleeding. No neonatal depression is observed with standard induction dosing. Larger doses can increase uterine tone, reduce uterine arterial perfusion, and lower maternal seizure threshold. In certain situations, ketamine may be used in smaller intravenous doses (<0.25 mg/kg) as a profound analgesic, but often causes unwanted hallucinations, which can be reduced with coadministration of benzodiazepines. Care should be taken if used for analgesia and conscious sedation so that repeated dosing does not result in loss of consciousness with an unprotected airway increasing the risk for pulmonary aspiration.


Muscle Relaxants


Uterine muscle tone is not affected by skeletal muscle relaxants, and in standard doses all classes of muscle relaxants are poorly transferred to the fetus. Succinylcholine 1 to 1.5 mg/kg intravenously has rapid onset (30-45 seconds) and short duration of action. After administration, it is hydrolyzed in the plasma by pseudocholinesterase and only small amounts cross to the fetus because it is highly ionized and poorly lipid soluble. It is undetectable in umbilical cord samples unless larger maternal doses are administered (2-3 mg/kg), and exceedingly high maternal doses (10 mg/kg) are needed to inadvertently create neonatal neuromuscular blockade. After succinylcholine administration, maternal muscle strength should always be monitored because prolonged weakness can occur if the hydrolytic enzyme has a reduced plasma concentration or an atypical form (such as in pseudocholinesterase deficiency) or if muscle weakness is exacerbated by prior administration of magnesium sulfate.


Rocuronium may be considered as an alternative to succinylcholine for muscle relaxation. It allows adequate relaxation for endotracheal intubation in less than 60 seconds at intravenous doses of 0.9 to 1.2 mg/kg. This is an attractive alternative to succinylcholine because even after an intravenous intubating dose of 0.9 to 1.2 mg/kg body weight, the neuromuscular blocking effect can rapidly be reversed by a large intravenous dose of sugammadex (12-16 mg/kg body weight), which makes the total duration of action shorter than after an equipotent dose of succinylcholine. Like succinylcholine, nondepolarizing muscle relaxants do not cross to the fetal circulation in amounts that would cause neonatal weakness. However, if large doses of nondepolarizing neuromuscular blockers are given over long periods, neonatal neuromuscular weakness can occur. Although cholinesterase inhibitors may be administered to the neonate, treatment is primarily respiratory support until the drug is eliminated. Neonatal elimination of muscle relaxants may take significantly longer than adult elimination.


In the case of administration of magnesium sulfate, a distinct potentiation of the effect of any nondepolarizing agents occurs, with subsequently prolonged recovery time. The choice and dosing of neuromuscular blocking drugs should therefore take into account the interaction with magnesium sulfate and the potential risk for muscle weakness resulting from residual neuromuscular block in the recovery room or postanesthesia care unit. As a consequence, neuromuscular monitoring based on an objective monitoring technique should be used to assess neuromuscular function in these patients.


Maintenance of General Anesthesia


After induction, general anesthesia is most frequently maintained with a volatile anesthetic agent with or without N 2 O. Volatile anesthetics help reduce the incidence of maternal recall. Although the MAC to prevent movement in response to a painful stimulus is reduced in pregnancy, electroencephalographic evidence suggests that anesthetic effects of a halogenated agent on the brain are similar in the pregnant and nonpregnant state. Volatile anesthetics are highly lipid soluble, have a low molecular weight, and are readily transferred to the fetus. Fetal concentrations depend on both the maternal plasma concentrations and duration of the anesthetic before delivery. After delivery, opioids, propofol, benzodiazepines, N 2 O, or a combination are administered and the halogenated anesthetic is typically reduced to 0.5 MAC. These additional intravenous drugs are administered only after the cord is clamped to prevent any transfer to the neonate and associated respiratory depression. Use of only volatile anesthetics at higher concentrations is associated with increased blood loss secondary to uterine atony because all volatile anesthetics negatively impact uterine muscle contraction.


General anesthesia for cesarean delivery is frequently used in cases of fetal distress because it is rapid and reliable. Clearly, a fetus depressed before delivery often becomes a depressed neonate. A Cochrane systematic review of uncomplicated cesarean deliveries comparing regional and general anesthesia concluded that “No significant difference was seen in terms of neonatal Apgar scores of six or less and of four or less at one and five minutes,” and the need for neonatal resuscitation with oxygen was not different between the two groups. The review noted neither method of anesthesia was superior for neonatal outcome.


If large concentrations of volatile anesthetics are administered for a prolonged time, neonatal flaccidity, cardiorespiratory depression, and decreased tone may be anticipated. If neonatal depression is due to volatile anesthetics, the infant should respond to assisted ventilation to facilitate exhalation of the anesthetics. Consequently, physicians able to assist with neonatal ventilation should be present at all cesarean deliveries performed under general anesthesia. In addition, communication to all perioperative physicians is critical if extended anesthetic time is anticipated before delivery. A prolonged time under general anesthesia prior to delivery can be anticipated by obstetricians in some patients such as those with significant scar tissue from prior surgeries or extreme obesity. Neonates may experience greater benefit from regional anesthesia in these scenarios.




Postcesarean Pain Control and Recovery


Pain after cesarean delivery is variable in intensity among patients. Excellent pain control after cesarean delivery can lead to improved maternal functional ability, enhanced recovery, decreased persistent opioid use, decreased incidence of chronic pain, and improved maternal-infant bonding. Postoperative pain control can be achieved with multimodal therapy. A typical strategy consists of neuraxial opioids, scheduled nonsteroidal antiinflammatory drugs, scheduled acetaminophen, and limited systemic opioids. Neuraxial opioids are considered the “gold standard” for effective postoperative pain control and have been shown to be superior to systemic opioids and transversus abdominis plane (TAP) block. Pruritus, nausea and vomiting, and respiratory depression are opioid-related side effects.


Alternative modes of analgesia are available and can be utilized if neuraxial opioids are contraindicated or general anesthesia is utilized. Peripheral nerve blocks include TAP, quadratus lumborum, and ilioinguinal-iliohypogastric blocks (see Chapter 46 ). A meta-analysis demonstrated TAP blocks significantly reduced postoperative pain intensity and decreased opioid consumption in women who did not receive intrathecal morphine. Quadratus lumborum blocks are gaining popularity in cesarean deliveries and may have advantages over TAP blocks. A randomized controlled trial comparing quadratus lumborum with TAP blocks found a reduction in morphine use in the quadratus lumborum group. Continuous wound infiltration with local anesthetics may also be beneficial for patients having general anesthesia.




Complications of Regional Anesthesia


In addition to the neuraxial drug–related complications noted previously, administration of neuraxial anesthesia can result in complications including PDPH, epidural or spinal hematoma, neurologic injury, or total spinal anesthesia (see also Chapter 45 ).


Postdural Puncture Headache


The earliest experiments with spinal cocaine resulted in severe PDPH. Leakage of spinal fluid is thought to result in vascular hyperemia, migraine physiology, and traction on pain-sensitive fibers. The headache associated with PDPH is postural in that it is worsened by standing and relieved by lying down. The incidence, severity, and duration of PDPH are related to the size of the needle and the shape of the tip. Spinal needles used for CSE technique range from 25 to 29 gauge and result in an incidence of PDPH of less than 1%. Epidural catheters are most commonly placed through a 17- or 18-gauge blunt-tipped needle. The incidence of unintentional dural puncture during labor epidural placement is 1% to 1.5%. The incidence of headache after an unintentional dural puncture with an epidural needle is reported at 30% to 80%.


In the setting of an unintended dural puncture with an epidural needle, an intrathecal catheter may be threaded, or the epidural needle may be removed and replaced at a different interspace. If an intrathecal catheter is placed, unintentional injection of an epidural anesthetic dose must be carefully avoided. Placement of the intrathecal catheter can provide labor analgesia and alleviates the need for multiple repeat epidural attempts with the potential of a second accidental dural puncture.


When diagnosing a PDPH, it is important to consider other causes of headache in the postpartum period. Assessing the patient for fever and nuchal rigidity is important because postdural-puncture meningitis can initially present with a headache. Early treatment of meningitis is important to prevent morbidity and mortality. Likewise, assessing the patient for hypertension is important to detect postpartum preeclampsia, which can present with a headache and requires rapid treatment to prevent maternal stroke. A thorough neurologic exam should also be performed because cerebral venous thrombosis, cranial subdural hematoma, and ischemic or hemorrhagic stroke can present as a postpartum headache. Benign headaches from tension, dehydration, sleep deprivation, caffeine withdrawal, or migraine should be considered prior to treating a PDPH.


PDPH is often initially treated conservatively. Given the similarity of symptoms to those of migraine headache, PDPHs have been treated with drugs that are useful in migraine with variable success. Caffeine can be minimally effective to treat the pain of a PDPH in the short-term likely because of its vasoconstrictive effects.


If the symptoms are severe enough to limit a patient’s activity then an epidural blood patch (EBP) should be considered. If there is evidence of cranial nerve involvement such as diplopia, then an EBP should be performed immediately. Note that the symptom of muffled hearing is common with PDPH and thought not to be a result of cranial nerve involvement, but instead, a decrease in middle ear pressure because the middle ear fluid is connected to the cranial CSF via the cochlear aqueduct. In one retrospective review of an EBP database, all parturients with PDPH experience relief after the EBP but 16.8% required two and 1.5% required three EBPs. Controversy exists over the optimum volume of blood to be injected during an EBP but studies support an attempt to administer 20 mL of autologous blood during the procedure.


Epidural Hematoma


The epidural space is highly vascular, and vessels can be punctured during neuraxial needle placement or catheter threading. However, with normal platelets and coagulation factors, epidural hematoma is extremely uncommon. The Serious Complication Repository (SCORE) Project reported the incidence of epidural hematoma to be 1 in 251,463 (95% CI, 1:46,090-1:10,142,861) and another large study found no cases of hematoma out of 79,837 obstetric epidural placements with an estimated upper limit of 1 in 4.6 × 10 −5. Although rare, back pain and persistent motor blockade are potential signs of epidural hematoma and should be thoroughly evaluated. Patients should be followed postpartum until complete resolution of the epidural blockade. Patients are at increased risk for developing a hematoma in the setting of coagulopathy and anticoagulation use. Following the guidelines of the American Society of Regional Anesthesia (ASRA) with consideration for the Society for Obstetric Anesthesia and Perinatology (SOAP) Consensus Statement ( Fig. 62.5 ) regarding anticoagulation therapy and neuraxial anesthesia in the obstetric patient is recommended. If an epidural hematoma is suspected, imaging should occur immediately with the goal of prompt epidural hematoma evacuation to avoid permanent neurologic injury.




Fig. 62.5


(A) Decision aid for urgent or emergent neuraxial procedures in the obstetric patient receiving unfractionated heparin. (B) Decision aid for urgent or emergent neuraxial procedures in the obstetric patient receiving LMWH. aPTT, activated partial thromboplastin time; GA, general anesthesia; LMWH, low-molecular-weight heparin; SEH, spinal epidural hematoma; SQ, subcutaneous; UFH, unfractionated heparin.

Reproduced with permission from Leffert L, Butwick A, Carvalho B, et al. The Society for Obstetric Anesthesia and Perinatology Consensus Statement on the Anesthetic Management of Pregnant and Postpartum Women Receiving Thromboprophylaxis or Higher Dose Anticoagulants. Anesth Analg. 2018;126:928–944.


Neurologic Injury


Direct spinal cord damage from an epidural or spinal needle placed for labor is exceedingly rare because obstetric neuraxial anesthesia procedures are typically performed below the level of the conus medullaris. It has, however, been described with spinal anesthesia attempts performed at unintentionally high levels resulting in spinal cord syrinx formation with injection. In a series of seven such cases, all of the patients who experienced this complication described pain on injection. Therefore if a patient complains of pain on injection of neuraxial medication, the proceduralist should stop injecting immediately .


Overall, damage attributed to direct neurologic injury has been estimated at 0.6 in 100,000 with epidural and 3 in 100,000 with spinal analgesia. Data from the SCORE project show an incidence of serious neurologic injury in the postpartum period to be 1 in 11,389 (95% CI, 1:7828-1:17,281) but only 1:35,923 (95% CI, 1:17,805-1:91,244) are thought to be related to anesthesia.


Evaluating postpartum lumbosacral neuropathies is a common part of an obstetric anesthesia practice. A large prospective study showed that 0.92% of women experienced postpartum lumbosacral or lower extremity nerve injury, the incidence of which was associated with nulliparity and prolonged second stage of labor, but not epidural analgesia use. Femoral and lateral femoral cutaneous neuropathies were the most common, likely from women in extreme hip flexion in the semi-Fowler position. Encouraging women to straighten their legs between pushes can restore blood flow to the nerves of the lumbar plexus. Most of these peripheral neuropathies heal with time but outpatient follow up with a neurologist to follow the course and rule out other causes should be considered.


Local Anesthetic Systemic Toxicity


The accidental injection of intravascular local anesthetic could result in the development of local anesthetic systemic toxicity (LAST). The development of LAST is rare in obstetrics secondary to the dilute local anesthetic solutions utilized for epidural analgesia and the use of lidocaine and 2-chloroprocaine for operative delivery. The SCORE project reported one maternal cardiac arrest from LAST after a TAP block. LAST as a result of TAP blocks in obstetrics has been reported multiple times and the use of no greater than 0.25% concentration of bupivacaine, adding 1:200,000 epinephrine, administering no greater than a 20 mL volume on each side, placement under ultrasound to be sure the injection is not intraperitoneal, and careful, slow, injection with intermittent aspiration is recommended. The treatment of LAST includes the administration of lipid emulsion in addition to basic and advanced cardiac life support.


Total Spinal Block


A total spinal block is a rare and life-threatening complication that occurs after excessive cephalic spread of local anesthetic in the CSF resulting in severe respiratory and cardiac compromise. It can occur after a single spinal injection or as a result of inadvertent intrathecal spread of epidural medication. Risk factors associated with high neuraxial blockade include obesity, spinal technique after failed epidural anesthesia, short stature, epidural after an accidental dural puncture, and spinal deformity.


Other Complications


When strict aseptic technique is used, infection is uncommon with spinal and epidural anesthesia. Nonetheless, postdural puncture meningitis and epidural abscess has been described and the ASA and ASRA recommend the following aseptic techniques during placement of neuraxial needles and catheter: removal of jewelry, hand washing, wearing of caps, wearing of masks covering both mouth and nose, use of sterile gloves, use of an antiseptic solution (e.g., chlorhexidine with alcohol), and sterile draping of the patient.


Postpartum back pain is common after childbirth regardless of whether neuraxial analgesia has been used. However, no evidence indicates that back pain is more common when neuraxial analgesia is used for labor. Several trials have identified an association between increased maternal temperature and epidural use as a secondary outcome. Attribution of causality is difficult in this setting and the etiology is not well understood but likely involves noninfectious inflammation.




Maternal Comorbidities


Hypertensive Disorders


Hypertensive disorders of pregnancy are among the most common causes of maternal morbidity and are associated with more frequent rates of maternal and fetal mortality. Hypertensive disorders of pregnancy complicate 5% to 10% of pregnancies worldwide and preeclampsia is diagnosed in 3% of pregnancies. The World Health Organization (WHO) has identified hypertension as the second most common cause of maternal death, accounting for 14% of mortality associated with pregnancy. Chronic hypertension may precede pregnancy and may or may not be complicated by superimposed preeclampsia.


Although some variations exist in hypertensive definitions worldwide, the following are used in the United States by the ACOG based on a working group in 2013. Gestational hypertension is defined as the onset of hypertension (systolic blood pressure [SBP] >140 mm Hg or diastolic blood pressure [DBP] >90 mm Hg) after 20 weeks’ gestation in a previously normotensive parturient without proteinuria. Preeclampsia is defined as hypertension (SBP >140 mm Hg or DBP >90 mm Hg) after 20 weeks’ gestation associated with proteinuria. Preeclampsia is diagnosed when urine protein excretion is greater than 300 mg in a 24 hour period or, alternatively, there is a protein/creatinine ratio of at least 0.3. In 2013, massive proteinuria (>5 g in 24 hours) and fetal growth restriction were eliminated as considerations of severe preeclampsia. In addition, the term mild preeclampsia is no longer used. Now only preeclampsia or preeclampsia with severe features are defined. Severe features of preeclampsia include an SBP of 160 mm Hg or greater or DBP of 110 mm Hg or greater on two separate occasions at least 4 hours apart while on bed rest; thrombocytopenia (platelet count less than 100,000/μL); impaired liver function with twice normal concentrations of liver enzymes; right upper quadrant pain; progressive renal insufficiency with serum creatinine greater than 1.1 mg/dL or a doubling of serum creatinine without other known renal disease; pulmonary edema; and new onset cerebral or visual abnormalities. In the absence of proteinuria, preeclampsia can be diagnosed with new onset hypertension (as previously defined) and the presence of a severe feature.


The combination of hemolysis, elevated liver enzyme, and low platelet count (HELLP) is considered preeclampsia with HELLP syndrome. Eclampsia is preeclampsia complicated by seizure activity. The incidence of preeclampsia has increased likely as a result of increases in maternal age and obesity, whereas the risk for eclampsia has decreased because of improved prenatal care and the use of prophylactic intravenous magnesium. The cause of preeclampsia is not known, but it is generally thought that placental insufficiency through a cascade of antiangiogenic factors (e.g., s-Flt) eventually leads to maternal generalized endothelial dysfunction. The cause of the placental insufficiency is likely variable and could include maternal or paternal genetics and/or environmental factors. Patients with preeclampsia have an elevated risk for cerebral hemorrhage, pulmonary edema, and coagulopathy. Current guidelines recommend treating SBP more than 160 mm Hg for prevention of intracerebral hemorrhage. Initial treatment typically includes intravenous labetalol, hydralazine, and/or oral nifedipine. Other considerations are increased airway edema with associated difficulty of intubation and increased rates of postpartum atony related to the use of magnesium sulfate. Methylergonovine (Methergine) should be used cautiously in patients with preeclampsia because it may lead to hypertensive crisis. Women with preeclampsia can be sensitive to both endogenous and exogenous catecholamines. Therefore careful administration of adrenergic agents is recommended.


Women with a diagnosis of preeclampsia should have their platelet count checked before initiation of regional anesthesia or removal of an epidural catheter. Coagulopathy is a contraindication to regional anesthesia. Although the risk for spinal hematoma is lower in pregnant women than in the elderly, one study found 68% of patients who had spinal hematomas after neuraxial blockade had preexisting coagulopathy. Lastly, despite the concerns for hypotension, spinal anesthesia is safe to administer in preeclamptic patients.


Coagulopathies


Thrombocytopenia complicates 10% of pregnancies as a result of several etiologic factors. Thrombocytopenia may be preexisting or can develop as a result of the pregnancy. As discussed earlier, thrombocytopenia that develops after 20 weeks’ gestation may be a sign of preeclampsia with HELLP syndrome. However, most thrombocytopenia that develops in pregnancy is benign, gestational thrombocytopenia. The platelet count is expected to decrease by approximately 10% in a normal pregnancy. Autoimmune thrombocytopenia, antiphospholipid syndrome, and liver disease are less common. Glucocorticoids and IV immunoglobulin can be used to elevate platelet counts in certain types of severe disease but require several days to be effective. No platelet count is universally accepted as safe for epidural placement. Most anesthesiologists agree that placement of an epidural in the setting of a platelet count greater than 100,000/mm 3 is safe and recent literature suggests lower thresholds may be safe. A retrospective observational study of cases from 14 diverse institutions combined with a systematic review evaluated 1524 parturients with a platelet count of less than 100,000/mm 3 and found the upper bound of the 95% CI for the risk of epidural hematoma for a platelet count of 0 to 49,000/mm 3 is 11%, 50,000 to 69,000/mm 3 is 3%, and 70,000 to 100,000/mm 3 is 0.2%. Of note, no cases of epidural hematomas requiring surgical decompression were identified in this cohort.


Women with von Willebrand disease are at increased risk for bleeding intrapartum and postpartum. Prophylactic treatment is recommended for women with von Willebrand factor (vWF) less than 50 international units/dL. Because of the multiple types and subtypes of von Willebrand disease that have different responses to therapy, it is imperative that hematologic studies be part of the management to help guide the most appropriate therapy. In type I von Willebrand disease, a partial reduction in the quantity of vWF is seen. Women with type I von Willebrand disease usually do not need prophylactic treatment because their vWF typically rises in the third trimester of pregnancy to greater than 50 international units/dL. Desmopressin can be used in women with type 1 von Willebrand disease to release vWF. Type 2 von Willebrand disease is characterized by a defect in the function of vWF, and facilitation of release is not helpful. Parturients with type 3 von Willebrand disease almost always require replacement of vWF before delivery, because they have almost no intrinsic vWF. Although concern should be elevated for epidural hematoma, women with normal factor levels can have regional anesthesia in the setting of a normal platelet count.


Deep venous thrombosis (DVT) and pulmonary embolism are more common in pregnancy as a result of hormonal changes. Significant risk factors are factor V Leiden, prothrombin G20210A, protein S, protein C and antithrombin deficiency, and antiphospholipid antibodies. Patients diagnosed with DVT or pulmonary embolism in pregnancy will need prolonged anticoagulation and will require delivery planning with a brief anticoagulation hiatus for regional anesthesia and delivery. Factor V Leiden is an abnormal variant of factor V that acts as a cofactor that allows activation of thrombin by factor Xa. The factor V Leiden variant cannot be easily degraded by activated protein C and thus leads to hypercoagulability. Patients with a factor V Leiden abnormality may be maintained on anticoagulants for prophylaxis or treatment of DVT and appropriate cessation of anticoagulants must occur prior to placement of neuraxial blockade.


Obesity


Maternal obesity (prepregnancy body mass index ≥30 kg/m 2 ) and metabolic syndrome cause gestational diabetes, newborn hyperglycemia, larger babies, prolonged labor, and cesarean delivery. Cesarean delivery for obese women holds greater risk of mortality. Sleep apnea is more common in obese parturients and is a risk factor for hypoventilation after treatment with systemic opioids and difficult tracheal intubation when general anesthesia is required. Morbidly obese parturients are at increased risk of longer first stage of labor and operative delivery. Epidural anesthesia placement in obese parturients is more difficult and takes longer and they are more likely to require a repeat procedure due to inadequate pain control or failure to achieve bilateral sensory blockade. An early anesthesia consultation is advised for morbidly obese women regardless of planned delivery mode.


Cardiac Disease


The leading cause of maternal mortality in the United States is cardiac disease. In planning an anesthetic for a delivery in a woman with congenital or acquired heart disease, the anesthesia provider must take into consideration the patient’s cardiac lesion or disease state; the normal physiologic changes of pregnancy, labor, and delivery; and the hemodynamic changes of the anesthetic itself. During pregnancy, labor, and delivery, regurgitant valvular lesions are generally tolerated better than stenotic valvular lesions. Pulmonary hypertension, severe left ventricular outflow tract obstruction, moderate to severe mitral stenosis, and cyanotic congenital heart disease all carry significant risk for maternal morbidity and mortality. Multiple risk factors have been identified as increasing a woman’s likelihood of major morbidity and mortality including prior stroke, low ejection fraction, aortopathy, and heart failure symptoms. The American Heart Association, the American College of Cardiology, and the European Society of Cardiology have classified certain conditions or cardiac lesions as high maternal or fetal risk. In comparison to other risk stratification systems, the modified WHO risk stratification system appears to perform best in predicting maternal morbidity or mortality during pregnancy and delivery. Such risk stratification informs the obstetric and anesthesia team as to what resources a parturient with heart disease may require, and thereby whether they need to transfer to a tertiary care facility for delivery.


Increased monitoring including 5-lead ECG or intraarterial blood pressure monitoring may be necessary for labor, especially in women who have a history of tachyarrhythmia or are hemodynamically tenuous. Epidural labor analgesia is recommended for women with heart disease to decrease catecholamine release and eliminate the increased cardiac output and tachycardia attributable to labor pain. The afterload reduction that occurs with epidural analgesia should be followed closely and may need to be countered with a carefully titrated α-adrenergic agonist to prevent tachycardia and ischemia. When cesarean delivery is indicated, it is important to tailor the anesthetic technique to each individual patient. Regional anesthesia is not necessarily contraindicated and should be considered for most patients.


Anticoagulation


Anticoagulation is often required for women with artificial heart valves, certain types of congenital heart disease, pulmonary hypertension, and cardiomyopathy. It is necessary to carefully time stopping anticoagulation to allow for epidural placement and birth while restarting before a clot can form. For this reason, women are usually maintained on heparin at the end of pregnancy, pending delivery, because of its quick offset. If IV unfractionated heparin was used, it should be stopped 4 to 6 hours before the procedure and normal coagulation status (PTT or activated clotting time [ACT]) documented before neuraxial instrumentation or removal of an epidural catheter. If a parturient is maintained on subcutaneous unfractionated heparin, the time interval required to stop prior to a neuraxial procedure corresponds to the dose and activated partial thromboplastin time (aPTT) that can be measured (see Fig. 62.5 A ). If conversion to intravenous heparin cannot take place before labor, warfarin should be stopped and neuraxial analgesia deferred until the PT is within normal limits and the INR is documented at less than 1.5. Low-molecular-weight heparin (LMWH) has been used frequently for prophylaxis of DVT in pregnancy. Unlike unfractionated heparin, the anticoagulant effects of LMWH cannot be reliably measured, and it is not reversible with protamine. It is recommended to wait a prespecified time interval prior to performing a neuraxial procedure, rather than ordering laboratory testing. The decision to place or remove a neuraxial block in a patient on LMWH should correspond to the individual dose and total daily dose (see Fig. 62.5 B ). Nonsteroidal antiinflammatory drugs do not in themselves increase risk for spinal hematoma, but can increase risk in combination with other anticoagulants. If labor begins before neuraxial analgesia can safely be given, intravenous remifentanil and N 2 O are alternatives in some settings.


Pulmonary Disease


As previously detailed in the section on pulmonary changes, adaptations in the respiratory system are required to meet the metabolic demands of the mother and fetus with increases in minute ventilation, decreased O 2 reserve, and increased airway edema most notable.


Asthma is characterized by reversible airway obstruction, airway hyperresponsiveness, and airway inflammation. It is the most common respiratory disease in pregnancy with considerable maternal morbidity. A prospective trial of 1739 pregnant asthmatic patients found those with mild asthma had an exacerbation rate of 12.6% and a hospitalization rate of 2.3%, those with moderate asthma had an exacerbation rate of 25.7% and a hospitalization rate of 6.8%, and those with severe asthma had an exacerbation rate of 51.9% and a hospitalization rate of 26.9%. Bronchodilators and antiinflammatory drugs are generally safe for the fetus and should be used in pregnancy to control asthma. A meta-analysis found that maternal asthma was associated with increased risk of maternal and placental complications including cesarean delivery, gestational diabetes, abruption, and hemorrhage.


Community-acquired pneumonia is the most common nonobstetric infectious complication that results in maternal death. During the 2009 H1N1 influenza epidemic, pregnant women were disproportionately affected. Pregnant women were at increased risk for hospitalization, ICU admission, and death. Preterm delivery is a significant complication of pneumonia even in the setting of antibiotic therapy. Aspiration of gastric contents is also more common during endotracheal intubation in pregnant than in nonpregnant patients because of laxity of the gastroesophageal junction combined with anatomic changes from the expanding uterus. Strict fasting recommendations, rapid-sequence tracheal intubation, and antacids are used before general anesthesia in pregnancy.


Cystic fibrosis is a common autosomal-dominant disorder in women of northern European origin. With improved medical care, women with cystic fibrosis are living to reproductive years and beyond. Pregnancy is uncommon in women with cystic fibrosis (216 pregnancies in 24,000 women), but when it occurs, careful multidisciplinary care is required. The disease is caused by a mutation in the cystic fibrous membrane conductance regulator in epithelial cells, which causes abnormalities in the lungs, pancreas, intestines, and hepatobiliary systems. The major issues in pregnancy are restrictive lung disease and diabetes.


Neurologic Disorders


Multiple sclerosis is a neuroinflammatory disorder that disproportionately affects young women. Pregnancy is associated with a decrease in the incidence of relapse, although the rate of relapse during the first 3 months postpartum is increased in comparison with the year before pregnancy. Multiple sclerosis is a disease of demyelination, and thus theoretic concern exists that local anesthetic toxicity may be enhanced. Cases of worsening symptoms after regional anesthesia have been reported; however, it is hard to impute causality in a relapsing and remitting disease. Nevertheless, the lowest effective concentration possible should be given and vasoconstrictive agents should be avoided. Some recommend epidural instead of intrathecal local anesthetic administration when possible.


Neurofibromatosis is an autosomal-dominant disorder that occurs in 1 in 3000 individuals, with variable manifestations. It is characterized by café-au-lait lesions on the skin, cutaneous neurofibromas, Lisch nodules of the iris, bone abnormalities, and tumors of the spinal cord and cranial nerves. Neurofibromas often grow in pregnancy. Disagreement exists as to whether neuraxial anesthesia is contraindicated in women with neurofibromatosis because of the incidence of vascular spinal tumors. Epidural hematoma has been reported in a woman with neurofibromatosis in the setting of a spinal tumor. The hormonal changes of pregnancy may cause tumor growth, and a knowledge of lesion location and current clinical symptoms is needed to avoid instrumentation of tumors and safely deliver neuraxial anesthesia.

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Mar 7, 2020 | Posted by in ANESTHESIA | Comments Off on Anesthesia for Obstetrics

Full access? Get Clinical Tree

Get Clinical Tree app for offline access