A Practical Approach To Obstetric Anesthesia
A Practical Approach To Obstetric Anesthesia
2nd edition
All rights reserved. This book is protected by copyright. No part of this book may be reproduced or
transmitted in any form or by any means, including as photocopies or scanned-in or other electronic copies,
or utilized by any information storage and retrieval system without written permission from the copyright
owner, except for brief quotations embodied in critical articles and reviews. Materials appearing in this
book prepared by individuals as part of their official duties as U.S. government employees are not covered
by the above-mentioned copyright. To request permission, please contact Wolters Kluwer at Two
Commerce Square, 2001 Market Street, Philadelphia, PA 19103, via email at [email protected], or via
our website at lww.com (products and services).
9 8 7 6 5 4 3 2 1
Printed in China
This work is provided “as is,” and the publisher disclaims any and all warranties, express or implied,
including any warranties as to accuracy, comprehensiveness, or currency of the content of this work.
This work is no substitute for individual patient assessment based on healthcare professionals’
examination of each patient and consideration of, among other things, age, weight, gender, current or prior
medical conditions, medication history, laboratory data, and other factors unique to the patient. The
publisher does not provide medical advice or guidance, and this work is merely a reference tool. Healthcare
professionals, and not the publisher, are solely responsible for the use of this work including all medical
judgments and for any resulting diagnosis and treatments.
Given continuous, rapid advances in medical science and health information, independent professional
verification of medical diagnoses, indications, appropriate pharmaceutical selections and dosages, and
treatment options should be made and healthcare professionals should consult a variety of sources. When
prescribing medication, healthcare professionals are advised to consult the product information sheet (the
manufacturer’s package insert) accompanying each drug to verify, among other things, conditions of use,
warnings and side effects and identify any changes in dosage schedule or contraindications, particularly if
the medication to be administered is new, infrequently used, or has a narrow therapeutic range. To the
maximum extent permitted under applicable law, no responsibility is assumed by the publisher for any
injury and/or damage to persons or property, as a matter of products liability, negligence law or otherwise,
or from any reference to or use by any person of this work.
LWW.com
Dedication
To the memory of Dr. Geraldine O’Sullivan
Preface
A PRACTICAL APPROACH TO OBSTETRIC ANESTHESIA, 2ND EDITION
This book would not have been possible without the commitment and hard work
of more than 40 contributors as well as the production staff at Wolters Kluwer.
Acknowledgments
I would like to dedicate this book to the memory of Sol Shnider, who started me
on my career in obstetric anesthesia; to my parents, Roy and Mary Lee, whose
resources and encouragement helped make my career in medicine a reality; to
my children, Charles and Katherine, who have honored me by pursuing careers
as physicians; and to my wife Mary, whose love and support I receive daily.
Curtis L. Baysinger
I would like to dedicate this book to my husband, Uli, my parents, and all of the
learners who study obstetric anesthesiology.
Brenda A. Bucklin
David R. Gambling
Contributors
Bryan S. Ahlgren, DO
Assistant Professor
University of Colorado School of Medicine
Aurora, Colorado
Curtis L. Baysinger, MD
Professor of Anesthesiology
Division of Obstetric Anesthesia
Department of Anesthesiology
Vanderbilt University School of Medicine
Nashville, Tennessee
Yaakov Beilin, MD
Professor of Anesthesiology and Obstetrics
Gynecology and Reproductive Science
Vice Chair for Quality
Department of Anesthesiology
Director, Obstetric Anesthesiology
Icahn School of Medicine at Mount Sinai
New York, New York
Jessica Booth, MD
Assistant Professor
Department of Anesthesiology
Wake Forest School of Medicine
Winston-Salem, North Carolina
Brenda A. Bucklin, MD
Professor of Anesthesiology
Assistant Dean, Clinical Core Curriculum
University of Colorado School of Medicine
Aurora, Colorado
William Camann, MD
Director, Obstetric Anesthesia Service
Brigham and Women’s Hospital
Associate Professor
Harvard Medical School
Boston, Massachusetts
Christopher R. Cambic, MD
Assistant Professor
Department of Anesthesiology
Northwestern University Feinberg School of Medicine
Chicago, Illinois
Laurie A. Chalifoux, MD
Assistant Professor of Anesthesiology
Northwestern University Feinberg School of Medicine
Chicago, Illinois
Debnath Chatterjee, MD
Associate Professor of Anesthesiology
Children’s Hospital Colorado/University of Colorado School of Medicine
Aurora, Colorado
Mary DiMiceli-Zsigmond, MD
Assistant Professor of Anesthesiology
Vanderbilt University School of Medicine
Nashville, Tennessee
Lior Drukker, MD
Lecturer
Department of Obstetrics and Gynecology
Shaare Zedek Medical Center
Hebrew University Medical School
Jerusalem, Israel
Tammy Y. Euliano, MD
Professor of Anesthesiology and Obstetrics & Gynecology
University of Florida College of Medicine
Gainesville, Florida
Roshan Fernando, MB, BCh, FRCA
Consultant Anesthetist
University College London Hospitals NHS Foundation Trust
London, United Kingdom
Andrew Geller, MD
Assistant Professor and Assistant Fellowship Director
Department of Anesthesiology
Cedars-Sinai Medical Center
Los Angeles, California
Stephanie R. Goodman, MD
Professor of Anesthesiology
Department of Anesthesiology
Columbia University Medical Center
New York, New York
Joy L. Hawkins, MD
Professor of Anesthesiology
Director of Obstetric Anesthesia
University of Colorado School of Medicine
Aurora, Colorado
Jennifer Hofer, MD
Assistant Professor
Department of Anesthesia & Critical Care
University of Chicago
Chicago, Illinois
Rachel M. Kacmar, MD
Assistant Professor
Department of Anesthesiology
University of Colorado School of Medicine
Aurora, Colorado
Ellen M. Lockhart, MD
Associate Professor and Vice Chairman
Department of Anesthesiology
Washington University School of Medicine
St. Louis, Missouri
Janine Malcolm, MD
Assistant Professor
Department of Endocrinology and Metabolism
University of Ottawa Hospital
Endocrinologist
Department of Endocrinology and Metabolism
The Ottawa Hospital
Ottawa, Ontario, Canada
Jill M. Mhyre, MD
Associate Professor of Anesthesiology
University of Arkansas for Medical Sciences
Little Rock, Arkansas
Rebecca D. Minehart, MD
Director, CA-1 Tutorial Simulation
Department of Anesthesia, Critical Care & Pain Medicine
Massachusetts General Hospital
Boston, Massachusetts
Dominique Moffitt, MD
Instructor in Anaesthesia
Harvard Medical School
Brigham and Women’s Hospital
Boston, Massachusetts
Richard A. Month, MD
Assistant Professor of Clinical Anesthesiology and Interim Chief of Obstetric
Anesthesia
Department of Anesthesiology and Critical Care
University of Pennsylvania Health System
Philadelphia, Pennsylvania
Uma Munnur, MD
Associate Professor
Baylor College of Medicine
Anesthesiologist
Ben Taub Hospital
Houston, Texas
Quisqueya T. Palacios, MD
Associate Professor
Department of Anesthesiology
Associate Professor
Department of Obstetrics and Gynecology
Baylor College of Medicine
Houston, Texas
Peter H. Pan, MD
Professor
Obstetrical and Gynecological Anesthesiology
Wake Forest Baptist Medical Center
Winston-Salem, North Carolina
Michael G. Richardson, MD
Associate Professor of Anesthesiology
Director, Obstetric Anesthesiology Fellowship Program
Vanderbilt University School of Medicine
Nashville, Tennessee
Barbara M. Scavone, MD
Professor
Department of Anesthesia & Critical Care
Professor
Department of Obstetrics & Gynecology
University of Chicago
Chicago, Illinois
Scott Segal, MD
Professor and Chair
Department of Anesthesiology
Wake Forest University School of Medicine
Winston-Salem, North Carolina
Hen Y. Sela, MD
Attending Physician
Department of Obstetrics and Gynecology
Shaare Zedek Medical Center
Hebrew University School of Medicine
Jerusalem, Israel
Adjunct Professor
Division of Maternal Fetal Medicine
Department of Obstetrics and Gynecology
Columbia University Medical Center
New York, New York
Yelena Spitzer
Instructor of Anesthesiology
Albert Einstein School of Medicine
Yeshiva University
New York, New York
John T. Sullivan, MD
Professor
Department of Anesthesiology
Northwestern University Feinberg School of Medicine
Chicago, Illinois
William J. Sullivan, QC, LLB, MCL
Adjunct Professor
Faculty of Medicine
University of British Columbia
Partner
Guild Yule LLP Barristers and Solicitors
Vancouver, British Columbia, Canada
Maya S. Suresh, MD
Professor & Chairman
Department of Anesthesiology
Baylor College of Medicine
Chief of Anesthesiology
Ben Taub Hospital
Houston, Texas
Andrea J. Traynor, MD
Clinical Associate Professor
Department of Anesthesiology
Stanford University School of Medicine
Stanford, California
Nathaen S. Weitzel, MD
Associate Professor
Department of Anesthesiology
University of Colorado Hospital
Aurora, Colorado
David Wlody, MD
Professor of Clinical Anesthesiology
SUNY, Downstate Medical Center
Brooklyn, New York
Cynthia A. Wong, MD
Professor and Vice Chair
Department of Anesthesiology
Northwestern University Feinberg School of Medicine
Chicago, Illinois
Mark I. Zakowski, MD
Associate Professor of Anesthesiology
Adjunct
Charles R. Drew University of Medicine and Science
Chief, Obstetric Anesthesiology
Cedars-Sinai Medical Center
Los Angeles, California
Kathryn J. Zuspan, MD
Obstetric Anesthesiologist
Edina, Minnesota
Contents
Preface
Acknowledgments
Contributors
4. Obstetric Medications
Ruchira Patel and Mrinalini Balki
24. Thrombophilias/Coagulopathies
James P.R. Brown and M. Joanne Douglas
Index
Pharmacology
and Physiology
Physiologic Changes of Pregnancy
Rachel M. Kacmar and Andrea J. Traynor
I. Cardiovascular system
A. Central hemodynamic changes
B. Electrocardiogram changes and rhythm disturbances
C. Aortocaval compression
II. Respiratory system
A. Arterial blood gases
B. Lung volumes and capacities and respiratory mechanics
C. Mechanisms of hypoxemia in pregnancy
D. Upper airway changes
E. Respiratory consequences of uncontrolled maternal pain
F. Oxygen delivery
III. Hematologic system
A. Dilutional anemia
B. Platelet count and function
C. Coagulation factors
D. Leukocytes and immune function
IV. Gastrointestinal system
A. Gastric position and pressure
B. Lower esophageal sphincter tone
C. Gastric secretion
D. Gastric emptying
V. Hepatic function
A. Increase in serum estrogen and progesterone
B. Hepatic blood flow
C. Increase in splanchnic, portal, and esophageal venous
pressure
D. Serum albumin concentration
VI. Renal system
A. Anatomic changes/alterations in renal blood flow
B. Changes in glomerular filtration rate/measures of renal
function
VII. Endocrine system
A. Thyroid function
B. Pancreatic function and glucose metabolism
C. Pituitary function
VIII. Musculoskeletal
A. Lumbar lordosis
B. Joint mobility
IX. Central nervous system
A. Inhalation anesthetics and minimum alveolar concentration
B. Neuraxial anesthesia/local anesthetics
X. Anesthetic implications of maternal physiologic changes during
pregnancy
A. Increased minute ventilation and reduced functional residual
capacity
B. Hypoxemia
C. Aortocaval compression
D. Upper airway changes
E. Endotracheal intubation
F. Use of muscle relaxants
G. Replacement of blood loss
H. Subarachnoid and epidural doses
I. Morbidity and mortality
KEYPOINTS
1. Aortocaval compression occurs as the uterus enlarges beyond the pelvis
at 16 to 20 weeks’ gestation and is responsible for decreased venous
return and cardiac output in the supine position.
2. Decreased functional residual capacity due to atelectasis from the
enlarging uterus and increased oxygen consumption render the parturient
particularly susceptible to periods of apnea.
3. Increases in progesterone result in decreased lower esophageal sphincter
tone and render the parturient at high risk for aspiration of gastric
contents.
4. Renal blood flow increases during pregnancy, resulting in increased
glomerular filtration rate. This leads to increased creatinine clearance and
decreased blood urea nitrogen (BUN) and creatinine (normal ~0.5 to 0.6
mg per dL at term).
5. Pain tolerance increases throughout pregnancy. Hormonal changes lead to
decreases in anesthetic requirements and an approximately 30% decrease
in minimum alveolar concentration (MAC).
REFERENCES
1. Scott DE. Anemia in pregnancy. Obstet Gynecol Annu. 1972;1:219–244.
2. Ueland K. Maternal cardiovascular dynamics. VII. Intrapartum blood volume changes. Am J Obstet
Gynecol. 1976;126: 671–677.
3. Recommendations to prevent and control iron deficiency in the United States. Centers for Disease
Control and Prevention. MMWR Recomm Rep. 1998;47:1–29.
4. Clark SL, Cotton DB, Lee W, et al. Central hemodynamic assessment of normal term pregnancy. Am J
Obstet Gynecol. 1989;161:1439–1442.
5. Clapp JF III, Capeless E. Cardiovascular function before, during, and after the first and subsequent
pregnancies. Am J Cardiol. 1997;80:1469–1473.
6. Kametas NA, McAuliffe F, Hancock J, et al. Maternal left ventricular mass and diastolic function
during pregnancy. Ultrasound Obstet Gynecol. 2001;18:460–466.
7. Schannwell CM, Zimmermann T, Schneppenheim M, et al. Left ventricular hypertrophy and diastolic
dysfunction in healthy pregnant women. Cardiology. 2002;97:73–78.
8. Gaiser R. Physiologic changes of pregnancy. In: Chestnut DH, Wong CA, Tsen LC, et al, eds.
Chestnut’s Obstetric Anesthesia: Principles and Practice. 5th ed. Philadelphia, PA: Elsevier Science,
Mosby; 2014:15–36.
9. Cutforth R, MacDonald CB. Heart sounds and murmurs in pregnancy. Am Heart J. 1966;71:741–747.
10. Shotan A, Ostrzega E, Mehra A, et al. Incidence of arrhythmias in normal pregnancy and relation to
palpitations, dizziness, and syncope. Am J Cardiol. 1997;79:1061–1064.
11. Lee SW, Khaw KS, Ngan Kee WD, et al. Haemodynamic effects from aortocaval compression at
different angles of lateral tilt in non-labouring term pregnant women. Br J Anaesth. 2012;109:950–
956.
12. Lyons HA, Antonio R. The sensitivity of the respiratory center in pregnancy and after the
administration of progesterone. Trans Assoc Am Physicians. 1959;72:173–180.
13. Jensen D, Duffin J, Lam YM, et al. Physiological mechanisms of hyperventilation during human
pregnancy. Respir Physiol Neurobiol. 2008;161:76–86.
14. Lim VS, Katz AI, Lindheimer MD. Acid-base regulation in pregnancy. Am J Physiol. 1976;231:1764–
1769.
15. Conklin KA. Maternal physiological adaptations during gestation, labor and the puerperium. Semin
Anesth. 1991;10: 221–234.
16. Crapo RO. Normal cardiopulmonary physiology during pregnancy. Clin Obstet Gynecol. 1996;39:3–
16.
17. Prowse CM, Gaensler EA. Respiratory and acid-base changes during pregnancy. Anesthesiology.
1965;26:381–392.
18. McClelland SH, Bogod DG, Hardman JG. Apnoea in pregnancy: an investigation using physiological
modelling. Anaesthesia. 2008;63:264–269.
19. Hagerdal M, Morgan CW, Sumner AE, et al. Minute ventilation and oxygen consumption during labor
with epidural analgesia. Anesthesiology. 1983;59:425–427.
20. Spätling L, Fallenstein F, Huch A, et al. The variability of cardiopulmonary adaptation to pregnancy at
rest and during exercise. Br J Obstet Gynaecol. 1992;99(suppl 8):1–40.
21. Kambam JR, Handte RE, Brown WU, et al. Effect of normal and preeclamptic pregnancies on the
oxyhemoglobin dissociation curve. Anesthesiology. 1986;65:426–427.
22. Burrows RF, Kelton JG. Incidentally detected thrombocytopenia in healthy mothers and their infants.
N Engl J Med. 1988;319:142–145.
23. Franchini M. Haemostasis and pregnancy. Thromb Haemost. 2006;95:401–413.
24. Lockwood CJ. Pregnancy-associated changes in the hemostatic system. Clin Obstet Gynecol.
2006;49:836–843.
25. Rosenkranz A, Hiden M, Leschnik B, et al. Calibrated automated thrombin generation in normal
uncomplicated pregnancy. Thromb Haemost. 2008;99:331–337.
26. Stirrat GM. Pregnancy and immunity. BMJ. 1994;308:1385–1386.
27. Shah S, Nathan L, Singh R, et al. E2 and not P4 increases NO release from NANC nerves of the
gastrointestinal tract: implications in pregnancy. Am J Physiol Regul Integr Comp Physiol.
2001;280:R1546–R1554.
28. Murray FA, Erskine JP, Fielding J. Gastric secretion in pregnancy. J Obstet Gynaecol Br Emp.
1957;64:373–381.
29. Cohen SE, Jasson J, Talafre ML, et al. Does metoclopramide decrease the volume of gastric contents
in patients undergoing cesarean section? Anesthesiology. 1984;61:604–607.
30. Wyner J, Cohen SE. Gastric volume in early pregnancy: effect of metoclopramide. Anesthesiology.
1982;57:209–212.
31. Wong CA, McCarthy RJ, Fitzgerald PC, et al. Gastric emptying of water in obese pregnant women at
term. Anesth Analg. 2007;105:751–755.
32. Whitehead EM, Smith M, Dean Y, et al. An evaluation of gastric emptying times in pregnancy and the
puerperium. Anaesthesia. 1993;48:53–57.
33. Carp H, Jayaram A, Stoll M. Ultrasound examination of the stomach contents of parturients. Anesth
Analg. 1992;74: 683–687.
34. Porter JS, Bonello E, Reynolds F. The influence of epidural administration of fentanyl infusion on
gastric emptying in labour. Anaesthesia. 1997;52:1151–1156.
35. Kelly MC, Carabine UA, Hill DA, et al. A comparison of the effect of intrathecal and extradural
fentanyl on gastric emptying in laboring women. Anesth Analg. 1997;85:834–838.
36. Zimmermann DL, Breen TW, Fick G. Adding fentanyl 0.0002% to epidural bupivacaine 0.125% does
not delay gastric emptying in laboring parturients. Anesth Analg. 1996;82:612–616.
37. American Society of Anesthesiologists. Practice guidelines for obstetric anesthesia: an updated report
by the American Society of Anesthesiologists Task Force on Obstetric Anesthesia. Anesthesiology.
2007;106:843–863.
38. Angel Garcia AL. Effect of pregnancy on pre-existing liver disease physiological changes during
pregnancy. Ann Hepatol. 2006;5:184–186.
39. Paech MJ, Scott K. Liver and renal disease. In: Gambling DR, Douglas MJ, McKay RSF, eds.
Obstetric Anesthesia and Uncommon Disorders. 2nd ed. Cambridge, United Kingdom: Cambridge
University Press; 2008:249–257.
40. Carter J. Liver function in normal pregnancy. Aust N Z J Obstet Gynaecol. 1990;30:296–302.
41. Bacq Y, Zarka O, Bréchot JF, et al. Liver function tests in normal pregnancy: a prospective study of
103 pregnant women and 103 matched controls. Hepatology. 1996;23:1030–1034.
42. Jeyabalan A, Lain KY. Anatomic and functional changes of the upper urinary tract during pregnancy.
Urol Clin North Am. 2007;34:1–6.
43. Jeyabalan A, Conrad KP. Renal function during normal pregnancy and preeclampsia. Front Biosci.
2007;12:2425–2437.
44. Klein P, Polidori D, Twito O, et al. Impaired decline in renal threshold for glucose during pregnancy—
a possible novel mechanism for gestational diabetes mellitus. Diabetes Metab Res Rev. 2014;30:140–
145.
45. Dafnis E, Sabatini S. The effect of pregnancy on renal function: physiology and pathophysiology. Am
J Med Sci. 1992;303: 184–205.
46. Marwaha RK, Chopra S, Gopalakrishnan S, et al. Establishment of reference range for thyroid
hormones in normal pregnant Indian women. BJOG. 2008;115:602–606.
47. Soldin OP, Tractenberg RE, Hollowell JG, et al. Trimester-specific changes in maternal thyroid
hormone, thyrotropin, and thyroglobulin concentrations during gestation: trends and associations
across trimesters in iodine sufficiency. Thyroid. 2004;14:1084–1090.
48. Casey BM, Dashe JS, Wells CE, et al. Subclinical hyperthyroidism and pregnancy outcomes. Obstet
Gynecol. 2006;107: 337–341.
49. Scheithauer BW, Sano T, Kovacs KT, et al. The pituitary gland in pregnancy: a clinicopathologic and
immunohistochemical study of 69 cases. Mayo Clin Proc. 1990;65:461–474.
50. Grattan DR, Steyn FJ, Kokay IC, et al. Pregnancy-induced adaptation in the neuroendocrine control of
prolactin secretion. J Neuroendocrinol. 2008;20:497–507.
51. Russell JA, Brunton PJ. Neuroactive steroids attenuate oxytocin stress responses in late pregnancy.
Neuroscience. 2006;138:879–889.
52. Chan MT, Mainland P, Gin T. Minimum alveolar concentration of halothane and enflurane are
decreased in early pregnancy. Anesthesiology. 1996;85:782–786.
53. Abboud TK, Sarkis F, Hung TT, et al. Effects of epidural anesthesia during labor on maternal plasma
beta-endorphin levels. Anesthesiology. 1983;59:1–5.
54. Datta S, Hurley RJ, Naulty JS, et al. Plasma and cerebrospinal fluid progesterone concentrations in
pregnant and nonpregnant women. Anesth Analg. 1986;65:950–954.
55. Christensen JH, Andreasen F, Jansen JA. Pharmacokinetics of thiopental in caesarian section. Acta
Anaesthesiol Scand. 1981;25:174–179.
56. Igarashi T, Hirabayashi Y, Shimizu R, et al. The fiberscopic findings of the epidural space in pregnant
women. Anesthesiology. 2000;92:1631–1636.
57. Kerr MG. The mechanical effects of the gravid uterus in late pregnancy. J Obstet Gynaecol Br
Commonw. 1965;72:513–529.
58. Flanagan HL, Datta S, Lambert DH, et al. Effect of pregnancy on bupivacaine-induced conduction
blockade in the isolated rabbit vagus nerve. Anesth Analg. 1987;66:123–126.
59. Bader AM, Datta S, Moller RA, et al. Acute progesterone treatment has no effect on bupivacaine-
induced conduction blockade in the isolated rabbit vagus nerve. Anesth Analg. 1990;71:545–548.
60. Ohel I, Walfisch A, Shitenberg D, et al. A rise in pain threshold during labor: a prospective clinical
trial. Pain. 2007;132(suppl 1):S104–S108.
61. Kodali BS, Chandrasekhar S, Bulich LN, et al. Airway changes during labor and delivery.
Anesthesiology. 2008;108:357–362.
62. Cortet M, Deneux-Tharaux C, Dupont C, et al. Association between fibrinogen level and severity of
postpartum haemorrhage: secondary analysis of a prospective trial. Br J Anaesth. 2012;108:984–989.
63. Abouleish EI. Postpartum tubal ligation requires more bupivacaine for spinal anesthesia than does
cesarean section. Anesth Analg. 1986;65:897–900.
64. Mhyre JM, Riesner MN, Polley LS, et al. A series of anesthesia-related maternal deaths in Michigan,
1985-2003. Anesthesiology. 2007;106:1096–1104.
65. Hawkins JL, Chang J, Palmer SK, et al. Anesthesia-related maternal mortality in the United States
1979-2002. Obstetr Gynecol. 2011;117:69–74.
66. D’Angelo R, Smiley RM, Riley ET, et al. Serious complications related to obstetric anesthesia: the
serious complication repository project of the Society for Obstetric Anesthesia and Perinatology.
Anesthesiology. 2014;120:1505–1512.
Uteroplacental Anatomy, Blood Flow, Respiratory Gas
Exchange, Drug Transfer, and Teratogenicity
Curtis L. Baysinger and Barton Staat
I. Anatomy
A. Anatomic and physiologic changes
B. Human placenta
C. Fetal circulation
II. Uteroplacental circulation
A. Placental circulatory development
B. Uterine blood flow
C. Umbilical blood flow
III. Respiratory gas exchange
A. Oxygen transfer
B. Carbon dioxide transfer
IV. Nutrient/drug transfer
A. Mechanisms of exchange
B. Drug transfer
C. Nutrient transfer
V. Teratogenicity
A. Classification of birth defects
B. Etiology of congenital malformations
C. Principles of teratology
D. Drug teratogenicity
KEYPOINTS
1. Placental growth depends on changes in the maternal circulation in
response to the metabolic demands associated with fetal growth.
2. The uterine artery primarily, and the ovarian artery secondarily, provide
maternal blood to the uterus and branch into the spiral arteries. These
provide blood to the intervillous space where maternal and fetal gas,
nutrient, and waste exchange occurs.
3. Successful placental and fetal development depends on maternal spiral
artery remodeling early in gestation, which converts the maternal
placental circulation into a low-resistance, high-flow circulatory system
that is maximally vasodilated.
4. Few mechanisms increase uterine blood flow; therefore, clinicians should
avoid interventions that decrease uterine blood flow.
5. Oxygen and carbon dioxide exchange are uterine blood flow dependent,
thus emphasizing the need to maintain adequate uterine perfusion for
adequate gas transfer.
6. Fetal compensatory mechanisms allow the fetus to tolerate reductions in
uterine blood flow for long periods of time without significant long-term
injury.
7. Most drugs cross the placenta by diffusion down a concentration gradient
from mother to fetus or vice versa. Increases in maternal–fetal gradient,
decreased maternal/increased fetal protein binding, lower molecular
weight, less ionization, and greater lipid solubility promote maternal to
fetal transfer.
8. Anesthetic agents are not teratogens. Identification of drugs that are weak
teratogens may require an extensive clinical experience in their use
before they can be identified.
B. Uterine blood flow (see Fig. 2.2). At term, uterine blood flow
(UBF) is approximately 700 mL per minute of which
approximately 70% to 90% passes through the intervillous space.
The rest supplies the metabolic demands of the myometrium.9
A. Oxygen transfer
1. The main determinant of oxygen delivery is maternal
blood flow, despite the barrier to oxygen diffusion.18 As a
result, increasing maternal PaO2 has little effect on increasing
fetal PaO2 (see Fig. 2.3).18 The normal placenta can
compensate for decreases in flow up to 50% without
decreasing fetal oxygen transfer.17
2. Oxygen content in fetal blood is enhanced by the high fetal
hemoglobin (Hgb) concentration (17 g per 100 mL fetus vs.
12 g per 100 mL mother) and higher affinity of fetal Hgb for
oxygen (P50 fetal Hgb is 18 mm Hg vs. P50 of 27 mm Hg
adult Hgb).19
3. The oxygen binding of Hgb is reduced by acidosis (Bohr
effect). As carbon dioxide diffuses from the fetus to the
mother, creating a relative maternal acidosis, maternal Hgb
releases oxygen more readily. Because the opposite is
occurring to fetal Hgb, this is often termed the “double Bohr
effect.”11 However, this effect accounts for only up to 8% of
oxygen transfer.20
4. Fetal compensatory measures to hypoxemia
a. The fetus has an oxygen reserve of only 42 mL; however,
irreversible fetal brain damage does not begin when
oxygen reserves are exhausted in 2 minutes but only after
approximately 10 minutes due to fetal compensatory
mechanisms that redistribute fetal cardiac output to vital
organs. A total cessation of placental blood flow occurs
rarely, except in cases of umbilical cord prolapse or acute
total placental abruption.21
b. Blood flow is redistributed to core organs (e.g., brain,
heart, placenta) by the release of vasopressin by the
fetus.22 Acute small decreases in uterine blood flow
(10%) cause increases in fetal catecholamines that aid in
fetal blood flow redistribution.22 (see Fig. 2.4).
C. Nutrient transfer
1. Glucose is transported by diffusion down the concentration
gradient from mother to fetus and by glucose transporters.
Regulation of transport is by a complicated signaling pathway
relying on mechanistic target of rapamycin complex
(mTORC) activation within the trophoblast.27
2. Amino acid transport relies on diffusion and an active
transport system linked to a Na+/K+ ATPase pump.27
3. Fatty acids transfer to the fetus by diffusion; however,
proteins to which free fatty acids bind are involved in an
active transport mechanism.27
V. Teratogenicity. The ACOG notes that severe congenital anomalies
are found in 3% of live births in the general population and 4.5% of
children manifest birth defects by age 5.28 The etiology of most
congenital malformations is unknown. Understanding how a drug
may cause a structural or functional defect relies on knowledge of the
six basic principles of teratology (see below). Knowledge of drug
teratogenicity is important because nearly 60% of women will take
medications other than vitamins during pregnancy.28 Birth defects
consist of malformations, deformations, and disruptions.
A. Classification of birth defects
1. Malformations are alterations of normal development that
occur as a result of an intrinsic abnormality in the
developmental process.
2. Deformations result from an abnormal mechanical force on
an otherwise normal fetus.
3. Disruptions are due to the disruption of an otherwise normal
developmental process.
B. Etiology of congenital malformations
1. Unknown: 65% to 75%
2. Genetic: 15% to 25%
3. Environmental: 10%
a. Environmental factors include drugs, chemicals, and
physical factors such as ionizing radiation and others.
Prescription medications are responsible for only a small
fraction of congenital malformations.29
C. Principles of teratology. The six principles of teratology form a
foundation of knowledge about the biologic plausibility of a
teratogen28:
1. Genetic susceptibility is the inherent resistance or
susceptibility of an organism to specific teratogens and
depends on the genotype of the fetus as well as the manner in
which it interacts with adverse environmental factors. Fetal
and maternal factors are likely to play a role. Inherited traits
can lead to increased sensitivity. Some of the factors that
affect susceptibility include:
a. Dose of exposure
b. Threshold dose
c. Pharmacokinetics and metabolism of drug/chemical
d. Pregnancy-related physiologic changes
e. Placental transport of substances.30
2. Timing of exposure can be critical when determining if an
effect on the fetus is possible.
a. Gestational age of 2 to 5 weeks is thought to be “all or
none.” Exposure at this time is likely to cause either
spontaneous abortion or no harm. Early in gestation, any
cell can be totipotential. If a single cell is damaged, it can
be replaced by one that is not damaged, but if a critical
number are damaged then the pregnancy is lost.
b. Gestational age of 5 to 10 weeks is critical for
organogenesis. This is the time when most organs are
developing and represents a fairly limited window for
teratogenicity.
c. After 10 weeks, most fetal organs grow and differentiate,
and exposure may decrease growth or decrease
differentiation.31
3. Mechanisms of teratogenesis
Teratogenic agents act in specific ways on developing cells
and tissues to initiate sequences of abnormal developmental
events. When a substance is suspected of being a teratogen, it
is necessary to ask whether there is biologic plausibility for
the production of the specific malformation by that substance.
4. Manifestations. A teratogenic agent is associated with
certain, specific malformations that have been previously
described. The manifestations of deviant development are
death, malformation, growth retardation, and functional
deficit.
5. Agent effects are often dependent on timing, dose, and
interactions.
a. Drugs. Many illicit drugs and alcohol are known
teratogens.29
b. Chemicals
c. Infections
Many infections are known to cause congenital
anomalies. The most common and well known are the
TORCH infections. TORCH refers to toxoplasmosis,
rubella, cytomegalovirus, herpes simplex, and others
including varicella.
d. Maternal diseases can also cause congenital anomalies
with the most common being diabetes mellitus. Poorly
controlled diabetes increases the risk of many fetal
anomalies, including congenital heart defects and sacral
agenesis.
6. Dose effect. The manifestations of abnormal development
increase in frequency and degree as dosage increases. There is
a dosage level below which malformations are not observed.
This is often extrapolated from animal models. A high risk of
teratogenicity is suspected if the dose-producing birth defects
in animals is less than tenfold higher than the maximum
human therapeutic dose. There is low risk if the teratogenic
dose in animals is >100-fold higher than the maximum human
therapeutic dose.32
D. Drug teratogenicity. Since 1975, the U.S. Food and Drug
Administration (FDA) has required that all drugs be
categorized on the basis of their ability to produce birth
defects. In December 2014, the FDA revised the standards for
reporting information about medication use during pregnancy.
This change occurred over several years in response to
widespread criticism of the previous letter classification system.
33 The old letter system (A, B, C, D, X) had serious
REFERENCES
1. Brent RL. Environmental causes of human congenital malformations: the pediatrician’s role in dealing
with these complex clinical problems caused by a multiplicity of environmental and genetic factors.
Pediatrics. 2004;113:957–968.
2. Asśali NS, Nuwayhid B, Zugaib M. Control of the uteroplacental circulation in health and disease. Eur
J Obstet Gynecol Reprod Biol. 1978;8:43–55.
3. Clapp JF III, Capeless E. Cardiovascular function before, during, and after the first and subsequent
pregnancies. Am J Cardiol. 1997;80:1469–1473.
4. Cross JC. Placental function in development and disease. Reprod Fertil Dev. 2006;18:71–76.
5. Syme M, Paxton JW, Keelan JA. Drug transfer and metabolism by the human placenta. Clin
Pharmacokinet. 2004;43:487–514.
6. Kingdom J, Huppertz B, Seaward G, et al. Development of the placental villous tree and its
consequences for fetal growth. Eur J Obstet Gynecol Reprod Biol. 2000;92:35–43.
7. Kaufmann P, Hans-George F. Placental development. In: Polin RA, Fox WW, Abman SH, eds. Fetal
and Neonatal Physiology. 3rd ed. Philadelphia, PA: W.B. Saunders; 2004:85–96.
8. Krause BJ, Hanson MA, Casanello P. Role of nitric oxide in placental vascular development and
function. Placenta. 2011;32:797–805.
9. Konje JC, Kaufmann P, Bell SC, et al. A longitudinal study of quantitative uterine blood flow with use
of color power angiography in appropriate for gestational age pregnancies. Am J Obstet Gynecol.
2001;185:608–613.
10. Laird MR, Faber JJ, Binder ND. Maternal placental blood flow is reduced in proportion to reduction in
uterine driving pressure. Pediatr Res. 1994;36:102–110.
11. Zakowski MI, Ramanathan S. Uteroplacental circulation and respiratory gas exchange. In: Suresh MS,
Segal S, Preston RL, et al, eds. Shnider and Levinson’s Anesthesia for Obstetrics. 5th ed. Philadelphia,
PA: Wolters Kluwer; 2013:19–45.
12. Urban G, Vergani P, Ghidini A, et al. State of the art: non-invasive ultrasound assessment of the
uteroplacental circulation. Semin Perinatol. 2007;31:232–239.
13. Sastry BV. Human placental cholinergic system. Biochem Pharmacol. 1997;53:1577–1586.
14. Ramasubramanian R, Johnson RF, Downing JW, et al. Hypoxemic fetoplacental vasoconstriction: a
graduated response to reduced oxygen conditions in the human placenta. Anesth Analg.
2006;103:439–442.
15. Tuuli M, Odibo AO. The role of serum markers and uterine artery Doppler in identifying at-risk
pregnancies. Clin Perinatol. 2011;38:1–19.
16. Wilkening RB, Meschia G. Fetal oxygen uptake, oxygenation, and acid-base balance as a function of
uterine blood flow. Am J Physiol. 1983;244:H749–H755.
17. Crawford JS. Placental physiology. In: Crawford JS, ed. Principles and Practice of Obstetric
Anesthesia. 5th ed. Boston, MA: Blackwell Science; 1984:101–131.
18. Ramanathan S, Gandhi S, Arismendy J, et al. Oxygen transfer from mother to fetus during cesarean
section under epidural anesthesia. Anesth Analg. 1982;61:576–581.
19. Parer JT. Uteroplacental physiology and exchange. In: Parer JT, ed. Handbook of Fetal Heart Rate
Monitoring. 2nd ed. Philadelphia, PA: W.B. Saunders; 1997:22–43.
20. Hill EP, Power GG, Longo LD. A mathematical model of carbon dioxide transfer in the placenta and
its interaction with oxygen. Am J Physiol. 1973;224:283–299.
21. Myers RE. Two patterns of perinatal brain damage and their conditions of occurrence. Am J Obstet
Gynecol. 1972;112:246–276.
22. Gu W, Jones CT, Parer JT. Metabolic and cardiovascular effects on fetal sheep of sustained reduction
of uterine blood flow. J Physiol. 1985;368:109–129.
23. Zakowski MI, Geller A. The placenta: anatomy, physiology, and transfer of drugs. In: Chestnut DH,
Wong CA, Tsen LC, et al, eds. Chestnut’s Obstetric Anesthesia: Principles and Practice. 5th ed.
Philadelphia, PA: Elsevier Mosby; 2014:55–74.
24. Myllynen P, Pasanen M, Pelkonen O. Human placenta: a human organ for developmental toxicology
research and biomonitoring. Placenta. 2005;26:361–371.
25. Pacifici GM, Nottoli R. Placental transfer of drugs administered to the mother. Clin Pharmacokinet.
1995;28:235–269.
26. Ní Mhuireachtaigh R, O’Gorman DA. Anesthesia in pregnant patients for nonobstetric surgery. J Clin
Anesth. 2006;18:60–66.
27. Larqué E, Ruiz-Palacios M, Koletzko B. Placental regulation of fetal nutrient supply. Curr Opin Clin
Nutr Metab Care. 2013;16:292–297.
28. Niebyl JR, Simpson JL. Drugs and environmental agents in pregnancy and lactation: embryology,
teratology, epidemiology. In: Gabbe SG, ed. Obstetrics: Normal and Problem Pregnancies. 5th ed.
Philadelphia, PA: Elsevier Science; 2007:184–214.
29. Adam MP, Polifka JE, Friedman JM. Evolving knowledge of the teratogenicity of medications in
human pregnancy. Am J Med Genet C Semin Med Genet. 2011;157C:175–182.
30. Gedeon C, Koren G. Designing pregnancy centered medications: drugs which do not cross the human
placenta. Placenta. 2006;27:861–868.
31. Cragan JD, Friedman JM, Holmes LB, et al. Ensuring the safe and effective use of medications during
pregnancy: planning and prevention through preconception care. Matern Child Health J.
2006;10:S129–S135.
32. Brent RL, Fawcett LB. Developmental toxicology, drugs, and fetal teratogenesis. In: Hobbins JC, ed.
Clinical Obstetrics: The Mother and Fetus. 3rd ed. Oxford, United Kingdom: Blackwell Science;
2007:217–291.
33. U.S. Food and Drug Administration. Content and format of labeling for human prescription drug and
biological products; requirements for pregnancy and lactation labeling.
https://www.federalregister.gov/articles/2014/12/04/2014-28241/content-and-format-of-labeling-for-
human-prescription-drug-and-biological-products-requirements-for. Accessed July 30, 2015.
34. Merlob P, Stahl B. Classification of drugs for teratogenic risk: an anachronistic way of counseling.
Teratology. 2002;66:61–62.
35. Bianca S. Drug use during pregnancy: are risk classifications more dangerous than the drugs? Lancet.
2003;362:329.
36. Cheek TG, Baird E. Anesthesia for nonobstetric surgery: maternal and fetal considerations. Clin
Obstet Gynecol. 2009;52:535–545.
37. American College of Obstetricians and Gynecologists. ACOG educational bulletin. Teratology.
Number 236. Int J Gynaecol Obstet. 1997;57:319–326.
38. Reproductive Toxicology Center. Reprotox. http://www.reprotox.org/. Accessed July 30, 2015.
39. TERIS (Teratogen Information System), University of Washington. Clinical tetralogy web: a resource
guide for clinicians. http://depts.washington.edu/terisweb/teris/. Accessed July 30, 2015.
40. Organization for Teratology Information Specialists. Welcome to MotherToBaby.
http://www.mothertobaby.org/. Accessed July 30, 2015.
41. American College of Obstetricians and Gynecologists. WEBTREATS: teratology/toxicology.
http://www.acog.org/About-ACOG/ACOG-Departments/Resource-Center/WEBTREATS-Teratology-
Toxicology. Accessed July 30, 2015.
Local Anesthetics and Toxicity
Jennifer Hofer and Barbara M. Scavone
I. Chemical structure
A. Local anesthetics in common clinical use
B. Amino-ester/amino-amide local anesthetics
C. Stereochemistry
II. Mechanism of action
A. Local anesthetic entry into the cell
B. Local anesthetic binding to the sodium channel
C. Local anesthetic dissociation from binding site
D. Local anesthetic effects on other membrane-bound proteins
III. Differential blockade
A. Physiologic basis
B. Anesthetic implications
IV. Additives
A. Bicarbonate
B. Epinephrine
C. Phenylephrine
D. Opioids
V. Effect of pregnancy on local anesthetic action
VI. Pharmacokinetics
A. Systemic absorption
B. Distribution
C. Clearance
D. Elimination
E. Amide protein binding
F. Local anesthetic continuous infusions
G. Effect of pregnancy on pharmacokinetics
H. Chronobiology
VII. Local anesthetics rapidly cross the placenta
VIII. Systemic toxicity
A. Incidence
B. Signs and symptoms
C. Effect of comorbidities on toxicity
D. Effect of pregnancy on toxicity
E. Prevention
F. Treatment
IX. Other types of reactions may occur
A. Neurotoxicity
B. TNS syndrome
C. Back pain
D. Myotoxic
E. Allergic reactions
KEYPOINTS
1. Lipid solubility, pKa, and protein binding all determine the clinical
profile of local anesthetics.
2. The pKa of any specific local anesthetic partially determines its speed of
onset, in that a molecule with a pKa closer to physiologic pH will be less
ionized and thus have a faster speed of onset than a similar molecule with
a higher pKa.
3. Increased lipid solubility facilitates entry of the local anesthetic molecule
into the cell membrane and increases local anesthetic potency.
4. Moderate lipid solubility aids departure of local anesthetics from the
binding site, but extreme lipid solubility (e.g., bupivacaine) favors
continued binding and increases duration of action.
5. Pregnancy alters local anesthetic neural blockade, susceptibility to
toxicity, and pharmacokinetics.
6. Prevention of local anesthetic systemic toxicity (LAST) includes (1)
frequent aspiration prior to injection, (2) slow incremental injection, (3)
limiting the total dose, and (4) use of a test dose.
7. Rapid treatment of convulsions and cardiovascular toxicity results in
decreased morbidity and mortality from local anesthetic overdose.
8. Treatment of severe local anesthetic toxicity includes lipid emulsion
(20%) therapy.
B. Anesthetic implications
1. Sympathetic blockade exceeds sensory blockade by several
dermatomes during spinal and epidural anesthesia, although
sympathetic block is not always complete.8–10
2. Sensory blockade exceeds motor blockade during spinal
and epidural anesthesia. Epidural labor analgesia with low
concentrations of local anesthetics can be used to provide pain
relief with minimal effects on maternal expulsive efforts.11
3. The Aδ fibers, associated with sharp or fast pain,
demonstrate more susceptibility to local anesthetic effects
than C fibers, associated with burning or slow pain.7
4. Cold temperature–sensing fibers demonstrate more
conduction blockade than pain-sensing fibers.12,13 The degree
of differential blockade is dependent on the concentration of
local anesthetic and volume. The use of less concentrated
solutions and greater volume often produces less motor block
but a more uniform sensory block.
5. A sensory level to pain, as opposed to temperature sensation,
should be used to gauge the adequacy of sensory blockade
during regional analgesia/anesthesia.
IV. Additives. Additives are combined with local anesthetics to achieve
various clinical effects.
A. Bicarbonate. As noted earlier, commonly used local anesthetics
are ionized because they are weak bases with pKa higher than
physiologic pH. Addition of bicarbonate to the local anesthetic
solution (1 mEq/mL) (8.4%) adjusts local anesthetic pH closer
to pKa and increases the ratio of the lipid-soluble form to the
protonated form. This results in less ionization, facilitates drug
entry, and hastens speed of onset.
B. Epinephrine. Epinephrine is an important additive to increase
local anesthetic activity. Besides intensifying local anesthetic–
induced anesthesia and analgesia, epinephrine prolongs the
duration of the block and reduces the systemic absorption of the
local anesthetic. Greater reliability and intensity of the block
are observed when epinephrine is added to epidurally
administered local anesthetics. Epinephrine has intrinsic analgesic
effects that are similar to drugs (e.g., clonidine) which produce
analgesia by stimulation of α2-adrenergic receptors in the spinal
cord. Although epinephrine has a bupivacaine dose-sparing effect
when used as an additive for epidural labor analgesia,14 it is
associated with increased motor block. Epinephrine causes
vasoconstriction and thus reduces clearance of drugs from the
intrathecal and epidural spaces into the central circulation.15
Epinephrine reduces mean peak plasma concentrations of
epidurally administered lidocaine and bupivacaine, which is
significant when considering the potential for local anesthetic
toxicity.16 Intrathecal epinephrine is administered in doses
between 50 and 200 μg and epidurally in doses of 1 to 5 μg per
mL of local anesthetic.
F. Treatment
1. Rapid treatment of convulsions and cardiovascular toxicity
results in decreased morbidity and mortality from local
anesthetic overdose.82 McCutchen and Gerancher83
administered lipid in a suspected case of bupivacaine
intoxication manifested by convulsions and early signs of
cardiovascular toxicity, consequently preventing progression
to full cardiovascular collapse. Therefore, the
anesthesiologist must be prepared for a rapid response to
LAST.
2. Because hypoxemia and/or acidosis enhance both the CNS
and cardiac toxicities of bupivacaine, initial resuscitative
measures must include attention to airway management,
oxygenation, and ventilation.84,85 The aim of ventilation is
to produce normocapnia not hypocapnia.78
3. Treatment of convulsions with small amounts of
benzodiazepines and/or barbiturates with or without
muscle relaxants will aid in airway management efforts and
decrease convulsion-mediated acidosis.
4. Current data support using amiodarone to treat
bupivacaine-induced severe ventricular dysrhythmias.78
Nine of 10 pigs with bupivacaine-induced severe
dysrhythmias survived after resuscitation including
amiodarone versus only 4 of 10 that received bretylium and 6
of 10 controls. The results were not significantly different
between groups, possibly due to small sample size.86 The
mechanism of amiodarone’s benefit in treating local
anesthetic toxicity is twofold: inhibition of outward potassium
channels that prolong repolarization and an antiadrenergic
effect.31,87
5. Lipid emulsions have been used successfully for prevention
and treatment of bupivacaine-induced cardiovascular collapse,
possibly because the lipophilic bupivacaine partitions into
the emulsion (the so-called “lipid sink” hypothesis).88–90 In
addition, lipid emulsion restores myocardial adenosine
triphosphate (ATP) via reversal of local anesthetic inhibition
of myocardial fatty acid oxidation. 31,71,91–93
a. Several case reports describe the successful use of lipid
emulsion during LAST in human patients, including one
description of a parturient.94–96 However, recurrence of
bupivacaine-induced cardiac arrest following lipid therapy
has also been described. This highlights the importance of
prolonged monitoring of patients after LAST.97
b. Optimal timing of lipid therapy remains unanswered.
Current recommendations include immediately
discontinuing administration of the local anesthetic,
treating hypoxia and acidosis with oxygen and ventilation,
and administering benzodiazepines to suppress
seizures.72,98 However, case reports support early lipid
emulsion administration at the first sign of CNS toxicity
to prevent progression to CV collapse,96,98,99 suggesting
improvement in mental status, agitation, or seizures when
lipid emulsion was administered early in the course of
LAST. (Weinberg100 explains that because lipid is not an
energy source for the brain, the reversal of neurologic
symptoms by lipid infusions supports the lipid sink
hypothesis rather than a direct metabolic effect on the
brain.)
c. See Figure 3.2 for recommendations regarding the use
of lipid emulsion for treatment of LAST. Practitioners
should ensure that lipid preparations are available
wherever regional anesthetics are administered.
6. The role of sympathomimetics, epinephrine, and vasopressin
in restoring hemodynamic stability is
controversial.73,78,100–103 The American Heart Association
Advanced Cardiovascular Life Support (ACLS)
Guidelines call for vasopressin administration along with
epinephrine
(http://circ.ahajournals.org/cgi/content/full/112/24_suppl/IV-
58). Although these are first-line drugs in the ACLS
algorithm, the etiology of cardiac arrest is heterogenous, and
when treating cardiac arrest from LAST, the role for
epinephrine and/or vasopressin may be more limited.100
a. In a rabbit model, epinephrine at moderate to high doses
was necessary for the return of spontaneous circulation;
however, the animals subsequently suffered hemodynamic
deterioration.103
b. In a rodent model comparing lipid emulsion alone versus
low-dose epinephrine plus lipid to treat cardiac toxicity, a
delayed but sustained response to spontaneous circulation
was witnessed in the lipid-only group. The return to
spontaneous circulation was faster with the addition of
low-dose epinephrine (10 μg/kg), but with higher
epinephrine doses (>10 μg/kg), resuscitation was
impaired, possibly due to delayed cardiovascular collapse
from hyperlactatemia.
c. The conclusion is that high-dose epinephrine may impair
recovery from local anesthetic-induced cardiac arrest;
however, smaller doses of epinephrine with lipids may
speed early resuscitation by increasing coronary perfusion
pressure and increasing systemic vascular resistance
without having adverse effects on long-term recovery.102
This conclusion is reflected in the 2012 checklist for
treatment of LAST published by the American Society of
Regional Anesthesia and Pain Medicine.15,100
7. In extreme cases, treatment of bupivacaine toxicity may
require cardiopulmonary bypass.104
ACKNOWLEDGMENT
The authors wish to thank Ms. Yvonne Kennedy for her assistance in the
preparation of this manuscript.
REFERENCES
1. Catterall WA, Mackie K. Local anesthetics. In: Brunton LL, Chabner BA, Knollmann BC, eds.
Goodman and Gilman’s The Pharmacological Basis of Therapeutics. 12th ed. New York, NY: The
McGraw-Hill; 2011:565–582.
2. Groban L. Central nervous system and cardiac effects from long-acting amide local anesthetic toxicity
in the intact animal model. Reg Anesth Pain Med. 2003;28:3–11.
3. Butterworth JF IV, Strichartz GR. Molecular mechanisms of local anesthesia: a review.
Anesthesiology. 1990;72:711–734.
4. Tucker GT. Pharmacokinetics of local anaesthetics. Br J Anaesth. 1986;58:717–731.
5. Catterall WA. From ionic currents to molecular mechanisms: the structure and function of voltage-
gated sodium channels. Neuron. 2000;26:13–25.
6. Lin Y, Liu S. Local anesthetics. In: Barash PG, Cullen BF, Stoelting RK, et al, eds. Handbook of
Clinical Anesthesia. 7th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2013:310–324.
7. Huang JH, Thalhammer JG, Raymond SA, et al. Susceptibility to lidocaine of impulses in different
somatosensory afferent fibers of rat sciatic nerve. J Pharmacol Exp Ther. 1997;282:802–811.
8. Cook PR, Malmqvist LA, Bengtsson M, et al. Vagal and sympathetic activity during spinal analgesia.
Acta Anaesthesiol Scand. 1990;34:271–275.
9. Malmqvist LA, Bengtsson M, Björnsson G, et al. Sympathetic activity and haemodynamic variables
during spinal analgesia in man. Acta Anaesthesiol Scand. 1987;31:467–473.
10. Chamberlain DP, Chamberlain BD. Changes in the skin temperature of the trunk and their relationship
to sympathetic blockade during spinal anesthesia. Anesthesiology. 1986;65:139–143.
11. Chestnut DH, Laszewski LJ, Pollack KL, et al. Continuous epidural infusion of 0.0625% bupivacaine-
0.0002% fentanyl during the second stage of labor. Anesthesiology. 1990;72:613–618.
12. Brull SJ, Greene NM. Time-courses of zones of differential sensory blockade during spinal anesthesia
with hyperbaric tetracaine or bupivacaine. Anesth Analg. 1989;69:342–347.
13. Brull SJ, Greene NM. Zones of differential sensory block during extradural anaesthesia. Br J Anaesth.
1991;66:651–655.
14. Polley LS, Columb MO, Naughton NN, et al. Effect of epidural epinephrine on the minimum local
analgesic concentration of epidural bupivacaine in labor. Anesthesiology. 2002;96:1123–1128.
15. Neal JM. Effects of epinephrine in local anesthetics on the central and peripheral nervous systems:
neurotoxicity and neural blood flow. Reg Anesth Pain Med. 2003;28:124–134.
16. Burm AG, van Kleef JW, Gladines MP, et al. Epidural anesthesia with lidocaine and bupivacaine:
effects of epinephrine on the plasma concentration profiles. Anesth Analg. 1986;65:1281–1284.
17. Sakura S, Sumi M, Sakaguchi Y, et al. The addition of phenylephrine contributes to the development
of transient neurologic symptoms after spinal anesthesia with 0.5% tetracaine. Anesthesiology.
1997;87:771–778.
18. Zhan Q, Huang S, Geng G, et al. Comparison of relative potency of intrathecal bupivacaine for motor
block in pregnant versus non-pregnant women. Int J Obstet Anesth. 2011;20:219–223.
19. Hirabayashi Y, Shimizu R, Saitoh K, et al. Cerebrospinal fluid progesterone in pregnant women. Br J
Anaesth. 1995;75:683–687.
20. Lee GY, Kim CH, Chung RK, et al. Spread of subarachnoid sensory block with hyperbaric
bupivacaine in second trimester of pregnancy. J Clin Anesth. 2009;21:482–485.
21. Hawkins JL, Chang J, Palmer SK, et al. Anesthesia-related maternal mortality in the United States:
1979-2002. Obstet Gynecol. 2011;117:69–74.
22. Butterworth JF IV, Walker FO, Lysak SZ. Pregnancy increases median nerve susceptibility to
lidocaine. Anesthesiology. 1990;72:962–965.
23. Flanagan HL, Datta S, Lambert DH, et al. Effect of pregnancy on bupivacaine-induced conduction
blockade in the isolated rabbit vagus nerve. Anesth Analg. 1987;66:123–126.
24. Popitz-Bergez FA, Leeson S, Thalhammer JG, et al. Intraneural lidocaine uptake compared with
analgesic differences between pregnant and nonpregnant rats. Reg Anesth. 1997;22:363–371.
25. Datta S, Lambert DH, Gregus J, et al. Differential sensitivities of mammalian nerve fibers during
pregnancy. Anesth Analg. 1983;62:1070–1072.
26. Hocking G, Wildsmith JA. Intrathecal drug spread. Br J Anaesth. 2004;93:568–578.
27. Abouleish EI. Postpartum tubal ligation requires more bupivacaine for spinal anesthesia than does
cesarean section. Anesth Analg. 1986;65:897–900.
28. Mayumi T, Dohi S, Takahashi T. Plasma concentrations of lidocaine associated with cervical, thoracic,
and lumbar epidural anesthesia. Anesth Analg. 1983;62:578–580.
29. Rosenberg PH, Veering BT, Urmey WF. Maximum recommended doses of local anesthetics: a
multifactorial concept. Reg Anesth Pain Med. 2004;29:564–575.
30. Burm AG, van der Meer AD, van Kleef JW, et al. Pharmacokinetics of the enantiomers of bupivacaine
following intravenous administration of the racemate. Br J Clin Pharmacol. 1994;38:125–129.
31. Dillane D, Finucane BT. Local anesthetic systemic toxicity. Can J Anaesth. 2010;57:368–380.
32. Burm AG, Vermeulen NP, van Kleef JW, et al. Pharmacokinetics of lignocaine and bupivacaine in
surgical patients following epidural administration. Simultaneous investigation of absorption and
disposition kinetics using stable isotopes. Clin Pharmacokinet. 1987;13:191–203.
33. Thomas JM, Schug SA. Recent advances in the pharmacokinetics of local anaesthetics. Long-acting
amide enantiomers and continuous infusions. Clin Pharmacokinet. 1999;36:67–83.
34. Richter O, Klein K, Abel J, et al. The kinetics of bupivacaine (Carbostesin) plasma concentrations
during epidural anesthesia following intraoperative bolus injection and subsequent continuous
infusion. Int J Clin Pharmacol Ther Toxicol. 1984;22:611–617.
35. Emanuelsson BM, Zaric D, Nydahl PA, et al. Pharmacokinetics of ropivacaine and bupivacaine during
21 hours of continuous epidural infusion in healthy male volunteers. Anesth Analg. 1995;81:1163–
1168.
36. Erichsen CJ, Sjövall J, Kehlet H, et al. Pharmacokinetics and analgesic effect of ropivacaine during
continuous epidural infusion for postoperative pain relief. Anesthesiology. 1996;84:834–842.
37. Burm AG, Stienstra R, Brouwer RP, et al. Epidural infusion of ropivacaine for postoperative analgesia
after major orthopedic surgery: pharmacokinetic evaluation. Anesthesiology. 2000;93:395–403.
38. Santos AC, Arthur GR, Lehning EJ, et al. Comparative pharmacokinetics of ropivacaine and
bupivacaine in nonpregnant and pregnant ewes. Anesth Analg, 1997;85:87–93.
39. Debon R, Chassard D, Duflo F, et al. Chronobiology of epidural ropivacaine: variations in the duration
of action related to the hour of administration. Anesthesiology. 2002;96:542–545.
40. Shafer SL, Lemmer B, Boselli E, et al. Pitfalls in chronobiology: a suggested analysis using intrathecal
bupivacaine analgesia as an example. Anesth Analg. 2010;111:980–998.
41. Simpson D, Curran MP, Oldfield V, et al. Ropivacaine: a review of its use in regional anaesthesia and
acute pain management. Drugs. 2005;65:2675–2717.
42. Mulroy MF. Systemic toxicity and cardiotoxicity from local anesthetics: incidence and preventive
measures. Reg Anesth Pain Med. 2002;27:556–561.
43. Hawkins JL, Koonin LM, Palmer SK, et al. Anesthesia-related deaths during obstetric delivery in the
United States, 1979-1990. Anesthesiology. 1997;86:277–284.
44. Bern S, Weinberg G. Local anesthetic toxicity and lipid resuscitation in pregnancy. Curr Opin
Anaesthesiol. 2011;24:262–267.
45. Brown DL, Ransom DM, Hall JA, et al. Regional anesthesia and local anesthetic-induced systemic
toxicity: seizure frequency and accompanying cardiovascular changes. Anesth Analg. 1995;81:321–
328.
46. Auroy Y, Narchi P, Messiah A, et al. Serious complications related to regional anesthesia: results of a
prospective survey in France. Anesthesiology. 1997;87:479–486.
47. Faccenda KA, Finucane BT. Complications of regional anaesthesia incidence and prevention. Drug
Saf. 2001;24:413–442.
48. Mather LE, Copeland SE, Ladd LA. Acute toxicity of local anesthetics: underlying pharmacokinetic
and pharmacodynamic concepts. Reg Anesth Pain Med. 2005;30:553–566.
49. Knudsen K, Beckman Suurküla M, Blomberg S, et al. Central nervous and cardiovascular effects of
i.v. infusions of ropivacaine, bupivacaine and placebo in volunteers. Br J Anaesth. 1997;78:507–514.
50. Rutten AJ, Nancarrow C, Mather LE, et al. Hemodynamic and central nervous system effects of
intravenous bolus doses of lidocaine, bupivacaine, and ropivacaine in sheep. Anesth Analg.
1989;69:291–299.
51. Chang DH, Ladd LA, Copeland S, et al. Direct cardiac effects of intracoronary bupivacaine,
levobupivacaine and ropivacaine in the sheep. Br J Pharmacol. 2001;132:649–658.
52. Reiz S, Häggmark S, Johansson G, et al. Cardiotoxicity of ropivacaine—a new amide local anaesthetic
agent. Acta Anaesthesiol Scand. 1989;33:93–98.
53. Graf BM. The cardiotoxicity of local anesthetics: the place of ropivacaine. Curr Top Med Chem.
2001;1:207–214.
54. Nau C, Strichartz GR. Drug chirality in anesthesia. Anesthesiology. 2002;97:497–502.
55. Moller R, Covino BG. Cardiac electrophysiologic properties of bupivacaine and lidocaine compared
with those of ropivacaine, a new amide local anesthetic. Anesthesiology. 1990;72:322–329.
56. Nancarrow C, Rutten AJ, Runciman WB, et al. Myocardial and cerebral drug concentrations and the
mechanisms of death after fatal intravenous doses of lidocaine, bupivacaine, and ropivacaine in the
sheep. Anesth Analg. 1989;69:276–283.
57. Feldman HS, Arthur GR, Covino BG. Comparative systemic toxicity of convulsant and
supraconvulsant doses of intravenous ropivacaine, bupivacaine, and lidocaine in the conscious dog.
Anesth Analg. 1989;69:794–801.
58. Clarkson CW, Hondeghem LM. Mechanism for bupivacaine depression of cardiac conduction: fast
block of sodium channels during the action potential with slow recovery from block during diastole.
Anesthesiology. 1985;62:396–405.
59. Morrison SG, Dominguez JJ, Frascarolo P, et al. A comparison of the electrocardiographic cardiotoxic
effects of racemic bupivacaine, levobupivacaine, and ropivacaine in anesthetized swine. Anesth Analg.
2000;90:1308–1314.
60. Polley LS, Columb MO, Naughton NN, et al. Relative analgesic potencies of ropivacaine and
bupivacaine for epidural analgesia in labor: implications for therapeutic indexes. Anesthesiology.
1999;90:944–950.
61. Capogna G, Celleno D, Fusco P, et al. Relative potencies of bupivacaine and ropivacaine for analgesia
in labour. Br J Anaesth. 1999;82:371–373.
62. Camorcia M, Capogna G, Columb MO. Minimum local analgesic doses of ropivacaine,
levobupivacaine, and bupivacaine for intrathecal labor analgesia. Anesthesiology. 2005;102:646–650.
63. D’Angelo R, James RL. Is ropivacaine less potent than bupivacaine? Anesthesiology. 1999;90:941–
943.
64. Van de Velde M, Dreelinck R, Dubois J, et al. Determination of the full dose-response relation of
intrathecal bupivacaine, levobupivacaine, and ropivacaine, combined with sufentanil, for labor
analgesia. Anesthesiology. 2007;106:149–156.
65. Stienstra R. The place of ropivacaine in anesthesia. Acta Anaesthesiol Belg. 2003;54:141–148.
66. Dony P, Dewinde V, Vanderick B, et al. The comparative toxicity of ropivacaine and bupivacaine at
equipotent doses in rats. Anesth Analg. 2000;91:1489–1492.
67. Groban L, Deal DD, Vernon JC, et al. Cardiac resuscitation after incremental overdosage with
lidocaine, bupivacaine, levobupivacaine, and ropivacaine in anesthetized dogs. Anesth Analg.
2001;92:37–43.
68. Yoshida M, Matsuda H, Fukuda I, et al. Sudden cardiac arrest during cesarean section due to epidural
anaesthesia using ropivacaine: a case report. Arch Gynecol Obstet. 2008;277:91–94.
69. Chazalon P, Tourtier JP, Villevielle T, et al. Ropivacaine-induced cardiac arrest after peripheral nerve
block: successful resuscitation. Anesthesiology. 2003;99:1449–1451.
70. Klein SM, Pierce T, Rubin Y, et al. Successful resuscitation after ropivacaine-induced ventricular
fibrillation. Anesth Analg. 2003;97:901–903.
71. Butterworth JF IV. Models and mechanisms of local anesthetic cardiac toxicity: a review. Reg Anesth
Pain Med. 2010;35:167–176.
72. Neal JM, Bernards CM, Butterworth JF, et al. ASRA practice advisory on local anesthetic systemic
toxicity. Reg Anesth Pain Med. 2010;35:152–161.
73. Neal JM, Mulroy MF, Weinberg GL, et al. American Society of Regional Anesthesia and Pain
Medicine checklist for managing local anesthetic systemic toxicity: 2012 version. Reg Anesth Pain
Med. 2012;37:16–18.
74. Morishima HO, Pedersen H, Finster M, et al. Bupivacaine toxicity in pregnant and nonpregnant ewes.
Anesthesiology. 1985;63:134–139.
75. Santos AC, Arthur GR, Wlody D, et al. Comparative systemic toxicity of ropivacaine and bupivacaine
in nonpregnant and pregnant ewes. Anesthesiology. 1995;82:734–740.
76. Santos AC, DeArmas PI. Systemic toxicity of levobupivacaine, bupivacaine, and ropivacaine during
continuous intravenous infusion to nonpregnant and pregnant ewes. Anesthesiology. 2001;95:1256–
1264.
77. Mulroy MF. Local anesthetics: helpful science, but don’t forget the basic clinical safety steps. Reg
Anesth Pain Med. 2005;30:513–515.
78. Weinberg GL. Current concepts in resuscitation of patients with local anesthetic cardiac toxicity. Reg
Anesth Pain Med. 2002;27:568–575.
79. Birnbach DJ, Chestnut DH. The epidural test dose in obstetric patients: has it outlived its usefulness?
Anesth Analg. 1999;88:971–972.
80. Guay J. The epidural test dose: a review. Anesth Analg. 2006;102:921–929.
81. Leighton BL, Norris MC, DeSimone CA, et al. The air test as a clinically useful indicator of
intravenously placed epidural catheters. Anesthesiology. 1990;73:610–613.
82. Feldman HS, Arthur GR, Pitkanen M, et al. Treatment of acute systemic toxicity after the rapid
intravenous injection of ropivacaine and bupivacaine in the conscious dog. Anesth Analg.
1991;73:373–384.
83. McCutchen T, Gerancher JC. Early intralipid therapy may have prevented bupivacaine-associated
cardiac arrest. Reg Anesth Pain Med. 2008;33:178–180.
84. Sage DJ, Feldman HS, Arthur GR, et al. Influence of lidocaine and bupivacaine on isolated guinea pig
atria in the presence of acidosis and hypoxia. Anesth Analg. 1984;63:1–7.
85. Heavner JE, Dryden CF Jr, Sanghani V, et al. Severe hypoxia enhances central nervous system and
cardiovascular toxicity of bupivacaine in lightly anesthetized pigs. Anesthesiology. 1992;77:142–147.
86. Haasio J, Pitkänen MT, Kyttä J, et al. Treatment of bupivacaine-induced cardiac arrhythmias in
hypoxic and hypercarbic pigs with amiodarone or bretylium. Reg Anesth. 1990;15:174–179.
87. Corcoran W, Butterworth J, Weller RS, et al. Local anesthetic-induced cardiac toxicity: a survey of
contemporary practice strategies among academic anesthesiology departments. Anesth Analg.
2006;103:1322–1326.
88. Weinberg GL, VadeBoncouer T, Ramaraju GA, et al. Pretreatment or resuscitation with a lipid infusion
shifts the dose-response to bupivacaine-induced asystole in rats. Anesthesiology. 1998;88:1071–1075.
89. Weinberg GL, Ripper R, Murphy P, et al. Lipid infusion accelerates removal of bupivacaine and
recovery from bupivacaine toxicity in the isolated rat heart. Reg Anesth Pain Med. 2006;31:296–303.
90. Weinberg G, Ripper R, Feinstein DL, et al. Lipid emulsion infusion rescues dogs from bupivacaine-
induced cardiac toxicity. Reg Anesth Pain Med. 2003;28:198–202.
91. Picard J, Meek T. Lipid emulsion to treat overdose of local anaesthetic: the gift of the glob.
Anaesthesia. 2006;61:107–109.
92. Shi K, Xia Y, Wang Q, et al. The effect of lipid emulsion on pharmacokinetics and tissue distribution
of bupivacaine in rats. Anesth Analg. 2013;116:804–809.
93. Kuo I, Akpa BS. Validity of the lipid sink as a mechanism for the reversal of local anesthetic systemic
toxicity: a physiologically based pharmacokinetic model study. Anesthesiology. 2013;118:1350–1361.
94. Rosenblatt MA, Abel M, Fischer GW, et al. Successful use of a 20% lipid emulsion to resuscitate a
patient after a presumed bupivacaine-related cardiac arrest. Anesthesiology. 2006;105:217–218.
95. Litz RJ, Popp M, Stehr SN, et al. Successful resuscitation of a patient with ropivacaine-induced
asystole after axillary plexus block using lipid infusion. Anaesthesia. 2006;61:800–801.
96. Spence AG. Lipid reversal of central nervous system symptoms of bupivacaine toxicity.
Anesthesiology. 2007;107:516–517.
97. Marwick PC, Levin AI, Coetzee AR. Recurrence of cardiotoxicity after lipid rescue from bupivacaine-
induced cardiac arrest. Anesth Analg. 2009;108:1344–1346.
98. Wolfe JW, Butterworth JF. Local anesthetic systemic toxicity: update on mechanisms and treatment.
Curr Opin Anaesthesiol. 2011;24:561–566.
99. Litz RJ, Roessel T, Heller AR, et al. Reversal of central nervous system and cardiac toxicity after local
anesthetic intoxication by lipid emulsion injection. Anesth Analg. 2008;106:1575–1577.
100. Weinberg GL. Treatment of local anesthetic systemic toxicity (LAST). Reg Anesth Pain Med.
2010;35:188–193.
101. Di Gregorio G, Schwartz D, Ripper R, et al. Lipid emulsion is superior to vasopressin in a rodent
model of resuscitation from toxin-induced cardiac arrest. Crit Care Med. 2009;37:993–999.
102. Hiller DB, Gregorio GD, Ripper R, et al. Epinephrine impairs lipid resuscitation from bupivacaine
overdose: a threshold effect. Anesthesiology. 2009;111:498–505.
103. Harvey M, Cave G, Prince G, et al. Epinephrine injection in lipid-based resuscitation from
bupivacaine-induced cardiac arrest: transient circulatory return in rabbits. Anesth Analg.
2010;111:791–796.
104. Long WB, Rosenblum S, Grady IP. Successful resuscitation of bupivacaine-induced cardiac arrest
using cardiopulmonary bypass. Anesth Analg. 1989;69:403–406.
105. Li DF, Bahar M, Cole G, et al. Neurological toxicity of the subarachnoid infusion of bupivacaine,
lignocaine or 2-chloroprocaine in the rat. Br J Anaesth. 1985;57:424–429.
106. Lambert LA, Lambert DH, Strichartz GR. Irreversible conduction block in isolated nerve by high
concentrations of local anesthetics. Anesthesiology. 1994;80:1082–1093.
107. Kanai Y, Katsuki H, Takasaki M. Lidocaine disrupts axonal membrane of rat sciatic nerve in vitro.
Anesth Analg. 2000;91:944–948.
108. Rigler ML, Drasner K, Krejcie TC, et al. Cauda equina syndrome after continuous spinal anesthesia.
Anesth Analg. 1991;72:275–281.
109. Schell RM, Brauer FS, Cole DJ, et al. Persistent sacral nerve root deficits after continuous spinal
anaesthesia. Can J Anaesth. 1991;38:908–911.
110. Auroy Y, Benhamou D, Bargues L, et al. Major complications of regional anesthesia in France: The
SOS Regional Anesthesia Hotline Service. Anesthesiology. 2002;97:1274–1280.
111. Zaric D, Christiansen C, Pace NL, et al. Transient neurologic symptoms (TNS) following spinal
anaesthesia with lidocaine versus other local anaesthetics. Cochrane Database Syst Rev. 2005;
(4):CD003006.
112. Hampl KF, Schneider MC, Pargger H, et al. A similar incidence of transient neurologic symptoms
after spinal anesthesia with 2% and 5% lidocaine. Anesth Analg. 1996;83:1051–1054.
113. Pollock JE, Liu SS, Neal JM, et al. Dilution of spinal lidocaine does not alter the incidence of transient
neurologic symptoms. Anesthesiology. 1999;90:445–450.
114. Tong D, Wong J, Chung F, et al. Prospective study on incidence and functional impact of transient
neurologic symptoms associated with 1% versus 5% hyperbaric lidocaine in short urologic
procedures. Anesthesiology. 2003;98:485–494.
115. Hampl KF, Schneider MC, Thorin D, et al. Hyperosmolarity does not contribute to transient radicular
irritation after spinal anesthesia with hyperbaric 5% lidocaine. Reg Anesth. 1995;20:363–368.
116. Pollock JE, Burkhead D, Neal JM, et al. Spinal nerve function in five volunteers experiencing transient
neurologic symptoms after lidocaine subarachnoid anesthesia. Anesth Analg. 2000;90:658–665.
117. Wong CA, Slavenas P. The incidence of transient radicular irritation after spinal anesthesia in obstetric
patients. Reg Anesth Pain Med. 1999;24:55–58.
118. Stevens RA, Urmey WF, Urquhart BL, et al. Back pain after epidural anesthesia with chloroprocaine.
Anesthesiology. 1993;78:492–497.
119. Zink W, Seif C, Bohl JR, et al. The acute myotoxic effects of bupivacaine and ropivacaine after
continuous peripheral nerve blockades. Anesth Analg. 2003;97:1173–1179.
120. Finucane BT. Allergies to local anesthetics—the real truth. Can J Anaesth. 2003;50:869–874.
121. Gall H, Kaufmann R, Kalveram CM. Adverse reactions to local anesthetics: analysis of 197 cases. J
Allergy Clin Immunol. 1996;97:933–937.
Obstetric Medications
Ruchira Patel and Mrinalini Balki
I. Tocolytic medications
A. β-Mimetic therapy
B. Calcium channel blockers
C. Magnesium sulfate
D. Cyclooxygenase (prostaglandin synthase) inhibitors
E. Nitroglycerin
F. Oxytocin antagonists (atosiban)
II. Uterotonic medications
A. Oxytocin
B. Carbetocin
C. Ergot alkaloids (ergonovine or methylergonovine)
D. Prostaglandins (F2α, E1, and E2 analog)
KEYPOINTS
1. β-Mimetic agents, calcium channel blockers, or nonsteroidal anti-
inflammatory drugs (NSAIDs) are the most commonly used tocolytics for
the short-term prolongation of pregnancy (up to 48 hours) to allow for the
administration of antenatal steroids or for women to be transferred to
tertiary care.
2. Magnesium sulfate is mainly indicated for the prevention and treatment
of seizures in severe preeclampsia and for fetal neuroprotection in
threatened preterm (<32 weeks) labor; however, it has a limited role in
tocolysis.
3. Oxytocin is considered the first-line agent for the treatment of postpartum
hemorrhage secondary to uterine atony, followed by ergonovine and
prostaglandins; however, a multimodal pharmacotherapy approach is
recommended.
4. The commonly encountered side effects of uterotonic drugs include
hypotension with oxytocin, hypertension with ergonovine, and
bronchospasm with carboprost.
C. Magnesium sulfate
1. Uses
a. Prevention and treatment of seizures in
preeclampsia/eclampsia. Magnesium sulfate is
recommended in patients with preeclampsia for whom
there is concern about the risk of eclampsia.13 The
Magpie trial demonstrated that women with
preeclampsia who were given magnesium had a 58%
lower risk of eclampsia than those allocated to receive
placebo.14 The number needed to treat to prevent one
seizure for women with severe preeclampsia was 63 (95%
CI, 38 to 181) and for those without severe preeclampsia
was 109 (95% CI, 72 to 225). The American College of
Obstetricians and Gynecologists (ACOG)15
recommends administration of magnesium sulfate to
women with eclampsia and preeclampsia with severe
features but not to those with mild preeclampsia
without any symptoms or gestational hypertension.
Magnesium sulfate is usually continued for 24 hours
postpartum.
b. Fetal neurodevelopment. Antenatal magnesium sulfate
therapy for women at risk for preterm birth is
established as being neuroprotective against motor
disorders for the preterm fetus.16,17 Magnesium therapy
given to women at risk for preterm birth substantially
reduced the risk of cerebral palsy in their children (RR
0.68; 95% CI, 0.54 to 0.87). The number of women
needed to treat to prevent one baby from developing
cerebral palsy was 63 (95% CI, 43 to 155). Two large
randomized controlled trials further support the use of
prenatal magnesium for preventing neurodisabilities in
preterm infants.18,19
c. Tocolysis to prolong pregnancy. Magnesium sulfate
inhibits uterine activity and has been used as a tocolytic
agent in women with preterm labor to prevent delivery.
However, its efficacy in preventing preterm delivery has
been shown on numerous occasions to be equivalent to
placebo. A recent Cochrane review found that magnesium
sulfate is ineffective at delaying birth or preventing
preterm birth, has no apparent advantages for a range of
neonatal and maternal outcomes as a tocolytic agent, and
its use for this indication may be associated with an
increased risk of total fetal, neonatal, or infant mortality.20
The ACOG, however, continues to support the short-term
use of magnesium for up to 48 hours to allow for the
administration of corticosteroids in women between 24
and 34 weeks’ gestation with preterm labor.21
CLINICAL PEARL Magnesium significantly improves
pulmonary and neurologic outcomes in neonates delivered preterm
and thus is used commonly in women with both preeclampsia who
often deliver preterm and in the treatment of preterm labor.
2. Mechanism of action
a. The anticonvulsant effect of magnesium sulfate is
attributed to its antagonistic action on the N-methyl-D-
aspartate (NMDA) receptor and direct cerebral
vasodilatation.
b. It competes with calcium for binding sites on the
sarcoplasmic reticulum and decreases intracellular
calcium levels. It hyperpolarizes the plasma membrane
and inhibits myosin light-chain kinase activity reducing
myometrial contractility (see Fig. 4.1).
c. It increases PGI2 production by vascular endothelium
resulting in smooth muscle relaxation and dilation.
3. Route of administration/dose
a. Magnesium sulfate is given as a bolus 4 to 6 g IV over 20
minutes and then as an infusion of 1 to 2 g per hour for
short-term (usually less than 48 hours) use in preterm
labor or fetal neuroprotection.
b. In the prevention and treatment of seizures, it is
administered in similar doses and continued for up to 24
hours postpartum.
4. Toxicity/side effects
a. Increasing plasma levels of magnesium are associated
with adverse effects (see Table 4.2). Because severe
adverse effects are seen above the plasma magnesium
level at which deep tendon reflexes are lost, patients
should be monitored by regularly checking reflexes.
b. Fetal cardiac or respiratory effects are not of concern
unless maternal toxicity is present.
c. It may prolong labor and increase the risk of PPH due to
its tocolytic effect.
d. Magnesium is renally excreted and hence toxicity is more
common in patients with renal impairment.
e. If magnesium toxicity occurs, the infusion should be
discontinued, and either 10 mL of calcium chloride 10%
or 30 mL of calcium gluconate 10% administered by slow
intravenous infusion.
5. Anesthetic considerations
a. Magnesium causes generalized muscle weakness; hence,
it is contraindicated in women with myasthenia gravis.
b. Magnesium increases the sensitivity to muscle relaxants.
A defasciculating dose of a nondepolarizing muscle
relaxant before succinylcholine administration should be
avoided. The intubating dose of succinylcholine does not
need to be altered; however, the maintenance dose of
nondepolarizing agents should be reduced.
c. Magnesium use may exacerbate hypotension caused by
hemorrhage or neuroaxial block due to a decrease in
systemic vascular resistance.
d. Magnesium has been shown to inhibit platelet function, to
decrease the amount of inhaled volatile agents required,
and to amplify the analgesic effect of opioids.
e. Magnesium toxicity can lead to respiratory depression and
cardiac arrest (see Table 4.2).
D. Cyclooxygenase (prostaglandin synthase) inhibitors
1. Drugs
a. Indomethacin
b. Sulindac
c. Ketorolac
2. Uses. Tocolysis to prolong pregnancy in threatened preterm
labor
a. The enzyme cyclooxygenase (COX), officially known as
prostaglandin-endoperoxide synthase, exists in two
isoforms: COX-1 synthesizes protective prostaglandins,
which are responsible for preserving the integrity of the
stomach lining, renal function, and platelet aggregation;
COX-2 is induced during the inflammatory process and
prostaglandins made by COX-2 are also important for
inducing uterine contractions during labor.22 NSAIDs
such as indomethacin can therefore delay premature labor
by inhibiting production of these prostaglandins.
b. Indomethacin is the most commonly used NSAID in
cases of threatened preterm labor. A Cochrane review
found that compared to placebo, indomethacin resulted in
a reduction in birth before 37 weeks’ gestation (RR 0.21;
95% CI, 0.07 to 0.62), an increase in gestational age
(weighted mean difference [WMD] 3.53 weeks; 95% CI,
1.13 to 5.92), and birth weight (WMD 716 g; 95% CI,
426 to 1,007). In addition, compared to other tocolytics,
COX inhibition resulted in a reduction in maternal drug
reactions requiring cessation of treatment. Surprisingly,
no differences were detected in neonatal outcomes.23
c. There is some data on the use of selective COX-2
inhibitors (nimesulide,24 celecoxib,25 rofecoxib26,27) for
the treatment of preterm labor; however, most of these
agents have been withdrawn or issued with black box
warnings because of the adverse cardiovascular risk and
severe fetal side effects associated with their use.28
3. Mechanism of action
a. NSAIDs inhibit COX (prostaglandin synthase) enzyme,
which reduces the synthesis of prostaglandins from the
precursor arachidonic acid (see Fig. 4.1). The extent of
enzyme inhibition varies among different NSAIDs, by
either general inhibition or specific inhibition of COX-2.
The synthesis of prostaglandins E2 and F2α, which are
potent uterine smooth muscle stimulants, is decreased.
b. Indomethacin is a nonspecific COX inhibitor.
4. Route of administration/dose
a. Indomethacin 50 to 100 mg oral or rectal loading dose is
followed by 25 to 50 mg every 4 to 6 hours.
b. It is recommended for use only before 32 weeks’
gestation and should only continue for 48 to 72 hours due
to the risk of severe fetal side effects.
5. Toxicity/side effects
a. The maternal gastrointestinal side effects associated with
the use of these medications include nausea, esophageal
reflux, gastritis, and emesis.
b. Platelet dysfunction is seen due to inhibition of
thromboxane A2.
c. Renal function may worsen in patients with renal
insufficiency from decreased renal blood flow. This is due
to inhibition of prostaglandin E2 and I2 activity.
d. There is an increased risk of cardiovascular side effects
such as myocardial infarction and stroke.
6. Anesthetic considerations
a. Anesthetic management is not significantly impacted.
b. Platelet inhibition is reversible and transient; therefore,
neuraxial anesthesia is not contraindicated.
E. Nitroglycerin
1. Uses. Nitroglycerin is used for multiple indications. It
induces rapid, profound, and short-duration uterine
relaxation for:
a. Fetal head entrapment
b. Extraction of second twin
c. Manual removal of placenta
d. Uterine inversion
e. External cephalic version
f. Preeclampsia
g. Tetanic contractions of uterus
2. Mechanism of action
a. Nitroglycerin is converted to nitric oxide, which activates
guanylate cyclase and increases cGMP. This in turn
inhibits calcium influx and relaxes smooth muscle (see
Fig. 4.1).
b. At low doses, nitroglycerin will dilate veins more than
arteries, thereby reducing preload, but at higher doses it
also dilates arteries, thereby reducing afterload.
3. Route of administration/dose
a. Nitroglycerin 50 μg IV followed by up to four additional
doses of 50 μg is used to treat most obstetric indications.
Aerosol form can be administered as a metered spray
sublingually in a dose of 400 μg each.
b. Sublingual tablet (0.3 to 0.6 mg) and continuous IV
infusion (5 to 15 μg per minute) are used for nonobstetric
indications, mainly cardiac disease.
c. Doses in excess of 1,500 μg have been administered
without significant adverse effects noted.
4. Toxicity/side effects
a. Nitroglycerin acts more on uterine than vascular smooth
muscle; however, vascular effects include transient
hypotension and reflex tachycardia.
b. Maternal headache and light-headedness can occur.
c. No significant fetal effects have been reported.
5. Anesthetic considerations
a. Nitroglycerin has a very short half-life and is useful when
short-term effect is required. For sustained effect, as for
the treatment of hypertension, multiple doses or an
infusion may be required.
b. Vascular effects can result in transient hypotension
and tachycardia; therefore, hemodynamic monitoring
is required.
c. Hypoxia, possibly due to pulmonary vasodilation and
blunting of protective hypoxic pulmonary
vasoconstriction, is common with continuous infusion of
nitroglycerin and has also been reported with bolus doses
in an obstetric patient.29
F. Oxytocin antagonist (atosiban)
1. Uses. Tocolysis to prolong pregnancy in threatened
preterm labor
a. Atosiban, an oxytocin antagonist, was originally
considered to be a promising tocolytic agent with a
favorable side effect profile. A Cochrane review found
atosiban to be as effective as β-receptor agonists for
preventing preterm birth within 48 hours of initiating
treatment (RR 0.89; 95% CI, 0.66 to 1.22). However, it
was not found superior, as a tocolytic agent, to placebo, β-
mimetics, or calcium channel blockers in terms of
pregnancy prolongation or neonatal outcomes.30
b. The use of atosiban was associated with a lower risk of
maternal side effects requiring cessation of treatment than
β-receptor agonists.30 However, in one study, atosiban
resulted in an increase in extremely preterm birth (before
28 weeks’ gestation) and infant deaths (up to 12 months),
which warrants caution with its use in women with
gestational age less than 28 weeks. This finding may be
confounded due to randomization of more women with
less than 26 weeks’ gestation to atosiban in the study and
small sample size.30
c. Although commonly used in Europe, the FDA declined to
approve the use of atosiban for tocolysis because of
concerns about the drug’s safety when used in fetuses less
than 28 weeks of gestation.31
2. Mechanism of action. It is a competitive inhibitor of
oxytocin at myometrial and decidual receptors (see Fig. 4.1).
3. Route of administration/dose. Atosiban 6.75 mg IV bolus is
followed by a 300 μg per minute infusion for 3 hours, then
100 μg per minute for up to 45 hours.
4. Toxicity/side effects
a. There are no major maternal side effects, and the overall
frequency of side effects is significantly less than that
reported for any other drug used for inhibition of preterm
labor.32 The sensitivity of myometrium to oxytocin
remains unaffected. For these reasons, it is considered a
second-line tocolytic after calcium channel blockers in
practices where it is available.
b. There is minimal placental transfer and no adverse fetal
side effects.
5. Anesthetic considerations. No known significant anesthetic
interactions have been reported.
II. Uterotonic medications
PPH is one of the major causes of mortality in pregnant women
worldwide and occurs in up to 5% of all deliveries. The etiology of
PPH can be classified into four main groups: uterine atony (tone);
retained products (tissue); vaginal, cervical, or uterine injury (trauma);
and coagulopathy (thrombin). The most common cause of PPH is
uterine atony, and the correct and prompt use of uterotonics can
prevent significant morbidity and mortality from hemorrhage. In
addition, these agents have other obstetric indications including
induction and augmentation of labor and termination of pregnancy.
A. Oxytocin
Oxytocin is a hormone produced in the hypothalamus and
secreted from the posterior lobe of the pituitary gland in a
pulsatile fashion. Its synthetic analog, Pitocin, is among the most
potent uterotonic agents known. Pitocin possesses fewer
antidiuretic hormone (ADH)-related side effects (water
intoxication) than oxytocin.
1. Uses
a. Induction and augmentation of labor. Synthetic oxytocin
is used to induce labor and produce periodic uterine
contractions first demonstrable at approximately 20 weeks
of gestation. There is an increase in responsiveness to
oxytocin as the gestational age advances, primarily due to
an increase in myometrial oxytocin binding sites.33
Higher concentrations of estrogen during pregnancy
increase the density and binding kinetics of the oxytocin
receptor and enhance uterine sensitivity to oxytocin.34
There is little change in myometrial sensitivity to
oxytocin from 34 weeks to term; however, once
spontaneous labor begins, uterine sensitivity to oxytocin
increases rapidly.35
b. Uterine atony and active management of third stage.
Oxytocin is the first-line agent in the prevention and
treatment of PPH. In a Cochrane review, prophylactic
oxytocin reduced the risk of blood loss exceeding 500
mL (RR 0.53; 95% CI, 0.38 to 0.74) and the need for
therapeutic uterotonic agents (RR 0.56; 95% CI, 0.36
to 0.87) compared with placebo.36 Active management
of the third stage of labor, involving prophylactic
administration of oxytocin before delivery of the placenta,
has been shown to reduce the incidence of PPH by
>60%.36
c. Contraction stress testing
A contraction stress test (CST) can be performed near the
end of pregnancy to determine how well the fetus will
cope with the contraction of childbirth. Oxytocin is given
intravenously to stimulate uterine contractions. The fetus
is monitored for any signs of distress once the parturient
experiences three contractions in 10 minutes. A positive
CST indicates high risk of fetal death due to hypoxia and
is a contraindication to labor. The CST is seldom
performed nowadays given the wide availability of other
tests of the fetal environment, such as the nonstress test
and biophysical profile measurements.
2. Mechanism of action. Oxytocin activates uterine G-protein-
coupled oxytocin receptors, which increases intracellular
calcium (via inositol triphosphate) and prostaglandin
production (via diacylglycerol). The increase in calcium and
prostaglandins induces uterine smooth muscle contraction
(see Fig. 4.2).
3. Route of administration/dose
a. Oxytocin 1 to 2 IU per minute IV infusion, titrated up to
32 IU per minute can be administered for
induction/augmentation of labor and CST.
b. For active management of the third stage of labor,
prophylactic oxytocin is routinely administered to reduce
uterine atony and PPH. A small loading dose of oxytocin
(ED 90 ± 0.35 IU; 95% CI, 0.18 to 0.52) has been
determined to be sufficient in producing adequate uterine
contractions during elective cesarean deliveries in
nonlaboring women and a similarly low loading dose (ED
90 ± 2.99 IU; 95% CI, 2.32 to 3.67) is required in
laboring women.37,38 This is followed by an infusion,
such as 20 to 40 units diluted in a liter of isotonic fluid IV
over 2 to 6 hours depending on the institutional policy.
For the treatment of PPH, slow boluses of oxytocin 5 IU
can be administered IV. It is preferable to administer this
drug as an infusion rather than boluses to minimize
hemodynamic side effects.
CLINICAL PEARL Small doses of oxytocin, often much less
than that used by many practitioners, are required to provide
adequate uterine contraction following elective, repeat cesarean
delivery; this is not true in women who have undergone a period of
labor where much larger doses are often required.
4. Toxicity/side effects
a. Maternal adverse effects include significant
hypotension, tachycardia, decreased free water
clearance, hyponatremia (although only seen when
administered in very high doses or with concurrent
hypotonic fluid administration), peripheral flushing,
nausea, emesis, and signs of myocardial ischemia.
b. Hemodynamic side effects are more pronounced when
oxytocin is administered as a rapid bolus (hypotension
is nearly always seen when administered as an IV
bolus of 5 to 10 units); hence, it should be used with
caution in those with cardiac disease or hemodynamic
instability. There are three reported cases of maternal
death resulting from cardiovascular collapse
secondary to an IV bolus of 10 IU oxytocin.39
c. Fetal effects, such as desaturation,40 hyperbilirubinemia,41
and retinal hemorrhage,42 have been reported following
oxytocin use.
5. Anesthetic considerations
a. Induction/augmentation of labor with oxytocin can induce
a state of uterine tetany resulting in nonreassuring fetal
heart rate patterns.43
b. Hypertonic uterine contractions and fetal bradycardia can
occur following CSE placement in women receiving an
infusion of oxytocin.43 If this occurs, discontinue
oxytocin infusion and consider IV terbutaline or
nitroglycerin to provide uterine relaxation.
c. The vasodilatory effects of oxytocin can produce
significant hypotension, especially when administered as
a bolus in patients with hypovolemia.
B. Carbetocin
1. Uses. Treatment of uterine atony for active management of
third stage of labor to reduce PPH
a. Carbetocin is a synthetic analog of the hormone oxytocin
with the advantage of having a longer half-life of 40
minutes compared to 4 to 10 minutes for oxytocin. The
Society of Obstetricians and Gynecologists of Canada
guidelines recommend carbetocin 100 µg over oxytocin
to prevent PPH in elective cesarean delivery and
vaginal delivery with one risk factor for PPH.44
b. A 2009 systematic review concluded that carbetocin is
not superior over currently available uterotonic agents
for the prevention of PPH, both in vaginal and
cesarean deliveries.45 According to a recent Cochrane
review, carbetocin significantly reduced the need for
additional uterotonics (RR 0.62; 95% CI, 0.44 to 0.88)
and the risk of PPH following cesarean deliveries
compared to oxytocin (RR 0.55; 95% CI, 0.31 to 0.95).
However, these results were limited by a small number of
studies and risk of bias, warranting further studies to
validate these findings.46 A dose finding study found that
the ED 90 of carbetocin at elective cesarean delivery was
14.8 µg (95% CI, 13.7 to 15.8); this is less than one-fifth
the recommended dose.47 A recent study found an ED 90
of carbetocin to be 120.5 µg (95% CI, 110.9 to 130.2;
99% CI, 107.8 to 133.2) in women with labor arrest;
however, there was a high incidence of dysrhythmias at
higher doses.48 They suggested uncertainty regarding the
efficacy of carbetocin in this patient population.
Therefore, further research is needed to establish the
appropriate dose, efficacy, and also the side effect profile
of carbetocin.
4. Toxicity/side effects
a. Methylergonovine can cause peripheral
vasoconstriction resulting in hypertension, increased
pulmonary artery pressures, pulmonary edema,
cerebral hemorrhage, and retinal damage.
b. Nausea and vomiting can occur in up to 20% of patients.
c. IV administration may lead to angina from coronary
artery vasospasm and cardiac arrest from myocardial
infarction.
5. Anesthetic considerations
a. Ergonovine should be avoided in patients with chronic
hypertension, preeclampsia, peripheral vascular disease,
and ischaemic heart disease.
b. Vasodilators should be available to treat hypertension and
antiemetics available to treat nausea and vomiting.
D. Prostaglandins (F2α, E1, and E2 analog)
1. Drugs
a. Carboprost (15-Methyl PGF2α)
b. Misoprostol (PGE1)
c. Dinoprostol (PGE2)
2. Uses
a. Induction of labor
(1) Induction of labor is one of the most commonly
performed obstetrical procedures in North America.
The first step in labor induction of women with an
unfavorable cervix is administration of dinoprostol or
misoprostol. This alone can initiate labor in many
women and obviate the need for oxytocin.
(2) A Cochrane review found that the use of dinoprostol
resulted in a reduction in unfavorable cervix after 12
to 24 hours (RR 0.41; 95% CI, 0.27 to 0.65), a
probable modest reduction in the rate of cesarean
deliveries (RR 0.91; 95% CI, 0.81 to 1.02), and a
possible lower rate of failing to achieve vaginal
delivery within 24 hours (RR 0.32; 95% CI, 0.02 to
4.83).52
(3) Misoprostol is an effective alternative to dinoprostol
preparations for cervical ripening and labor
induction.53 The ACOG has indicated that the use
of misoprostol appears safe and efficacious when
used as a cervical ripening and/or labor induction
agent.54 However, misoprostol is not advised for
cervical ripening or labor induction in women in the
third trimester with prior uterine incisions due to the
risk of uterine rupture.55
b. Treatment of uterine atony
(1) Misoprostol is less effective than oxytocin or
ergometrine for active management of the third stage
of labor and is associated with a higher risk of severe
PPH compared to conventional injectable uterotonics
(RR 1.33; 95% CI, 1.16 to 1.52).56
(2) Carboprost is the most common prostaglandin
used for uterine atony and is typically
administered after oxytocin and ergometrine. It is
the only prostaglandin currently available that
can be administered parenterally.
3. Mechanism of action
a. Prostaglandins cause dissolution of collagen bundles and
an increase in the submucosal water content of the cervix,
which leads to a cervical state that is associated with a
greater success of labor induction with oxytocin.57
b. Prostaglandins increase myometrial calcium
concentration, which causes the uterus to contract (see
Fig. 4.2). This may initiate labor or in the postpartum
period prevent uterine atony and PPH.
4. Route of administration/dose
a. Misoprostol (Cytotec) can be administered via vaginal,
rectal, or oral route for labor induction in doses of 50 to
400 μg (depending on the gestational age), every 4 hours
to a maximum of 6 doses.
b. For prevention or treatment of PPH, misoprostol 600 to
1,000 μg can be administered either orally, sublingually,
vaginally, or rectally.
c. Dinoprostone (Cervidil) 10 mg is inserted in the posterior
fornix for labor induction. It can be replaced after 12
hours and removed at the onset of active labor.
d. Carboprost (Hemabate) 250 μg can be administered IM or
intramyometrial for uterine atony, repeated every 15
minutes up to a total dose of 2 mg.
5. Toxicity/side effects
a. All prostaglandins can cause nausea, vomiting, diarrhea,
shivering, and fever.
b. Carboprost (15-Methyl PGF2α) causes increased
systemic and pulmonary vascular resistance and can
cause bronchoconstriction and hypoxemia due to
intrapulmonary shunting and ventilation–perfusion
mismatch.58
c. PGE2 administration on the other hand causes decreased
systemic vascular resistance and blood pressure, increased
cardiac output, and relaxes the gastroesophageal
sphincter, potentially increasing the risk of passive
regurgitation.
d. Misoprostol (PGE1) can cause uterine hyperstimulation
when administered for labor induction but appears to have
minimal effect on maternal hemodynamics.
6. Anesthetic considerations
a. The use of prostaglandins to induce labor in patients
attempting vaginal birth after previous cesarean delivery
can increase the risk of uterine rupture.
b. Carboprost should be used with caution in patients with
reactive airway disease, and avoided if cardiac disease
and pulmonary hypertension are present.
c. An antiemetic and antidiarrheal may be needed after
carboprost administration.
REFERENCES
1. American College of Obstetricians and Gynecologists, Committee on Practice Bulletins–Obstetrics.
ACOG Practice Bulletin no. 127: management of preterm labor. Obstet Gynecol. 2012;119:1308–
1317.
2. Neilson JP, West HM, Dowswell T. Betamimetics for inhibiting preterm labour. Cochrane Database
Syst Rev. 2014;(2):CD004352.
3. U.S. Food and Drug Administration. FDA Drug Safety Communication: New warnings against use of
terbutaline to treat preterm labor. http://www.fda.gov/Drugs/DrugSafety/ucm243539.htm. Accessed
January 26, 2015.
4. Yaju Y, Nakayama T. Effectiveness and safety of ritodrine hydrochloride for the treatment of preterm
labour: a systematic review. Pharmacoepidemiol Drug Saf. 2006;15:813–822.
5. Lamont RF. The pathophysiology of pulmonary oedema with the use of beta-agonists. BJOG.
2000;107:439–444.
6. Flenady V, Wojcieszek AM, Papatsonis DN, et al. Calcium channel blockers for inhibiting preterm
labour and birth. Cochrane Database Syst Rev. 2014;(6):CD002255.
7. Smith P, Anthony J, Johanson R. Nifedipine in pregnancy. BJOG. 2000;107:299–307.
8. Cornette J, Duvekot J, Roos-Hesselink J, et al. Maternal and fetal haemodynamic effects of nifedipine
in normotensive pregnant women. BJOG. 2011;118:510–540.
9. van Veen AJ, Pelinck MJ, van Pampus MG, et al. Severe hypotension and fetal death due to tocolysis
with nifedipine. BJOG. 2005;112:509–510.
10. Impey L. Severe hypotension and fetal distress following sublingual administration of nifedipine to a
patient with severe pregnancy induced hypertension at 33 weeks. Br J Obstet Gynaecol.
1993;100:959–961.
11. Feldman S, Karalliedde L. Drug interactions with neuromuscular blockers. Drug Saf. 1996;15:261–
273.
12. Csapo AI, Puri CP, Tarro S, et al. Deactivation of the uterus during normal and premature labor by the
calcium antagonist nicardipine. Am J Obstet Gynecol. 1982;142:483–491.
13. Duley L, Gülmezoglu AM, Henderson-Smart DJ. Magnesium sulphate and other anticonvulsants for
women with pre-eclampsia. Cochrane Database Syst Rev. 2003;(2):CD000025.
14. Altman D, Carroli G, Duley L, et al. Do women with pre-eclampsia, and their babies, benefit from
magnesium sulphate? The Magpie Trial: a randomised placebo-controlled trial. Lancet.
2002;359:1877–1890.
15. American College of Obstetricians and Gynecologists. Hypertension in pregnancy. Report of the
American College of Obstetricians and Gynecologists’ Task Force on Hypertension in Pregnancy.
Obstet Gynecol. 2013;122:1122–1131.
16. Doyle LW, Crowther CA, Middleton P, et al. Magnesium sulphate for women at risk of preterm birth
for neuroprotection of the fetus. Cochrane Database Syst Rev. 2009;(1):CD004661.
17. Doyle LW, Crowther CA, Middleton P, et al. Antenatal magnesium sulfate and neurologic outcome in
preterm infants: a systematic review. Obstet Gynecol. 2009;113:1327–1333.
18. Rouse DJ, Hirtz DG, Thom E, et al. A randomized, controlled trial of magnesium sulfate for the
prevention of cerebral palsy. N Engl J Med. 2008;359:895–905.
19. Marret S, Marpeau L, Follet-Bouhamed C, et al. Effect of magnesium sulphate on mortality and
neurologic morbidity of the very-preterm newborn (of less than 33 weeks) with two-year neurological
outcome: results of the prospective PREMAG trial. Gynecol Obstet Fertil. 2008;36:278–288.
20. Crowther CA, Hiller JE, Doyle LW. Magnesium sulphate for preventing preterm birth in threatened
preterm labour. Cochrane Database Syst Rev. 2002;(4):CD001060.
21. American College of Obstetricians and Gynecologists Committee on Obstetric Practice Society for
Maternal-Fetal Medicine. Committee Opinion No. 573: magnesium sulfate use in obstetrics. Obstet
Gynecol. 2013;122:727–728.
22. Vane JR, Bakhle YS, Botting RM. Cyclooxygenases 1 and 2. Annu Rev Pharmacol Toxicol.
1998;38:97–120.
23. King J, Flenady V, Cole S, et al. Cyclo-oxygenase (COX) inhibitors for treating preterm labour.
Cochrane Database Syst Rev. 2005;(2):CD001992.
24. Locatelli A, Vergani P, Bellini P, et al. Can a cyclo-oxygenase type-2 selective tocolytic agent avoid
the fetal side effects of indomethacin? BJOG. 2001;108:325–326.
25. Stika CS, Gross GA, Leguizamon G, et al. A prospective randomized safety trial of celecoxib for
treatment of preterm labor. Am J Obstet Gynecol. 2002;187:653–660.
26. McWhorter J, Carlan SJ, OLeary TD, et al. Rofecoxib versus magnesium sulfate to arrest preterm
labor: a randomized trial. Obstet Gynecol. 2004;103:923–930.
27. Groom KM, Shennan AH, Jones BA, et al. TOCOX—a randomised, double-blind, placebo-controlled
trial of rofecoxib (a COX-2-specific prostaglandin inhibitor) for the prevention of preterm delivery in
women at high risk. BJOG. 2005;112:725–730.
28. Loudon JA, Groom KM, Bennett PR. Prostaglandin inhibitors in preterm labour. Best Pract Res Clin
Obstet Gynaecol. 2003;17:731–744.
29. Saroa R, Sachan S, Palta S, et al. Obstetric use of nitroglycerin: anesthetic implications. Saudi J
Anaesth. 2013;7:350–352.
30. Flenady V, Reinebrant HE, Liley HG, et al. Oxytocin receptor antagonists for inhibiting preterm
labour. Cochrane Database Syst Rev. 2014;(6):CD004452.
31. Romero R, Sibai BM, Sanchez-Ramos L, et al. An oxytocin receptor antagonist (atosiban) in the
treatment of preterm labor: a randomized, double-blind, placebo-controlled trial with tocolytic rescue.
Am J Obstet Gynecol. 2000;182:1173–1183.
32. Gyetvai K, Hannah ME, Hodnett ED, et al. Tocolytics for preterm labor: a systematic review. Obstet
Gynecol. 1999;94:869–877.
33. Fuchs AR, Fuchs F, Husslein P, et al. Oxytocin receptors in the human uterus during pregnancy and
parturition. Am J Obstet Gynecol. 1984;150:734–741.
34. Zeeman GG, Khan-Dawood FS, Dawood MY. Oxytocin and its receptor in pregnancy and parturition:
current concepts and clinical implications. Obstet Gynecol. 1997;89:873–883.
35. Calderyro-Barcia R, Sereno JA. The response of human uterus to oxytocin throughout pregnancy. In:
Caldeyro-Barcia R, Heller H, eds. Oxytocin. London, United Kingdom: Pergamon Press; 1959:177–
202.
36. Westhoff G, Cotter AM, Tolosa JE. Prophylactic oxytocin for the third stage of labour to prevent
postpartum haemorrhage. Cochrane Database Syst Rev. 2013;(10):CD001808.
37. Carvalho JC, Balki M, Kingdom J, et al. Oxytocin requirements at elective cesarean delivery: a dose-
finding study. Obstet Gynecol. 2004;104:1005–1010.
38. Balki M, Ronayne M, Davies S, et al. Minimum oxytocin dose requirement after cesarean delivery for
labor arrest. Obstet Gynecol. 2006;107:45–50.
39. Balki M, Tsen L. Oxytocin protocols for cesarean delivery. Int Anesthesiology Clin. 2014;52:48–66.
40. Simpson KR, James DC. Effects of oxytocin-induced uterine hyperstimulation during labor on fetal
oxygen status and fetal heart rate patterns. Am J Obstet Gynecol. 2008;199:34.e1–e5.
41. Beazley JM, Alderman B. Neonatal hyperbilirubinaemia following the use of oxytocin in labour. Br J
Obstet Gynaecol. 1975;82:265–271.
42. Schoenfeld A, Buckman G, Nissenkorn I, et al. Retinal hemorrhages in the newborn following labor
induced by oxytocin or dinoprostone. Arch Ophthalmol. 1985;103:932–934.
43. Tsen LC, Balki M. Oxytocin protocols during caesarean delivery: time to acknowledge the risk/benefit
ratio. Int J Obstet Anesth. 2010;19:243–245.
44. Leduc D, Senikas V, Lalonde AB, et al. Active management of the third stage of labour: prevention
and treatment of postpartum hemorrhage. J Obstet Gynaecol Can. 2009;235:980–993.
45. Peters NC, Duvekot J. Carbetocin for the prevention of postpartum hemorrhage: a systematic review.
Obstet Gynecol Surv. 2009;64:129–135.
46. Su LL, Chong YS, Samuel M. Carbetocin for preventing postpartum haemorrhage. Cochrane
Database Syst Rev. 2012;(2):CD005457.
47. Khan M, Balki M, Ahmed I, et al. Carbetocin at elective cesarean delivery: a sequential allocation trial
to determine the minimum effective dose. Can J Anaesth. 2014;61:242–248.
48. Nguyen-Lu N, Carvalho JC, Farine D, et al. Carbetocin at cesarean delivery for labour arrest: a
sequential allocation trial to determine the effective dose. Can J Anaesth. 2015;62:866–874.
49. Moertl MG, Friedrich S, Kraschl J, et al. Haemodynamic effects of carbetocin and oxytocin given as
intravenous bolus on women undergoing caesarean delivery: a randomized trial. BJOG.
2011;118:1349–1356.
50. Su LL, Chong YS, Samuel M. Oxytocin agonists for preventing postpartum haemorrhage. Cochrane
Database Syst Rev. 2007;(3):CD005457.
51. Liabsuetrakul T, Choobun T, Peeyananjarassri K, et al. Prophylactic use of ergot alkaloids in the third
stage of labor. Cochrane Database Syst Rev. 2007;(2):CD005456.
52. Thomas J, Fairclough A, Kavanagh J, et al. Vaginal prostaglandin (PGE2 and PGF2a) for induction of
labour at term. Cochrane Database Syst Rev. 2014;(6):CD003101.
53. Hofmeyr GJ, Gülmezoglu AM, Pileggi C. Vaginal misoprostol for cervical ripening and induction of
labour. Cochrane Database Syst Rev. 2010;(10):CD000941.
54. American College of Obstetricians and Gynecologists Committee on Practice Bulletins—Obstetrics.
ACOG Practice Bulletin No. 107: induction of labor. Obstet Gynecol. 2009;114:386–397.
55. American College of Obstetricians and Gynecologists Committee on Practice Bulletins—Obstetrics.
ACOG Practice Bulletin No. 115: vaginal birth after previous cesarean delivery. Obstet Gynecol.
2010;116:450–463.
56. Tunçalp Ö, Hofmeyr GJ, Gülmezoglu AM. Prostaglandins for preventing postpartum haemorrhage.
Cochrane Database Syst Rev. 2012;(8):CD000494.
57. Keirse MJ. Natural prostaglandins for induction of labor and preinduction cervical ripening. Clin
Obstet Gynecol. 2006;49:609–626.
58. Hankins G, Berryman G, Scott R, et al. Maternal arterial desaturation with 15-methyl prostaglandin F2
alpha for uterine atony. Obstet Gynecol. 1988;3:367–370.
Antepartum
Considerations
Ethical and Legal Considerations in Obstetric
Anesthesia
M. Joanne Douglas and William J. Sullivan
I. Introduction to ethics
II. Informed consent
A. Background
B. Can laboring women give informed consent?
C. Implied consent
D. Presentation of information and risk
E. Purpose of the informed consent discussion
F. Withholding information
G. Refusal to be informed
III. Other consent issues
A. Delegation of informed consent
B. Refusal/withdrawal of consent
C. Obtaining consent from minors
D. Written consent
E. Birth plans—the Ulysses directive
F. Exceptions to informed consent
IV. Professional negligence: The law
V. Informed consent: The law
VI. Litigation specific to obstetric anesthesia
VII. Disclosure and apology
VIII. Maternal autonomy and fetal beneficence
Summary
KEYPOINTS
1. Ethics: Ethics are a vital component of all health care. Anesthesiologists
and patients may differ in their perception as to optimal treatment,
creating an ethical dilemma. Before making a medical or ethical decision,
all of the facts must be ascertained.
2. Informed consent: Informed consent requires consent be given and the
consent be informed. Informed consent honors, and in law, enforces the
woman’s autonomy. Consenting is a process, and written consent by itself
is not enough. Women have the right to give consent, withdraw consent,
refuse consent, and to delegate that right to another. Women lose that
right if incapable, but informed consent is still required.
3. Communication: Good communication is an essential part of ethics and
informed consent.
4. The law: Informed consent, in law, enforces patient autonomy. The
patient must consent to the proposed treatment and that consent must be
informed. Failure to obtain consent will normally constitute a battery or
an assault. Failure to properly inform will normally constitute negligence.
The patient must be capable to give informed consent. Even if the patient
is incapable, informed consent is still required unless it is an emergency
and there is no one present legally to consent.
5. Maternal autonomy and fetal beneficence: Occasionally, the interests
of the woman and her fetus may differ, presenting the physician with a
dilemma. In most circumstances, legally and ethically, the woman’s
autonomy should be respected.
I. Introduction to ethics
A. Ethical decision making in health care in the Western world is
based mainly on the application of the four prima facie ethical
principles:
1. Autonomy (choice)
2. Nonmaleficence (do no harm)
3. Beneficence (prevent harm, remove harm)
4. Justice (be fair, treat like cases alike) to the facts of the
particular health care case.1 This method replaced the older
way of “doctor knows best” decision-making (strong
paternalism).
B. Remember that not all choices are ethical in nature. Whether
to use a 16 or 19 gauge needle for an epidural may require a
decision, but the decision is solely a medical one.
C. Ethical decision-making is not done in a vacuum. The facts are
essential. Knowing which facts are relevant can only be
determined after ascertaining all of the facts available. This
includes not only the patient’s pertinent medical facts but also
social, economic, and cultural facts that may influence the
decision. An ethical dilemma may seem to appear when one does
not know all of the facts but may disappear once sufficient facts
are available.
D. Questions to ask when assessing the facts
1. What will the proposed treatment accomplish?
2. What happens if there is no treatment?
3. Are there alternatives to the proposed treatment?
4. What are the risks and benefits of the proposed treatment, the
alternate treatments, and no treatment?
5. What does the patient want? (This is where nonmedical facts
enter into the decision-making.)
6. What other considerations are there in the case?
E. Jonsen et al.2 suggest setting out the facts under the headings of
“medical indications,” “patient preferences” (autonomy), “quality
of life” (including with and without treatment), and “contextual
features” (other factors such as religion and allocation of
resources).
F. The facts will identify the possible choices of action. It may be
that when applying the ethical principles to the choices, the
principles will align with one choice and there is no ethical
dilemma. The patient chooses an epidural for labor pain
(autonomy), the recommended treatment (beneficence and
nonmaleficence), and therefore no dilemma exists (provided the
ethical principle of justice is not wronged).
G. However, the ethical principles may conflict. If more than one
principle applies and, in choosing one, the other(s) cannot be
followed, there is an ethical dilemma. An example is the laboring
parturient who is in pain. The physician wants to relieve the pain
(beneficence) and the parturient refuses (autonomy). The result is,
whichever one is chosen, the other is excluded. When facing an
ethical dilemma, the challenge for the practitioner is determining
which ethical principle to follow.
H. Each of the prima facie principles is applied to the relevant
facts. The principle that discloses the stronger obligation, or as
stated by Beauchamp and Childress,1 the “strongest right” on
those facts is the principle to be followed. Notwithstanding this,
the law is very clear on the importance of autonomy. “Every
human being of adult years and of sound mind has a right to
determine what shall be done with his own body and a surgeon
who performs an operation without his patient’s consent commits
an assault, for which he is liable in damages.”3
II. Informed consent
A. Background
1. Informed consent underlines and legally enforces the
principle of patient autonomy—the woman chooses
(hopefully in consultation with the anesthesiologist) what she
wants after she has been fully informed. This legal
requirement consists of two separate parts. The first is the
requirement of consent.
2. Consent must be:
a. Voluntary
b. For the proposed procedure and who will do it
c. Given by a capable patient
3. Care must be taken not to confuse lack of knowledge of the
world, intellectual disability, or mental illness or instability
with a lack of capability to consent to health care. If the
woman understands:
a. The nature and purpose of the proposed treatment
b. The condition for which the procedure is proposed
c. The risks and benefits of the procedure
d. The consequences of consenting or not consenting
e. That the procedure applies to her situation, then, unless
there is law in a particular jurisdiction to the contrary, she
has capability to give or withhold consent
C. Implied consent
This can occur from either the action or the words of a patient
indicating consent. A note as to how that determination was made
should be added to the patient’s chart.
3. Informed consent may not come from the woman, but it must
come from someone with the authority to give it. The law as
to who has that authority will vary from jurisdiction to
jurisdiction. As well, in some jurisdictions (where they are
legally binding), the authority may come from an advance
directive, whereas in others, it may come from a court order.
IV. Professional negligence: The law
A. Professional negligence occurs when the physician does a
procedure and in doing so does it to a lesser standard than the
recognized standard of care for that procedure causing injury to
the patient.
B. To succeed, the patient must establish negligence, which is all of
the following:
1. Duty (usually a given)
2. Breach of the standard of care. In most jurisdictions, the
standard of care is that of a reasonable practitioner in that
specialty. One should be acquainted with the definition of
standard of care in the jurisdiction in which they practice.
3. Damages (the patient suffered an injury)
4. Causation (the injury was caused by the breach of standard of
care)
V. Informed consent: The law
A. Failure to obtain informed consent occurs
1. First, consent to do the procedure is not obtained. To succeed,
the patient must only establish that no consent was given.
Damages will be awarded even if there is no injury. The fact
that the physician met the standard of care in doing the
procedure is immaterial.
2. Second, the patient is insufficiently informed. To succeed, the
patient must establish negligence, which is all of the
following :
a. Duty—usually a given
b. Breach of the standard of care which in most jurisdictions
is what would a reasonable patient want to know. In some
others, it is what would a reasonable physician tell a
patient, and in others, what a reasonable patient in that
patient’s circumstances would want to know.1,39
c. Damages (the patient suffered an injury)
d. Causation (If the patient would nevertheless have had the
procedure even if fully informed, there is no causation.)
VI. Litigation specific to obstetric anesthesia
A. The American Society of Anesthesiologists (ASA) Closed
Claims Project related to obstetric anesthesia40 found that:
1. The proportion of claims due to substandard care decreased
after 1990.
2. Claims related to newborn death or brain damage resulted
from delays in providing anesthetic care for emergency
deliveries and poor communication between obstetrician and
anesthesiologist.
3. Maternal morbidity/mortality resulted from a delayed
response to an unexpected event such as a high neuraxial
block and/or substandard care.
B. Metzner et al.41 recommended the following to improve
practice and decrease the risk of litigation:
1. Better communication between patient and anesthesiologist
including informed consent and appropriate follow-up
2. Following practice guidelines concerning decision-to-incision
interval; better communication between obstetrics and
anesthesia
3. Earlier diagnosis and treatment of anesthetic complications
(e.g., high block, difficult intubation)
4. Improved treatment of maternal disease, such as hemorrhage
C. Although this Closed Claims report did not discuss whether
informed consent was an issue in the claims, a study from the
United Kingdom found that 10% of all claims related to regional
anesthesia (obstetric and nonobstetric) asserted that there was a
lack of consent.42
VII. Disclosure and apology
SUMMARY
It is important to understand the ethical duty to respect autonomy. That ethical
duty is enforced by the legal requirement for informed consent. Failure to
properly obtain informed consent may well result in a damage award against a
physician. Decisions, whether medical or ethical, cannot be made in a vacuum.
Know the facts of the case.
REFERENCES
1. Beauchamp TL, Childress JF. Principles of Biomedical Ethics. 7th ed. Oxford, United Kingdom:
Oxford University Press; 2013.
2. Jonsen AR, Siegler M, Winslade WJ. Clinical Ethics. 6th ed. New York, NY: McGraw-Hill
Companies, Inc.; 2006.
3. Schloendorff v Society of New York Hospital, 105, NE 92 (NY 1914).
4. Black JD, Cyna AM. Issues of consent for regional analgesia in labour: a survey of obstetric
anaesthetists. Anaesth Intensive Care. 2006;34:254–260.
5. Pattee C, Ballantyne M, Milne B. Epidural analgesia for labour and delivery: informed consent issues.
Can J Anaesth. 1997;44:918–923.
6. Jackson A, Henry R, Avery N, et al. Informed consent for labour epidurals: what labouring women
want to know. Can J Anesth. 2000;47:1068–1073.
7. Jackson GNB, Sensky T, Reide P, et al. The capacity to consent to epidural analgesia in labour. Int J
Obstet Anesth. 2011;20:269–270.
8. Affleck PJ, Waisel DB, Cusick MJ, et al. Recall of risks following labor epidural analgesia. J Clin
Anesth. 1998;10:141–144.
9. White LA, Gorton P, Wee MYK, et al. Written information about epidural analgesia for women in
labour: did it improve knowledge? Int J Obstet Anesth. 2003;12:93–97.
10. Swan HD, Borshoff DC. Informed consent—recall of risk information following epidural analgesia in
labour. Anaesth Intens Care. 1994;22:139–141.
11. Gerancher JC, Grice SC, Dewan DM, et al. An evaluation of informed consent prior to epidural
analgesia for labor and delivery. Int J Obstet Anesth. 2000;9:168–173.
12. Hoehner PJ. Ethical aspects of informed consent in obstetric anesthesia—new challenges and
solutions. J Clin Anesth. 2003;15:587–600.
13. Fortescue C, Wee MYK, Malhotra S, et al. Is preparation for emergency obstetric anaesthesia
adequate? A maternal questionnaire survey. Int J Obstet Anesth. 2007;16:336–340.
14. Straessle R, Gilliard N, Frascarolo P, et al. Is a pre-anaesthetic information form really useful? Acta
Anaesthesiol Scand. 2011;55:517–523.
15. Smedstad KG, Beilby W. Informed consent for epidural analgesia in labour. Can J Anesth.
2000;47:1055–1059.
16. Jenkins K, Baker AB. Consent and anaesthetic risk. Anaesthesia. 2003;58:962–984.
17. DiGiovanni LM. Ethical issues in obstetrics. Obstet Gynecol Clin N Am. 2010;37:345–357.
18. American Society of Anesthesiologists. ASA Committee on Professional Liability. Manual on
Professional Liability. Park Ridge, IL: American Society of Anesthesiologists; 2010.
19. Kelly GD, Blunt C, Moore PAS, et al. Consent for regional anaesthesia in the United Kingdom: what
is material risk? Int J Obstet Anesth. 2004;13:71–74.
20. Bethune L, Harper N, Lucas DN, et al. Complications of obstetric regional analgesia: how much
information is enough? Int J Obstet Anesth. 2004;13:30–34.
21. Plaat F, McGlennan A. Women in the 21st century deserve more information: disclosure of material
risk in obstetric anaesthesia. Int J Obstet Anesth. 2004;13:69–70.
22. Jackson GNB, Robinson PN, Lucas DN, et al. What mothers know, and want to know, about the
complications of general anaesthesia. Acta Anaesthesiol Scand. 2012;56:585–588.
23. Zollo RA, Lurie SJ, Epstein R, et al. Patterns of communication during the preanesthesia visit.
Anesthesiology. 2009;111:971–978.
24. Broaddus BM, Chandrasekhar S. Informed consent in obstetric anesthesia. Anesth Analg.
2011;112:912–915.
25. Waisel DB. Let the patient drive the informed consent process: ignore legal requirements. Anesth
Analg. 2011;113:13–15.
26. Drew NC, Salmon P, Webb L. Mothers’, midwives’ and obstetricians’ views on the features of
obstetric care which influence satisfaction with childbirth. Br J Obstet Gynaecol. 1989;96:1084–1088.
27. Slusarenko P, Noble WH. Epidural anaesthesia: concerns regarding informed consent. Can Anaesth
Soc J. 1985;32:681–682.
28. Lanigan C, Reynolds F. Risk information supplied by obstetric anaesthetists in Britain and Ireland to
mothers awaiting elective caesarean section. Int J Obstet Anesth. 1995;4:7–13.
29. Epstein RM, Korones DN, Quill TE. Withholding information from patients—when less is more. N
Engl J Med. 2010;362:380–381.
30. Weiniger CF, Elchalal U, Sprung CL, et al. Holy consent—a dilemma for medical staff when maternal
consent is withheld for emergency caesarean section. Int J Obstet Anesth. 2006;15:145–148.
31. Simon GR, Wilkins CJ, Smith I. Sevoflurane induction for emergency caesarean section: two case
reports in women with needle phobia. Int J Obstet Anesth. 2002;11:296–300.
32. Marco AP. Informed consent for surgical anesthesia care: has the time come for separate consent?
Anesth Analg. 2010;110:280–282.
33. Marcucci C, Seagull FJ, Loreck D, et al. Capacity to give surgical consent does not imply capacity to
give anesthesia consent: implications for anesthesiologists. Anesth Analg. 2010;110:596–600.
34. Meisel A, Kuczewski M. Legal and ethical myths about informed consent. Arch Intern Med.
1996;156:2521–2526.
35. Brooks H, Sullivan WJ. The importance of patient autonomy at birth. Int J Obstet Anesth.
2002;11:196–203.
36. Burcher P. The Ulysses contract in obstetrics: a woman’s choices before and during labour. J Med
Ethics. 2013;39:27–30.
37. Scott WE. Ethics in obstetric anaesthesia. Anaesthesia. 1996;51:717–718.
38. Thornton J, Moore M. Women who request epidural analgesia in labour should always be given it:
opposer. Int J Obstet Anesth. 1995;4:41–43.
39. American College of Obstetricians and Gynecologists Committee on Ethics. ACOG Committee
Opinion No. 439: informed consent. Obstet Gynecol. 2009;114:401–408.
40. Davies JM, Posner KL, Lee LA, et al. Liability associated with obstetric anesthesia: a closed claims
analysis. Anesthesiology. 2009;110:131–139.
41. Metzner J, Posner KL, Lam MS, et al. Closed claims’ analysis. Best Pract Res Clin Anaesthesiol.
2011;25:263–276.
42. Szypula K, Ashpole KJ, Bogod D, et al. Litigation related to regional anaesthesia: an analysis of
claims against the NHS in England 1995-2007. Anaesthesia. 2010;65:443–452.
43. Hall MA, Bobinski MA, Orentlicher D. Health Care Law and Ethics. 7th ed. New York, NY: Wolters
Kluwer; 2007:284–285.
44. Frenkel DN, Liebman CB. Words that heal. Ann Intern Med. 2004;140:482–483.
45. Van Dusen V, Spies A. Professional apology: dilemma or opportunity? Am J Pharm Educ.
2003;67(4):3.
46. American College of Obstetricians and Gynecologists Committee on Ethics. ACOG Committee
Opinion No. 321: maternal decision making, ethics, and the law. Obstet Gynecol. 2005;106:1127–
1137.
47. Dobson v Dobson, 2 SCR 753 (1999).
48. Re MB. 2 FCR 541 (1997).
49. Winnipeg Child and Family Service v DFG, 3 SCR 925 (1997).
50. Pemberton v Tallahassee, 66 F Supp. 2d, 1247 (ND Fla 1999).
51. Re AC, 573 A 2d, 1235 (DC App 1990).
Nonobstetric Surgery during Pregnancy
Joy L. Hawkins and Debnath Chatterjee
I. Incidence and anesthetic concerns specific to pregnancy
A. Alterations in maternal physiology
B. Maintenance of fetal oxygenation
C. Prevention and treatment of preterm labor
II. Teratogenicity of anesthetic agents
III. Preoperative plan and counseling
IV. Intraoperative anesthetic management
V. Postoperative care
VI. Special situations
A. Trauma
B. Neurosurgical procedures
C. Cardiac surgery requiring cardiopulmonary bypass
D. Laparoscopic surgeries
E. Fetal interventions
Summary
KEYPOINTS
1. A wide variety of nonobstetric surgeries have been performed
successfully during pregnancy with good maternal and fetal outcomes. A
perioperative team approach that includes obstetric consultation,
surgeons, anesthesiology, neonatology, and nursing is key.
2. Physiologic changes of pregnancy will alter preoperative and
intraoperative anesthetic techniques, but no anesthetic agents need be
avoided during pregnancy. No anesthetic medications have been shown to
have any teratogenic effects in humans at any gestational age.
3. Fetal heart rate monitoring should be done preand postoperatively, but
intraoperative monitoring may also provide useful information for
positioning and cardiorespiratory management and may influence the
decision to deliver a viable fetus.
4. Intraoperative and postoperative pneumatic compression devices and
early postoperative ambulation are recommended to prevent deep venous
thrombosis. Optimizing postoperative pain control is critical to
mobilization.
SUMMARY
In conclusion, surgery may be necessary during pregnancy. Anesthesiologists
should reassure the mother that anesthetic drugs and techniques themselves will
not put the fetus or the pregnancy at risk. Prevention of preterm labor is the
greatest concern and may require perioperative monitoring and tocolysis.
Effective postoperative pain management without sedation will aid in early
diagnosis and treatment of preterm labor and assist with early mobilization to
prevent thromboembolic complications.
REFERENCES
1. Reitman E, Flood P. Anaesthetic considerations for non-obstetric surgery during pregnancy. Br J
Anaesth. 2011;107(suppl 1): i72–i78.
2. Koren G, Bologa M, Long D, et al. Perception of teratogenic risk by pregnant women exposed to
drugs and chemicals during the first trimester. Am J Obstet Gynecol. 1989;160:1190–1194.
3. Chohan L, Kilpatrick C. Laparoscopy in pregnancy: a literature review. J Clin Obstet Gynecol.
2009;52:557–569.
4. Lo WY, Friedman JM. Teratogenicity of recently introduced medications in human pregnancy. Obstet
Gynecol. 2002;100:465–473.
5. Mazze RI, Fujinaga M, Baden JM. Halothane prevents nitrous oxide teratogenicity in Sprague-Dawley
rats; folinic acid does not. Teratology. 1988;38:121–127.
6. Mazze RI, Källén B. Reproductive outcome after anesthesia and operation during pregnancy: a
registry study of 5405 cases. Am J Obstet Gynecol. 1989;161:1178–1185.
7. Rosenberg L, Mitchell AA, Parsells JL, et al. Lack of relation of oral clefts to diazepam use during
pregnancy. N Engl J Med. 1983;309:1282–1285.
8. Shiono PH, Mills JL. Oral clefts and diazepam use during pregnancy. N Engl J Med. 1984;311:919–
920.
9. Dolovich LR, Addis A, Vaillancourt JM, et al. Benzodiazepine use in pregnancy and major
malformations or oral cleft: meta-analysis of cohort and case-control studies. BMJ. 1997;317:839–
843.
10. Buhimschi CS, Weiner CP. Medications in pregnancy and lactation (Parts 1 and 2). Obstet Gynecol.
2009;113:166–188 and 417–432.
11. Boivin JF. Risk of spontaneous abortion in women occupationally exposed to anaesthetic gases: a
meta-analysis. Occup Environ Med. 1997;54:541–548.
12. Ikonomidou C, Bosch F, Miksa M, et al. Blockade of NMDA receptors and apoptotic
neurodegeneration in the developing brain. Science. 1999;283:70–74.
13. Ikonomidou C, Bittigau P, Ishimaru MJ, et al. Ethanol-induced apoptotic neurodegeneration and fetal
alcohol syndrome. Science. 2000;287:1056–1060.
14. Jevtovic-Todorovic V, Hartman RE, Izumi Y, et al. Early exposure to common anesthetic agents
causes widespread neurodegeneration in the developing rat brain and persistent learning deficits. J
Neurosci. 2003;23:876–882.
15. Olney JW, Young C, Wozniak DF, et al. Anesthesia-induced developmental neuroapoptosis: does it
happen in humans? Anesthesiology. 2004;101:273–275.
16. Jevtovic-Todorovic V, Absalom AR, Blomgren K, et al. Anaesthetic neurotoxicity and neuroplasticity:
an expert group report and statement based on the BJA Salzburg Seminar. Br J Anaesth.
2013;111:143–151.
17. Palmer SK, Van Norman GA, Jackson SL. Routine pregnancy testing before elective anesthesia is not
an American Society of Anesthesiologists standard. Anesth Analg. 2009;108:1715–1716.
18. Hong JY. Adnexal mass surgery and anesthesia during pregnancy: a 10-year retrospective review. Int J
Obstet Anesth. 2006;15:212–216.
19. American College of Obstetricians and Gynecologists Committee on Obstetric Practice. ACOG
Committee Opinion No. 474: nonobstetric surgery during pregnancy. Obstet Gynecol. 2011;117:420–
421. http://www.asahq.org. Accessed 9/22/15.
20. Immer-Bansi A, Immer FF, Henle S, et al. Unnecessary emergency caesarean section due to silent
CTG during anaesthesia? Br J Anaesth. 2001;87:791–793.
21. Brown HL. Trauma in pregnancy. Obstet Gynecol. 2009;114:147–160.
22. Baysinger CL. Imaging during pregnancy. Anesth Analg. 2010;110:863–867.
23. Wang LP, Paech MJ. Neuroanesthesia for the pregnant woman. Anesth Analg. 2008;107:193–200.
24. Piotin M, de Souza Filho CB, Kothimbakam R, et al. Endovascular treatment of acutely ruptured
intracranial aneurysms in pregnancy. Am J Obstet Gynecol. 2001;185:1261–1262.
25. Khandelwal M, Rasanen J, Ludormirski A, et al. Evaluation of fetal and uterine hemodynamics during
maternal cardiopulmonary bypass. Obstet Gynecol. 1996;88:667–671.
26. John AS, Gurley F, Schaff HV, et al. Cardiopulmonary bypass during pregnancy. Ann Thoracic Surg.
2011;91:1191–1197.
27. Reynolds JD, Booth JV, de la Fuente S, et al. A review of laparoscopy for non-obstetric-related
surgery during pregnancy. Curr Surg. 2003;60:164–173.
28. Uemura K, McClaine RJ, de la Fuente SG, et al. Maternal insufflation during the second trimester
equivalent produces hypercapnia, acidosis, and prolonged hypoxia in fetal sheep. Anesthesiology.
2004;101:1332–1338.
29. Pearl J, Price R, Richardson W, et al. The Society of American Gastrointestinal Endoscopic Surgeons
(SAGES) guidelines for diagnosis, treatment, and use of laparoscopy for surgical problems during
pregnancy. Surg Endosc. 2011;25:3479–3492. doi:10.1007/s00464-011-1927-3.
30. Lin EE, Tran KM. Anesthesia for fetal surgery. Semin Pediatr Surg. 2013;22:50–55.
31. Marwan A, Crombleholme TM. The EXIT procedure: principles, pitfalls, and progress. Semin Pediatr
Surg. 2006;15:107–115.
32. Rychik J, Tian Z, Cohen MS, et al. Acute cardiovascular effects of fetal surgery in the human.
Circulation. 2004;110:1549–1556.
33. Cass DL, Olutoye OO, Cassady CI, et al. EXIT-to-resection for fetuses with large lung masses and
persistent mediastinal compression near birth. J Ped Surg. 2013;48:138–144.
34. Adzick NS, Thom EA, Spong CY, et al. A randomized trial of prenatal versus postnatal repair of
myelomeningocele. N Engl J Med. 2011;364:993–1004.
35. Ferschl M, Ball R, Lee H, et al. Anesthesia for in utero repair of myelomeningocele. Anesthesiology.
2013;118:1211–1223.
36. Golombeck K, Ball RH, Lee H, et al. Maternal morbidity after maternal-fetal surgery. Am J Obstet
Gynecol. 2006;194:834–839.
Labor and Delivery
Fetal Assessment and Monitoring
Michael G. Richardson, Mary DiMiceli-Zsigmond, and David R.
Gambling
I.Physiologic basis of fetal monitoring
II.Electronic fetal monitoring
III.Interpretation of electronic fetal monitoring
IV. Antepartum fetal monitoring
V. Intrapartum fetal surveillance
A. Physiology
B. Intrapartum electronic fetal monitoring–nomenclature and
interpretation
C. Classification of electronic fetal monitoring tracings
D. Management of abnormal electronic fetal monitoring patterns
and in utero resuscitation
E. Intermittent auscultation
VI. Medication and anesthetic effects on fetal surveillance
A. Maternally administered medications
B. Regional anesthesia
C. General anesthesia
KEYPOINTS
1. The goal of antepartum and intrapartum fetal monitoring is to improve
neonatal outcomes, namely to reduce neonatal mortality and neurologic
morbidity.
2. A working knowledge of EFM allows the obstetric anesthesiologist to
anticipate problems and communicate effectively with the obstetrician or
perinatologist.
3. The physiologic basis underlying antepartum fetal monitoring is fetal
dependence on continuous delivery of adequate amounts of oxygen via a
pathway of transfer steps from maternal to fetal circulations. Any process
that interferes with this transfer of oxygen can lead to fetal hypoxia and
acidosis.
4. Electronic fetal monitoring (EFM) has a low positive predictive value,
and there has been considerable interindividual variation as to how a fetal
heart rate (FHR) pattern is interpreted.
5. Other forms of antepartum surveillance include nonstress test, contraction
stress test, biophysical profile, and umbilical artery Doppler velocimetry.
They help to diagnose fetal abnormalities, monitor fetal well-being, and
determine optimal time for delivery.
6. In recent years, renewed attention has been aimed at standardizing
definitions, interpretation of EFM, interventions, and underlying
physiology, with the hope of enhancing the effectiveness and safety of
fetal monitoring and its contribution to better neonatal outcomes.
7. Continuous EFM during labor is associated with a reduction in neonatal
seizures but no difference in rates of cerebral palsy or infant mortality.
However, continuous EFM is associated with an increase in cesarean and
instrumental vaginal deliveries.
8. It is important to understand how certain medications and anesthetic
techniques can affect fetal physiology, either directly or indirectly. This
will, in turn, impact on the ability to interpret EFM appropriately and act
accordingly.
REFERENCES
1. American College of Obstetricians and Gynecologists. ACOG Practice Bulletin No. 145: antepartum
fetal surveillance. Obstet Gynecol. 2014;124:182–192.
2. American College of Obstetricians and Gynecologists. ACOG Practice Bulletin No. 106: intrapartum
fetal heart rate monitoring: nomenclature, interpretation, and general management principles. Obstet
Gynecol. 2009;114:192–202.
3. Liston R, Sawchuck D, Young D. Fetal health surveillance: antepartum and intrapartum consensus
guideline. J Obstet Gynaecol Can. 2007;29:S3–S56.
4. National Collaborating Centre for Women’s and Children’s Health. Intrapartum Care: Care of Healthy
Women and Their Babies During Childbirth, Clinical Guideline 190. London, United Kingdom:
National Institute for Health and Care Excellence; 2014.
5. Royal Australian and New Zealand College of Obstetricians and Gynaecologists. Intrapartum Fetal
Surveillance, Clinical Guideline. 3rd ed. Victoria, Australia: The Royal Australian and New Zealand
College of Obstetricians and Gynaecologists; 2014.
6. Clark SL, Hankins GD. Temporal and demographic trends in cerebral palsy—fact and fiction. Am J
Obstet Gynecol. 2003;188:628–633.
7. Graham EM, Ruis KA, Hartman AL, et al. A systematic review of the role of intrapartum hypoxia-
ischemia in the causation of neonatal encephalopathy. Am J Obstet Gynecol. 2008;199:587–595.
8. Costantine MM, Saade GR. The first cesarean: role of “fetal distress” diagnosis. Semin Perinatol.
2012;36:379–383.
9. Grimes DA, Schulz KF. Uses and abuses of screening tests. Lancet. 2002;359:881–884.
10. Grimes DA, Peipert JF. Electronic fetal monitoring as a public health screening program: the
arithmetic of failure. Obstet Gynecol. 2010;116:1397–1400.
11. Declercq ER, Sakala C, Corry MP, Applebaum S. Listening to Mothers II: report of the Second
National U.S. Survey of Women’s Childbearing Experiences: conducted January–February 2006 for
Childbirth Connection by Harris Interactive in partnership with Lamaze International. J Perinat Educ.
2007;16:15–17. doi:10.1624/105812407X244778.
12. Miller DA, Miller LA. Electronic fetal heart rate monitoring: applying principles of patient safety. Am
J Obstet Gynecol. 2012;206:278–283.
13. Baschat AA. Fetal growth restriction—from observation to intervention. J Perinat Med. 2010;38:239–
246.
14. Fletcher AJ, Gardner DS, Edwards CM, et al. Development of the ovine fetal cardiovascular defense
to hypoxemia towards full term. Am J Physiol Heart Circ Physiol. 2006;291:H3023–H3034.
15. Thompson JL, Kuller JA, Rhee EH. Antenatal surveillance of fetal growth restriction. Obstet Gynecol
Surv. 2012;67:554–565.
16. Sureau C. Historical perspectives: forgotten past, unpredictable future. Baillieres Clin Obstet
Gynaecol. 1996;10:167–184.
17. National Institute of Child Health and Human Development Research Planning Workshop. Electronic
fetal heart rate monitoring: research guidelines for interpretation. National Institute of Child Health
and Human Development Research Planning Workshop. Am J Obstet Gynecol. 1997;177:1385–1390.
18. 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.
19. American College of Obstetricians and Gynecologists. ACOG Practice Bulletin No. 116: management
of intrapartum fetal heart rate tracings. Obstet Gynecol. 2010;116:1232–1240.
20. Clark SL, Nageotte MP, Garite TJ, et al. Intrapartum management of category II fetal heart rate
tracings: towards standardization of care. Am J Obstet Gynecol. 2013;209:89–97.
21. Parer JT, Ikeda T. A framework for standardized management of intrapartum fetal heart rate patterns.
Am J Obstet Gynecol. 2007;197:26.e1–26.e6.
22. Parer JT, Hamilton EF. Comparison of 5 experts and computer analysis in rule-based fetal heart rate
interpretation. Am J Obstet Gynecol. 2010;203:451.e1–451.e7.
23. Coletta J, Murphy E, Rubeo Z, et al. The 5-tier system of assessing fetal heart rate tracings is superior
to the 3-tier system in identifying fetal acidemia. Am J Obstet Gynecol. 2012;206:226.e1–226.e5.
24. Sadaka A, Furuhashi M, Minami H, et al. Observation on validity of the five-tier system for fetal heart
rate pattern interpretation proposed by Japan Society of Obstetricians and Gynecologists. J Matern
Fetal Neonatal Med. 2011;24:1465–1469.
25. Garite TJ, Simpson KR. Intrauterine resuscitation during labor. Clin Obstet Gynecol. 2011;54:28–39.
26. Westgate JA, Wibbens B, Bennet L, et al. The intrapartum deceleration in center stage: a physiologic
approach to the interpretation of fetal heart rate changes in labor. Am J Obstet Gynecol.
2007;197:236.e1–236.e11.
27. Reddy UM, Filly RA, Copel JA. Prenatal imaging: ultrasonography and magnetic resonance imaging.
Obstet Gynecol. 2008;112:145–157.
28. Clark SL, Sabey P, Jolley K. Nonstress testing with acoustic stimulation and amniotic fluid volume
assessment: 5973 tests without unexpected fetal death. Am J Obstet Gynecol. 1989;160:694–697.
29. Miller DA, Rabello YA, Paul RH. The modified biophysical profile: antepartum testing in the 1990s.
Am J Obstet Gynecol. 1996;174:812–817.
30. Freeman RK, Anderson G, Dorchester W. A prospective multi-institutional study of antepartum fetal
heart rate monitoring. I. Risk of perinatal mortality and morbidity according to antepartum fetal heart
rate test results. Am J Obstet Gynecol. 1982;143:771–777.
31. Devoe LD. Antenatal fetal assessment: contraction stress test, nonstress test, vibroacoustic stimulation,
amniotic fluid volume, biophysical profile, and modified biophysical profile—an overview. Semin
Perinatol. 2008;32:247–252.
32. Ray M, Freeman R, Pine S, et al. Clinical experience with the oxytocin challenge test. Am J Obstet
Gynecol. 1972;114:1–9.
33. Manning FA, Harman CR, Morrison I, et al. Fetal assessment based on fetal biophysical profile
scoring. IV. An analysis of perinatal morbidity and mortality. Am J Obstet Gynecol. 1990;162:703–
709.
34. Manning FA, Snijders R, Harman CR, et al. Fetal biophysical profile score. VI. Correlation with
antepartum umbilical venous fetal pH. Am J Obstet Gynecol. 1993;169:755–763.
35. Vintzileos AM, Nochimson DJ, Guzman ER, et al. Intrapartum electronic fetal heart rate monitoring
versus intermittent auscultation: a meta-analysis. Obstet Gynecol. 1995;85:149–155.
36. Morrow RJ, Adamson SL, Bull SB, et al. Effect of placental embolization on the umbilical arterial
velocity waveform in fetal sheep. Am J Obstet Gynecol. 1989;161:1055–1060.
37. Mandruzzato GP, Bogatti P, Fischer L, et al. The clinical significance of absent or reverse end-diastolic
flow in the fetal aorta and umbilical artery. Ultrasound Obstet Gynecol. 1991;1:192–196.
38. Karsdorp VH, van Vugt JM, van Geijn HP, et al. Clinical significance of absent or reversed end
diastolic velocity waveforms in umbilical artery. Lancet. 1994;344:1664–1668.
39. Vasconcelos RP, Brazil Frota Aragão JR, Costa Carvalho FH, et al. Differences in neonatal outcome in
fetuses with absent versus reverse end-diastolic flow in umbilical artery Doppler. Fetal Diagn Ther.
2010;28:160–166.
40. Valcamonico A, Danti L, Frusca T, et al. Absent end-diastolic velocity in umbilical artery: risk of
neonatal morbidity and brain damage. Am J Obstet Gynecol. 1994;170:796–801.
41. Trudinger BJ, Cook CM, Giles WB, et al. Fetal umbilical artery velocity waveforms and subsequent
neonatal outcome. Br J Obstet Gynaecol. 1991;98:378–384.
42. Yoon BH, Romero R, Roh CR, et al. Relationship between the fetal biophysical profile score,
umbilical artery Doppler velocimetry, and fetal blood acid-base status determined by cordocentesis.
Am J Obstet Gynecol. 1993;169:1586–1594.
43. Soregaroli M, Bonera R, Danti L, et al. Prognostic role of umbilical artery Doppler velocimetry in
growth-restricted fetuses. J Matern Fetal Neonatal Med. 2002;11:199–203.
44. Alfirevic Z, Stampalija T, Gyte GM. Fetal and umbilical Doppler ultrasound in high-risk pregnancies.
Cochrane Database Syst Rev. 2010;(1):CD007529.
45. Bricker L, Neilson JP. Routine Doppler ultrasound in pregnancy. Cochrane Database Syst Rev. 2000;
(2):CD001450.
46. Hankins GD, Miller DA. A review of the 2008 NICHD Research Planning Workshop:
recommendations for fetal heart rate terminology and interpretation. Clin Obstet Gynecol. 2011;54:3–
7.
47. Sholapurkar SL. Critical evaluation of American categorization of fetal heart rate (FHR) decelerations
and three tier classification—shortcomings, contradictions, remedies and need for debate. Open J
Obstet Gynecol. 2013;3:362–370.
48. Ugwumadu A. Are we (mis)guided by current guidelines on intrapartum fetal heart rate monitoring?
Case for a more physiological approach to interpretation. BJOG. 2014;121:1063–1070.
49. Hill JB, Chauhan SP, Magann EF, et al. Intrapartum fetal surveillance: review of three national
guidelines. Am J Perinatol. 2012;29:539–550.
50. Higuchi H, Takagi S, Zhang K, et al. Effect of lateral tilt angle on the volume of the abdominal aorta
and inferior vena cava in pregnant and nonpregnant women determined by magnetic resonance
imaging. Anesthesiology. 2015;122:286–293.
51. Palmer CM. Tilting at aortocaval compression. Anesthesiology. 2015;122:231–232.
52. Alfirevic Z, Devane D, Gyte GM. Continuous cardiotocography (CTG) as a form of electronic fetal
monitoring (EFM) for fetal assessment during labour. Cochrane Database Syst Rev. 2013;
(5):CD006066.
53. Devane D, Lalor JG, Daly S, et al. Cardiotocography versus intermittent auscultation of fetal heart on
admission to labour ward for assessment of fetal wellbeing. Cochrane Database Syst Rev. 2012;
(2):CD005122.
54. Hill JB, Alexander JM, Sharma SK, et al. A comparison of the effects of epidural and meperidine
analgesia during labor on fetal heart rate. Obstet Gynecol. 2003;102:333–337.
55. Sekhavat L, Behdad S. The effects of meperidine analgesia during labor on fetal heart rate. Int J
Biomed Sci. 2009;5:59–62.
56. Nelson KE, Eisenach JC. Intravenous butorphanol, meperidine, and their combination relieve pain and
distress in women in labor. Anesthesiology. 2005;102:1008–1013.
57. Evron S, Glezerman M, Sadan O, et al. Remifentanil: a novel systemic analgesic for labor pain. Anesth
Analg. 2005;100:233–238.
58. Douma MR, Verwey RA, Kam-Endtz CE, et al. Obstetric analgesia: a comparison of patient-
controlled meperidine, remifentanil, and fentanyl in labour. Br J Anaesth. 2010;104:209–215.
59. Verdurmen KM, Renckens J, van Laar JO, et al. The influence of corticosteroids on fetal heart rate
variability: a systematic review of the literature. Obstet Gynecol Surv. 2013;68:811–824.
60. Duffy CR, Odibo AO, Roehl KA, et al. Effect of magnesium sulfate on fetal heart rate patterns in the
second stage of labor. Obstet Gynecol. 2012;119:1129–1136.
61. Hallak M, Martinez-Poyer J, Kruger ML, et al. The effect of magnesium sulfate on fetal heart rate
parameters: a randomized, placebo-controlled trial. Am J Obstet Gynecol. 1999;181:1122–1127.
62. Robins K, Lyons G. Supraventricular tachycardia in pregnancy. Br J Anaesth. 2004;92:140–143.
63. Chen RJ, Huang SC, Chow SN. Paroxysmal supraventricular tachycardia during pregnancy and
postpartum period. Int J Gynaecol Obstet. 1994;44:279–280.
64. Gowda RM, Khan IA, Mehta NJ, et al. Cardiac arrhythmias in pregnancy: clinical and therapeutic
considerations. Int J Cardiol. 2003;88:129–133.
65. Elkayam U, Goodwin TM. Adenosine therapy for supraventricular tachycardia during pregnancy. Am
J Cardiol. 1995;75:521–523.
66. Abdelhak Y, Roque H, Young BK. Terbutaline: effects on the fetal heart at term. J Perinat Med.
2011;40:69–71.
67. Clarke VT, Smiley RM, Finster M. Uterine hyperactivity after intrathecal injection of fentanyl for
analgesia during labor: a cause of fetal bradycardia? Anesthesiology. 1994;81:1083.
68. Gaiser RR, McHugh M, Cheek TG, et al. Predicting prolonged fetal heart rate deceleration following
intrathecal fentanyl/bupivacaine. Int J Obstet Anesth. 2005;14:208–211.
69. Patel NP, El-Wahab N, Fernando R, et al. Fetal effects of combined spinal-epidural vs epidural labour
analgesia: a prospective, randomised double-blind study. Anaesthesia. 2014;69:458–467.
70. Gambling DR, Bender M, Faron S, et al. Prophylactic intravenous ephedrine to minimize fetal
bradycardia after combined spinal-epidural analgesia: a randomized controlled study. Can J Anesth.
2015;62:1201–1208. doi:10.1007/s12630-015-0450-8.
71. Mardirosoff C, Dumont L, Boulvain M, et al. Fetal bradycardia due to intrathecal opioids for labour
analgesia: a systematic review. BJOG. 2002;109:274–281.
72. Abrão KC, Francisco RP, Miyadahira S, et al. Elevation of uterine basal tone and fetal heart rate
abnormalities after labor analgesia: a randomized controlled trial. Obstet Gynecol. 2009;113:41–47.
73. Van de Velde M, Teunkens A, Hanssens M, et al. Intrathecal sufentanil and fetal heart rate
abnormalities: a double-blind, double placebo-controlled trial comparing two forms of combined
spinal epidural analgesia with epidural analgesia in labor. Anesth Analg. 2004;98:1153–1159.
74. St. Amant MS, Koffel B, Malinow AM. The effects of epidural opioids on fetal heart rate variability
when coadministered with 0.25% bupivacaine for labor analgesia. Am J Perinatol. 1998;15:351–356.
75. Cigarini I, Kaba A, Bonnet F, et al. Epidural clonidine combined with bupivacaine for analgesia in
labor. Effects on mother and neonate. Reg Anesth. 1995;20:113–120.
76. Rosen MA. Paracervical block for labor analgesia: a brief historic review. Am J Obstet Gynecol.
2002;186:S127–S130.
77. Junttila EK, Karjalainen PK, Ohtonen PP, et al. A comparison of paracervical block with single-shot
spinal for labour analgesia in multiparous women: a randomised controlled trial. Int J Obstet Anesth.
2009;18:15–21.
78. LeFevre ML. Fetal heart rate pattern and postparacervical fetal bradycardia. Obstet Gynecol.
1984;64:343–346.
79. Palomäki O, Huhtala H, Kirkinen P. A comparative study of the safety of 0.25% levobupivacaine and
0.25% racemic bupivacaine for paracervical block in the first stage of labor. Acta Obstet Gynecol
Scand. 2005;84:956–961.
80. Clark RB, Brown MA, Lattin DL. Neostigmine, atropine, and glycopyrrolate: does neostigmine cross
the placenta? Anesthesiology. 1996;84:450–452.
Maternal Infection and Fever
Rebecca D. Minehart, William Camann, and Scott Segal
I. Fever in pregnancy
A. General considerations
B. Neonatal outcome
II. Noninfectious fever in parturients
A. Noninfectious fever and epidural analgesia for labor
B. Mechanisms
III. Infectious causes of fever in parturients
A. Chorioamnionitis
B. Respiratory tract infection: pneumonia
C. Respiratory tract infection: influenza
D. Urinary tract infection
E. Postpartum infection (endometritis)
F. Group B streptococcus
G. Cytomegalovirus
H. Hepatitis
I. HIV
J. Herpes simplex virus (HSV-1 and HSV-2)
K. Miscellaneous infections
IV. Sepsis and septic shock
A. General considerations
B. Treatment
C. Anesthetic management
V. Neuraxial anesthesia for the febrile parturient
A. Risk
B. IV antibiotics
C. Aseptic technique
KEYPOINTS
1. Whatever the cause of maternal fever, it is clear that higher maternal
temperatures can adversely impact neonatal outcomes.
2. Multiple studies of various designs have confirmed the excess fever in
women receiving epidural analgesia in labor.
3. The effect of epidural-associated fever, as opposed to infection, has not
been definitively linked to neonatal brain injury.
4. Postpartum, the anesthesia provider must be prepared to treat possible
uterine atony and hemorrhage, which can occur after vaginal delivery or
cesarean delivery (CD) in pregnancies complicated by chorioamnionitis.
5. If there are serious complications of influenza, supportive care is
warranted on an individual basis, paying particular attention to
intravascular volume status during neuraxial anesthesia because
dehydration may occur.
6. Without evidence of coagulopathy, neuraxial anesthesia can be
administered in parturients with hepatitis. However, in patients with
severe liver disease, metabolism of local anesthetics is also of concern
because of hepatic biotransformation of amino-amide local anesthetics.
7. One of the biggest concerns of administering neuraxial anesthesia to
patients with HIV infection is whether these patients are at increased risk
for infectious complications. Safe administration of neuraxial anesthesia
requires standard precautions and careful avoidance of needlestick injury.
8. Consensus is that neuraxial anesthesia is safe for secondary or re-
activated HSV infection, provided there are no active herpetic lesions at
the site of needle insertion.
9. Patients with varicella zoster may present challenges in pain management
because the pain of zoster is difficult to control and requires careful
evaluation of all available modalities.
10. The Guidelines for the International Surviving Sepsis Campaign call for
administration of high-dose intravenous antibiotics within 1 hour of
admission for anyone with suspected sepsis.
11. Handwashing; jewelry removal; and use of sterile gloves (to supplement
handwashing and not as a substitute), fresh mask, and cap have been
shown to reduce the incidence of microbial contamination of the work
area but not directly to reduce nosocomial infection.
B. Mechanisms
1. The mechanisms for the increased temperature are
incompletely understood. Formerly, thermoregulatory
factors including ambient temperature, decreased sweating, or
decreased hyperventilation were implicated. However, with
the realization that the gradual rise in temperature is an
averaging artifact, it is likely that the relevant mechanisms are
those that explain fever in the 20% to 30% of women who
develop it. The most likely cause is noninfectious (sterile)
inflammation, discussed in the following sections.
2. Inflammatory markers have been evaluated in parturients
with and without fever who received epidurals. Goetzl et al.24
demonstrated increased serum levels of inflammatory
cytokines (IL-6, IL-8) in maternal and fetal cord blood
samples in those with fever who had received epidural
analgesia. The levels were higher at baseline in those women
who eventually developed a fever, and the levels rose with
longer exposure to epidural analgesia.
3. Placental inflammation is consistently observed but is not
infectious. In reviewing placental findings in large cohorts of
women who self-selected epidural analgesia, placental
inflammation was consistently observed in the febrile
groups.25,26 More importantly, assiduous efforts to culture
bacteria or detect bacterial DNA failed to demonstrate
infection in all but rare cases.27 Similarly, prophylactic broad-
spectrum antibiotics did not alter placental inflammation or
maternal fever in a randomized trial.28
4. Anti-inflammatory agents moderate epidural-associated
fever. Goetzl et al.29 randomized women with epidural
analgesia to receive high-dose systemic methylprednisolone
or placebo. Steroid treatment prevented fever and increases in
maternal and fetal IL-6 but at the cost of nearly 10% neonatal
bacteremia. Similar effects were seen with epidural steroids30
but not with the weak anti-inflammatory agent. Prophylactic
acetamenophen is also ineffective in preventing maternal
fever.31
The effect of epidural-associated fever, as opposed to
infection, has not been definitively linked to neonatal brain
injury. Nonetheless, because the pathophysiology is likely
similar, and because maternal inflammation consistently
reproduces newborn brain injuries in experimental models,
continued efforts to elucidate the mechanisms and potential
ways to modulate these effects are indicated.
2. Chronic hepatitis
a. Importance of screening. Chronic hepatitis can be a
multisystem disorder with complications such as
cirrhosis, hepatic failure, hepatocellular carcinoma, or
even death. Parturients who are carriers or have a history
of chronic hepatitis should be fully examined (by both
physical examination and laboratory studies, including
liver function tests) to assess severity of the disease.
Specific vaccines and immunoglobulins are available for
hepatitis A and B. All pregnant women should undergo
screening for hepatitis B (as recommended by the
Centers for Disease Control and Prevention and the
ACOG65) because vertical transmission can lead to
neonatal hepatitis. HB immune globulin and antiviral
therapy with lamivudine have shown some promise in
reducing vertical transmission.
b. Neonatal transmission. In addition to these
considerations, neonatal immunoprophylaxis should be
administered to attenuate neonatal infection. Without
prophylaxis, the risk of infection is 10% to 20% when the
mother is hepatitis B surface-antigen positive. If the
mother is hepatitis B virus core component e-antigen
positive, the risk of neonatal infection is approximately
85%.65 Maternal (i.e., vertical) transmission of
hepatitis B infection to the neonate remains an
important public health concern.
3. Anesthetic considerations
Anesthetic concerns typically involve assessment of
severity of the hepatitis, whether acute or chronic.
Parturients should be evaluated for clotting abnormalities in
the presence of severe liver dysfunction. Clotting factors
should be replaced before administration of neuraxial
anesthesia.
a. Neuraxial anesthesia
Without evidence of coagulopathy, neuraxial anesthesia
can be administered. In patients with severe liver disease,
metabolism of local anesthetics is also of concern
because of the hepatic biotransformation of amino-amide
local anesthetics. In patients with severe disease, there is
reduced blood flow to the liver and liver function is poor.
This may result in significantly higher blood levels of
amino-amide local anesthetics and reduced rate of
degradation. Hepatic pseudocholinesterase
concentrations can also be decreased in patients with
liver disease. This may decrease the clearance of 2-
chloroprocaine and other amino-ester local anesthetics.
Theoretically, engorgement of the epidural venous plexus
may be greater in the parturient with portal hypertension
and esophageal varices, compared to a normal pregnancy,
perhaps increasing the risk of intravascular placement,
enhanced local anesthetic spread, or epidural hematoma.
Experts generally recommend careful administration of
drugs usually administered during epidural anesthesia.64
Theoretically, spinal anesthesia may be more
advantageous because of the small dose of local
anesthetic administered. However, there may be concerns
about rapid onset sympathetic blockade in the presence of
hypovolemia.
b. General anesthesia
In cases complicated by coagulopathy, severe
hemorrhage, or umbilical cord prolapse, general
anesthesia may be necessary. The distribution and
metabolism of medications may be altered and should be
taken into account. Intravascular volume should be
evaluated with consideration for invasive monitoring
when ascites and/or cardiovascular compromise are
present. Rapid sequence induction can be facilitated
with a variety of induction agents, but the choice of agent
should be based on the patient’s hemodynamic status.
Although liver disease may affect pseudocholinesterase
concentrations and delay metabolism of
succinylcholine, this is of minimal clinical concern.
Because of potential delayed clearance of opioids, they
should be administered judiciously. Finally, it is important
to avoid hypoxia and reduced hepatic blood flow during
general anesthesia to not worsen liver function.64 (see
Chapter 27 for further detail on anesthetic considerations
for liver disease.)
I. Human immunodeficiency virus
1. General considerations
In the early part of the 21st century, the fastest growing
population of newly infected individuals with HIV were
women of reproductive age, but fortunately, the rate of
infection in women decreased from 2009 to 2013, the most
recently reported data.66 In light of rapidly evolving standards
of care for HIV-infected individuals, a website
(aidsinfo.nih.gov) (last accessed April 8, 2015) has been
created and should be accessed by those caring for such
patients. Transmission of HIV from mother to fetus is
thought to occur 30% of the time in utero in women not
receiving antiretroviral medication. It is estimated that 70%
of cases of vertical transmission occur during labor and
delivery in nonbreastfed infants.67 Many women are given
antiretroviral treatments during labor to decrease this risk, and
women with high viral loads often undergo CD. This strategy
has been successful in preventing vertical transmission in
approximately 99% of cases.68,69
2. Systemic manifestations
With the widespread adoption of highly active antiretroviral
therapy (HAART), HIV and its progression to AIDS no
longer primarily manifests as opportunistic infection but still
can involve multiple organ systems. Common anesthetic
considerations include careful evaluation of all organ systems
affected. There may be abnormalities of pulmonary,
gastrointestinal, hematologic, cardiovascular, endocrine,
and/or renal systems. In addition, the conditions that led to
HIV infection in the parturient (most notably substance abuse)
may complicate anesthetic and obstetrical management. The
progression of disease in terms of CD4+ cell count or HIV-1
RNA levels may be helpful in predicting mortality after
surgery, although this relationship may be less valid in the era
of HAART.70
a. Neurologic disease
Neurologic involvement occurs early during primary
HIV infection.71 During the initial infection, patients may
complain of headache, photophobia, and retro-orbital
pain. Cognitive or affective disorders may also be
observed. In addition, some cases may be complicated by
meningoencephalitis as well as cranial and peripheral
neuropathies. Others may be asymptomatic but still have
cerebral spinal fluid abnormalities. During the latent
phase of infection, patients may present with a
demyelinating neuropathy, similar to Guillain-Barré
syndrome. This is presumed to be autoimmune in nature.
It usually responds to IV immunoglobulin or
plasmapheresis. The late stages of HIV infection are
manifest by profound neurologic deterioration in
nearly all patients. These complications can include
meningitis, diffuse encephalopathy, focal brain disorders,
myelopathy, peripheral neuropathy, AIDS dementia
complex, inflammatory myopathy, and autonomic
neuropathy.72
b. Pulmonary complications are not caused directly by the
HIV but result from opportunistic infections associated
with the disease. The most common infective agent is
Pneumocystis jiroveci (formerly carinii). This fungal
pathogen is responsible for a pneumonia causing a clinical
presentation similar to adult respiratory distress syndrome
(i.e., severe hypoxemia and diffuse interstitial infiltrates
on chest radiograph). Respiratory failure requiring
intubation was formerly and nearly always fatal; in
the era of improved ICU care, mortality is still high
but has been reduced to approximately 50% overall
with a Pneumocystis mortality of approximately
10%.73,74 Other pulmonary diseases include reactivation
of latent tuberculosis (TB), bacterial infections (e.g., S.
pneumoniae, H. influenzae), and other fungal infections
(e.g., Aspergillus, Cryptococcus, Coccidiodes).
c. Gastrointestinal involvement
Large quantities of lymphoid tissue in the gut contribute
to viral replication in this organ system. Nearly all
patients with HIV infection complain of gastrointestinal
disturbances during the disease. These abnormalities can
include esophagitis, severe diarrhea, or hepatobiliary
disease. A variety of pathogens are responsible for these
disorders, including HSV, Candida albicans, CMV,
Mycobacterium avium, TB, and Cryptosporidia. HAART
toxicity also contributes to gastrointestinal symptoms.75
d. Hematologic disorders can result from HIV infection of
peripheral cell lines as well as antiretroviral agents that
produce hematologic toxicity. Cytopenias (most notably
leukopenia) are the most common abnormalities and may
manifest as anemia or thrombocytopenia. HAART has
been successful at preventing or reversing many of these
abnormalities.76
e. Cardiovascular disease
Although overt cardiovascular disease is uncommon in
these patients, lymphocytic infiltration of the myocardium
is present in up to 50% of cases.77 Pericarditis is the most
prevalent disorder, but others include pulmonary
hypertension, focal myocarditis, and infective
endocarditis.
f. Endocrine disorders typically result from HIV infection
of endocrine tissue, opportunistic infections, or
antiretroviral therapy.78 Involvement of the pituitary,
adrenal, and thyroid glands at autopsy is common, but
clinical evidence of disease is rare. Other endocrine
involvement can result in either hypoglycemia or
hyperglycemia from antiretroviral therapies (e.g., protease
inhibitors, pentamidine).
g. Renal disease
Sepsis, dehydration, and drug toxicity contribute to
the risk of acute renal failure and chronic renal
insufficiency in these patients. In addition, patients with
HIV infection may develop renal failure from HIV-
associated immune complex nephropathy. Antiretroviral
drug therapy is associated with renal toxicity over time
and may be more common than direct HIV effects in the
HAART era.79
3. Anesthetic considerations
a. Neuraxial anesthesia
(1) Infectious risk. One of the biggest concerns of
administering neuraxial anesthesia to patients
with HIV infection is whether these patients are at
increased risk for infectious complications.
Although there is no evidence suggesting that these
patients are at increased risk for such complications,
strict aseptic technique must be maintained. Besides
adherence to the precautions for safe administration
of neuraxial anesthesia recommended by the
American Society of Regional Anesthesia (ASRA)
(see Chapter 10),80 the American Society of
Anesthesiologists (ASA) Subcommittee on
Infection Control has recommended use of standard
precautions and careful avoidance of needlestick
injury.81 Other concerns are related to the
administration of neuraxial anesthesia in patients who
may develop neurologic deficits in the future.
Although CNS involvement occurs very early in
the course of HIV disease and the deficits will not
likely be temporally related to the neuraxial
anesthetic, this should not be a concern when
contemplating neuraxial anesthesia.
K. Miscellaneous infections
1. Lyme disease. Twelve strains of the spirochete Borrelia
burgdorferi sensu lato cause Lyme disease, which is the most
common vector-borne infection in the United States. It is
transmitted by the bites of deer ticks (Ixodes scapularis) as
well as Western black-legged ticks (Ixodes pacificus).
Common presentation of Lyme disease is a “bull’s-eye” rash
(erythema migrans) which spreads outward from the initial
site of inoculation and may be accompanied by fever, malaise,
fatigue, myalgias, arthralgias, and headaches. Some
parturients may be asymptomatic; however, others may
experience more serious cardiac and neurologic
manifestations. Fetal risk with maternal infection is largely
unknown and appears low, with no evidence of congenital
transmission.93 Antimicrobial treatment is warranted in
maternal Lyme disease, although the first-line drug,
doxycycline is contraindicated in pregnancy and cefuroxime
or amoxicillin are the preferred alternatives.93 Anesthetic
considerations in the parturient with Lyme disease focus
on severity of disease, specifically cardiac and neurologic
involvement. If there are indications of central or
peripheral nervous system involvement, it may be wise to
avoid neuraxial anesthetic techniques, although there are
case reports of successful spinal anesthesia in infected
parturients.
2. Listeria. Listeria monocytogenes is the gram-positive rod
bacterium responsible for listeriosis. Listeriosis is rare in the
nonpregnant population, but infection is increased 20-fold
during pregnancy.94 Listeria survive at temperatures used
for refrigeration and may be transmitted in many ready-to-eat
food products that have been properly refrigerated.95 Many
parturients are advised to avoid such food products during
their pregnancy. Common signs and symptoms of listeriosis
include fever, headache, myalgias, and gastrointestinal
symptoms (including diarrhea). Rarely, severe maternal
complications (e.g., death) may occur from Listeria
infections but are mainly related to underlying
immunocompromised conditions. Fetal and neonatal
infection can be devastating, with a mortality rate of
approximately 20%.96 Prompt antimicrobial treatment must
be instituted to improve fetal survival.
3. Parvovirus B19 is the virus responsible for erythema
infectiosum (fifth disease) as well as transient aplastic crisis
in patients with hemoglobinopathies. Transmission is
primarily through hand-to-mouth contact and respiratory
secretions. Symptoms may include fever, sore throat, reticular
rash, and peripheral arthropathy, with the latter two occurring
up to several weeks after the initial infection. Disease can be
mild in parturients, even those who are additionally
immunocompromised.97 Fetal infection with parvovirus B19
may result in fetal hydrops (due to myocarditis and/or
anemia) or fetal loss, especially if maternal infection occurs in
the first 20 weeks of gestation. Intrauterine transfusion of the
fetus may be required.97
4. Syphilis is caused by infection with the spirochete Treponema
pallidum, most commonly transmitted through sexual contact.
Syphilis infections are on the rise with increased HIV
infections and IV drug abuse. Eighty percent of women who
are currently infected are of reproductive age. Pregnancy
does not typically alter the course of syphilis, but syphilis
may be transmitted from mother to fetus, resulting in
intrauterine growth restriction, preterm birth, stillbirth,
neonatal death, and congenital infection.98 Serologic
testing during prenatal visits and treatment of syphilis with
penicillin helps to reduce the maternal–fetal transmission rate
from 70%–100% to 1%–2%.99 There are few specific
anesthetic considerations in parturients with syphilis
unless there is severe late stage disease affecting the CNS or
aorta. However, parturients presenting with syphilis often
have had no or poor prenatal care, and infection may be a
marker for cocaine use.100
5. Tuberculosis. Despite a decreasing prevalence in the United
States, TB continues to be a significant problem in urban
areas and among non–US-born populations.101,102 TB is
spread primarily through aerosolized droplets (typically
produced by coughing) that enter the respiratory system.
Although pregnancy does not appear to change the course of
TB, active TB and the emergence of multidrug-resistant M.
tuberculosis can pose serious problems to parturients,
neonates, and health care providers.102 In urban areas, it is
routine for pregnant women to be screened for TB by purified
protein derivative (PPD) skin testing during their prenatal
visits. A follow-up chest x-ray should be performed for
positive PPD results. This will document presence of active
disease. Active TB should be treated during pregnancy with
first-line therapy including isoniazid, rifampin, and/or
ethambutol.103 All are considered to be safe for the fetus, but
isoniazid is associated with an increased incidence of hepatitis
during pregnancy; therefore, liver function tests must be
followed closely. It has been suggested that extrapulmonary
TB, possibly of the placenta, uterus, or genital tract, is
associated with increased maternal and fetal morbidity as well
as increased risk of congenital or neonatal TB.104 Caregivers
should take extra precautions to avoid exposure to infection in
the presence of active TB (such as placing the parturient in a
negative-pressure room and having exposed persons wear
N95 particulate respirator masks).
6. Varicella zoster virus (VZV) is a member of the herpes virus
family. There are two distinct forms of the disease. Varicella,
or chickenpox, is a febrile, systemic disease associated with
generalized pruritic vesicles, whereas herpes zoster (or
“shingles”) is a reactivation of the latent virus, which
resides in sensorineural ganglia. In cases of shingles, the
patient experiences eruption of painful vesicles in a unilateral
dermatomal distribution. Primary varicella can also present
with encephalitis or pneumonia (in 20% to 25% of primary
infections occurring as an adult), with occasional rapid
respiratory failure in the latter. If varicella pneumonia occurs,
it must be treated with IV acyclovir. Previously, varicella
pneumonia carried a 36% to 41% mortality rate in
parturients.However, introduction of antiviral agents (e.g.,
acyclovir) as well as earlier and improved recognition
have decreased maternal mortality to 13% to 14%.105 If
primary varicella occurs, the natural progression of both
neonatal varicella and varicella in the parturient may be
attenuated by administration of varicella zoster immune
globulin (VZIG), which must be administered within 96
hours of exposure to the virus. If acute primary varicella is
present at the time of delivery, there is current debate about
how to best manage anesthesia. Because viremia may be
present for up to 2 weeks following onset of cutaneous
lesions, some authors recommend avoiding neuraxial
anesthesia. However, others would support its use due to the
high risk of varicella pneumonia during the course of the
disease, and there have been published case reports of
uneventful outcome after spinal anesthesia,106 perhaps after
administration of VZIG.88 If neuraxial anesthesia is
administered to a patient with primary or recurrent VZV
infection, spinal or epidural needles should probably not
be introduced through cutaneous lesions. Patients with
zoster may also present challenges in pain management
because the pain of zoster is difficult to control and requires
careful evaluation of all available modalities.
C. Anesthetic management
If delivery becomes necessary, neuraxial anesthetic techniques
may not be a viable choice due to aberrant hemodynamic,
respiratory, and coagulation parameters. When administering
general anesthesia, it is important to choose induction agents
which support the cardiovascular system, such as etomidate or
ketamine. These agents will not prolong the time from induction
to delivery, avoiding further compromise to the fetus. In addition,
if a severe intraabdominal infection is suspected, it may be
prudent to avoid succinylcholine because it may cause
hyperkalemia in this setting.117 Rocuronium may be preferred for
rapid sequence induction due to its rapid onset of action. Up to
40% of pregnant patients with septic shock may need to undergo
surgery.109
V. Neuraxial anesthesia for the febrile parturient
Neuraxial anesthesia is generally preferred to general anesthesia in
obstetrics, with reduced mortality and serious morbidity compared to
general anesthesia. If general anesthesia is required for seriously
infected patients, the risk of hemodynamic instability, airway
compromise, and electrolyte abnormalities (discussed earlier) should
be considered. Neuraxial anesthesia in febrile patients poses
additional considerations.
A. Risk
There is some concern about causing infection while performing
neuraxial anesthetic techniques in febrile parturients. However, in
most cases, these techniques are safe when performed after
administration of appropriate antimicrobial agents. In a classic
paper by Carp and Bailey,118 treatment with a single dose of
gentamicin before dural puncture in rats with E. coli bacteremia
appeared to eliminate the risk of infection; however, it is unclear
if this applies to the febrile obstetric population. Most spinal or
epidural infections appear to be related to surgical procedures,
hematogenous spread of infectious agents, prolonged
catheterization, compromised immune status, and/or a lapse in
sterile technique, but many cases are spontaneous in origin. The
overall incidence of epidural abscess formation in parturients
receiving neuraxial anesthesia is unknown, largely due to the
rarity of occurrence and lack of centralized reporting.
However, two large prospectively collected audits, one in the
United Kingdom119 and one in the United States120 calculated the
risk between approximately 1:60,000 and 1:300,000, albeit with
very wide confidence intervals given the rarity of the outcome.
The most common pathogens associated with spinal or epidural
abscesses are S. aureus (50%), various streptococci (15%), and
gram-negative rods (15% to 20%).121 Organisms cultured from
anesthesiologists’ nasopharynx or skin have been implicated in
some case reports.
B. IV antibiotics
It is reasonable to require administration of appropriate IV
antibiotics before placement of a neuraxial anesthetic in a
febrile parturient who appears ill and possibly bacteremic.122
However, the dose and timing of such treatment is currently
unknown. In addition, there are no current guidelines surrounding
maximum temperature, white blood cell counts, or other clinical
signs, which one would clearly contraindicate neuraxial
anesthesia. In a woman with florid sepsis, cardiovascular,
respiratory, and hematologic concerns may preclude the use of
neuraxial anesthesia, but the risks and benefits of alternatives
must be assessed and weighed individually.
C. Aseptic technique
11 The ideal aseptic technique for regional anesthesia is
controversial (see Chapter 10). There have been questions about
the use of maximal aseptic precautions that are similar to those
used during central venous access placement. Handwashing;
jewelry removal; and use of sterile gloves (to supplement
handwashing and not as a substitute), fresh mask, and cap have
been shown to reduce the incidence of microbial contamination of
the work area but not directly to reduce nosocomial infection. The
ASRA consensus statement80 and ASA Task Force report122
acknowledge the importance of these precautions.
REFERENCES
1. Kuczkowski KM, Reisner LS. Anesthetic management of the parturient with fever and infection. J
Clin Anesth. 2003;15:478–488.
2. Macaulay JH, Bond K, Steer PJ. Epidural analgesia in labor and fetal hyperthermia. Obstet Gynecol.
1992;80:665–669.
3. Herbst A, Wølner-Hanssen P, Ingemarsson I. Risk factors for fever in labor. Obstet Gynecol.
1995;86:790–794.
4. Petrova A, Demissie K, Rhoads GG, et al. Association of maternal fever during labor with neonatal
and infant morbidity and mortality. Obstet Gynecol. 2001;98:20–27.
5. Lieberman E, Eichenwald E, Mathur G, et al. Intrapartum fever and unexplained seizures in term
infants. Pediatrics. 2000;106:983–988.
6. Lieberman E, Lang J, Richardson DK, et al. Intrapartum maternal fever and neonatal outcome.
Pediatrics. 2000;105:8–13.
7. Segal S. Labor epidural analgesia and maternal fever. Anesth Analg. 2010;111:1467–1475.
8. Lieberman E, Lang JM, Frigoletto F, et al. Epidural analgesia, intrapartum fever, and neonatal sepsis
evaluation. Pediatrics. 1997;99:415–419.
9. Philip J, Alexander JM, Sharma SK, et al. Epidural analgesia during labor and maternal fever.
Anesthesiology. 1999;90:1271–1275.
10. Kaul B, Vallejo M, Ramanathan S, et al. Epidural labor analgesia and neonatal sepsis evaluation rate: a
quality improvement study. Anesth Analg. 2001;93:986–990.
11. Impey L, Greenwood C, MacQuillan K, et al. Fever in labour and neonatal encephalopathy: a
prospective cohort study. BJOG. 2001;108:594–597.
12. Impey LW, Greenwood CE, Black RS, et al. The relationship between intrapartum maternal fever and
neonatal acidosis as risk factors for neonatal encephalopathy. Am J Obstet Gynecol. 2008;198:49.e41–
e46.
13. Wu YW, Escobar GJ, Grether JK, et al. Chorioamnionitis and cerebral palsy in term and near-term
infants. JAMA. 2003;290:2677–2684.
14. Wu YW, Colford JM Jr. Chorioamnionitis as a risk factor for cerebral palsy: a meta-analysis. JAMA.
2000;284:1417–1424.
15. Dammann O, Drescher J, Veelken N. Maternal fever at birth and non-verbal intelligence at age 9 years
in preterm infants. Dev Med Child Neurol. 2003;45:148–151.
16. Zerbo O, Iosif AM, Walker C, et al. Is maternal influenza or fever during pregnancy associated with
autism or developmental delays? Results from the CHARGE (CHildhood Autism Risks from Genetics
and Environment) study. J Autism Dev Disord. 2013;43:25–33.
17. Camann WR, Hortvet LA, Hughes N, et al. Maternal temperature regulation during extradural
analgesia for labour. Br J Anaesth. 1991;67:565–568.
18. Fusi L, Steer PJ, Maresh MJ, et al. Maternal pyrexia associated with the use of epidural analgesia in
labour. Lancet. 1989;1:1250–1252.
19. Goetzl L, Rivers J, Zighelboim I, et al. Intrapartum epidural analgesia and maternal temperature
regulation. Obstet Gynecol. 2007;109:687–690.
20. Mantha VR, Vallejo MC, Ramesh V, et al. Maternal and cord serum cytokine changes with continuous
and intermittent labor epidural analgesia: a randomized study. ScientificWorldJournal.
2012;2012:607938.
21. Mantha VR, Vallejo MC, Ramesh V, et al. The incidence of maternal fever during labor is less with
intermittent than with continuous epidural analgesia: a randomized controlled trial. Int J Obstet
Anesth. 2008;17:123–129.
22. Tian F, Wang K, Hu J, et al. Continuous spinal anesthesia with sufentanil in labor analgesia can induce
maternal febrile responses in puerperas. Int J Clin Exp Med. 2013;6:334–341.
23. Wong CA, Scavone BM, Peaceman AM, et al. The risk of cesarean delivery with neuraxial analgesia
given early versus late in labor. N Engl J Med. 2005;352:655–665.
24. Goetzl L, Evans T, Rivers J, et al. Elevated maternal and fetal serum interleukin-6 levels are associated
with epidural fever. Am J Obstet Gynecol. 2002;187:834–838.
25. Dashe JS, Rogers BB, McIntire DD, et al. Epidural analgesia and intrapartum fever: placental
findings. Obstet Gynecol. 1999;93:341–344.
26. Vallejo MC, Kaul B, Adler LJ, et al. Chorioamnionitis, not epidural analgesia, is associated with
maternal fever during labour. Can J Anaesth. 2001;48:1122–1126.
27. Riley LE, Celi AC, Onderdonk AB, et al. Association of epidural-related fever and noninfectious
inflammation in term labor. Obstet Gynecol. 2011;117:588–595.
28. Sharma SK, Rogers BB, Alexander JM, et al. A randomized trial of the effects of antibiotic
prophylaxis on epidural-related fever in labor. Anesth Analg. 2014;118:604–610.
29. Goetzl L, Zighelboim I, Badell M, et al. Maternal corticosteroids to prevent intrauterine exposure to
hyperthermia and inflammation: a randomized, double-blind, placebo-controlled trial. Am J Obstet
Gynecol. 2006;195:1031–1037.
30. Wang LZ, Hu XX, Liu X, et al. Influence of epidural dexamethasone on maternal temperature and
serum cytokine concentration after labor epidural analgesia. Int J Gynaecol Obstet. 2011;113:40–43.
31. Goetzl L, Rivers J, Evans T, et al. Prophylactic acetaminophen does not prevent epidural fever in
nulliparous women: a double-blind placebo-controlled trial. J Perinatol. 2004;24:471–475.
32. Knight M, Kurinczuk J. Introduction and methodology. In: Knight M, Kenyon S, Brocklehurst P, et al.,
eds. Saving Lives, Improving Mothers’ Care: Lessons Learned to Inform Future Maternity Care from
the UK and Ireland Confidential Enquiries into Maternal Deaths and Morbidity 2009-2012. Oxford,
United Kingdom: National Perinatal Epidemiology Unit, University of Oxford; 2014:1–8.
33. Newton ER. Chorioamnionitis and intraamniotic infection. Clin Obstet Gynecol. 1993;36:795–808.
34. Duff P, Gibbs RS, Sweet RL. Maternal and fetal infections. In: Creasy RK, Resnik R, Iams JD, et al.,
eds. Maternal-Fetal Medicine: Principles and Practice. 6th ed. Philadelphia, PA: Saunders-Elsevier;
2009:739–796.
35. Qureshi F, Jacques SM, Bendon RW, et al. Candida funisitis: a clinicopathologic study of 32 cases.
Pediatr Dev Pathol. 1998;1:118–124.
36. Gibbs RS, Castillo MS, Rodgers PJ. Management of acute chorioamnionitis. Am J Obstet Gynecol.
1980;136:709–713.
37. Yoder PR, Gibbs RS, Blanco JD, et al. A prospective, controlled study of maternal and perinatal
outcome after intra-amniotic infection at term. Am J Obstet Gynecol. 1983;145:695–701.
38. Roberts DJ, Celi AC, Riley LE, et al. Acute histologic chorioamnionitis at term: nearly always
noninfectious. PLoS One. 2012;7:e31819.
39. Goodman EJ, DeHorta E, Taguiam JM. Safety of spinal and epidural anesthesia in parturients with
chorioamnionitis. Reg Anesth. 1996;21:436–441.
40. Satin AJ, Maberry MC, Leveno KJ, et al. Chorioamnionitis: a harbinger of dystocia. Obstet Gynecol.
1992;79:913–915.
41. Kim MJ, Romero R, Gervasi MT, et al. Widespread microbial invasion of the chorioamniotic
membranes is a consequence and not a cause of intra-amniotic infection. Lab Invest. 2009;89:924–
936.
42. Shimoya K, Taniguchi T, Matsuzaki N, et al. Chorioamnionitis decreased incidence of respiratory
distress syndrome by elevating fetal interleukin-6 serum concentration. Hum Reprod. 2000;15:2234–
2240.
43. Van Marter LJ, Dammann O, Allred EN, et al. Chorioamnionitis, mechanical ventilation, and postnatal
sepsis as modulators of chronic lung disease in preterm infants. J Pediatr. 2002;140:171–176.
44. Wu YW. Systematic review of chorioamnionitis and cerebral palsy. Ment Retard Dev Disabil Res Rev.
2002;8:25–29.
45. Alexander JM, McIntire DM, Leveno KJ. Chorioamnionitis and the prognosis for term infants. Obstet
Gynecol. 1999;94:274–278.
46. Rouse DJ, Landon M, Leveno KJ, et al. The Maternal-Fetal Medicine Units cesarean registry:
chorioamnionitis at term and its duration-relationship to outcomes. Am J Obstet Gynecol.
2004;191:211–216.
47. Sarkar SS, Bhagat I, Bhatt-Mehta V, et al. Does maternal intrapartum antibiotic treatment prolong the
incubation time required for blood cultures to become positive for infants with early-onset sepsis? Am
J Perinatol. 2015;32:357–362.
48. Bader AM, Gilbertson L, Kirz L, et al. Regional anesthesia in women with chorioamnionitis. Reg
Anesth. 1992;17:84–86.
49. Brito V, Niederman MS. Pneumonia complicating pregnancy. Clin Chest Med. 2011;32:121–132.
50. Benedetti TJ, Valle R, Ledger WJ. Antepartum pneumonia in pregnancy. Am J Obstet Gynecol.
1982;144:413–417.
51. Fiore AE, Shay DK, Broder K, et al. Prevention and control of seasonal influenza with vaccines:
recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm
Rep. 2009;58(RR-8):1–52.
52. Haberg SE, Trogstad L, Gunnes N, et al. Risk of fetal death after pandemic influenza virus infection or
vaccination. N Engl J Med. 2013;368:333–340.
53. Hansen C, Desai S, Bredfeldt C, et al. A large, population-based study of 2009 pandemic influenza A
virus subtype H1N1 infection diagnosis during pregnancy and outcomes for mothers and neonates. J
Infect Dis. 2012;206:1260–1268.
54. Bacak SJ, Callaghan WM, Dietz PM, et al. Pregnancy-associated hospitalizations in the United States,
1999–2000. Am J Obstet Gynecol. 2005;192:592–597.
55. Hill JB, Sheffield JS, McIntire DD, et al. Acute pyelonephritis in pregnancy. Obstet Gynecol.
2005;105:18–23.
56. Smaill F, Vazquez JC. Antibiotics for asymptomatic bacteriuria in pregnancy. Cochrane Database Syst
Rev. 2007;(2):CD000490.
57. Chaim W, Bashiri A, Bar-David J, et al. Prevalence and clinical significance of postpartum
endometritis and wound infection. Infect Dis Obstet Gynecol. 2000;8:77–82.
58. Smaill FM, Grivell RM. Antibiotic prophylaxis versus no prophylaxis for preventing infection after
cesarean section. Cochrane Database Syst Rev. 2014;(10):CD007482.
59. Mackeen AD, Packard RE, Ota E, et al. Antibiotic regimens for postpartum endometritis. Cochrane
Database Syst Rev. 2015;(2):CD001067.
60. Shipp TD, Zelop C, Cohen A, et al. Post-cesarean delivery fever and uterine rupture in a subsequent
trial of labor. Obstet Gynecol. 2003;101:136–139.
61. Verani JR, McGee L, Schrag SJ; Division of Bacterial Diseases, National Center for Immunization and
Respiratory Diseases, Centers for Disease Control and Prevention. Prevention of perinatal group B
streptococcal disease—revised guidelines from CDC, 2010. MMWR Recomm Rep. 2010;59(RR-10):1–
36.
62. Hamilton ST, van Zuylen W, Shand A, et al. Prevention of congenital cytomegalovirus complications
by maternal and neonatal treatments: a systematic review. Rev Med Virol. 2014;24:420–433.
63. Rac MW, Sheffield JS. Prevention and management of viral hepatitis in pregnancy. Obstet Gynecol
Clin North Am. 2014;41:573–592.
64. Wax DB, Beilin Y, Frolich M. Liver disease. In: Chestnut DH, Wong CA, Tsen LC, et al., eds.
Chestnut’s Obstetric Anesthesia: Principles and Practice. 5th ed. Philadelphia, PA: Elsevier;
2014:1068–1080.
65. American College of Obstetricians and Gynecologists. ACOG Practice Bulletin No. 86: viral hepatitis
in pregnancy. Obstet Gynecol. 2007;110:941–956.
66. Centers for Disease Control and Prevention. HIV surveillance report, vol. 25.
http://www.cdc.gov/hiv/library/reports/surveillance/2013/surveillance_Report_vol_25.html. Accessed
March 10, 2015.
67. De Cock KM, Fowler MG, Mercier E, et al. Prevention of mother-to-child HIV transmission in
resource-poor countries: translating research into policy and practice. JAMA. 2000;283:1175–1182.
68. Centers for Disease Control and Prevention. HIV among pregnant women, infants, and children.
http://www.cdc.gov/hiv/risk/gender/pregnantwomen/facts/index.html. Accessed March 10, 2015.
69. Siegfried N, van der Merwe L, Brocklehurst P, et al. Antiretrovirals for reducing the risk of mother-to-
child transmission of HIV infection. Cochrane Database Syst Rev. 2011;(7):CD003510.
70. King JT Jr, Perkal MF, Rosenthal RA, et al. Thirty-day postoperative mortality among individuals with
HIV infection receiving antiretroviral therapy and procedure-matched, uninfected comparators. JAMA
Surg. 2015;150:343–351. doi: 10.1001/jamasurg.2014.2257.
71. Michaels J, Sharer LR, Epstein LG. Human immunodeficiency virus type 1 (HIV-1) infection of the
nervous system: a review. Immunodefic Rev. 1988;1:71–104.
72. Spudich S, Gonzalez-Scarano F. HIV-1-related central nervous system disease: current issues in
pathogenesis, diagnosis, and treatment. Cold Spring Harb Perspect Med. 2012;2:a007120.
73. Miller RF, Huang L, Walzer PD. Pneumocystis pneumonia associated with human immunodeficiency
virus. Clin Chest Med. 2013;34:229–241.
74. Miller RF, Allen E, Copas A, et al. Improved survival for HIV infected patients with severe
Pneumocystis jirovecii pneumonia is independent of highly active antiretroviral therapy. Thorax.
2006;61:716–721.
75. Al Anazi AR. Gastrointestinal opportunistic infections in human immunodeficiency virus disease.
Saudi J Gastroenterol. 2009;15:95–99.
76. Choi SY, Kim I, Kim NJ, et al. Hematological manifestations of human immunodeficiency virus
infection and the effect of highly active antiretroviral therapy on cytopenia. Korean J Hematol.
2011;46:253–257.
77. Cheitlin M. Cardiovascular complications of HIV infection. In: Sande M, Volberding P, eds. The
Medical Management of AIDS. 5th ed. Philadelphia, PA: WB Saunders; 1999:275–284.
78. Kibirige D, Ssekitoleko R. Endocrine and metabolic abnormalities among HIV-infected patients: a
current review. Int J STD AIDS. 2013;24:603–611.
79. Rosenberg AZ, Naicker S, Winkler CA, et al. HIV-associated nephropathies: epidemiology, pathology,
mechanisms and treatment. Nat Rev Nephrol. 2015;11:150–160.
80. Hebl JR. The importance and implications of aseptic techniques during regional anesthesia. Reg
Anesth Pain Med. 2006;31:311–323.
81. American Society of Anesthesiologists Committee on Occupational Health Task Force on Infection
Control. Recommendations for infection control for the practice of anesthesiology (third edition).
https://www.asahq.org/~/media/sites/asahq/files/public/resources/asa committees/recommendations-
for-infection-control-for-the-practice-of-anesthesiology-%281%29.pdf?la=en. Accessed March 10,
2015.
82. Tom DJ, Gulevich SJ, Shapiro HM, et al. Epidural blood patch in the HIV-positive patient. Review of
clinical experience. San Diego HIV Neurobehavioral Research Center. Anesthesiology. 1992;76:943–
947.
83. Evron S, Glezerman M, Harow E, et al. Human immunodeficiency virus: anesthetic and obstetric
considerations. Anesth Analg. 2004;98:503–511.
84. Panel on Treatment of HIV-Infected Pregnant Women and Prevention of Perinatal Transmission.
Recommendations for use of antiretroviral drugs in pregnant HIV-1-infected women for maternal
health and interventions to reduce perinatal HIV transmission in the United States.
http://aidsinfo.nih.gov/contentfiles/lvguidelines/PerinatalGL.pdf. Accessed March 10, 2015.
85. Roberts CM, Pfister JR, Spear SJ. Increasing proportion of herpes simplex virus type 1 as a cause of
genital herpes infection in college students. Sex Transm Dis. 2003;30:797–800.
86. Brown ZA, Vontver LA, Benedetti J, et al. Effects on infants of a first episode of genital herpes during
pregnancy. N Engl J Med. 1987;317:1246–1251.
87. Bader AM, Camann WR, Datta S. Anesthesia for cesarean delivery in patients with herpes simplex
virus type-2 infections. Reg Anesth. 1990;15:261–263.
88. Brown ZA, Wald A, Morrow RA, et al. Effect of serologic status and cesarean delivery on
transmission rates of herpes simplex virus from mother to infant. JAMA. 2003;289:203–209.
89. Hollier LM, Wendel GD. Third trimester antiviral prophylaxis for preventing maternal genital herpes
simplex virus (HSV) recurrences and neonatal infection. Cochrane Database Syst Rev. 2008;
(1):CD004946.
90. Davies PW, Vallejo MC, Shannon KT, et al. Oral herpes simplex reactivation after intrathecal
morphine: a prospective randomized trial in an obstetric population. Anesth Analg. 2005;100:1472–
1476.
91. Boyle RK. Herpes simplex labialis after epidural or parenteral morphine: a randomized prospective
trial in an Australian obstetric population. Anaesth Intensive Care. 1995;23:433–437.
92. Bauchat JR. Focused review: neuraxial morphine and oral herpes reactivation in the obstetric
population. Anesth Analg. 2010;111:1238–1241.
93. Shapiro ED. Clinical practice. Lyme disease. N Engl J Med. 2014;370:1724–1731.
94. Gellin BG, Broome CV, Bibb WF, et al. The epidemiology of listeriosis in the United States—1986.
Listeriosis Study Group. Am J Epidemiol. 1991;133:392–401.
95. Ramaswamy V, Cresence VM, Rejitha JS, et al. Listeria—review of epidemiology and pathogenesis. J
Microbiol Immunol Infect. 2007;40:4–13.
96. Allerberger F, Huhulescu S. Pregnancy related listeriosis: treatment and control. Expert Rev Anti Infect
Ther. 2015;13:395–403.
97. Crane J, Mundle W, Boucoiran I, et al. Parvovirus B19 infection in pregnancy. J Obstet Gynaecol Can.
2014;36:1107–1116.
98. Qin J, Yang T, Xiao S, et al. Reported estimates of adverse pregnancy outcomes among women with
and without syphilis: a systematic review and meta-analysis. PLoS One. 2014;9:e102203.
99. Hook EW III, Marra CM. Acquired syphilis in adults. N Engl J Med. 1992;326:1060–1069.
100. Nanda D, Feldman J, Delke I, et al. Syphilis among parturients at an inner city hospital: association
with cocaine use and implications for congenital syphilis rates. N Y State J Med. 1990;90:488–490.
101. Mathad JS, Gupta A. Tuberculosis in pregnant and postpartum women: epidemiology, management,
and research gaps. Clin Infect Dis. 2012;55:1532–1549.
102. Laibl VR, Sheffield JS. Tuberculosis in pregnancy. Clin Perinatol. 2005;32:739–747.
103. Loto OM, Awowole I. Tuberculosis in pregnancy: a review. J Preg. 2012;2012:379271.
104. Jana N, Vasishta K, Saha SC, et al. Obstetrical outcomes among women with extrapulmonary
tuberculosis. N Engl J Med. 1999;341:645–649.
105. Harger JH, Ernest JM, Thurnau GR, et al. Risk factors and outcome of varicella-zoster virus
pneumonia in pregnant women. J Infect Dis. 2002;185:422–427.
106. Brown NW, Parsons AP, Kam PC. Anaesthetic considerations in a parturient with varicella presenting
for caesarean section. Anaesthesia. 2003;58:1092–1095.
107. Cono J, Cragan JD, Jamieson DJ, et al. Prophylaxis and treatment of pregnant women for emerging
infections and bioterrorism emergencies. Emerg Infect Dis. 2006;12:1631–1637.
108. Maharaj D. Puerperal pyrexia: a review. Part I. Obstet Gynecol Surv. 2007;62:393–399.
109. Mabie WC, Barton JR, Sibai B. Septic shock in pregnancy. Obstet Gynecol. 1997;90:553–561.
110. Timezguid N, Das V, Hamdi A, et al. Maternal sepsis during pregnancy or the postpartum period
requiring intensive care admission. Int J Obstet Anesth. 2012;21:51–55.
111. Martin SR, Foley MR. Intensive care in obstetrics: an evidence-based review. Am J Obstet Gynecol.
2006;195:673–689.
112. Nduka OO, Parrillo JE. The pathophysiology of septic shock. Crit Care Clin. 2009;25:677–702.
113. American College of Obstetricians and Gynecologists. ACOG Technical Bulletin: septic shock.
Number 204. Int J Gynaecol Obstet. 1995;50:71–79.
114. Cantwell R, Clutton-Brock T, Cooper G, et al. Saving Mothers’ Lives: reviewing maternal deaths to
make motherhood safer: 2006-2008. The Eighth Report of the Confidential Enquiries into Maternal
Deaths in the United Kingdom. BJOG. 2011;118(suppl 1):1–203.
115. Dellinger RP, Levy MM, Rhodes A, et al. Surviving sepsis campaign: international guidelines for
management of severe sepsis and septic shock: 2012. Crit Care Med. 2013;41:580–637.
116. Lucas DN, Robinson PN, Nel MR. Sepsis in obstetrics and the role of the anaesthetist. Int J Obstet
Anesth. 2012;21:56–67.
117. Kohlschutter B, Baur H, Roth F. Suxamethonium-induced hyperkalaemia in patients with severe intra-
abdominal infections. Br J Anaesth. 1976;48:557–562.
118. Carp H, Bailey S. The association between meningitis and dural puncture in bacteremic rats.
Anesthesiology. 1992;76:739–742.
119. Cook TM, Counsell D, Wildsmith JA. Major complications of central neuraxial block: report on the
Third National Audit Project of the Royal College of Anaesthetists. Br J Anaesth. 2009;102:179–190.
120. D’Angelo R, Smiley RM, Riley ET, et al. Serious complications related to obstetric anesthesia: the
serious complication repository project of the Society for Obstetric Anesthesia and Perinatology.
Anesthesiology. 2014;120:1505–1512.
121. Schroeder TH, Krueger WA, Neeser E, et al. Spinal epidural abscess—a rare complication after
epidural analgesia for labour and delivery. Br J Anaesth. 2004;92:896–898.
122. American Society of Anesthesiologists Task Force on Infectious Complications Associated with
Neuraxial Techniques. Practice advisory for the prevention, diagnosis, and management of infectious
complications associated with neuraxial techniques: a report by the American Society of
Anesthesiologists Task Force on Infectious Complications Associated with Neuraxial Techniques.
Anesthesiology. 2010;112:530–545.
Non-neuraxial Analgesic Techniques
Wint Mon, Roshan Fernando, and Geraldine O’Sullivan
I. Nonpharmacologic methods of pain relief in labor
A. Antenatal health education
B. Support during labor (birth partners and doulas)
C. Relaxation techniques—music and breathing exercises
D. Complementary therapies
E. Hydrotherapy
F. Transcutaneous electrical nerve stimulation
G. Sterile water injection
II. Non-neuraxial pharmacologic methods of pain relief
A. Inhalation techniques
B. Non-opioid analgesia and sedatives
C. Opioid analgesia
KEYPOINTS
1. Most women choose pain relief methods based on their expectations of
labor pain.
2. Pregnant women should have access to results from the best available
research about the effectiveness and possible side effects of both
pharmacologic and nonpharmacologic pain relief measures.
3. A calm, soothing environment should be provided to improve a woman’s
laboring experience.
4. Lumbosacral massage has been shown to reduce back pain during labor.
5. Hypnobirthing has become a popular pain relief method in recent years,
although there is no current evidence to support its use as labor analgesia.
6. Acupuncture has been shown to reduce pain intensity during labor and
delivery.
7. When using inhalation analgesia, it is important that a woman remains
conscious to avoid regurgitation and aspiration of stomach contents.
8. Sedative anxiolytics are not recommended for use during labor because
they can readily cross the placenta and cause significant side effects for
both the mother and the neonate.
9. Systemic opioids can be given as nurse-controlled analgesia in the form
of intermittent intravenous or intramuscular boluses or as intravenous
patient-controlled analgesia (PCA).
10. Due to its fast onset and offset, remifentanil is suitable to treat the
cyclical pain of uterine contractions. However, continuous pulse oximetry
monitoring and one-to-one nursing care are essential when remifentanil is
in use.
MANY WOMEN EXPERIENCE SEVERE PAIN DURING LABOR. There are many forms
of pain relief available today, and these can be subdivided into neuraxial and
non-neuraxial analgesic techniques. Most women employ a number of ways to
cope with labor pain. Neuraxial analgesia techniques provide the most effective
pain relief and have the least systemic effects on the mother and the baby.
However, some women prefer to use less invasive techniques for pain relief.
Many women choose labor pain relief methods based on their expectations
of labor and labor pain.1 A systematic review by Hodnett2 specifically addressed
the issue of women’s views on the experience of childbirth in relation to
intrapartum analgesia. Four factors were found to be the most important for
women’s experiences of childbirth. These were (1) personal expectations, (2) the
amount of support from caregivers, (3) the quality of the caregiver–patient
relationship, and (4) the involvement in decision making. Therefore, it is
essential that maternity care providers have a discussion with women about their
expectation of birth and labor pain and provide them with evidence-based
information about all pain relief methods. In this chapter, we describe the
nonpharmacologic methods and the non-neuraxial pharmacologic methods of
pain relief in labor and current evidence surrounding their use.
I. Nonpharmacologic methods of pain relief in labor
A. Antenatal health education
1. Health education is an integral part of antenatal care. Delivery
of health education ranges from online resources to
classroom-style workshops designed to inform pregnant
women and their partners about labor, delivery, and care of
the newborn. A systematic survey of peer-reviewed studies
published between 1996 and 2006 found that women
preferred a small group learning environment where they
could talk to each other as well as the educator and relate
information to their individual circumstances.3
2. Antenatal classes cover aspects of labor pain, coping
strategies, and pain relief options available for pregnant
women. The first U.S. survey of women’s childbearing
experiences published in 2004 by the Maternity Center
Association suggested that women should have access to the
results of the best available research about the effectiveness
and possible side effects of both pharmacologic and
nonpharmacologic pain relief measures.4 In a randomized trial
carried out by Maimburg et al.,5 women who received health
education in the antenatal period were less likely to request
epidural analgesia in labor. However, they were still likely to
request some form of parenteral analgesia.
B. Support during labor (birth partners and doulas)
1. Most women choose to have a birthing partner during labor. A
birthing partner can be any close relative or friend. Some
women employ doulas for support during labor and delivery.
Doula originates from the ancient Greek word meaning
“female servant” and is a nonmedical person who supports a
woman during labor and after childbirth. Birth partners and
doulas can not only give emotional support by listening and
giving reassurance to laboring women but also provide
physical support such as massage and help with positioning.
2. A Cochrane review by Hodnett et al.6 assessed the effects of
continuous, one-to-one intrapartum support compared to usual
care. Twenty-two trials involving 15,288 women were
included in the analysis. Women allocated to continuous
support were more likely to have a spontaneous vaginal birth
and were less likely to have intrapartum analgesia or report
dissatisfaction. Moreover, their labors were shorter and they
were less likely to have a cesarean delivery (CD) or an
instrumental vaginal birth. Women who had continuous
support were also less likely to have neuraxial analgesia or a
baby with a low 5-minute Apgar score.
C. Relaxation techniques—music and breathing exercises
1. Music
An increase in the use of mobile devices allows women to
listen to their music of choice in delivery rooms. Hosseini et
al.7 investigated the effect of music therapy on labor pain and
progress in primigravid parturients. The study showed that
music caused a statistically significant decrease in pain
scores. Another randomized trial also demonstrated that music
therapy during labor decreased postpartum anxiety and pain
and increased the satisfaction with childbirth experience.8
2. Breathing exercises
Breathing exercises are widely discussed in antenatal classes.
Deep breathing, “belly” breathing, and patterned breathing are
the examples of popular breathing techniques used during
labor and delivery. One controlled trial of breathing and
relaxation techniques was included in a systematic review of
complementary and alternative therapies for pain management
in labor.9 Women were randomly assigned to an experimental
group, who received “respiratory autogenic training”
(progressive muscle relaxation and focused slow breathing),
or a control group, who received a traditional
“psychoprophylaxis.” There was no difference between the
groups in women’s experience of pain, instrumental delivery
rate, augmentation with oxytocin, and neonatal Apgar scores.
D. Complementary therapies
The use of complementary medicine has become popular with
many women, who are keen to avoid pharmacologic or invasive
methods of pain relief in labor. The Complementary Medicine
Field of the Cochrane Collaboration gives the definition of
complementary medicine as “practices and ideas which are
outside the domain of conventional medicine in several
countries.”10 The majority of the complementary therapies are
delivered by trained therapists, who may not be available in all
obstetric hospitals.
1. Massage and reflexology
a. Massage is commonly used to relax tense soft tissues and
calm the individual. In labor, a lumbosacral area massage
may be used for back pain and a light abdominal massage
may help during contraction. Many theories have been
proposed to explain the mechanisms by which massage
relieves pain in labor. One of them is the gate control
theory of pain (see Fig. 9.1).11 It has also been suggested
that massage decreases the release of cortisol and
norepinephrine levels12 and increases the serotonin
level.13 A systematic review by Smith et al.14 included six
studies evaluating the effects of massage on labor pain. A
reduction in pain score was reported in the massage
therapy group when compared to the group who received
usual care during the first stage of labor (standardized
mean difference [SMD] −0.82; 95% confidence interval
[CI], −1.17 to −0.47; four trials; 225 women).
2. Hypnosis
a. Hypnosis is the induction of a deeply relaxed state in
which a person loses the power of voluntary action and is
highly responsive to suggestions. Once in this state,
sometimes called a hypnotic trance, the patients are given
therapeutic suggestions to encourage changes in behavior
or relief of symptoms. “Hypnobirthing” has become a
popular trend in recent years. Hypnobirthing programs
teach women self-hypnosis, relaxation, and breathing
techniques to help manage the stress and pain associated
with labor. The anterior cingulate gyrus has been
demonstrated on positron emission tomography to be one
of the sites affected by the hypnotic modulation of pain.16
b. A Cochrane review of hypnosis included seven trials
involving a total of 1,213 women. Six of the seven trials
evaluated antenatal hypnotherapy. Only one trial provided
hypnotherapy during labor. There were no significant
differences between the hypnosis group and the control
group for the primary outcomes: use of pharmacologic
pain relief (average risk ratio [RR] 0.63; 95% CI, 0.39 to
1.01; six studies; 1,032 women), spontaneous vaginal
birth (average RR 1.35; 95% CI, 0.93 to 1.96; four
studies; 472 women), or satisfaction with pain relief (RR
1.06; 95% CI, 0.94 to 1.20; one study; 264 women).17 A
large randomized controlled, single-blinded trial (1,222
healthy nulliparous women), carried out in Denmark, also
did not find any differences in the use of epidural
analgesia or pain experience across three study groups: a
self-hypnosis group, a relaxation group, and a usual care
group.18
3. Aromatherapy
a. Aromatherapy is a complementary therapy using essential
oils obtained from distillation of plant materials such as
roots, leaves, bark, seeds, and flowers. Essential oils can
be used through massage and aromatherapy diffusers or
vaporizers. The two commonly used oils in labor are
lavender and frankincense. Essential oils are thought to
increase the body’s own sedative, stimulant, and relaxing
substances.
b. A systematic review performed by Smith et al.19 included
two trials (535 women) evaluating the use of
aromatherapy in labor. The trials found no difference
between the aromatherapy and control groups for the
primary outcomes of pain intensity, assisted vaginal
delivery, CD, and admission to neonatal intensive care
unit.
E. Hydrotherapy
1. Hydrotherapy, in the form of immersion in the birthing pool
or bath, is used worldwide to promote relaxation and to
reduce pain and anxiety in laboring women. For women
without any antenatal or intrapartum complications,
hydrotherapy is deemed to be safe.26 Immersion is associated
with decreases in neuroendocrine responses in healthy adults.
At a thermoneutral temperature of 34.5°C, during rest and
exercise, immersion has been associated with decreases in
levels of plasma norepinephrine27 and epinephrine.28 The
National Institute for Health and Care Excellence29 guideline,
published in 2007, recommended that the temperature of the
woman and the water should be monitored every hour to
ensure that the woman is comfortable and not febrile. The
temperature of the water should not be above 37.5°C.
2. Nutter et al.30 published an integrated analysis of peer-
reviewed literature on water birth in 2014. Thirty-eight
studies, including 2 randomized controlled trials and 36
observational studies, were reviewed. Their aggregate results
found that water birth was associated with high levels of
maternal satisfaction with pain relief. It was also linked to
decreased incidence of episiotomy and severe perineal tears.
Neonatal mortality rates were found to be low and similar in
women who used water immersion during labor and those
who did not use water immersion. Another review included
12 trials (3,243 women).31 Results for the first stage of labor
showed that there was a significant reduction in the epidural,
spinal, and paracervical analgesia use among the women
allocated to water immersion compared to the controls (478 of
1,254 vs. 529 of 1,245; RR 0.90; 95% CI, 0.82 to 0.99; six
trials). There were no differences in the rates of assisted
vaginal deliveries (RR 0.86; 95% CI, 0.71 to 1.05; seven
trials), cesarean deliveries (RR 1.21; 95% CI, 0.87 to 1.68;
eight trials), perineal trauma, or maternal infection. There
were also no differences for Apgar scores less than seven at 5
minutes (RR 1.58; 95% CI, 0.63 to 3.93; five trials), neonatal
unit admissions (RR 1.06; 95% CI, 0.71 to 1.57; three trials),
or neonatal infection rates (RR 2.00; 95% CI, 0.50 to 7.94;
five trials). However, there is considerable heterogeneity for
some outcomes in this review. Authors recommended further
research to be done.
F. Transcutaneous electrical nerve stimulation
1. Transcutaneous electrical nerve stimulation (TENS) is a
technique whereby low-voltage pulsatile electrical impulses,
varying in frequency and intensity, are administered through
electrodes. For pain in the first stage of labor, electrodes are
usually placed 2 cm over the T10–L1 dermatomes on either
side of the spinous processes. A second set of electrodes is
placed over the S2–S4 dermatomal areas for the pain relief in
the second stage. The mechanism of action of TENS is based
on the gate control theory of pain (see Fig. 9.1).11 It has also
been reported that there are marked increases in β-endorphin
and met-enkephalin with low-frequency TENS.32
2. A review of 14 studies involving 1,256 women found that
there was little difference in satisfaction with pain relief or in
pain ratings between TENS and control groups. On the other
hand, women receiving TENS to acupuncture points were less
likely to report severe pain (RR 0.41; 95% CI, 0.32 to 0.55).33
In a Cochrane review involving 25 studies, Dowswell et al.34
reported that there was little difference in pain ratings
between the TENS and control groups. The majority of
women, who used TENS, reported that they would use it
again in a future labor.33
b. Diamorphine
(1) Diamorphine is a synthetic diacetylated analogue of
morphine. It is a prodrug and is hydrolyzed by
esterases in plasma, brain, and liver to active
metabolites, such as 6-monoacetylmorphine. The
latter is responsible for most of diamorphine’s
analgesic activity. Diamorphine is twice as potent as
morphine. It acts on the μ-opioid receptors to exert its
analgesic effects. It is usually given intramuscularly
for labor pain. The dose ranges from 5 to 10 mg. The
onset of action is 5 to 10 minutes after the
intramuscular injection. It lasts for approximately 90
minutes. It is more lipid-soluble than morphine and
readily crosses the placenta. It may cause euphoria
and respiratory depression in mothers and neonatal
respiratory depression when administered in high
doses.
(2) A UK-wide survey carried out by Tuckey et al.54
reported that 84.4% of responding units used
meperidine and 34.1% diamorphine. Prophylactic
antiemetics were coadministered in 73.7% of units.
McInnes et al.55 compared the intramuscular injection
of diamorphine with patient-controlled intravenous
diamorphine. Women in the patient-controlled
diamorphine group used significantly less
diamorphine than those in the intramuscular
diamorphine group. However, the PCA group was
significantly more likely to report that they were very
dissatisfied with the use of diamorphine and were
significantly more likely to opt out of the trial before
the birth of the baby.
CLINICAL PEARL More studies need to be done to compare
the efficacy and side effect profile of diamorphine with those of
other systemic opioids.
c. Morphine
(1) Morphine is a μ-opioid receptor agonist. It is now
infrequently used for labor analgesia. It can be given
every 4 hours intravenously (0.05 to 0.1 mg per kg)
or intramuscularly (0.1 to 0.2 mg per kg). If given
intravenously, its onset of action is 2 to 3 minutes
with a peak effect at 10 to 20 minutes. If an
intramuscular route is used, the onset of action takes
20 to 40 minutes with a peak effect at 1 to 2 hours.
Morphine is metabolized in the liver to an inactive
metabolite, morphine-3-glucuronide, and the active
metabolite, morphine-6-glucuronide. Both
metabolites are excreted by the kidneys and have
elimination half-lives of up to 4.5 hours. Like many
other opioids, morphine readily crosses the placenta.
The elimination half-life in neonates is a lot longer
than in adults. Morphine has predictable side effects,
such as respiratory depression, pruritus, nausea, and
vomiting. Respiratory depression can also be present
in the neonates.
(2) A Cochrane review by Ullman et al.56 included two
trials, which compared intravenous morphine and
intravenous meperidine. One of the studies reported
that the morphine group was less satisfied with pain
relief than the meperidine group (RR 0.87; 95% CI,
0.78 to 0.98). A second trial also reported that the
women in the intravenous morphine group were more
likely to request a second dose of analgesia when
compared to those in the meperidine group.
CLINICAL PEARL There is currently no evidence to support
the use of morphine for labor analgesia.
d. Fentanyl
(1) Fentanyl is a phenylpiperidine derivative, which is
highly selective for the μ-opioid receptor. It is known
to be 100 times as potent as morphine and 800 times
as potent as meperidine. In labor, fentanyl is usually
given intravenously in the form of PCA when
neuraxial analgesia is contraindicated or unavailable.
Its onset of action is 2 to 4 minutes with a short
duration of action (30 to 60 minutes). In small doses,
fentanyl undergoes rapid redistribution. However, in
repeated doses, the drug may accumulate. Fentanyl is
metabolized by the liver into inactive metabolites,
which are excreted by the kidneys. Its context-
sensitive half-life increases with an increase in the
duration of infusion. It can readily cross the placenta
and has a potential to cause neonatal respiratory
depression.
(2) Miyakoshi et al.57 performed a retrospective study
evaluating the use of patient-controlled intravenous
fentanyl in labor. Women who received fentanyl had
significantly longer labors and an increased need of
oxytocin augmentation when compared with the
women who received no analgesia. Neonatal
outcomes (i.e., Apgar score <7 at 1 minute or 5
minutes and umbilical artery pH <7.20) were
comparable between two groups irrespective of the
mode of delivery. Of the women who expressed their
satisfaction, 72% (48 of 67) exhibited “excellent” or
“good” for pain relief with patient-controlled
fentanyl.
CLINICAL PEARL Current evidence suggests that fentanyl
provides good pain relief in labor. Because its context-sensitive
half-life increases with prolonged use, both the mother and the baby
must be appropriately monitored for a period of time after stopping
a prolonged intravenous infusion.
e. Remifentanil
(1) Remifentanil is an ultrashort-acting anilidopiperidine
derivative. It acts selectively on the μ-opioid
receptors. Its onset of action is approximately 1
minute. It is hydrolyzed rapidly by tissue and plasma
esterases into inactive metabolites and has a context-
sensitive half-life of 3 minutes irrespective of the
duration of infusion. Due to its fast onset and offset,
remifentanil is suitable to treat the cyclical pain of
uterine contractions. It is typically administered in the
form of PCA when epidural analgesia is
contraindicated or unavailable. Bolus doses range
from 20 to 40 μg with a lockout time of 2 to 3
minutes. An example of a remifentanil PCA protocol
is described in Figure 9.4. The rapid elimination of
remifentanil reduces the risk of respiratory
depression; however, there have been case reports of
respiratory arrest and cardiovascular arrest in
laboring women who received remifentanil PCA.58,59
It is of paramount importance to monitor these
women closely with “one-to-one” midwifery or
nursing care. Continuous oxygen saturation
monitoring must be applied and means to give
supplementary oxygen with full resuscitation
equipment must be available. Clinical studies
evaluating the analgesic effect and maternal and fetal
outcomes of remifentanil are described in the
following text and in Tables 9.2 and 9.3.53,60–79
(2) Remifentanil versus other systemic opioids. A
meta-analysis carried out by Schnabel et al.60
included 12 randomized controlled trials. Women
who received remifentanil PCA had a significantly
lower rate of conversion to epidural analgesia, a
lower mean pain score at 1 hour, and a higher
satisfaction score when compared to the women who
had meperidine. Serious maternal or fetal adverse
outcomes were not reported.
(3) Remifentanil patient-controlled analgesia versus
epidural analgesia. Liu et al.61 published a meta-
analysis of randomized controlled trials comparing
remifentanil PCA with epidural analgesia. Five
eligible studies were included in this meta-analysis.
The authors reported that women in the remifentanil
PCA group had higher VAS pain score when
compared to those in the epidural group at 1 hour and
2 hours. There was no statistical difference in the
incidence of nausea, vomiting, pruritus, and umbilical
artery pH values between the two groups. A
randomized controlled trial carried out by Stocki et
al.62 also reported that remifentanil PCA was inferior
to epidural analgesia for the provision of labor
analgesia. Mean respiratory rate was lower in the
remifentanil PCA group (18 ± 4 vs. 21 ± 4 breaths per
minute in the epidural group, P = .03). Mean SpO2
was also lower in the remifentanil PCA group (96.8%
± 1.4 vs. 98.4 ± 1.2 in the epidural group, P < .0001).
Moreover, nine apnea events occurred in five women
receiving remifentanil. Apgar scores and neonatal
respiratory outcomes were similar in both groups.62
This trial reinforces the safety concerns surrounding
the use of remifentanil PCA. Obstetric nurses should
be appropriately trained to recognize apnea and other
respiratory complications associated with
remifentanil use.
f. Alfentanil
(1) Alfentanil is a synthetic phenylpiperidine derivative,
which is structurally similar to fentanyl. It has 10% to
20% of fentanyl’s potency. It is highly bound to
protein and is less lipophilic than fentanyl. The onset
of action is 1 minute, with a shorter duration than
fentanyl. Alfentanil is metabolized by the liver and
excreted in the urine. Context-sensitive half-life after
4 hours of infusion is 60 minutes. Bolus dose of 10
μg per kg is usually used. It is not frequently used for
labor analgesia.
(2) When alfentanil PCA was compared with fentanyl
PCA, women in the fentanyl group reported lower
pain scores on average, although the difference was
not statistically significant. There were no
statistically significant differences for any secondary
outcomes such as nausea, CD rate, and naloxone
administration.80
g. Sufentanil
(1) Sufentanil is also a synthetic phenylpiperidine
derivative. It is reported to be 6 to 10 times more
potent than fentanyl. It has a rapid onset and short
duration of action. It is mostly metabolized by the
liver, although a small proportion of drug is
metabolized in the small intestines. It is excreted in
the urine and feces. Its terminal half-life is
approximately 2.5 hours. Sufentanil is mainly used
for neuraxial analgesia and is infrequently used
parentally for labor pain. Bolus dose ranges from 1 to
3 μg per kg.
(2) Camann et al.81 compared the intrathecal, epidural,
and intravenous sufentanil for labor analgesia.
Twenty-four women were given sufentanil 10 μg
either intrathecally, epidurally, or intravenously
during active labor. The median duration of analgesia
(median, IQR) was 84 (70 to 92) minutes in the
intrathecal group, 30 (23 to 32) minutes in the
epidural group, and 34 (17 to 30) minutes in the
intravenous route (P < .0001). The intrathecal group
also showed rapid and significant decrease in VAS
scores. The authors concluded that epidural and
intravenous administration of 10 μg of sufentanil did
not provide satisfactory analgesia.81
h. Codeine phosphate
(1) Codeine phosphate is a naturally occurring opioid. It
is usually given orally or intramuscularly. Dose
ranges from 30 to 60 mg given every 6 hours. It
remains a popular analgesic drug during peripartum
period, especially in early labor and postpartum
period. The incidence of exposure to codeine during
pregnancy is 5% to 6%.82 Although it is a weaker
opioid than morphine, it can still predispose mothers
and babies to opioid-related side effects, such as
drowsiness and respiratory depression.
(2) Nezvalová-Henriksen et al.83 compared the
pregnancy outcomes of 2,666 women who used
codeine in pregnancy with the outcomes of 65,316
women who used no opioids in pregnancy. Codeine
use anytime during pregnancy was found to be
associated with an increased planned CD rate
(adjusted odds ratio [OR] 1.4; 95% CI, 1.2 to 1.7; P <
.0001). Third-trimester use was associated with an
increase in emergency CD rate (adjusted OR 1.5;
95% CI, 1.3 to 1.8; P < .0001) and postpartum
hemorrhage (adjusted OR 1.3; 95% CI, 1.1 to 1.5; P
< .0001). No significant associations with other
adverse pregnancy outcomes were found.83 Because
more effective analgesic drugs are available, routine
use of codeine is not recommended for active labor.
(3) The U.S. Food and Drug Administration (FDA) has
issued a safety alert regarding codeine use in nursing
mothers and in young children. Respiratory
depression and death related to ultrarapid metabolism
of codeine to morphine have been reported in
children who received codeine in the postoperative
period following tonsillectomy and/or adenoidectomy
and had evidence of being ultrarapid metabolizers of
codeine. Moreover, deaths have also occurred in
nursing infants, who were exposed to high levels of
morphine in breast milk, because their mothers were
ultrarapid metabolizers of codeine. Some individuals
are ultrarapid metabolizers due to a specific CYP2D6
genotype. The prevalence of this CYP2D6 phenotype
varies widely among different ethnic groups. It has
been estimated at 0.5% to 1% in Chinese and
Japanese; 0.5% to 1% in Hispanics; 1% to 10% in
Caucasians; 3% in African Americans; and 16% to
28% in North Africans, Ethiopians, and Arabs.
Ultrarapid metabolizers convert codeine into its
active metabolite, morphine, more rapidly and
completely than other people. This rapid conversion
results in higher than expected serum morphine levels
predisposing them to life-threatening or fatal
respiratory depression or experience signs of
overdose. The FDA, therefore, advises that health
care providers should choose the lowest effective
dose for the shortest period of time and inform
patients and caregivers about these risks and the signs
of morphine overdose when prescribing codeine-
containing drugs.84
i. Tramadol
(1) Tramadol is a synthetic analogue of codeine. It is
weak agonist at all opioid receptors with 20-fold
preference for μ-opioid receptors. It is also known to
inhibit neuronal reuptake of norepinephrine, and it
potentiates the release of serotonin. It is given orally,
intramuscularly, or intravenously in labor. Dose
ranges from 50 to 100 mg every 6 hours. It is
metabolized in the liver by demethylation. Its
elimination half-life is 4 to 6 hours. In equianalgesic
doses to morphine, tramadol causes less respiratory
and cardiovascular depression than morphine.
Tramadol is a proconvulsant and should be avoided
in patients with a history of epilepsy.
(2) Shetty et al.85 compared the efficacy of intramuscular
tramadol and pentazocine in the first stage of labor.
Sixty-five patients were divided into the pentazocine
group (30 mg pentazocine) and the tramadol group (1
mg per kg tramadol). There was no significant
difference in the analgesic effect between two groups.
Neither of the analgesics was effective toward the
end of the first stage. However, in the tramadol
group, the majority of women (55%) rated pain as
severe, whereas in the pentazocine group, the
majority of women (60%) rated pain as moderately
severe. There were not many side effects with either
of the drugs in the given dosage.85 Another
randomized controlled trial compared the efficacy of
intramuscular tramadol (100 mg) and meperidine (50
mg) in active labor. The duration of labor was shorter
in the tramadol group, for the first stage (190 vs. 140
minutes, P < .0001) and for the second stage (33 vs.
25 minutes, P = .001). There were no differences in
VAS scores for pain at 10 minutes and 1 hour
between two groups. Women in the meperidine group
had lower VAS pain scores than those in the tramadol
group in the second stage of labor (8 vs. 9; P = .009).
There was a significantly higher incidence of nausea
and vomiting (35% vs. 15%; P = .003) and
drowsiness (80% vs. 29%; P < .0001) in the
meperidine group.86
j. Oxycodone
(1) Oxycodone is a μ-opioid receptor agonist. It has
increasingly been used to treat acute pain and can be
given orally or intravenously. Oral bioavailability
ranges from 60% to 87%. Its onset of action is
between 15 and 30 minutes, and it has duration of
action of 4 to 6 hours, although the analgesic effect of
controlled release formulations can last up to 12
hours. Most of the drug is metabolized in the liver,
whereas the rest is excreted by the kidneys. The
plasma half-life is 3 to 5 hours.
(2) Kokki et al.87 carried out an open-labeled study to
evaluate the maternal pharmacokinetic properties of
intravenous oxycodone in the first stage of labor.
Maternal plasma oxycodone concentration was found
to have a median half-life of 2.6 hours (ranges from
1.8 to 2.8). Oxycodone concentrations in the
umbilical plasma were similar to levels in maternal
plasma at the time of birth. The authors concluded
that maternal elimination half-life of intravenous
oxycodone was significantly shorter than the
elimination half-life reported in nonpregnant women
and that maternal plasma oxycodone correlated well
with neonatal umbilical concentrations at the time of
birth.87 More studies need to be done to evaluate the
efficacy of oxycodone to treat labor pain.
SUMMARY
It is essential to provide pain relief to laboring women in a timely manner. We
have described common nonpharmacologic and non-neuraxial pharmacologic
analgesic techniques and have cited the latest evidence supporting their use.
Neuraxial analgesia provides the most effective form of pain relief to laboring
women. The benefits and risks of non-neuraxial techniques should be discussed
with the parturient in whom either neuraxial analgesia is contraindicated or in
those who prefer a non-neuraxial technique.
REFERENCES
1. Lally JE, Murtagh MJ, Macphail S, et al. More in hope than expectation: a systematic review of
women’s expectations and experience of pain relief in labour. BMC Med. 2008;6:7.
2. Hodnett ED. Pain and women’s satisfaction with the experience of childbirth: a systematic review. Am
J Obstet Gynecol. 2002;186:S160–S172.
3. Nolan ML. Information giving and education in pregnancy: a review of qualitative studies. J Perinat
Educ. 2009;18:21–30.
4. Corry MP. Recommendations from Listening to Mothers: the first national U.S. survey of women’s
childbearing experiences. Birth. 2004;31:61–65.
5. Maimburg RD, Vaeth M, Dürr J, et al. Randomised trial of structured antenatal training sessions to
improve the birth process. BJOG. 2010;117:921–928.
6. Hodnett ED, Gates S, Hofmeyr GJ, et al. Continuous support for women during childbirth. Cochrane
Database Syst Rev. 2013;(7):CD003766.
7. Hosseini SE, Bagheri M, Honarparvaran N. Investigating the effect of music on labor pain and
progress in the active stage of first labor. Eur Rev Med Pharmacol Sci. 2013;17:1479–1487.
8. Simavli S, Kaygusuz I, Gumus I, et al. Effect of music therapy during vaginal delivery on postpartum
pain relief and mental health. J Affect Disord. 2014;156:194–199.
9. Smith CA, Collins CT, Cyna AM, et al. Complementary and alternative therapies for pain
management in labour. Cochrane Database Syst Rev. 2006;(4):CD003521.
10. Berman BM. Cochrane complementary medicine field. About the Cochrane Collaboration (Fields).
2006;(1):CE000052.
11. Melzack R, Wall PD. Pain mechanisms: a new theory. Science. 1965;150:971–979.
12. Field T. Pregnancy and labor massage. Expert Rev Obstet Gynecol. 2010;5:177–181.
13. Field T. Massage therapy effects. Am Psychol. 1998;53:1270–1281.
14. Smith CA, Levett KM, Collins CT, et al. Massage, reflexology and other manual methods for pain
management in labour. Cochrane Database Syst Rev. 2012;(2):CD009290.
15. Dolatian M, Hasanpour A, Montazeri S, et al. The effect of reflexology on pain intensity and duration
of labor on primiparas. Iran Red Crescent Med J. 2011;13:475–479.
16. Faymonville ME, Laureys S, Degueldre C, et al. Neural mechanisms of antinociceptive effects of
hypnosis. Anesthesiology. 2000;92:1257–1267.
17. Madden K, Middleton P, Cyna AM, et al. Hypnosis for pain management during labour and childbirth.
Cochrane Database Syst Rev. 2012;(11):CD009356.
18. Werner A, Uldbjerg N, Zachariae R, et al. Self-hypnosis for coping with labour pain: a randomised
controlled trial. BJOG. 2013;120:346–353.
19. Smith CA, Collins CT, Crowther CA. Aromatherapy for pain management in labour. Cochrane
Database Syst Rev. 2011;(7):CD009215.
20. Stux G, Pomeranz B. Basics of Acupuncture. Berlin, Germany: Springer Verlag; 1995.
21. Pomeranz B, Stux G. Scientific Bases of Acupuncture. Berlin, Germany: Springer Verlag; 1989.
22. Ng LKY, Katims JJ, Lee MHM. Acupuncture: a neuromodulation technique for pain control. In:
Arnoff GM, ed. Evaluation and Treatment of Chronic Pain. 2nd ed. Baltimore, MD: Williams &
Wilkins; 1992:291–298.
23. Carr D, Lythgoe J. Use of acupuncture during labour. Pract Midwife. 2014;17:10, 12–15.
24. Chen Y, Zhang X, Fang Y, et al. Analyzing the study of using acupuncture in delivery in the past ten
years in China. Evid Based Complement Alternat Med. 2014;2014:672508.
25. Smith CA, Collins CT, Crowther CA, et al. Acupuncture or acupressure for pain management in
labour. Cochrane Database Syst Rev. 2011;(7):CD009232.
26. Benfield RD, Herman J, Katz VL, et al. Hydrotherapy in labor. Res Nurs Health. 2001;24:57–67.
27. Johansen LB, Pump B, Warberg J, et al. Preventing hemodilution abolishes natriuresis of water
immersion in humans. Am J Physiol. 1998;275:R879–R888.
28. Grossman E, Goldstein DS, Hoffman A, et al. Effects of water immersion on sympathoadrenal and
dopa-dopamine systems in humans. Am J Physiol. 1992;262:R993–R999.
29. National Institute for Health and Care Excellence. Intrapartum Care: Care of Healthy Women and
Their Babies during Childbirth. London, United Kingdom: National Institute for Health and Care
Excellence; 2007.
30. Nutter E, Meyer S, Shaw-Battista J, et al. Waterbirth: an integrative analysis of peer-reviewed
literature. J Midwifery Womens Health. 2014;59:286–319.
31. Cluett ER, Burns E. Immersion in water in labour and birth. Cochrane Database Syst Rev. 2009;
(2):CD000111.
32. Clement-Jones V, McLoughlin L, Tomlin S, et al. Increased beta-endorphin but not met-enkephalin
levels in human cerebrospinal fluid after acupuncture for recurrent pain. Lancet. 1980;2:946–949.
33. Bedwell C, Dowswell T, Neilson JP, et al. The use of transcutaneous electrical nerve stimulation
(TENS) for pain relief in labour: a review of the evidence. Midwifery. 2011;27:e141–e148.
34. Dowswell T, Bedwell C, Lavender T, et al. Transcutaneous electrical nerve stimulation (TENS) for
pain relief in labour. Cochrane Database Syst Rev. 2009;(2):CD007214.
35. Derry S, Straube S, Moore RA, et al. Intracutaneous or subcutaneous sterile water injection compared
with blinded controls for pain management in labour. Cochrane Database Syst Rev. 2012;
(1):CD009107.
36. King TL, Wong CA. Nitrous oxide for labor pain: is it a laughing matter? Anesth Analg. 2014;118:12–
14.
37. Maze MJ, Fujinaga M. Recent advances in understanding the actions and toxicity of nitrous oxide.
Anaesthesia. 2000;55:311–314.
38. Likis FE, Andrews JC, Collins MR, et al. Nitrous oxide for the management of labor pain: a
systematic review. Anesth Analg. 2014;118:153–167.
39. Klomp T, van Poppel M, Jones L, et al. Inhaled analgesia for pain management in labour. Cochrane
Database Syst Rev. 2012;(9):CD009351.
40. Yeo ST, Holdcroft A, Yentis SM, et al. Analgesia with sevoflurane during labour: I. Determination of
the optimum concentration. Br J Anaesth. 2007;98:105–109.
41. Yeo ST, Holdcroft A, Yentis SM, et al. Analgesia with sevoflurane during labour: II. Sevoflurane
compared with Entonox for labour analgesia. Br J Anaesth. 2007;98:110–115.
42. McGuinness C, Rosen M. Enflurane as an analgesic in labour. Anaesthesia. 1984;39:24–26.
43. Ross JA, Tunstall ME. Simulated use of premixed 0.25% isoflurane in 50% nitrous oxide and 50%
oxygen. Br J Anaesth. 2002;89:820–824.
44. Abboud TK, Swart F, Zhu J, et al. Desflurane analgesia for vaginal delivery. Acta Anaesthesiol Scand.
1995;39:259–261.
45. Abd-El-Maeboud KH, Elbohoty AE, Mohammed WE, et al. Intravenous infusion of paracetamol for
intrapartum analgesia. J Obstet Gynaecol Res. 2014;40:2152–2157.
46. Elbohoty AE, Abd-Elrazek H, Abd-El-Gawad M, et al. Intravenous infusion of paracetamol versus
intravenous pethidine as an intrapartum analgesic in the first stage of labor. Int J Gynaecol Obstet.
2012;118:7–10.
47. Joel S, Joselyn A, Cherian VT, et al. Low-dose ketamine infusion for labor analgesia: a double-blind,
randomized, placebo controlled clinical trial. Saudi J Anaesth. 2014;8:6–10.
48. McAuley DM, O’Neill MP, Moore J, et al. Lorazepam premedication for labour. Br J Obstet
Gynaecol. 1982;89:149–154.
49. Bucklin BA, Hawkins JL, Anderson JR, et al. Obstetric anesthesia workforce survey: twenty-year
update. Anesthesiology. 2005;103:645–653.
50. McIntosh DG, Rayburn WF. Patient-controlled analgesia in obstetrics and gynecology. Obstet
Gynecol. 1991;78:1129–1135.
51. Wee MY, Tuckey JP, Thomas PW, et al. A comparison of intramuscular diamorphine and
intramuscular pethidine for labour analgesia: a two-centre randomised blinded controlled trial. BJOG.
2014;121:447–456.
52. Leong WL, Sng BL, Sia AT. A comparison between remifentanil and meperidine for labor analgesia: a
systematic review. Anesth Analg. 2011;113:818–825.
53. Douma MR, Verwey RA, Kam-Endtz CE, et al. Obstetric analgesia: a comparison of patient-
controlled meperidine, remifentanil, and fentanyl in labour. Br J Anaesth. 2010;104:209–215.
54. Tuckey JP, Prout RE, Wee MY. Prescribing intramuscular opioids for labour analgesia in consultant-
led maternity units: a survey of UK practice. Int J Obstet Anesth. 2008;17:3–8.
55. McInnes RJ, Hillan E, Clark D, et al. Diamorphine for pain relief in labour: a randomised controlled
trial comparing intramuscular injection and patient-controlled analgesia. BJOG. 2004;111:1081–1089.
56. Ullman R, Smith LA, Burns E, et al. Parenteral opioids for maternal pain relief in labour. Cochrane
Database Syst Rev. 2010;(9):CD007396.
57. Miyakoshi K, Tanaka M, Morisaki H, et al. Perinatal outcomes: intravenous patient-controlled
fentanyl versus no analgesia in labor. J Obstet Gynaecol Res. 2013;39:783–789.
58. Bonner JC, McClymont W. Respiratory arrest in an obstetric patient using remifentanil patient-
controlled analgesia. Anaesthesia. 2012;67:538–540.
59. Marr R, Hyams J, Bythell V. Cardiac arrest in an obstetric patient using remifentanil patient-controlled
analgesia. Anaesthesia. 2013;68:283–287.
60. Schnabel A, Hahn N, Broscheit J, et al. Remifentanil for labour analgesia: a meta-analysis of
randomised controlled trials. Eur J Anaesthesiol. 2012;29:177–185.
61. Liu ZQ, Chen XB, Li HB, et al. A comparison of remifentanil parturient-controlled intravenous
analgesia with epidural analgesia: a meta-analysis of randomized controlled trials. Anesth Analg.
2014;118:598–603.
62. Stocki D, Matot I, Einav S, et al. A randomized controlled trial of the efficacy and respiratory effects
of patient-controlled intravenous remifentanil analgesia and patient-controlled epidural analgesia in
laboring women. Anesth Analg. 2014;118:589–597.
63. Volmanen P, Akural E, Raudaskoski T, et al. Comparison of remifentanil and nitrous oxide in labour
analgesia. Acta Anaesthesiol Scand. 2005;49:453–458.
64. Blair JM, Dobson GT, Hill DA, et al. Patient controlled analgesia for labour: a comparison of
remifentanil with pethidine. Anaesthesia. 2005;60:22–27.
65. Blair JM, Hill DA, Fee JP. Patient controlled analgesia for labour using remifentanil: a feasibility
study. Br J Anaesth. 2001;87:415–420.
66. Volmanen P, Akural EI, Raudaskoski T, et al. Remifentanil in obstetric analgesia: a dose-finding study.
Anesth Analg. 2002;94:913–917.
67. Thurlow JA, Laxton CH, Dick A, et al. Remifentanil by patient-controlled analgesia compared with
intramuscular meperidine for pain relief in labour. Br J Anaesth. 2002;88:374–378.
68. Volikas I, Butwick A, Wilkinson C, et al. Maternal and neonatal side-effects of remifentanil patient-
controlled analgesia in labour. Br J Anaesth. 2005;95:504–509.
69. Evron S, Glezerman M, Sadan O, et al. Remifentanil: a novel systemic analgesic for labor pain. Anesth
Analg. 2005;100:233–238.
70. Buehner U, Broadbent JR, Chesterfield B. Remifentanil patient-controlled analgesia for labour: a
complete audit cycle. Anaesth Intensive Care. 2011;39:666–670.
71. Jost A, Ban B, Kamenik M. Modified patient-controlled remifentanil bolus delivery regimen for
labour pain. Anaesthesia. 2013;68:245–252.
72. Balki M, Kasodekar S, Dhumne S, et al. Remifentanil patient-controlled analgesia for labour:
optimizing drug delivery regimens. Can J Anaesth. 2007;54:626–633.
73. Tveit TO, Halvorsen A, Seiler S, et al. Efficacy and side effects of intravenous remifentanil patient-
controlled analgesia used in a stepwise approach for labour: an observational study. Int J Obstet
Anaesth. 2013;22:19–25.
74. Ng TK, Cheng BC, Chan WS, et al. A double-blind randomised comparison of intravenous patient-
controlled remifentanil with intramuscular pethidine for labour analgesia. Anaesthesia. 2011;66:796–
801.
75. Volikas I, Male D. A comparison of pethidine and remifentanil patient-controlled analgesia in labour.
Int J Obstet Anesth. 2001;10:86–90.
76. Stourac P, Suchomelova H, Stodulkova M, et al. Comparison of parturient-controlled remifentanil
with epidural bupivacain and sufentanil for labour analgesia: randomised controlled trial. Biomed Pap
Med Fac Univ Palacky Olomouc Czech Repub. 2014;158:227–232.
77. Douma MR, Middledorp JM, Verwey RA, et al. A randomised comparison of intravenous remifentanil
patient-controlled analgesia with epidural ropivacaine/sufentanil during labour. Int J Obstet Anesth.
2011;20:118–123.
78. Volmanen P, Sarvela J, Akural EI, et al. Intravenous remifentanil vs. epidural levobupivacaine with
fentanyl for pain relief in early labour: a randomised, controlled, double-blinded study. Acta
Anaesthesiol Scand. 2008;52:249–255.
79. Marwah R, Hassan S, Carvalho JC, et al. Remifentanil versus fentanyl for intravenous patient-
controlled labour analgesia: an observational study. Can J Anaesth. 2012;59:246–254.
80. Morley-Forster PK, Reid DW, Vandeberghe H. A comparison of patient-controlled analgesia fentanyl
and alfentanil for labour analgesia. Can J Anaesth. 2000;47:113–119.
81. Camann WR, Denney RA, Holby ED, et al. A comparison of intrathecal, epidural, and intravenous
sufentanil for labor analgesia. Anesthesiology. 1992;77:884–887.
82. Palanisamy A, Bailey CR. Codeine in mothers and children: where are we now? Anaesthesia.
2014;69:655–660.
83. Nezvalová-Henriksen K, Spigset O, Nordeng H. Effects of codeine on pregnancy outcome: results
from a large population-based cohort study. Eur J Clin Pharmacol. 2011;67:1253–1261.
84. U.S. Food and Drug Administration. Codeine product labeling changes.
http://www.fda.gov/Safety/MedWatch/SafetyInformation/ucm356221.htm. Accessed July 25, 2015.
85. Shetty J, Vishalakshi A, Pandey D. Labour analgesia when epidural is not a choice: tramadol versus
pentazocine. ISRN Obstet Gynecol. 2014;2014:930349.
86. Khooshideh M, Shahriari A. A comparison of tramadol and pethidine analgesia on the duration of
labour: a randomised clinical trial. Aust N Z J Obstet Gynaecol. 2009;49:59–63.
87. Kokki M, Franco MG, Raatikainen K, et al. Intravenous oxycodone for pain relief in the first stage of
labour—maternal pharmacokinetics and neonatal exposure. Basic Clin Pharmacol Toxicol.
2012;111:182–188.
88. Nelson KE, Eisenach JC. Intravenous butorphanol, meperidine, and their combination relieve pain and
distress in women in labor. Anesthesiology. 2005;102:1008–1013.
89. Frank M, McAteer EJ, Cattermole R, et al. Nalbuphine for obstetric analgesia. A comparison of
nalbuphine with pethidine for pain relief in labour when administered by patient-controlled analgesia
(PCA). Anaesthesia. 1987;42:697–703.
Choice of Neuraxial Analgesia and Local Anesthetics
Dominique Moffitt and Arvind Palanisamy
I. Pain pathways during labor
A. Severity of labor pain
B. First stage of labor
C. Second stage of labor
D. Effective blockade in both stages of labor
II. Neuraxial anatomy in pregnancy
A. Reduction in intervertebral gap
B. Widening and rotation of the pelvis
C. Higher level of the apex of thoracic kyphosis
D. Engorgement of epidural veins
E. Difficult identification of the ligamentum flavum
III. Techniques for neuraxial analgesia during labor
A. Indications
B. Contraindications
C. Rationale for choice of technique
D. Preparation
E. Description of technique
F. Epidural test dose
G. Choice of local anesthetics
H. Maintenance of analgesia
I. Analgesia for vaginal delivery
J. Ambulation during labor
IV. Side effects and complications
A. Hypotension
B. Treatment for pruritus
C. Failed analgesia
D. Unintended dural puncture (“wet tap”)
E. Back pain
F. Excessive motor block, subdural, and high/total spinal block
G. Urinary retention
H. Maternal hyperthermia
I. Fetal heart rate abnormalities
J. Accidental IV injection
K. Meningitis
L. Epidural hematoma and abscess
M. Neurologic deficits
KEYPOINTS
1. Pain associated with labor is known to be the worst pain that a woman
can expect to experience in her lifetime.
2. Pain relief during labor and delivery is an essential part of good obstetric
care.
3. Anatomic and physiologic changes occur throughout pregnancy and add
to the complexity of administering neuraxial analgesia during labor.
4. Maternal request, in the absence of a medical contraindication, is
sufficient indication for initiation of labor analgesia.
5. Neuraxial analgesia is widely accepted as the most effective form of labor
analgesia.
6. Choice of neuraxial technique is at the discretion of the anesthesiologist
and is based on the patient’s medical history and clinical condition.
7. The goal of neuraxial analgesia is to provide effective and safe pain
control while improving patient satisfaction and without imposing a
negative effect on obstetric outcomes.
8. Labor analgesia is generally safe, and the risk of serious complications is
extremely low.
2. Obstetric considerations
a. When surgical intervention is very likely (e.g.,
nonreassuring fetal status), some anesthesiologists in the
past incorrectly favored the epidural technique for labor in
order to ensure the epidural catheter was functional in
order to avoid general anesthesia. A large retrospective
study confirmed that epidurals placed as part of a
CSE are more reliable than those placed alone.5 This
has been confirmed more recently by a large
randomized trial comparing CSE with traditional
epidural catheters.6
b. The choice of analgesic method in early labor is
controversial, but recent evidence favors the CSE
technique over systemic opioids or conventional epidural,
citing faster progress of labor as an advantage.7,8 The goal
of neuraxial analgesia is to provide effective and safe pain
control while improving patient satisfaction and without
imposing a negative effect on obstetric outcomes.
c. In active labor, CSE offers somewhat faster onset of
analgesia than a conventional epidural. A randomized
controlled trial in a private practice setting compared CSE
and traditional epidural analgesia in 800 term parturients.6
They found that patients receiving CSE had better pain
scores during the first stage of labor and required fewer
top-ups by the anesthesiologist, an important
consideration when anesthesia manpower is limited. In
contrast, a recent Cochrane Systematic Review concluded
that there is “little basis for offering CSE over epidurals in
labor, with no difference in overall maternal satisfaction
despite a slightly faster onset with CSE.”9 Collectively,
the effect may be relatively minimal when modern high-
volume, low-concentration epidural analgesia is used with
an overall advantage of the CSE of no more than 5 to 10
minutes.
d. In advanced labor, particularly in multiparous patients,
the CSE technique is often favored due to its rapid onset
of sacral analgesia.10
e. Ambulation may be facilitated by the CSE technique.
However, very low dose conventional epidurals have been
shown to confer similar motor strength and balance;
therefore, this advantage is minimal.
f. Preexisting fetal heart abnormalities may influence the
anesthesiologist to choose a conventional epidural
technique due to the association of intrathecal opioid
injections (and the CSE technique) with FHR
abnormalities (e.g., fetal bradycardia).11
The issue of FHR patterns before and after CSE and
standard epidural techniques has been addressed in two
recent publications.12,13
3. Anesthetic considerations
a. Anticipated difficult airway may encourage early
placement of an indwelling epidural catheter. Poorly
functioning catheters should be replaced early during
labor and not waiting until the emergency cesarean
delivery (CD) has been called.
b. Early (i.e., before onset of labor or maternal request)
insertion of a spinal or epidural catheter for obstetric (e.g.,
twin gestation or preeclampsia) or anesthetic indications
(e.g., anticipated difficult airway or obesity) may be
advantageous to reduce the need for general anesthesia if
an emergent procedure becomes necessary.14 In addition,
cases of impending coagulopathy and decreasing platelet
counts (e.g., hemolysis, elevated liver enzymes, low
platelets [HELLP] syndrome) may warrant early
placement.
c. The rate of postdural puncture headache (PDPH) with
CSE is low, approximately 1% or less with modern
pencil-point needles. The overall risk of dural puncture
with an epidural needle may be reduced by the CSE
technique.5
d. Deliberate continuous spinal techniques are rarely used
due to the increased risk of PDPH. However, in extreme
morbid obesity and selected other situations in which
slow-onset surgical anesthesia (e.g., aortic stenosis) is
desired, this technique may be appropriate. The inherent
dangers of having a woman labor with an indwelling
intrathecal catheter limit the use of this practice in many
hospitals. These dangers include mistakingly injecting a
dose of local anesthetic appropriate for an epidural into
the intrathecal space with the risk of high block leading to
respiratory arrest and cardiovascular collapse. These
catheters need to be well labeled and all care providers
informed that the catheter is intrathecal.
1. Epidural technique
a. The epidural tray. A commercial or self-prepared kit
should contain all materials needed for the block. Most
commercially available epidural kits contain a 17-G
epidural needle (either Tuohy or Weiss), a 19-G or 20-G
epidural catheter and ampules of saline and local
anesthetics (usually lidocaine 1% and lidocaine 1.5% with
epinephrine 1:200,000 for local infiltration and use as a
test dose, respectively), skin prep solutions (povidone-
iodine or chlorhexidine), sterile drape, gauze wipes, and
transparent dressing.
b. Sterile precautions. Hand washing, jewelry removal, and
the use of sterile gloves (to supplement hand washing, and
not as a substitute) are important sterile precautions, fresh
mask, and cap have been shown to reduce the incidence
of microbial contamination. Basic components of aseptic
technique are often breached. The American Society of
Regional Anesthesia (ASRA) consensus statement17 (see
Table 10.4) and ASA Taskforce in Obstetric Anesthesia14
acknowledge the importance of these procedures but
remain equivocal regarding the use of gowns. Alcohol-
based chlorhexidine solution has been convincingly
shown to provide superior germicidal properties
compared to povidone-iodine (Betadine) skin
preparation.17 Although it has been shown that
chlorhexidine solutions have faster and stronger
bactericidal effects than povidone-iodine, chlorhexidine is
not U.S. Food and Drug Administration (FDA)-approved
for use as a disinfectant solution before neuraxial
techniques due to controversy about neurotoxicity in
animal models; nevertheless, this agent is widely used for
skin preparation before neuraxial anesthesia.
d. Techniques
(1) Loss of resistance. This commonly employed
method to identify the lumbar epidural space is based
on the tactile sensation of a sudden “give way” when
the soft tissue resistance offered by the ligamentum
flavum is overcome when the needle tip enters the
epidural space. A specially designed plastic or glass
LOR syringe, customized to reduce frictional and
resistive forces between the syringe and the plunger,
is attached to the hub of the epidural needle.
(a) Air versus saline. Although the
anesthesiologist’s preference may assume
primary importance, increasing evidence points
to the superiority of saline-based techniques
(saline alone, saline with a bubble of air) over air
for location of the epidural space.21 The cited
advantages include better sensory appreciation,
decreased incidence of patchy blocks, and
elimination of the risks of epidural venous air
embolism and pneumocephalus headache
(should an accidental dural puncture occur).
Some practitioners use air alone for the CSE
technique so that any clear fluid coming back
through the spinal needle can only be
cerebrospinal fluid (CSF); and not a depot of
saline or local anesthetic in the epidural space).
If using air, it is important not to empty the
syringe into the epidural space after LOR.
(b) Intermittent versus continuous pressure.
Application of “intermittent” or “continuous”
pressure to the syringe plunger is an integral
component of the LOR technique (see Fig. 10.3).
Purported advantages of the continuous
technique include faster identification of the
epidural space and possible reduced incidence of
accidental dural puncture. Purported advantages
of the intermittent technique include more
control over needle advancement (two hands on
the needle) and also reduced risk of dural
puncture.
REFERENCES
1. Traynor AJ, Tran ZV, Aragon M, et al. Obstetric anesthesia workforce survey: 30-year update. Anesth
Analg. In press.
2. Melzack R. The myth of painless childbirth (the John J. Bonica lecture). Pain. 1984;19:321–337.
3. American College of Obstetricians and Gynecologists. ACOG Committee Opinion No. 295: pain
relief during labor. Obstet Gynecol. 2004;104:213.
4. American College of Obstetricians and Gynecologists. ACOG Committee Opinion No. 339: analgesia
and cesarean delivery rates. Obstet Gynecol. 2006;107:1487–1488.
5. Pan PH, Bogard TD, Owen MD. Incidence and characteristics of failures in obstetric neuraxial
analgesia and anesthesia: a retrospective analysis of 19,259 deliveries. Int J Obstet Anesth.
2004;13:227–233.
6. Gambling D, Berkowitz J, Farrell TR, et al. A randomized controlled comparison of epidural analgesia
and combined spinal-epidural analgesia in a private practice setting: pain scores during first and
second stages of labor and at delivery. Anesth Analg. 2013;116:636–643.
7. Wong CA, Scavone BM, Peaceman AM, et al. The risk of cesarean delivery with neuraxial analgesia
given early versus late in labor. N Engl J Med. 2005;352:655–665.
8. Tsen LC, Thue B, Datta S, et al. Is combined spinal-epidural analgesia associated with more rapid
cervical dilation in nulliparous patients when compared with conventional epidural analgesia?
Anesthesiology. 1999;91:920–925.
9. Simmons SW, Taghizadeh N, Dennis AT, et al. Combined spinal-epidural versus epidural analgesia in
labour. Cochrane Database Syst Rev. 2012;(10):CD003401.
10. Abouleish A, Abouleish E, Camann W. Combined spinal-epidural analgesia in advanced labour. Can J
Anaesth. 1994;41: 575–578.
11. Mardirosoff C, Dumont L, Boulvain M, et al. Fetal bradycardia due to intrathecal opioids for labour
analgesia: a systematic review. BJOG. 2002;109:274–281.
12. Patel NP, El-Wahab N, Fernando R, et al. Fetal effects of combined spinal-epidural vs epidural labour
analgesia: a prospective, randomised double-blind study. Anaesthesia. 2014;69:458–467.
13. Gambling DR, Bender M, Faron S, et al. Prophylactic intravenous ephedrine to minimize fetal
bradycardia after combined spinal-epidural labour analgesia: a randomized controlled study. Can J
Anaesth. 2015;62:1201–1208.
14. American Society of Anesthesiologists Task Force on Obstetric Anesthesia. Practice guidelines for
obstetric anesthesia: an updated report by the American Society of Anesthesiologists Task Force on
Obstetric Anesthesia. Anesthesiology. 2007;106:843–863.
15. Kodali BS, Chandrasekhar S, Bulich LN, et al. Airway changes during labor and delivery.
Anesthesiology. 2008;108:357–362.
16. Harney D, Moran CA, Whitty R, et al. Influence of posture on the incidence of vein cannulation
during epidural catheter placement. Eur J Anaesthesiol. 2005;22:103–106.
17. Hebl JR. The importance and implications of aseptic techniques during regional anesthesia. Reg
Anesth Pain Med. 2006;31:311–323.
18. Broadbent CR, Maxwell WB, Ferrie R, et al. Ability of anaesthetists to identify a marked lumbar
interspace. Anaesthesia. 2000;55:1122–1126.
19. Chakraverty R, Pynsent P, Isaacs K. Which spinal levels are identified by palpation of the iliac crests
and the posterior superior iliac spines? J Anat. 2007;210:232–236.
20. Sahota JS, Carvalho JC, Balki M, et al. Ultrasound estimates for midline epidural punctures in the
obese parturient: paramedian sagittal oblique is comparable to transverse median plane. Anesth Analg.
2013;116:829–835.
21. Shenouda PE, Cunningham BJ. Assessing the superiority of saline versus air for use in the epidural
loss of resistance technique: a literature review. Reg Anesth Pain Med. 2003;28:48–53.
22. Carabuena JM, Mitani AM, Liu X, et al. The learning curve associated with the epidural technique
using the Episure AutoDetect™ versus conventional glass syringe: an open-label, randomized,
controlled, crossover trial of experienced anesthesiologists in obstetric patients. Anesth Analg.
2013;116:145–154.
23. Hollway TE, Telford RJ. Observations on deliberate dural puncture with a Tuohy needle: depth
measurements. Anaesthesia. 1991;46:722–724.
24. Jaime F, Mandell GL, Vallejo MC, et al. Uniport soft-tip, open-ended catheters versus multiport firm-
tipped close-ended catheters for epidural labor analgesia: a quality assurance study. J Clin Anesth.
2000;12:89–93.
25. Segal S, Eappen S, Datta S. Superiority of multi-orifice over single-orifice epidural catheters for labor
analgesia and cesarean delivery. J Clin Anesth. 1997;9:109–112.
26. Beilin Y, Bernstein HH, Zucker-Pinchoff B. The optimal distance that a multiorifice epidural catheter
should be threaded into the epidural space. Anesth Analg. 1995;81:301–304.
27. Campbell DC, Camann WR, Datta S. The addition of bupivacaine to intrathecal sufentanil for labor
analgesia. Anesth Analg. 1995;81:305–309.
28. Hamilton CL, Riley ET, Cohen SE. Changes in the position of epidural catheters associated with
patient movement. Anesthesiology. 1997;86:778–784.
29. Arkoosh VA, Palmer CM, Yun EM, et al. A randomized, double-masked, multicenter comparison of
the safety of continuous intrathecal labor analgesia using a 28-gauge catheter versus continuous
epidural labor analgesia. Anesthesiology. 2008;108:286–298.
30. Cappiello E, O’Rourke N, Segal S, et al. A randomized trial of dural puncture epidural technique
compared with the standard epidural technique for labor analgesia. Anesth Analg. 2008;107:1646–
1651.
31. Thomas JA, Pan PH, Harris LC, et al. Dural puncture with a 27-gauge Whitacre needle as part of a
combined spinal-epidural technique does not improve labor epidural catheter function. Anesthesiology.
2005;103:1046–1051.
32. Davies JM, Posner KL, Lee LA, et al. Liability associated with obstetric anesthesia: a closed claims
analysis. Anesthesiology. 2009;110:131–139.
33. Guay J. The epidural test dose: a review. Anesth Analg. 2006;102:921–929.
34. Leighton BL, Norris MC, Sosis M, et al. Limitations of epinephrine as a marker of intravascular
injection in laboring women. Anesthesiology. 1987;66:688–691.
35. Breen TW, Shapiro T, Glass B, et al. Epidural anesthesia for labor in an ambulatory patient. Anesth
Analg. 1993;77:919–924.
36. Lyons G, Columb M, Hawthorne L, et al. Extradural pain relief in labour: bupivacaine sparing by
extradural fentanyl is dose dependent. Br J Anaesth. 1997;78:493–497.
37. Panni MK, Segal S. Local anesthetic requirements are greater in dystocia than in normal labor.
Anesthesiology. 2003;98:957–963.
38. Polley LS, Columb MO, Lyons G, et al. The effect of epidural fentanyl on the minimum local
analgesic concentration of epidural chloroprocaine in labor. Anesth Analg. 1996;83:987–990.
39. Polley LS, Columb MO, Naughton NN, et al. Relative analgesic potencies of ropivacaine and
bupivacaine for epidural analgesia in labor: implications for therapeutic indexes. Anesthesiology.
1999;90:944–950.
40. Roelants F, Lavand’homme PM, Mercier-Fuzier V. Epidural administration of neostigmine and
clonidine to induce labor analgesia: evaluation of efficacy and local anesthetic-sparing effect.
Anesthesiology. 2005;102:1205–1210.
41. Wong CA, Ratliff JT, Sullivan JT, et al. A randomized comparison of programmed intermittent
epidural bolus with continuous epidural infusion for labor analgesia. Anesth Analg. 2006;102:904–
909.
42. van der Vyver M, Halpern S, Joseph G. Patient-controlled epidural analgesia versus continuous
infusion for labour analgesia: a meta-analysis. Br J Anaesth. 2002;89:459–465.
43. Capogna G, Camorcia M, Stirparo S, et al. Programmed intermittent epidural bolus versus continuous
epidural infusion for labor analgesia: the effects on maternal motor function and labor outcome. A
randomized double-blind study in nulliparous women. Anesth Analg. 2011;113:826–831.
44. Lim Y, Sia AT, Ocampo CE. Comparison of computer integrated patient controlled epidural analgesia
vs. conventional patient controlled epidural analgesia for pain relief in labour. Anaesthesia.
2006;61:339–344.
45. Vallejo MC, Firestone LL, Mandell GL, et al. Effect of epidural analgesia with ambulation on labor
duration. Anesthesiology. 2001;95:857–861.
46. Cyna AM, Andrew M, Emmett RS, et al. Techniques for preventing hypotension during spinal
anaesthesia for caesarean section. Cochrane Database Syst Rev. 2006;(4):CD002251.
47. Iatrou CA, Dragoumanis CK, Vogiatzaki TD, et al. Prophylactic intravenous ondansetron and
dolasetron in intrathecal morphine-induced pruritus: a randomized, double-blinded, placebo-controlled
study. Anesth Analg. 2005;101:1516–1520.
48. Bauer ME, Kountanis JA, Tsen LC, et al. Risk factors for failed conversion of labor epidural analgesia
to cesarean delivery anesthesia: a systematic review and meta-analysis of observational trials. Int J
Obstet Anesth. 2012;21:294–309.
49. Beilin Y, Zahn J, Bernstein HH, et al. Treatment of incomplete analgesia after placement of an
epidural catheter and administration of local anesthetic for women in labor. Anesthesiology.
1998;88:1502–1506.
50. Russell IF. A prospective controlled study of continuous spinal analgesia versus repeat epidural
analgesia after accidental dural puncture in labour. Int J Obstet Anesth. 2012;21:7–16.
51. Howell CJ, Dean T, Lucking L, et al. Randomised study of long term outcome after epidural versus
non-epidural analgesia during labour. BMJ. 2002;325:357.
52. D’Angelo R, Smiley RM, Riley E, et al. Serious complications related to obstetric anesthesia: the
Serious Complication Repository project of the Society for Obstetric Anesthesia and Perinatology.
Anesthesiology. 2014;120:1505–1512.
53. Liang CC, Wong SY, Tsay PT, et al. The effect of epidural analgesia on postpartum urinary retention in
women who deliver vaginally. Int J Obstet Anesth. 2002;11:164–169.
54. Goetzl L, Zighelboim I, Badell M, et al. Maternal corticosteroids to prevent intrauterine exposure to
hyperthermia and inflammation: a randomized, double-blind, placebo-controlled trial. Am J Obstet
Gynecol. 2006;195:1031–1037.
55. Weinberg G. Lipid rescue resuscitation from local anaesthetic cardiac toxicity. Toxicol Rev.
2006;25:139–145.
56. Baer ET. Post-dural puncture bacterial meningitis. Anesthesiology. 2006;105:381–393.
57. Wong CA. Nerve injuries after neuraxial anesthesia and their medicolegal implications. Best Pract Res
Clin Obstet Gynaecol. 2010;24:367–381.
58. Ruppen W, Derry S, McQuay H, et al. Incidence of epidural hematoma, infection, and neurologic
injury in obstetric patients with epidural analgesia/anesthesia. Anesthesiology. 2006;105:394–399.
Ultrasound and Echocardiographic Techniques in
Obstetric Anesthesia
Laurie A. Chalifoux and John T. Sullivan
I. Introduction
II. Focused cardiac ultrasound
A. Introduction
B. Techniques
C. Clinical applications
D. FoCUS training, certification, and program maintenance
III. Pulmonary ultrasound
A. Introduction
B. Technique
C. Applications
IV. Ultrasound-guided neuraxial anesthesia
A. Introduction
B. Technique
C. Applications
V. Ultrasound-guided regional anesthesia (transversus abdominis
plane block)
A. Introduction
B. Technique
C. Application
VI. Intracranial pressure measurement (optic nerve sheath diameter)
A. Introduction
B. Technique
C. Applications
VII. Gastric volume measurement
A. Introduction
B. Technique
C. Applications
VIII. Pelvic/abdominal ultrasound
A. Introduction
B. Technique
C. Applications
IX. Airway examination
A. Introduction
B. Technique
C. Applications
X. Lower extremity vein ultrasound
A. Introduction
B. Technique
C. Applications
KEYPOINTS
1. Applications of point of care ultrasound are rapidly expanding in
obstetric management.
2. Focused cardiac ultrasound can be used as a powerful extension of the
physical examination to evaluate a variety of clinical conditions including
hypotension, dyspnea, and cardiac arrest.
3. Pulmonary ultrasound can serve as a rapid and more sensitive measure of
lung water and pneumothorax than radiography, which can be used to
guide management.
4. Ultrasound-guided neuraxial blockade may improve efficiency of block
placement and has applications for training.
5. Gastric ultrasound can be used to qualitatively assess gastric volume and
content, which may inform timing of decisions for nonelective obstetric
procedures.
I. Introduction
A. The application of ultrasound technology is rapidly changing the
practice of medicine in providing a safe, noninvasive tool to aid
in diagnosis, guide therapy, and assist with procedures.
B. Reduced costs and increased portability have led to increased
point of care applications of ultrasound.
C. Ultrasound has a long track record of safe use including in the
first trimester of pregnancy.1
D. The two principal skills required for point of care ultrasound are
image acquisition and interpretation. There is general agreement
that point of care ultrasound is valuable for qualitative assessment
for the purpose of rapidly narrowing differential diagnoses. There
is more debate about the appropriateness of quantitative
assessment in the domain of point of care ultrasound.
E. Ultrasound measurements have generally been demonstrated to be
accurate and reproducible.2 Point of care ultrasound is often
performed in less than ideal conditions (dynamic clinical
environment, patient movement, less training, and oversight) that
may compromise some quantitative validity.
F. With expansion of the use of ultrasound, there are challenges in
providing training to a broader group of users while ensuring
adequate quality of application.
G. Elements of a quality point of care ultrasound program include
using a structured approach to examination, documenting findings
in the medical record, storing images and videos, and maintaining
a quality assurance process.3
H. Most ultrasound machines have standard features to adjust gain,
scan depth, freeze images, and conduct basic measurements.
Additional available features include time-motion imaging (M-
mode), color Doppler, and cardiac calculations.
I. In selecting ultrasound probes, it is important to understand the
relationship between frequency and image granularity/depth of
penetration. Deeper structures (>6 cm depth, e.g., abdomen and
chest) require low frequency ultrasound (2 to 5 MHz); however,
some imaging quality is sacrificed. Superficial structures (<6 cm
depth, e.g., pleura, peripheral nerves) can be imaged with higher
frequency ultrasound (5 to 8 MHz), resulting in sharp imaging.4
Choose an ultrasound probe based on intended structures to be
imaged (see Table 11.1).
3. Contractility
a. Biventricular systolic function can be determined
qualitatively using any cardiac view, but the parasternal
views are the most accessible in the pregnant patient.
b. Using the PSAX view, contractility of the concentric left
ventricle can be qualitatively assessed rapidly.
c. The PLAX provides another option for assessing
contractility including opportunities for more quantitative
measurements using ejection fraction calculations.
4. FoCUS protocols
a. A systematic approach to the FoCUS examination has
been advocated to increase its accuracy and improve its
diagnostic capability.3
b. Several specific protocols and algorithms have been
developed for the peripartum patient, although no
individual protocol has been demonstrated to be
diagnostically superior or associated with improved
clinical outcomes.
c. Two commonly referenced protocols are the FoCUS
assessed transthoracic echocardiography (FATE)9 and
rapid obstetric screening echocardiography (ROSE).10
d. FATE was originally developed for critical care
application but has been adapted to a wide range of
clinical situations.9
e. The FATE examination includes the following steps: (1)
rule out obvious pathology, (2) assess wall thickness and
dimension of cardiac chamber, (3) assess contractility, (4)
assess pleura, and (5) relate these findings to the clinical
situation.9,11
f. Four basic views are utilized for FATE: SLAX, A4CH,
PSAX, and PLSX, with a basic pleural examination,
making FATE relatively simple and easy to learn.9,11
g. The ROSE scan was the first published FoCUS protocol
adapted to pregnant women.
h. The ROSE protocol incorporates qualitative and
quantitative evaluations using PSAX, PLAX, A4CH, and
A5CH views. The apical 5-chamber view (not shown in
Fig. 11.1) is a more anterior 4-chamber scan that includes
the left ventricular outflow tract.
i. The ROSE protocol also includes more advanced
quantitative elements of echocardiography such as cardiac
output, ventricular regional wall motion, valve
interrogation, and diastolic function that have been
deemed outside of the scope of basic FoCUS.3
C. Clinical applications
1. Hypotension
a. Unexplained hypotension is common in obstetric
anesthetic practice, and diagnosis can be difficult
considering the prevalence of both neuraxial anesthesia
and hemorrhage.
b. In other clinical domains, the use of FoCUS has been
shown to rapidly narrow broad differential diagnoses12
and reduce inappropriate therapy (e.g., fluid
administration in congestive heart failure [CHF])13 in
patients who are hypotensive.
c. One study of 184 patients presenting with nontraumatic,
undifferentiated hypotension were randomized to receive
early versus delayed “goal-directed cardiac ultrasound”
(i.e., FoCUS). Physicians who utilized FoCUS were able
to significantly narrow their differential diagnoses (see
Fig. 11.3) and were also more likely to choose the correct
final diagnosis as “most likely” on the differential list.12
2. Dyspnea
a. Symptoms of shortness of breath, tachypnea, oxygen
desaturation, or auscultative findings, alone or in
combination, are common in pregnancy or immediately
postpartum. FoCUS, preferably in combination with lung
ultrasound, may assist diagnosis and management.
b. FoCUS has been recommended as a first-line diagnostic
intervention for the pregnant patient with shortness of
breath or pulmonary edema.14
c. Differentiating pulmonary from cardiogenic pathology
may be of particular value. Qualitative, side-by-side
evaluation of left- and right-sided cardiac systolic
function and filling volumes in this setting is useful to
rule out some diagnoses related to volume overload or
cardiac failure or diminish the likelihood of others such as
embolism.
3. Chest pain
a. There may be a limited role for FoCUS in the setting of
chest pain in pregnancy. As an adjunct to physical
examination, FoCUS can provide a rapid screening tool to
identify some rare, life-threatening causes such as
myocardial ischemia or infarction, pleural or pericardial
effusions, right ventricular strain associated with
pulmonary embolism, or aortic dissection.15
b. Comprehensive echocardiography is an American Society
of Echocardiography class I recommendation to evaluate
chest pain when electrocardiograms (the diagnostic gold
standard) are inconclusive.16 However, assessment of
segmental wall motion abnormalities is an advanced
echocardiographic skill and probably falls outside the
scope of FoCUS.3
4. Cardiac arrest
a. Cardiac arrest in pregnancy occurs at a rate of
approximately 1 in 12,000 admissions,17 with the most
common etiologies being hemorrhage, heart failure,
embolism, and sepsis.
b. FoCUS offers potential diagnostic and therapeutic
advantages in managing some of these conditions.
c. There is also evidence suggesting that FoCUS may aid
resuscitation management post-arrest in nonpregnant
patients by detecting potentially reversible mechanical
causes of cardiac arrest (e.g., profound hypovolemia,
embolism, tension pneumothorax, or tamponade) as well
as confirming cardiac standstill.
d. The subcostal 4-chamber view can be initiated during
advanced cardiac life support (ACLS) chest
compressions, with higher quality images acquired during
brief pauses to confirm pulses.
e. The use of FoCUS during cardiac arrest changes
management, is more accurate than physical examination
or electrocardiography in determining mechanical cardiac
function, and may facilitate better outcomes; however, its
use has not yet been shown to actually improve
outcomes.3
REFERENCES
1. Barnett SB. Routine ultrasound scanning in first trimester: what are the risks? Semin Ultrasound CT
MR. 2002;23:387–391.
2. Belfort MA, Rokey R, Saade GR, et al. Rapid echocardiographic assessment of left and right heart
hemodynamics in critically ill obstetric patients. Am J Obstet Gynecol. 1994;171:884–892.
3. Via G, Hussain A, Wells M, et al. International evidence-based recommendations for focused cardiac
ultrasound. J Am Soc Echocardiogr. 2014;27:683.e1–e33.
4. Moore CL, Copel JA. Point-of-care ultrasonography. N Engl J Med. 2011;364:749–757.
5. Sandham JD, Hull RD, Brant RF, et al. A randomized, controlled trial of the use of pulmonary-artery
catheters in high-risk surgical patients. N Engl J Med. 2003;348:5–14.
6. Berenholtz SM, Lubomski LH, Weeks K, et al. Eliminating central line-associated bloodstream
infections: a national patient safety imperative. Infect Control Hosp Epidemiol. 2014;35:56–62.
7. Spencer KT, Kimura BJ, Korcarz CE, et al. Focused cardiac ultrasound: recommendations from the
American Society of Echocardiography. J Am Soc Echocardiogr. 2013;26:567–581.
8. Dennis A, Stenson A. The use of transthoracic echocardiography in postpartum hypotension. Anesth
Analg. 2012;115: 1033–1037.
9. Jensen MB, Sloth E, Larsen KM, et al. Transthoracic echocardiography for cardiopulmonary
monitoring in intensive care. Eur J Anaesthesiol. 2004;21:700–707.
10. Dennis AT. Transthoracic echocardiography in obstetric anaesthesia and obstetric critical illness. Int J
Obstet Anesth. 2011;20:160–168.
11. Holm JH, Frederiksen CA, Juhl-Olsen P, et al. Perioperative use of focus assessed transthoracic
echocardiography (FATE). Anesth Analg. 2012;115:1029–1032.
12. Jones AE, Tayal VS, Sullivan DM, et al. Randomized, controlled trial of immediate versus delayed
goal-directed ultrasound to identify the cause of nontraumatic hypotension in emergency department
patients. Crit Care Med. 2004;32: 1703–1708.
13. Ferrada P, Evans D, Wolfe L, et al. Findings of a randomized controlled trial using limited
transthoracic echocardiogram (LTTE) as a hemodynamic monitoring tool in the trauma bay. J Trauma
Acute Care Surg. 2014;76:31–37; discussion 37–38.
14. Cantwell R, Clutton-Brock T, Cooper G, et al. Saving Mothers’ Lives: reviewing maternal deaths to
make motherhood safer: 2006-2008. The Eighth Report of the Confidential Enquiries into Maternal
Deaths in the United Kingdom. BJOG. 2011;118(suppl 1):1–203.
15. Labovitz AJ, Noble VE, Bierig M, et al. Focused cardiac ultrasound in the emergent setting: a
consensus statement of the American Society of Echocardiography and American College of
Emergency Physicians. J Am Soc Echocardiogr. 2010;23:1225–1230.
16. Douglas PS, Khandheria B, Stainback RF, et al. ACCF/ASE/ACEP/ASNC/SCAI/SCCT/SCMR 2007
appropriateness criteria for transthoracic and transesophageal echocardiography: a report of the
American College of Cardiology Foundation Quality Strategic Directions Committee Appropriateness
Criteria Working Group, American Society of Echocardiography, American College of Emergency
Physicians, American Society of Nuclear Cardiology, Society for Cardiovascular Angiography and
Interventions, Society of Cardiovascular Computed Tomography, and the Society for Cardiovascular
Magnetic Resonance endorsed by the American College of Chest Physicians and the Society of
Critical Care Medicine. J Am Coll Cardiol. 2007;50:187–204.
17. Mhyre JM, Tsen LC, Einav S, et al. Cardiac arrest during hospitalization for delivery in the United
States, 1998-2011. Anesthesiology. 2014;120:810–818.
18. Dennis AT, Castro J, Carr C, et al. Haemodynamics in women with untreated pre-eclampsia.
Anaesthesia. 2012;67: 1105–1118.
19. World Health Organization Guidelines Review Committee. WHO Recommendations for Prevention
and Treatment of Pre-Eclampsia and Eclampsia. Geneva, Switzerland: World Health Organization;
2011.
20. Kuklina EV, Ayala C, Callaghan WM. Hypertensive disorders and severe obstetric morbidity in the
United States. Obstet Gynecol. 2009;113:1299–1306.
21. Berg CJ, Mackay AP, Qin C, et al. Overview of maternal morbidity during hospitalization for labor
and delivery in the United States: 1993-1997 and 2001-2005. Obstet Gynecol. 2009;113:1075–1081.
22. Campbell OM, Graham WJ, Lancet Maternal Survival Series steering group. Strategies for reducing
maternal mortality: getting on with what works. Lancet. 2006;368:1284–1299.
23. Druzin MS, Shields L, Peterson, N. Preeclampsia Toolkit: Improving health care response to
preeclampsia: a California toolkit to transform maternity care.
https://www.cmqcc.org/preeclampsia_toolkit. Accessed December 2, 2014.
24. Dennis AT, Solnordal CB. Acute pulmonary oedema in pregnant women. Anaesthesia. 2012;67:646–
659.
25. Zieleskiewicz L, Contargyris C, Brun C, et al. Lung ultrasound predicts interstitial syndrome and
hemodynamic profile in parturients with severe preeclampsia. Anesthesiology. 2014;120:906–914.
26. Zhang M, Liu ZH, Yang JX, et al. Rapid detection of pneumothorax by ultrasonography in patients
with multiple trauma. Crit Care. 2006;10:R112.
27. Bouhemad B, Zhang M, Lu Q, et al. Clinical review: bedside lung ultrasound in critical care practice.
Crit Care. 2007;11:205.
28. Martindale JL, Noble VE, Liteplo A. Diagnosing pulmonary edema: lung ultrasound versus chest
radiography. Eur J Emerg Med. 2013;20:356–360.
29. Lichtenstein DA, Mezière G, Lascols N, et al. Ultrasound diagnosis of occult pneumothorax. Crit Care
Med. 2005;33: 1231–1238.
30. Lichtenstein D, Mezière G, Biderman P, et al. The “lung point”: an ultrasound sign specific to
pneumothorax. Intensive Care Med. 2000;26:1434–1440.
31. Facchini C, Malfatto G, Giglio A, et al. Lung ultrasound and transthoracic impedance for noninvasive
evaluation of pulmonary congestion in heart failure [published online ahead of print January 7, 2015].
J Cardiovasc Med. doi:10.2459/JCM.0000000000000226.
32. Lichtenstein D, Goldstein I, Mourgeon E, et al. Comparative diagnostic performances of auscultation,
chest radiography, and lung ultrasonography in acute respiratory distress syndrome. Anesthesiology.
2004;100:9–15.
33. Liteplo AS, Marill KA, Villen T, et al. Emergency thoracic ultrasound in the differentiation of the
etiology of shortness of breath (ETUDES): sonographic B-lines and N-terminal pro-brain-type
natriuretic peptide in diagnosing congestive heart failure. Acad Emerg Med. 2009;16:201–210.
34. Sciscione AC, Ivester T, Largoza M, et al. Acute pulmonary edema in pregnancy. Obstet Gynecol.
2003;101:511–515.
35. Zieleskiewicz L, Lagier D, Contargyris C, et al. Lung ultrasound-guided management of acute
breathlessness during pregnancy. Anaesthesia. 2013;68:97–101.
36. Perlas A, Chaparro LE, Chin KJ. Lumbar neuraxial ultrasound for spinal and epidural anesthesia: a
systematic review and meta-analysis [published online ahead of print December 9, 2014]. Reg Anesth
Pain Med. doi:10.1097/AAP.0000000000000184.
37. Chin KJ, Perlas A, Chan V, et al. Ultrasound imaging facilitates spinal anesthesia in adults with
difficult surface anatomic landmarks. Anesthesiology. 2011;115:94–101.
38. Karmakar MK, Li X, Ho AM, et al. Real-time ultrasound-guided paramedian epidural access:
evaluation of a novel in-plane technique. Br J Anaesth. 2009;102:845–854.
39. Grau T, Leipold RW, Conradi R, et al. Ultrasound imaging facilitates localization of the epidural space
during combined spinal and epidural anesthesia. Reg Anesth Pain Med. 2001;26:64–67.
40. Chin KJ, Chan V. Ultrasonography as a preoperative assessment tool: predicting the feasibility of
central neuraxial blockade. Anesth Analg. 2010;110:252–253.
41. Vallejo MC, Phelps AL, Singh S, et al. Ultrasound decreases the failed labor epidural rate in resident
trainees. Int J Obstet Anesth. 2010;19:373–378.
42. Halpern SH, Banerjee A, Stocche R, et al. The use of ultrasound for lumbar spinous process
identification: a pilot study. Can J Anaesth. 2010;57:817–822.
43. Balki M, Lee Y, Halpern S, et al. Ultrasound imaging of the lumbar spine in the transverse plane: the
correlation between estimated and actual depth to the epidural space in obese parturients. Anesth
Analg. 2009;108:1876–1881.
44. Petersen PL, Mathiesen O, Torup H, et al. The transversus abdominis plane block: a valuable option
for postoperative analgesia? A topical review. Acta Anaesthesiol Scand. 2010;54:529–535.
45. McDonnell JG, Curley G, Carney J, et al. The analgesic efficacy of transversus abdominis plane block
after cesarean delivery: a randomized controlled trial. Anesth Analg. 2008;106:186–191.
46. Costello JF, Moore AR, Wieczorek PM, et al. The transversus abdominis plane block, when used as
part of a multimodal regimen inclusive of intrathecal morphine, does not improve analgesia after
cesarean delivery. Reg Anesth Pain Med. 2009;34:586–589.
47. Belavy D, Cowlishaw PJ, Howes M, et al. Ultrasound-guided transversus abdominis plane block for
analgesia after caesarean delivery. Br J Anaesth. 2009;103:726–730.
48. Griffiths JD, Barron FA, Grant S, et al. Plasma ropivacaine concentrations after ultrasound-guided
transversus abdominis plane block. Br J Anaesth. 2010;105:853–856.
49. Weiss E, Jolly C, Dumoulin JL, et al. Convulsions in 2 patients after bilateral ultrasound-guided
transversus abdominis plane blocks for cesarean analgesia. Reg Anesth Pain Med. 2014;39:248–251.
50. Kelly SMC, Malhotra RK. Ultrasound-guided transversus abdominis plane blocks for analgesia post
cesarean section. J Com Eff Res. 2011;1:35–38.
51. Hansen HC, Helmke K. Validation of the optic nerve sheath response to changing cerebrospinal fluid
pressure: ultrasound findings during intrathecal infusion tests. J Neurosurg. 1997;87:34–40.
52. Dubourg J, Javouhey E, Geeraerts T, et al. Ultrasonography of optic nerve sheath diameter for
detection of raised intracranial pressure: a systematic review and meta-analysis. Intensive Care Med.
2011;37:1059–1068.
53. Dubost C, Le Gouez A, Jouffroy V, et al. Optic nerve sheath diameter used as ultrasonographic
assessment of the incidence of raised intracranial pressure in preeclampsia: a pilot study.
Anesthesiology. 2012;116:1066–1071.
54. Frontera JA, Ahmed W. Neurocritical care complications of pregnancy and puerperum. J Crit Care.
2014;29:1069–1081.
55. American Society of Anesthesiologists Task Force on Obstetric Anesthesia. Practice guidelines for
obstetric anesthesia: an updated report by the American Society of Anesthesiologists Task Force on
Obstetric Anesthesia. Anesthesiology. 2007;106:843–863.
56. O’Sullivan G, Liu B, Hart D, et al. Effect of food intake during labour on obstetric outcome:
randomised controlled trial. BMJ. 2009;338:b784.
57. Cubillos J, Tse C, Chan VW, et al. Bedside ultrasound assessment of gastric content: an observational
study. Can J Anaesth. 2012;59:416–423.
58. Arzola C, Cubillos J, Perlas A, et al. Interrater reliability of qualitative ultrasound assessment of
gastric content in the third trimester of pregnancy. Br J Anaesth. 2014;113:1018–1023.
59. Scalea TM, Rodriguez A, Chiu WC, et al. Focused Assessment with Sonography for Trauma (FAST):
results from an international consensus conference. J Trauma. 1999;46:466–472.
60. Antonelli E, Morales MA, Dumps P, et al. Sonographic detection of fluid collections and postoperative
morbidity following cesarean section and hysterectomy. Ultrasound Obstet Gynecol. 2004;23:388–
392.
61. Ashar T, Ladner H. Trauma: the FAST approach: an introduction to bedside trauma ultrasound. Isr J
Emerg Med. 2006;6: 43–51.
62. Koskas M, Nizard J, Salomon LJ, et al. Abdominal and pelvic ultrasound findings within 24 hours
following uneventful cesarean section. Ultrasound Obstet Gynecol. 2008;32:520–526.
63. Lousquy R, Morel O, Soyer P, et al. Routine use of abdominopelvic ultrasonography in severe
postpartum hemorrhage: retrospective evaluation in 125 patients. Am J Obstet Gynecol.
2011;204:232.e1–e6.
64. Hui CM, Tsui BC. Sublingual ultrasound as an assessment method for predicting difficult intubation: a
pilot study. Anaesthesia. 2014;69:314–319.
65. Hsieh KS, Lee CL, Lin CC, et al. Secondary confirmation of endotracheal tube position by ultrasound
image. Crit Care Med. 2004;32(suppl 9):S374–S377.
66. Werner SL, Smith CE, Goldstein JR, et al. Pilot study to evaluate the accuracy of ultrasonography in
confirming endotracheal tube placement. Ann Emerg Med. 2007;49:75–80.
67. Sustić A, Kovac D, Zgaljardić Z, et al. Ultrasound-guided percutaneous dilatational tracheostomy: a
safe method to avoid cranial misplacement of the tracheostomy tube. Intensive Care Med.
2000;26:1379–1381.
68. Heit JA, Kobbervig CE, James AH, et al. Trends in the incidence of venous thromboembolism during
pregnancy or postpartum: a 30-year population-based study. Ann Intern Med. 2005;143:697–706.
69. Blaivas M. Ultrasound in the detection of venous thromboembolism. Crit Care Med. 2007;35(suppl
5):S224–S234.
Impact of Neuraxial Analgesia on Obstetric Outcomes
Christopher R. Cambic and Cynthia A. Wong
I.Introduction
II.Effects of neuraxial analgesia on the progress of labor
III. Duration of the first stage of labor
IV. Duration of the second stage of labor
V. Instrumental vaginal delivery
VI. Cesarean delivery
VII. Influence of oxytocin augmentation and ambulation on labor
outcomes
VIII. Impact of neuraxial analgesia on maternal fever rates
IX. Impact of neuraxial analgesia on breastfeeding success rates
X. Conclusion
KEYPOINTS
1. Neuraxial analgesia is the most effective form of labor analgesia.
Additionally, it provides physiologic and safety benefits to both mother
and fetus.
2. Effective neuraxial analgesia may increase the risk of instrumental
vaginal delivery and prolong the second stage of labor. The impact of
these outcomes on maternal and fetal well-being remains unclear.
3. Neuraxial analgesia does not increase the risk of cesarean delivery
compared with systemic opioid analgesia. Moreover, the initiation of
labor analgesia during the latent phase of labor does not increase the risk
of cesarean delivery or prolong the duration of labor.
4. Neuraxial labor analgesia is associated with maternal temperature
elevation in some women. The etiology is unclear and further research is
indicated.
5. The impact of neuraxial labor analgesia on breastfeeding success rates is
not clear and warrants further inquiry.
I. Introduction
A. Neuraxial analgesia is the most effective form of pain relief
during labor in current clinical practice.1
B. The use of neuraxial labor analgesia has increased since the
1970s.2
1. In the United States, 2008 data from 27 states demonstrated
that 61% of women who underwent a singleton vaginal
delivery received neuraxial analgesia.3
2. In the United Kingdom, approximately 34% of parturients
received neuraxial analgesia or anesthesia for delivery in 2012
to 2013.4
C. Factors influencing the rate of neuraxial analgesia include3,5:
1. Availability of anesthesia providers
2. Race/ethnicity
3. Personal and cultural expectations
4. Information and advice provided to pregnant women by other
health care providers
5. Obstetric complications
D. Neuraxial analgesia provides benefits to the mother, fetus/neonate,
and the parturient’s partner (see Table 12.1).6–13
CLINICAL PEARL Neuraxial analgesia provides superior pain
relief to the mother during labor and delivery and conveys other
significant benefits.
X. Conclusion
A. Neuraxial analgesia has the potential to positively and negatively
affect labor outcomes.
B. No single medication or technique exists that can universally
manage labor pain for all patients and circumstances. Anesthesia
providers need to assess each patient’s individual needs in order
to provide safe and effective labor analgesia.
REFERENCES
1. Jones L, Othman M, Dowswell T, et al. Pain management for women in labour: an overview of
systematic reviews. Cochrane Database Syst Rev. 2012;(3):CD009234.
2. Bucklin BA, Hawkins JL, Anderson JR, et al. Obstetric anesthesia workforce survey: twenty-year
update. Anesthesiology. 2005;103:645–653.
3. Osterman MJ, Martin JA. Epidural and spinal anesthesia use during labor: 27-state reporting area,
2008. Natl Vital Stat Rep. 2011;59:1–16.
4. Philip J, Alexander JM, Sharma SK, et al. Epidural analgesia during labor and maternal fever.
Anesthesiology. 1999;90:1271–1275.
5. Paech MJ. The King Edward Memorial Hospital 1,000 mother survey of methods of pain relief in
labour. Anaesth Intensive Care. 1991;19:393–399.
6. Capogna G, Camorcia M, Stirparo S. Expectant fathers’ experience during labor with or without
epidural analgesia. Int J Obstet Anesth. 2007;16:110–115.
7. Levinson G, Shnider SM, DeLorimier AA, et al. Effects of maternal hyperventilation on uterine blood
flow and fetal oxygenation and acid-base status. Anesthesiology. 1974;40:340–347.
8. Noble AD, Craft IL, Bootes JA, et al. Continuous lumbar epidural analgesia using bupivicaine: a study
of the fetus and newborn child. J Obstet Gynaecol Br Commonw. 1971;78:559–563.
9. Peabody JL. Transcutaneous oxygen measurement to evaluate drug effects. Clin Perinatol.
1979;6:109–121.
10. Shnider SM, Abboud TK, Artal R, et al. Maternal catecholamines decrease during labor after lumbar
epidural anesthesia. Am J Obstet Gynecol. 1983;147:13–15.
11. Smith CV, Rayburn WF, Allen KV, et al. Influence of intravenous fentanyl on fetal biophysical
parameters during labor. J Matern Fetal Med. 1996;5:89–92.
12. Wong CA, Scavone BM, Peaceman AM, et al. The risk of cesarean delivery with neuraxial analgesia
given early versus late in labor. N Engl J Med. 2005;352:655–665.
13. Yoo KY, Lee J, Kim HS, et al. The effects of opioids on isolated human pregnant uterine muscles.
Anesth Analg. 2001;92:1006–1009.
14. Hess PE, Pratt SD, Soni AK, et al. An association between severe labor pain and cesarean delivery.
Anesth Analg. 2000;90:881–886.
15. Alexander JM, Sharma SK, McIntire DD, et al. Intensity of labor pain and cesarean delivery. Anesth
Analg. 2001;92:1524–1528.
16. Ohel G, Gonen R, Vaida S, et al. Early versus late initiation of epidural analgesia in labor: does it
increase the risk of cesarean section? A randomized trial. Am J Obstet Gynecol. 2006;194:600–605.
17. Wang F, Shen X, Guo X, et al. Epidural analgesia in the latent phase of labor and the risk of cesarean
delivery: a five-year randomized controlled trial. Anesthesiology. 2009;111:871–880.
18. Anim-Somuah M, Smyth RM, Jones L. Epidural versus non-epidural or no analgesia in labour.
Cochrane Database Syst Rev. 2011;(12):CD000331.
19. Sharma SK, McIntire DD, Wiley J, et al. Labor analgesia and cesarean delivery: an individual patient
meta-analysis of nulliparous women. Anesthesiology. 2004;100:142–148.
20. Gambling D, Berkowitz J, Farrell TR, et al. A randomized controlled comparison of epidural analgesia
and combined spinal-epidural analgesia in a private practice setting: pain scores during first and
second stages of labor and at delivery. Anesth Analg. 2013;116:636–643.
21. Tsen LC, Thue B, Datta S, et al. Is combined spinal-epidural analgesia associated with more rapid
cervical dilation in nulliparous patients when compared with conventional epidural analgesia?
Anesthesiology. 1999;91:920–925.
22. Campbell DC, Zwack RM, Crone LA, et al. Ambulatory labor epidural analgesia: bupivacaine versus
ropivacaine. Anesth Analg. 2000;90:1384–1389.
23. van der Vyver M, Halpern S, Joseph G. Patient-controlled epidural analgesia versus continuous
infusion for labour analgesia: a meta-analysis. Br J Anaesth. 2002;89:459–465.
24. George RB, Allen TK, Habib AS. Intermittent epidural bolus compared with continuous epidural
infusions for labor analgesia: a systematic review and meta-analysis. Anesth Analg. 2013;116:133–
144.
25. Sultan P, Murphy C, Halpern S, et al. The effect of low concentrations versus high concentrations of
local anesthetics for labour analgesia on obstetric and anesthetic outcomes: a meta-analysis. Can J
Anaesth. 2013;60:840–854.
26. McGaughey HS Jr, Corey EL, Eastwood D, et al. Effect of synthetic anesthetics on the spontaneous
motility of human uterine muscle in vitro. Obstet Gynecol. 1962;19:233–240.
27. Fanning RA, Campion DP, Collins CB, et al. A comparison of the inhibitory effects of bupivacaine
and levobupivacaine on isolated human pregnant myometrium contractility. Anesth Analg.
2008;107:1303–1307.
28. Abrão KC, Francisco RP, Miyadahira S, et al. Elevation of uterine basal tone and fetal heart rate
abnormalities after labor analgesia: a randomized controlled trial. Obstet Gynecol. 2009;113:41–47.
29. Van de Velde M, Teunkens A, Hanssens M, et al. Intrathecal sufentanil and fetal heart rate
abnormalities: a double-blind, double placebo-controlled trial comparing two forms of combined
spinal epidural analgesia with epidural analgesia in labor. Anesth Analg. 2004;98:1153–1159.
30. Patel NP, El-Wahab N, Fernando R, et al. Fetal effects of combined spinal-epidural vs epidural labour
analgesia: a prospective, randomised double-blind study. Anaesthesia. 2014;69:458–467.
31. Abboud TK, David S, Nagappala S, et al. Maternal, fetal, and neonatal effects of lidocaine with and
without epinephrine for epidural anesthesia in obstetrics. Anesth Analg. 1984;63:973–979.
32. Craft JB Jr, Epstein BS, Coakley CS. Effect of lidocaine with epinephrine versus lidocaine (plain) on
induced labor. Anesth Analg. 1972;51:243–246.
33. Cheek TG, Samuels P, Miller F, et al. Normal saline i.v. fluid load decreases uterine activity in active
labour. Br J Anaesth. 1996;77:632–635.
34. Rahm VA, Hallgren A, Högberg H, et al. Plasma oxytocin levels in women during labor with or
without epidural analgesia: a prospective study. Acta Obstet Gynecol Scand. 2002;81:1033–1039.
35. Reitman E, Conell-Price J, Evansmith J, et al. β2-adrenergic receptor genotype and other variables that
contribute to labor pain and progress. Anesthesiology. 2011;114:927–939.
36. Terkawi AS, Jackson WM, Thiet MP, et al. Oxytocin and catechol-O-methyltransferase receptor
genotype predict the length of the first stage of labor. Am J Obstet Gynecol. 2012;207:184.e1–184.e8.
37. Zhang J, Landy HJ, Branch DW, et al. Contemporary patterns of spontaneous labor with normal
neonatal outcomes. Obstet Gynecol. 2010;116:1281–1287.
38. Laughon SK, Branch DW, Beaver J, et al. Changes in labor patterns over 50 years. Am J Obstet
Gynecol. 2012;206:419.e1–419.e9.
39. Cheng YW, Shaffer BL, Nicholson JM, et al. Second stage of labor and epidural use: a larger effect
than previously suggested. Obstet Gynecol. 2014;123:527–535.
40. Rouse DJ, Weiner SJ, Bloom SL, et al. Second-stage labor duration in nulliparous women: relationship
to maternal and perinatal outcomes. Am J Obstet Gynecol. 2009;201:357.e1–351.e7.
41. Laughon SK, Berghella V, Reddy UM, et al. Neonatal and maternal outcomes with prolonged second
stage of labor. Obstet Gynecol. 2014;124:57–67.
42. American College of Obstetrics and Gynecology Committee on Practice Bulletins—Obstetrics. ACOG
Practice Bulletin Number 49, December 2003: Dystocia and augmentation of labor. Obstet Gynecol.
2003;102:1445–1454.
43. Fraser WD, Marcoux S, Krauss I, et al. Multicenter, randomized, controlled trial of delayed pushing
for nulliparous women in the second stage of labor with continuous epidural analgesia. The PEOPLE
(Pushing Early or Pushing Late with Epidural) Study Group. Am J Obstet Gynecol. 2000;182:1165–
1172.
44. Tuuli MG, Frey HA, Odibo AO, et al. Immediate compared with delayed pushing in the second stage
of labor: a systematic review and meta-analysis. Obstet Gynecol. 2012;120:660–668.
45. Gambling DR, Sharma SK, Ramin SM, et al. A randomized study of combined spinal-epidural
analgesia versus intravenous meperidine during labor: impact on cesarean delivery rate.
Anesthesiology. 1998;89:1336–1344.
46. Sharma SK, Alexander JM, Messick G, et al. Cesarean delivery: a randomized trial of epidural
analgesia versus intravenous meperidine analgesia during labor in nulliparous women. Anesthesiology.
2002;96:546–551.
47. Comparative Obstetric Mobile Epidural Trial Study Group UK. Effect of low-dose mobile versus
traditional epidural techniques on mode of delivery: a randomised controlled trial. Lancet.
2001;358:19–23.
48. Wassen MM, Zuijlen J, Roumen FJ, et al. Early versus late epidural analgesia and risk of instrumental
delivery in nulliparous women: a systematic review. BJOG. 2011;118:655–661.
49. Impey L, MacQuillan K, Robson M. Epidural analgesia need not increase operative delivery rates. Am
J Obstet Gynecol. 2000;182:358–363.
50. Yancey MK, Pierce B, Schweitzer D, et al. Observations on labor epidural analgesia and operative
delivery rates. Am J Obstet Gynecol. 1999;180:353–359.
51. Segal S, Su M, Gilbert P. The effect of a rapid change in availability of epidural analgesia on the
cesarean delivery rate: a meta-analysis. Am J Obstet Gynecol. 2000;183:974–978.
52. Collis RE, Davies DW, Aveling W. Randomised comparison of combined spinal-epidural and standard
epidural analgesia in labour. Lancet. 1995;345:1413–1416.
53. Simmons SW, Taghizadeh N, Dennis AT, et al. Combined spinal-epidural versus epidural analgesia in
labour. Cochrane Database Syst Rev. 2012;(10):CD003401.
54. James KS, McGrady E, Quasim I, et al. Comparison of epidural bolus administration of 0.25%
bupivacaine and 0.1% bupivacaine with 0.0002% fentanyl for analgesia during labour. Br J Anaesth.
1998;81:507–510.
55. Capogna G, Camorcia M, Stirparo S, et al. Programmed intermittent epidural bolus versus continuous
epidural infusion for labor analgesia: the effects on maternal motor function and labor outcome. A
randomized double-blind study in nulliparous women. Anesth Analg. 2011;113:826–831.
56. Lieberman E, Davidson K, Lee-Parritz A, et al. Changes in fetal position during labor and their
association with epidural analgesia. Obstet Gynecol. 2005;105:974–982.
57. Baghestan E, Irgens LM, Børdahl PE, et al. Trends in risk factors for obstetric anal sphincter injuries
in Norway. Obstet Gynecol. 2010;116:25–34.
58. Richter HE, Brumfield CG, Cliver SP, et al. Risk factors associated with anal sphincter tear: a
comparison of primiparous patients, vaginal births after cesarean deliveries, and patients with previous
vaginal delivery. Am J Obstet Gynecol. 2002;187:1194–1198.
59. Jangö H, Langhoff-Roos J, Rosthøj S, et al. Modifiable risk factors of obstetric anal sphincter injury in
primiparous women: a population-based cohort study. Am J Obstet Gynecol. 2014;210:59.e1–59.e6.
60. Sharma SK, Sidawi JE, Ramin SM, et al. Cesarean delivery: a randomized trial of epidural versus
patient-controlled meperidine analgesia during labor. Anesthesiology. 1997;87:487–494.
61. Ramin SM, Gambling DR, Lucas MJ, et al. Randomized trial of epidural versus intravenous analgesia
during labor. Obstet Gynecol. 1995;86:783–789.
62. Lieberman E, Cohen A, Lang J, et al. Maternal intrapartum temperature elevation as a risk factor for
cesarean delivery and assisted vaginal delivery. Am J Public Health. 1999;89:506–510.
63. Thorp JA, Hu DH, Albin RM, et al. The effect of intrapartum epidural analgesia on nulliparous labor:
a randomized, controlled, prospective trial. Am J Obstet Gynecol. 1993;169:851–858.
64. Bell AF, White-Traut R, Medoff-Cooper B. Neonatal neurobehavioral organization after exposure to
maternal epidural analgesia in labor. J Obstet Gynecol Neonatal Nurs. 2010;39:178–190.
65. Lawrence A, Lewis L, Hofmeyr GJ, et al. Maternal positions and mobility during first stage labour.
Cochrane Database Syst Rev. 2013;(8):CD003934.
66. Gupta JK, Hofmeyr GJ, Shehmar M. Position in the second stage of labour for women without
epidural anaesthesia. Cochrane Database Syst Rev. 2012;(5):CD002006.
67. Kemp E, Kingswood CJ, Kibuka M, et al. Position in the second stage of labour for women with
epidural anaesthesia. Cochrane Database Syst Rev. 2013;(1):CD008070.
68. Segal S. Labor epidural analgesia and maternal fever. Anesth Analg. 2010;111:1467–1475.
69. Goetzl L, Rivers J, Zighelboim I, et al. Intrapartum epidural analgesia and maternal temperature
regulation. Obstet Gynecol. 2007;109:687–690.
70. Herbst A, Wølner-Hanssen P, Ingemarsson I. Risk factors for fever in labor. Obstet Gynecol.
1995;86:790–794.
71. Soper DE, Mayhall CG, Dalton HP. Risk factors for intraamniotic infection: a prospective
epidemiologic study. Am J Obstet Gynecol. 1989;161:562–566.
72. Goetzl L, Evans T, Rivers J, et al. Elevated maternal and fetal serum interleukin-6 levels are associated
with epidural fever. Am J Obstet Gynecol. 2002;187:834–838.
73. Goetzl L, Zighelboim I, Badell M, et al. Maternal corticosteroids to prevent intrauterine exposure to
hyperthermia and inflammation: a randomized, double-blind, placebo-controlled trial. Am J Obstet
Gynecol. 2006;195:1031–1037.
74. Riley LE, Celi AC, Onderdonk AB, et al. Association of epidural-related fever and noninfectious
inflammation in term labor. Obstet Gynecol. 2011;117:588–595.
75. Sharma SK, Rogers BB, Alexander JM, et al. A randomized trial of the effects of antibiotic
prophylaxis on epidural-related fever in labor. Anesth Analg. 2014;118:604–610.
76. Goetzl L, Cohen A, Frigoletto F Jr, et al. Maternal epidural analgesia and rates of maternal antibiotic
treatment in a low-risk nulliparous population. J Perinatol. 2003;23:457–461.
77. Lieberman E, Lang JM, Frigoletto F Jr, et al. Epidural analgesia, intrapartum fever, and neonatal
sepsis evaluation. Pediatrics. 1997;99:415–419.
78. Greenwell EA, Wyshak G, Ringer SA, et al. Intrapartum temperature elevation, epidural use, and
adverse outcome in term infants. Pediatrics. 2012;129:e447–e454.
79. Loftus JR, Hill H, Cohen SE. Placental transfer and neonatal effects of epidural sufentanil and fentanyl
administered with bupivacaine during labor. Anesthesiology. 1995;83:300–308.
80. Wieczorek PM, Guest S, Balki M, et al. Breastfeeding success rate after vaginal delivery can be high
despite the use of epidural fentanyl: an observational cohort study. Int J Obstet Anesth. 2010;19:273–
277.
81. Beilin Y, Bodian CA, Weiser J, et al. Effect of labor epidural analgesia with and without fentanyl on
infant breast-feeding: a prospective, randomized, double-blind study. Anesthesiology. 2005;103:1211–
1217.
Anesthetic Considerations for Women Receiving
Cesarean Delivery
Robert R. Gaiser
I. Background
II. Indications for cesarean delivery
III. Surgical considerations
A. Technical aspects
IV. Complications of cesarean delivery
A. Anesthetic complications
B. Surgical complications
C. Subsequent pregnancy risk
V. Preoperative considerations
A. Preoperative assessment
B. Consent
C. Blood products
D. Aspiration prophylaxis
E. Supplemental oxygen
F. Other preparation
G. Intraoperative medications
VI. Anesthetic techniques
A. Basic considerations
B. Epidural anesthesia for cesarean delivery
C. Spinal anesthesia for cesarean delivery
D. Combined spinal-epidural anesthesia for cesarean delivery
E. General anesthesia for cesarean delivery
VII. Postoperative management
VIII. Conclusions
KEYPOINTS
1. Cesarean delivery (CD) is the most commonly performed surgical
procedure.
2. In women receiving neuraxial anesthesia, high neuraxial block is a
leading cause of maternal mortality.
3. Hemorrhage, heart failure, amniotic fluid embolism, and sepsis are
common etiologies for cardiac arrest during hospitalization for delivery.
4. Obstetric blood loss is often underestimated.
5. Maternal history and risk of hemorrhage should guide decision making
about whether a type and screen or a type and cross match should be
obtained prior to CD.
6. Before cesarean delivery, practitioners should consider the timely
administration of nonparticulate antacids, H2-receptor antagonists, and/or
metoclopramide for aspiration prophylaxis.1
7. Uncomplicated patients who are scheduled for elective cesarean delivery
may have moderate amounts of clear fluids 2 hours and greater prior to
induction of anesthesia.1
8. The patient undergoing elective cesarean delivery or postpartum tubal
sterilization should not eat solid food for 6-8 hours beforehand,
depending on the composition of the food (e.g., fat content).1
9. Antibiotic prophylaxis should be administered within 1 hour of the
surgical incision.
10. Although phenylephrine is associated with fewer metabolic effects on the
fetus, either phenylephrine or ephedrine may be used for the prevention
and treatment of hypotension associated with anesthetic administration.
11. The decision to use a specific anesthetic technique for CD depends on the
situation and patient factors such as severe thrombocytopenia. The
obstetrician determines the urgency of the case.
12. Regardless of the anesthetic technique, all patients are maintained in left
uterine displacement until delivery of the neonate.
13. The administration of a bolus of crystalloid intravenously prior to and
during the initiation of neuraxial anesthesia may decrease the incidence
of hypotension but does not reliably prevent hypotension.
14. Combined spinal-epidural anesthesia may be advantageous when the
duration of CD is expected to outlast the spinal anesthetic.
15. Failed intubation, failed ventilation and oxygenation, and pulmonary
aspiration of gastric contents remain leading anesthesia-related causes of
maternal death, but there are new concerns of hypoventilation or airway
obstruction during extubation, emergence, or recovery.
I. Background
A. Cesarean delivery (CD) remains the most frequently performed
surgical procedure in the United States.
1. Beginning in 1998, the CD rate has increased every year until
2009. In 2009, it peaked at 32.9% of all births. The total rate
declined in 2010 to 32.8% and remained stable for 2011 and
2012.2
2. Recently, the American College of Obstetricians and
Gynecologists (ACOG) and the Society for Maternal and
Fetal Medicine issued a joint consensus statement: “The rapid
increase in the rate of cesarean births without evidence of
concomitant decreases in maternal or neonatal morbidity or
mortality raises significant concern that cesarean delivery is
overused. Therefore, it is important for health care providers
to understand . . . the safe and appropriate opportunities to
prevent overuse of cesarean delivery, particularly primary
cesarean delivery.” 3
This document includes 19 recommendations to decrease the
CD rate.
3. Approximately 60% of all CDs are primary (first-time CD).
Although in many cases, a prior cesarean does not necessitate
a subsequent CD; rates of trial of labor after cesarean
(TOLAC) have declined in recent years.
4. In 2012, the states with the lowest CD rate were Utah and
Idaho, whereas Louisiana and Florida had the highest rates.2
5. The increase in the use of epidural analgesia for labor is not
the reason for this increase in the incidence of CD, even if
administered early in the course of labor.4
B. Although CD rates have increased worldwide, rates vary by
country and are related to maternal, fetal, medicolegal,
socioeconomic, and obstetric factors.
C. Cesarean delivery on maternal request (CDMR) is a recent
consideration in obstetric practice. It is defined as a primary
prelabor CD on maternal request in the absence of any maternal
or fetal indications. Estimates suggest that these requests do not
contribute substantially to the increased CD rates. The ACOG
Committee on Obstetric Practice has concluded “that in the
absence of maternal or fetal indications for cesarean delivery, a
plan for vaginal delivery is safe and appropriate and should be
recommended to patients. In cases in which cesarean delivery on
maternal request is planned, delivery should not be performed
before a gestational age of 39 weeks. Cesarean delivery on
maternal request should not be motivated by the unavailability of
effective pain management. Cesarean delivery on maternal
request particularly is not recommended for women desiring
several children, given that the risks of placenta previa, placenta
accreta, and gravid hysterectomy increase with each cesarean
delivery.”5
D. “Obstetricians may accede to a patient’s request for CDMR.
However, obstetricians are not obliged ethically or professionally
to do so, and if the patient and clinician cannot agree on an
intended route of delivery, ACOG indicates that referral to
another health care practitioner is appropriate.”6
E. As compared to planned vaginal delivery, planned CD without
obstetric or fetal indications may be associated with lower rates of
hemorrhage and transfusion, surgical complications, and maternal
incontinence in the first year following delivery. However, it is
associated with a higher rate of neonatal respiratory morbidity
compared with vaginal delivery when delivery is earlier than 39
to 40 weeks of gestation.5
F. A recent meta-analysis of 157 eligible randomized controlled
trials (n = 31,085) evaluated whether the risk of cesarean was
higher following induction of labor. The results suggested that the
risk of CD was 12% lower when induction occurred in term and
post-term gestations. In addition, the risk of fetal death or
admission to the neonatal intensive care unit was decreased and
there was no impact on maternal death.7
B. Surgical complications
1. Intraoperative surgical complications occur in 12% to 15% of
CDs.15
a. The most common complication is hemorrhage.
Hemorrhage results from uterine atony, uterine
lacerations, or broad ligament hematomas.
b. The incidence of transfusion for primary CD is 3.2%,
with the median transfused volume being 2 units. The
majority of blood for primary CD is transfused
postoperatively. The incidence of transfusion for repeat
CD is 2.2%, with the majority of transfusions occurring
postoperatively. The median amount of blood transfused
is 2 units. Risk factors for transfusion include placenta
previa and preoperative anemia.16
c. The need for transfusion is not great due to the increase in
blood volume accompanying pregnancy, but an atonic
uterus can lose up to 2 L of blood in 5 minutes.
Furthermore, obstetric blood loss is often underestimated.
d. General anesthesia for CD is associated with a higher
blood loss than neuraxial anesthesia. However, this
increase in blood loss is NOT associated with an increase
in the need for blood transfusion.17
e. Table 13.2 lists the suggested resources that should be
available for management of obstetric hemorrhage.
f. Uterine or uterocervical lacerations are the next most
common complications.
g. Other complications include bladder laceration, fetal
laceration, and hysterectomy.
2. Postoperative complications include anemia, fever, urinary
tract infection, urinary retention, endometritis, thrombosis,
ileus, and wound infection.18
V. Preoperative considerations
A. Preoperative assessment
All patients who will undergo CD require a detailed history and
physical examination. The physical examination must be as
comprehensive as necessary but particular attention should be
paid to examination of the airway and the back. According to the
American Society of Anesthesiologists (ASA) Practice
Guidelines for Obstetric Anesthesia, “The anesthesiologist should
conduct a focused history and physical examination before
providing anesthesia care.”1
D. Aspiration prophylaxis
1. All parturients undergoing CD should receive aspiration
prophylaxis preoperatively including an H2-receptor blocker,
metoclopramide, and a nonparticulate antacid.1 Aspiration
prophylaxis is a recommendation of the ASA Practice
Guidelines for Obstetric Anesthesia.1
2. Parturients in labor are at an increased risk for aspiration
during general anesthesia. Gastric emptying is slowed with
the onset of painful contractions and parenteral opioids.
Epidural and intrathecal fentanyl can also impair gastric
emptying. If a patient aspirates, the patient is at risk for the
development of pneumonitis. In 1946, Mendelson reported 66
cases of aspiration of stomach contents during obstetric
anesthesia.22 Of these patients, 5 patients aspirated solid
material; 21 patients were subsequently diagnosed as having
aspirated; and 40 patients aspirated liquid material. The
aspiration of solid materials definitely increases the risk of
aspiration pneumonitis. For liquid material, the risk of
aspiration pneumonitis is dependent on the pH of the solution
(increased if pH<2.5) and volume of the solution (increased if
the volume is >25 mL). The administration of above
mentioned aspiration prophylaxis is thought to reduce the risk
for aspiration pneumonitis.23
3. “The oral intake of modest amounts of clear liquids may be
allowed for uncomplicated laboring patients. The
uncomplicated patient undergoing elective cesarean delivery
may have modest amounts of clear liquids up to 2 h before
induction of anesthesia. Examples of clear liquids include, but
are not limited to, water, fruit juices without pulp, carbonated
beverages, clear tea, black coffee, and sports drinks. The
volume of liquid ingested is less important than the presence
of particulate matter in the liquid ingested. However, patients
with additional risk factors for aspiration (e.g., morbid
obesity, diabetes, difficult airway) or patients at increased risk
for operative delivery (e.g., nonreassuring fetal heart rate
pattern) may have further restrictions of oral intake,
determined on a case-by-case basis.”1
4. “Solid foods should be avoided in laboring patients. The
patient undergoing elective surgery (e.g., scheduled cesarean
delivery or postpartum tubal ligation) should undergo a
fasting period for solids of 6–8 h depending on the type of
food ingested (e.g., fat content).”1
E. Supplemental oxygen
The routine administration of supplemental oxygen during CD
has been questioned because there is evidence that it may be
ineffective and, in some cases, detrimental because of the
conversion of oxygen to free radical oxygen species. Current
evidence suggests that supplementary oxygen given to healthy
term pregnant women during elective CD under neuraxial
anesthesia is associated with higher maternal and neonatal oxygen
levels (maternal SpO2, PaO2, UaPO2, and UvPO2) and higher
levels of oxygen free radicals. However, the intervention appears
neither beneficial or harmful to the neonate’s short-term clinical
outcome as assessed by Apgar scores.24
F. Other preparation
The ASA Practice Guidelines for Obstetric Anesthesia state that
the operating room for CD must have the same equipment as for
regular surgery.1 The ASA standards for basic monitoring apply
to the care of patients undergoing CD. The incidence of failed
intubation for CD is 1:224 general anesthetics. Whether the use of
videolaryngoscopy on the incidence of failed intubation is
unclear.25 A difficult airway cart as well as supplies for massive
hemorrhage and malignant hyperthermia should also be readily
available (see Chapter 14, Difficult Airway Management in the
Pregnant Patient).
G. Intraoperative medications
1. Prophylactic antibiotics are administered to reduce the risk
of endometritis, wound infections, urinary tract infections,
and fever. ACOG recommends administration of a first-
generation cephalosporin or other narrow-spectrum antibiotic
within 1 hour of the surgical incision.26 However, a recent
database review of over 1 million women who underwent CD
examined rates of antibiotic use and determined that only
60% of patients received antibiotics on the day of surgery.27
The study revealed a large variation by geographic region but
no influence of age, race, or insurance status.
2. Vasopressor administration
a. A vasopressor must be readily available. Either
phenylephrine or ephedrine is an acceptable choice.
b. The initial thought that ephedrine was the preferred agent
for the treatment of hypotension during CD was based on
animal models in which α-agonists decreased uterine
blood flow but mixed agonists had no effect. Subsequent
studies did not demonstrate an effect on uterine blood
flow from α-agonists.
c. A statistically significant difference in improved umbilical
arterial pH was found for phenylephrine, but the clinical
importance of this small difference is uncertain. The
difference is not due to an effect on uterine blood flow but
because ephedrine crosses the placenta and stimulates the
release of fetal catecholamines. The increase in
catecholamine levels leads to an increase in oxygen
consumption and an increase in lactate concentration.28
d. The effect of ephedrine on umbilical arterial pH may also
be a result of genetic susceptibility in certain fetuses.
Those infants who do develop an acidosis tend to have
similar genetics.29
e. Phenylephrine is typically administered by continuous
infusion, especially when spinal anesthesia is
administered. The infusion is initiated at the time of
administration of the intrathecal medications. There is no
difference in umbilical arterial pH when phenylephrine is
administered as a continuous infusion or as intermittent
boluses. With a continuous infusion, there is a lower
incidence of maternal nausea and vomiting.30
f. Either phenylephrine or ephedrine is an acceptable choice
for the treatment of hypotension. If multiple doses are
required, phenylephrine is the preferred drug. If the
maternal heart rate is low, as may occur with spinal
anesthesia with a T1–T2 sensory level, ephedrine is the
better choice as phenylephrine may slow the maternal
heart rate further. Rather than the choice of drug, the most
important factor is the rapid correction of hypotension
because uteroplacental perfusion is proportionate to blood
pressure. However, this has recently been questioned. In
one study of 919 mothers who underwent CD, over 30%
of patients who experienced a ≤30% decrease in maternal
mean arterial pressure for greater than 5 minutes had no
adverse effect on the neonate.31
g. In another quantitative, systematic review of studies
comparing phenylephrine and ephedrine was conducted, 7
randomized controlled trials were identified for a total of
292 patients. There was no difference between
phenylephrine and ephedrine in the ability to correct
maternal hypotension, but a higher incidence of maternal
bradycardia occurred if phenylephrine was used. In regard
to the neonate, there was no difference in the incidence of
true fetal acidosis, but neonates whose mothers received
phenylephrine had higher umbilical arterial pH values.32
h. Most recently, one study has suggested that
norepinephrine may be useful in this setting.33
d. Paresthesia
(1) Spinal anesthesia is performed at interspaces lower
than L2 to prevent trauma to the spinal cord. A
paresthesia may occur during placement of the spinal
needle or injection of the anesthetic as the needle
contacts the cauda equina. Patients may complain of
pain or a shock shooting into the lower extremity.
(2) For persistent paresthesias, the needle should be
removed and placed at another interspace.
e. Cardiac arrest
(1) Unexpected cardiac arrest has been reported in
patients receiving spinal anesthesia. It generally
occurs in patients with high sensory levels in which
the cardiac accelerator fibers (T1–T4) have been
blocked.
(2) The early use of epinephrine in the resuscitation of
patients experiencing cardiac arrest from spinal
anesthesia improves survival.53
VIII. Conclusions
A. The number of CDs in the United States has stabilized. However,
with a CD rate of one-third of all births in the United States, it
represents the most commonly performed surgical procedure.
B. Epidural analgesia does not increase the risk of CD. If an epidural
catheter had been placed for labor, it may be used for CD. If the
catheter appears to be nonfunctional, it is important to stop
injecting the catheter before large volumes are administered to
prevent a high sensory level from administration of spinal
anesthetic.
C. If general anesthesia is chosen, rapid sequence induction should
be used because parturients are at risk for aspiration. Parturients
require the same amount of anesthesia as nonpregnant individuals
to prevent awareness. Postpartum bleeding is greater with general
anesthesia, although there is no difference in the need for
transfusion.
D. General, spinal, epidural, and CSE anesthesia are used for CD.
Each has advantages and disadvantages. The decision to use a
particular anesthetic must be individualized and will, in part, be
dependent on the institution and the level of training of the
surgeons. In addition, the choice will be dependent on the
urgency of the situation and the maternal condition.
REFERENCES
1. American Society of Anesthesiologists Task Force on Obstetric Anesthesia. Practice guidelines for
obstetric anesthesia: an updated report by the American Society of Anesthesiologists Task Force on
Obstetric Anesthesia. Anesthesiology. 2007;106:843–863.
2. Osterman MJ, Martin JA. Primary cesarean delivery rates, by state: results from the revised birth
certificate: 2006-2012. Natl Vital Stat Rep. 2014;63:1–11.
3. American College of Obstetricians and Gynecologists, Society for Maternal-Fetal Medicine. Obstetric
Care Consensus No. 1: safe prevention of the primary cesarean delivery. Obstet Gynecol.
2014;123:693–711.
4. Wong CA, Scavone BM, Peaceman AM, et al. The risk of cesarean delivery with neuraxial analgesia
given early versus late in labor. N Engl J Med. 2005;352:655–665.
5. American College of Obstetricians and Gynecologists. ACOG Committee Opinion No. 559: cesarean
delivery on maternal request. Obstet Gynecol. 2013;121:904–907.
6. Ecker J. Elective cesarean delivery on maternal request. JAMA. 2013;309:1930–1936.
7. Mishanina E, Rogozinska E, Thatthi T, et al. Use of labour induction and risk of cesarean delivery: a
systematic review and meta-analysis. CMAJ. 2014;186:665–673.
8. Osterman MJ, Martin JA. Changes in cesarean delivery rates by gestational age: United States, 1996-
2011. NCHS Data Brief. 2013;124:1–8.
9. McAdams RM. Time to implement delayed cord clamping. Obstet Gynecol. 2014;123:549–552.
10. Hung KJ, Berg O. Early skin-to-skin after cesarean to improve breastfeeding. MCN Am J Matern
Child Nurs. 2011;36:318–324.
11. Bamigboye AA, Hofmeyr GJ. Closure versus non-closure of the peritoneum at caesarean section:
short- and long-term outcomes. Cochrane Database Syst Rev. 2014;8:CD000163.
12. Hawkins JL, Chang J, Palmer SK, et al. Anesthesia-related maternal mortality in the United States:
1979-2002. Obstet Gynecol. 2011;117:69–74.
13. Creanga AA, Berg CJ, Syverson C, et al. Pregnancy-related mortality in the United States, 2006-2010.
Obstet Gynecol. 2015;125:5–12.
14. Mhyre JM, Tsen LC, Einav S, et al. Cardiac arrest during hospitalization for delivery in the United
States, 1998-2011. Anesthesiology. 2014;120:810–818.
15. Hammad IA, Chauhan SP, Magann EF, et al. Peripartum complications with cesarean delivery: a
review of Maternal-Fetal Medicine Units Network publications. J Mat Fetal Neonatal Med.
2014;27:463–474.
16. Rouse DJ, MacPherson C, Landon M, et al. Blood transfusion and cesarean delivery. Obstet Gynecol.
2006;108:891–897.
17. Heesen M, Hofmann T, Klöhr S, et al. Is general anaesthesia for caesarean section associated with
postpartum haemorrhage? Systematic review and meta-analysis. Acta Anaesthesiol Scand.
2013;57:1092–1102.
18. Zelop C, Heffner LJ. The downside of cesarean delivery: short- and long-term complications. Clin
Obstet Gynecol. 2004;47:386–393.
19. Silver RM, Landon MB, Rouse DJ, et al. Maternal morbidity associated with multiple repeat cesarean
deliveries. Obstet Gynecol. 2006;107:1226–1232.
20. Leeman LM, Beagle M, Espey E, et al. Diminishing availability of trial of labor after cesarean
delivery in New Mexico hospitals. Obstet Gynecol. 2013;122:242–247.
21. Lydon-Rochelle M, Holt VL, Easterling TR, et al. Risk of uterine rupture during labor among women
with a prior cesarean delivery. N Engl J Med. 2001;345:3–8.
22. Mendelson CL. The aspiration of stomach contents into the lungs during obstetric anesthesia. Am J
Obstet Gynecol. 1946;52:191–205.
23. Paranjothy S, Griffiths JD, Broughton HK, et al. Interventions at caesarean section for reducing the
risk of aspiration pneumonitis. Cochrane Database Syst Rev. 2014;2:CD004943.
24. Chatmongkolchart S, Prathep S. Supplemental oxygen for caesarean section during regional
anesthesia. Cochrane Database Syst Rev. 2013;6:CD006161.
25. Quinn AC, Milne D, Columb M, et al. Failed tracheal intubation in obstetric anaesthesia: 2 yr national
case-control study in the UK. Br J Anaesth. 2013;110:74–80.
26. American College of Obstetricians and Gynecologists. ACOG Practice Bulletin No. 120: use of
prophylactic antibiotics in labor and delivery. Obstet Gynecol. 2011;117:1472–1483.
27. Brubaker SG, Friedman AM, Cleary KL, et al. Patterns of use and predictors of receipt of antibiotics
in women undergoing cesarean delivery. Obstet Gynecol. 2014;124:338–344.
28. Saravanan S, Kocarev M, Wilson RC, et al. Equivalent dose of ephedrine and phenylephrine in the
prevention of post-spinal hypotension in caesarean section. Br J Anaesth. 2006;96:95–99.
29. Landau R, Liu SK, Blouin JL, et al. The effect of maternal and fetal beta2-adrenoceptor and nitric
oxide synthase genotype on vasopressor requirement and fetal acid-base status during spinal
anesthesia for cesarean delivery. Anesth Analg. 2011;112:1432–1437.
30. Doherty A, Ohashi Y, Downey K, et al. Phenylephrine infusion versus bolus regimens during cesarean
delivery under spinal anesthesia: a double-blind randomized clinical trial to assess hemodynamic
changes. Anesth Analg. 2012;115:1343–1350.
31. Maayan-Metzger A, Schushan-Eisen I, Todris L, et al. Maternal hypotension during elective cesarean
section and short-term neonatal outcome. Am J Obstet Gynecol. 2010;202:56.e1–e5.
32. Lee A, Ngan Kee WD, Gin T. A quantitative, systematic review of randomized controlled trials of
ephedrine versus phenylephrine for the management of hypotension during spinal anesthesia for
cesarean delivery. Anesth Analg. 2002;94:920–926.
33. Ngan Kee WD, Lee SW, Ng FF, et al. Randomized double-blinded comparison of norepinephrine and
phenylephrine for maintenance of blood pressure during spinal anesthesia for cesarean delivery.
Anesthesiology. 2015;122:736–745.
34. American College of Obstetricians and Gynecologists. ACOG Practice Bulletin: Clinical Management
Guidelines for Obstetrician-Gynecologists Number 76, October 2006: postpartum hemorrhage. Obstet
Gynecol. 2006;108:1039–1047.
35. Maayan-Metzger A, Schushan-Eisen I, Todris L, et al. The effect of time intervals on neonatal
outcome in elective cesarean delivery at term under regional anesthesia. Int J Gynaecol Obstet.
2010;111:224–228.
36. Torloni MR, Betran AP, Souza JP, et al. Classifications for cesarean section: a systematic review. PLoS
One. 2011;6:e14566.
37. Nor NM, Russell IF. Assessing blocks after spinal anaesthesia for elective caesarean section: how
different questions affect findings from the same stimulus. Int J Obstet Anesth. 2013;22:294–297.
38. Auroy Y, Benhamou D, Bargues L, et al. Major complications of regional anesthesia in France: the
SOS regional anesthesia hotline service. Anesthesiology. 2002;97:1274–1280.
39. Evans RW, Armon C, Frohman EM, et al. Assessment: prevention of post-lumbar puncture headaches:
report of the Therapeutics and Technology Assessment Subcommittee of the Academy of Neurology.
Neurology. 2000;55:909–914.
40. Russell IF. A prospective controlled study of continuous spinal analgesia versus repeat epidural
analgesia after accidental dural puncture in labour. Int J Obstet Anesth. 2012;21:7–16.
41. Lee LA, Posner KL, Domino KB, et al. Injuries associated with regional anesthesia in the 1980s and
1990s: a closed claims analysis. Anesthesiology. 2004;101:143–152.
42. Howell CJ, Dean T, Lucking L, et al. Randomised study of long term outcome after epidural versus
non-epidural analgesia during labour. BMJ. 2002;325:357.
43. Karambelkar DJ, Ramanathan S. 2-Chloroporcoaine antagonism of epidural morphine analgesia. Acta
Anaesthesiol Scand. 1997;41:774–778.
44. Colonna-Romano P, Lingaraju N, Godfrey SD, et al. Epidural test dose and intravascular injection in
obstetrics: sensitivity, specificity, and lowest effective dose. Anesth Analg. 1992;75:372–376.
45. Brose WG, Cohen SE. Epidural lidocaine for cesarean section: effect of varying epinephrine
concentration. Anesthesiology. 1988;69:936–940.
46. Palmer CM, Nogami WM, Van Maren G, et al. Postcesarean epidural morphine: a dose-response
study. Anesth Analg. 2000;90:887–891.
47. Boyle RK. A review of anatomical and immunological links between epidural morphine and herpes
simplex labialis in obstetric patients. Anaesth Intensive Care. 1995;23:425–432.
48. Lambert DH, Hurley RJ, Hertwig L, et al. Role of needle gauge and tip configuration in the production
of lumbar puncture headache. Reg Anesth. 1997;22:66–72.
49. Visalyaputra S, Rodanant O, Somboonviboon W, et al. Spinal versus epidural anesthesia for cesarean
delivery in severe preeclampsia: a prospective randomized, multicenter study. Anesth Analg.
2005;101:862–868.
50. Carvalho B, Collins J, Drover DR, et al. ED(50) and ED(95) of intrathecal bupivacaine in morbidly
obese patients undergoing cesarean delivery. Anesthesiology. 2011;114:529–535.
51. Dyer RA, Farina Z, Jouber IA, et al. Crystalloid preload versus rapid crystalloid administration after
induction of spinal anesthesia (coload) for elective caesarean section. Anaesth Intensive Care.
2004;32:351–357.
52. Reynolds F, Seed PT. Anaesthesia for caesarean section and neonatal acid-base status: a meta-analysis.
Anaesthesia. 2005;60:636–653.
53. Kopp SL, Horlocker TT, Warner ME, et al. Cardiac arrest during neuraxial anesthesia: frequency and
predisposing factors associated with survival. Anesth Analg. 2005;100:855–865.
54. Aouad MT, Siddik SS, Jalbout MI, et al. Does pregnancy protect against intrathecal lidocaine-induced
transient neurologic symptoms? Anesth Analg. 2001;92:401–404.
55. Palmer CM, Emerson S, Volgoropolous D, et al. Dose-response relationship of intrathecal morphine
for postcesarean analgesia. Anesthesiology. 1999;90:437–444.
56. Blumgart CH, Ryall D, Dennison B, et al. Mechanism of extension of spinal anaesthesia by extradural
injection of local anaesthetic. Br J Anaesth. 1992;69:457–460.
57. Pant M, Fong R, Scavone B. Prevention of peri-induction hypertension in preeclamptic patients: a
focused review. Anesth Analg. 2014;119:1350–1356.
58. Mhyre JM, Riesner MN, Polley LS, et al. A series of anesthesia-related maternal deaths in Michigan,
1985-2003. Anesthesiology. 2007;106:1096–1104.
59. Ueyama H, Hagihira S, Takashina M, et al. Pregnancy does not enhance volatile anesthetic sensitivity
on the brain: an electroencephalographic analysis study. Anesthesiology. 2010;113:577–584.
60. American College of Obstetricians and Gynecologists. Guidelines for Perinatal Care. 7th ed.
Washington, DC: American College of Obstetricians and Gynecologists; 2012:109–110, 160, 192–
194, 248.
61. McMorrow RC, Ni Mhuircheartaigh RJ, Ahmed KA, et al. Comparison of transversus abdominis
plane block vs spinal morphine for pain relief after caesarean section. Br J Anaesth. 2011;106:706–
712.
62. Horlocker TT, Burton AW, Connis RT, et al. Practice guidelines for the prevention, detection, and
management of respiratory depression associated with neuraxial opioid administration.
Anesthesiology. 2009;110:218–230.
Difficult Airway Management in the Pregnant Patient
Uma Munnur and Maya S. Suresh
I. Introduction
II. Definitions
A. Difficult airway
B. Difficult face mask ventilation or supraglottic airway
ventilation
C. Difficult supraglottic airway placement
D. Difficult laryngoscopy
E. Difficult tracheal intubation
F. Failed intubation
III. Goals and preparation for airway management during pregnancy
A. Goals to eliminate airway-related mortality
B. Preparation
IV. Incidence of difficult and failed intubation
A. Risk of failed intubation in obstetric patients
B. Factors contributing to the difficult airway
V. Anesthesia-related morbidity and mortality
A. Experience in the United States
B. Experience in the United Kingdom
VI. Maternal deaths and airway-related issues following emergence
A. US data
B. Data from the United Kingdom
VII. Anatomic and physiologic changes during pregnancy contributing
in difficult airway management
A. Airway changes
B. Respiratory changes
C. Cardiovascular changes
D. Gastrointestinal changes
E. Recommendations for airway management: implications of
the physiologic changes of pregnancy
VIII. Airway assessment
A. History
B. Physical examination
C. Specific individual tests for assessment of difficult tracheal
intubation
IX. Morbid obesity in pregnancy and the airway
A. Prevalence of morbid obesity
B. Technical challenges
C. Increased anesthetic risk
X. Aspiration of gastric contents
A. Aspiration-related death
B. Risk of difficult mask ventilation and aspiration after repeat
attempts at intubation
XI. Anesthetic management in obstetric patients with a predicted
difficult airway
A. American Society of Anesthesiologists practice guidelines
B. Management of the parturient with a predicted difficult
airway undergoing labor or operative delivery, where airway
management is not necessary
C. Management of the predicted difficult airway in obstetric
patient where airway management is needed: emphasis on
awake intubation
XII. Management of pregnant patient with an unanticipated difficult
airway
A. Step 1: first attempt at tracheal intubation
B. Step 2: second tracheal intubation/best attempt—difficult
laryngoscopy/difficult intubation
C. Step 3: maintenance of oxygenation/ventilation—failed
intubation
D. Step 4: management of the “cannot intubate/cannot ventilate”
situation
E. Step 5: management of pregnant patients with a critical
airway (increasing hypoxemia)
XIII. Extubation and postanesthesia care unit airway issues
XIV. Conclusion
KEYPOINTS
1. Airway-related complications associated with failed endotracheal
intubation and the inability to oxygenate/ventilate after induction of
general anesthesia for cesarean delivery are associated with increased
rates of maternal morbidity and mortality.
2. Attention should be focused on history, physical examination, and airway
evaluation for predictors of difficult intubation and difficult ventilation.
3. Heightened awareness of general anesthesia–related maternal morbidity
and mortality has led to a widespread adoption of neuraxial techniques
for both labor and cesarean delivery, and improvements in the
management of difficult airway has led to a significant decrease in
airway-related deaths.
4. Emergency airway management following failed tracheal intubation in
obstetrics presents challenges for the anesthesia provider.
5. A well-thought-out airway rescue plan, within the framework of a
predetermined difficult airway algorithm, is required prior to the
induction of anesthesia in the parturient.
6. Protocols to reduce anesthesia risks for women undergoing labor, during
induction of general anesthesia for cesarean delivery, during anesthetic
emergence, and postoperatively should be established by each institution
where obstetric anesthesia is provided.
7. All anesthesia providers should acquire and maintain advanced airway
management skills including cricothyroidotomy.
I. Introduction
Airway-related complications associated with failed endotracheal
intubation and the inability to ventilate/oxygenate after induction of
general anesthesia (GA) for cesarean delivery (CD) are associated
with increased rates of maternal morbidity and mortality. Anatomic
and physiologic changes during pregnancy place the parturient at
higher risk for airway-related complications; therefore, the parturient
undergoing GA is at greater risk for airway-related problems
compared to the nonpregnant patient. Obesity, estimated to affect over
50% of the pregnant population, also increases the risk for difficult
intubation and ventilation. As the prevalence of high-risk obstetric
patients increases, anesthesia providers will likely encounter more
clinical situations where neuraxial anesthesia may be exceedingly
difficult or contraindicated, making airway management exceedingly
challenging. In many countries, the recognition that adverse maternal
anesthetic outcomes are often associated with difficult airway
management has led to a dramatic decline in the use of GA for both
elective and emergency CD.
Increased focus on improving patient safety in anesthesia by
anesthesiologists has led to tremendous advances in airway
management in recent years. These include (1) introduction and
revision of the American Society of Anesthesiologists (ASA) Task
Force on Difficult Airway Management (DAM)—recommendations
for management of the difficult airway,1 (2) considerable advances in
comprehensive airway management, (3) improvement and increases
in the number of airway devices available to aid in airway
management, and (4) an increase in publications worldwide
concerning advanced airway management. These improvements have
led to a decline in the incidence of airway-related perioperative
morbidity during anesthesia and surgery in the general surgical and
obstetrical populations.2
II. Definitions. There is no consensus on a standard definition of the
difficult airway in the available literature. The following are
definitions used by the ASA Task Force on Difficult Airway
Management1:
A. Difficult airway. Clinical situation in which a conventionally
trained anesthesiologist experiences difficulty with face mask
ventilation of the upper airway, difficulty with tracheal intubation,
or both leading to hypoxemia.
B. Difficult face mask ventilation or supraglottic airway
ventilation
1. Successful face mask ventilation provides anesthesia
providers with a rescue technique during unsuccessful
attempts at laryngoscopy and unanticipated difficult airway
situations.
2. A grading scale for mask ventilation consisting of four
categories (Grades 1 to 4): Grade 1: easy ventilation by mask,
Grade 2: ventilation by mask with oral airway/adjuvant with
or without muscle relaxant, Grade 3: difficult ventilation
(inadequate, unstable, or requiring two providers) with or
without muscle relaxant, Grade 4: unable to mask ventilate
with or without muscle relaxant. Grades 3 and 4 define
difficult mask ventilation and impossible mask ventilation,
respectively.
3. Supraglottic airway (SGA) equipment includes the laryngeal
mask airway (LMA), intubating LMA (ILMA), and laryngeal
tubes.
4. Difficult SGA ventilation
a. “Difficult SGA” ventilation occurs when the
anesthesiologist is unable to provide adequate ventilation
because of one or more of the following problems: (1)
inadequate mask or SGA seal, (2) excessive gas leak, or
(3) excessive resistance to the ingress or egress of gas.
b. Signs of inadequate ventilation include (but are not
limited to) absent or inadequate chest movement, absent
or inadequate breath sounds, auscultatory signs of severe
obstruction, cyanosis, gastric air entry or dilatation,
decreasing or inadequate oxygen saturation (SpO2),
absent or inadequate exhaled carbon dioxide as detected
by end-tidal carbon dioxide (ETCO2) monitoring, absent
or inadequate spirometry measures of exhaled gas flow,
and hemodynamic changes associated with hypoxemia or
hypercarbia (e.g., hypertension, tachycardia,
dysrhythmia).
C. Difficult supraglottic airway placement. SGA placement that
requires multiple attempts.
D. Difficult laryngoscopy. It is not possible to visualize any portion
of the vocal cords after multiple attempts at conventional
laryngoscopy.
E. Difficult tracheal intubation. The ASA Task Force describes
difficult intubation as requiring multiple attempts.
F. Failed intubation. This has occurred when placement of the
endotracheal tube (ETT) fails after multiple attempts. The ASA
Task Force on Difficult Airway Management definition may not
be applicable in the obstetrical patient undergoing CD in which
GA is used for urgent delivery of the neonate. A common practice
in such obstetrical patients is to induce GA in a rapid sequence
manner using a single dose of succinylcholine to accomplish the
intubation. A more appropriate definition of failed intubation in
obstetrical patients may be an inability to secure the airway after
administration of a single dose of succinylcholine followed by no
more than two attempts at intubation using conventional
laryngoscopy or an alternative device to assist with the tracheal
intubation.
III. Goals and preparation for airway management during
pregnancy
A. Goals to eliminate airway-related mortality. The impact of
adverse maternal and fetal outcomes due to anesthesia-related
airway mishap or suboptimal management can have devastating
effects on the patient’s family and result in financial liability for
anesthesia providers. Recently announced safe motherhood
initiatives and United Nations Millennium Development Goals,
which emphasize improved maternal health, focus on eliminating
anesthesia-related maternal mortality, particularly airway-related
mortality. Obstetric anesthesia providers should:
1. Understand the impact of anatomic and physiologic changes
during pregnancy and the impact on airway management (see
Chapter 1).
2. Balance the urgency of delivering the baby while ensuring
maternal safety.
3. Understand the importance of prioritizing and establishing
oxygenation and ventilation, using a number of airway rescue
devices.
4. Prevent pulmonary aspiration, particularly during the use of
SGA devices.
5. Recognize a difficult airway and have effective strategies to
manage a parturient with a difficult airway undergoing labor
or CD. The ultimate goal as stated in the Safe Motherhood
Initiatives is to prevent and eliminate all maternal deaths,
including deaths associated with difficult airway management.
B. Preparation. With these goals in mind, the obstetric anesthesia
provider should implement the following steps:
1. Learn the predictors of difficult airway management.
2. Assess a patient for risk factors that predispose to airway-
related complications.
3. Have a well-thought-out airway rescue plan, within the
framework of a predetermined difficult airway algorithm.
4. Have a difficult airway cart immediately available in the labor
and delivery area.
5. Understand the importance of preventing aspiration of gastric
contents when using an SGA.
6. Acquire and maintain advanced airway management skills
including cricothyroidotomy.
2. Cesarean delivery
a. Neuraxial anesthesia
(1) Neuraxial anesthesia is the most common mode of
anesthesia utilized for CD in developed countries. For
elective or urgent CD in women with a known
difficult airway and no prior neuraxial block,
neuraxial anesthesia is usually preferable, provided
there is adequate time, no acute maternal or fetal
compromise, and no contraindication to neuraxial
anesthesia. Neuraxial techniques are safe and
predictable in women with difficult airways,8
especially for the elective or emergent CD if airway
intervention is deemed unnecessary. A single-shot
spinal, continuous epidural anesthesia, combined
spinal-epidural (CSE), or continuous spinal
anesthesia (CSA) can be used.42
(2) The use of neuraxial anesthesia in patients with a
recognized difficult airway does not always eliminate
the problem of difficult airway management. The
danger of neuraxial anesthesia in a patient with
known or suspected difficult airway is that failure to
provide an adequate block, prolonged surgery, or
uncontrolled hemorrhage could result in the need for
immediate induction of GA under suboptimal
conditions. In such a situation, airway equipment to
help manage the airway should be immediately
available.
b. Spinal anesthesia. Spinal anesthesia is often
administered for a CD regardless of airway status. With
administration of an appropriate dose of local anesthetic
for spinal anesthesia, a high motor block with respiratory
insufficiency can almost always be avoided.
c. Epidural anesthesia
(1) When a CD is nonemergent, epidural anesthesia can
be used. The advantages of epidural anesthesia
include gradual titration of block, avoidance of major
hemodynamic and respiratory compromise, and the
ability to extend the anesthetic until surgery is
completed.
(2) In a patient with an epidural catheter in situ, adequate
function of the catheter must be confirmed before
surgery. If anesthesia is inadequate, and if time
permits, the epidural catheter should be replaced.
Bloom et al.43 reported that failed neuraxial
anesthesia requiring conversion to GA occurred more
commonly with an epidural than spinal or CSE (4.3%
vs. 2.1% and 1.7%, respectively).
d. Spinal anesthesia after failed epidural blockade. The
administration of spinal anesthesia after a failed epidural
is associated with an increased risk for high or total spinal
anesthesia. However, with adequate precautions, the use
of spinal anesthesia in this setting has several advantages,
including rapid onset and reliable surgical anesthesia. The
recommended precautions, when spinal anesthesia is
planned after a failed epidural, are (1) no epidural bolus
administered in the 30 minutes preceding spinal
anesthesia, (2) decrease the dose of intrathecal local
anesthetic by 20% to 30%, and (3) maintain the sitting
position for 1 to 2 minutes after intrathecal injection
followed by supine positioning with left uterine
displacement.44,45
REFERENCES
1. Apfelbaum JL, Hagberg CA, Caplan RA, et al. Practice guidelines for management of the difficult
airway: an updated report by the American Society of Anesthesiologists Task Force on Management
of the Difficult Airway. Anesthesiology. 2013;118:251–270.
2. Davies JM, Posner KL, Lee LA, et al. Liability associated with obstetric anesthesia: a closed claims
analysis. Anesthesiology. 2009;110:131–139.
3. Lyons G. Failed intubation: six years’ experience in a teaching maternity unit. Anaesthesia.
1985;40:759–762.
4. Samsoon GL, Young JR. Difficult tracheal intubation: a retrospective study. Anaesthesia.
1987;42:487–490.
5. Kheterpal S, Healy D, Aziz MF, et al. Incidence, predictors, and outcome of difficult mask ventilation
combined with difficult laryngoscopy: a report from the multicenter perioperative outcomes group.
Anesthesiology. 2013;119:1360–1369.
6. Palanisamy A, Mitani AA, Tsen LC. General anesthesia for cesarean delivery at a tertiary care hospital
from 2000 to 2005: a retrospective analysis and 10-year update. Int J Obstet Anesth. 2011;20:10–16.
7. Bucklin BA. Gerard W. Ostheimer “what’s new in obstetric anesthesia” lecture. Anesthesiology.
2006;104:865–871.
8. Hawkins JL, Koonin LM, Palmer SK, et al. Anesthesia-related deaths during obstetric delivery in the
United States: 1979–1990. Anesthesiology. 1997;86:277–284.
9. Hawkins JL, Chang J, Palmer SK, et al. Anesthesia-related maternal mortality in the United States:
1979–2002. Obstet Gynecol. 2011;117:69–74.
10. Chadwick HS. Obstetric anesthesia closed claims update II.
http://depts.washington.edu/asaccp/sites/default/files/pdf /Click%20here%20for%20_74.pdf Accessed
November 4, 2015.
11. Cooper GM, McClure JH. Anaesthesia chapter from Saving Mothers’ Lives: reviewing maternal
deaths to make pregnancy safer. Br J Anaesth. 2008;100:17–22.
12. Quinn AC, Milne D, Columb M, et al. Failed tracheal intubation in obstetric anaesthesia: 2 yr national
case-control study in the UK. Br J Anaesth. 2013;110:74–80.
13. Cook TM, Woodall N, Harper J, et al; for the Fourth National Audit Project. Major complications of
airway management in the UK: results of the Fourth National Audit Project of the Royal College of
Anaesthetists and the Difficult Airway Society, part 2: intensive care and emergency departments. Br J
Anaesth. 2011;106:632–642.
14. Mhyre JM, Riesner MN, Polley LS, et al. A series of anesthesia-related maternal deaths in Michigan:
1985–2003. Anesthesiology. 2007;106:1096–1104.
15. McClure JH, Cooper GM, Clutton-Brock TH; for Centre for Maternal and Child Enquiries. Saving
Mothers’ Lives: reviewing maternal deaths to make motherhood safer: 2006–2008: a review. Br J
Anaesth. 2011;107:127–132.
16. Kodali BS, Chandrasekhar S, Bulich LN, et al. Airway changes during labor and delivery.
Anesthesiology. 2008;108: 357–362.
17. American Society of Anesthesiologists Task Force on Obstetric Anesthesia. Practice guidelines for
obstetric anesthesia: an updated report by the American Society of Anesthesiologists Task Force on
Obstetric Anesthesia. Anesthesiology. 2007;106:843–863.
18. Pilkington S, Carli F, Dakin MJ, et al. Increase in Mallampati score during pregnancy. Br J Anaesth.
1995;74:638–642.
19. Takenaka I, Aoyama K, Kadoya T. Mandibular protrusion test for prediction of difficult mask
ventilation. Anesthesiology. 2001;94:935.
20. Calder I, Calder J, Crockard HA. Difficult direct laryngoscopy in patients with cervical spine disease.
Anaesthesia. 1995;50:756–763.
21. Chou HC, Wu TL. Mandibulohyoid distance in difficult laryngoscopy. Br J Anaesth. 1993;71:335–
339.
22. Gupta S, Pareek S, Dulara SC. Comparison of two methods for predicting difficult intubation in
obstetric patients. Middle East J Anesthesiol. 2003;17:275–285.
23. Wilson ME, Spiegelhalter D, Robertson JA, et al. Predicting difficult intubation. Br J Anaesth.
1988;61:211–216.
24. Murphy MF, Walls RM. The difficult and failed airway. In: Walls RA, ed. Manual of Emergency
Airway Management. Philadelphia, PA: Lippincott Williams & Wilkins; 2000:31–39.
25. Morgan BM, Magni V, Goroszenuik T. Anaesthesia for emergency caesarean section. Br J Obstet
Gynaecol. 1990;97: 420–424.
26. Langeron O, Masso E, Huraux C, et al. Prediction of difficult mask ventilation. Anesthesiology.
2000;92:1229–1236.
27. Felton E, Suresh M, Wali A. Survey questionnaire: difficult airway management during emergent
cesarean section and availability of difficult airway equipment in the labor and delivery suite: a
comparison between academic and private practice hospitals. Anesthesiology. 2005;103:A583.
28. Shiga T, Wajima Z, Inoue T, et al. Predicting difficult intubation in apparently normal patients: a meta-
analysis of bedside screening test performance. Anesthesiology. 2005;103:429–437.
29. Kheterpal S, Han R, Tremper KK, et al. Incidence and predictors of difficult and impossible mask
ventilation. Anesthesiology. 2006;105:885–891.
30. Pathi A, Esen U, Hildreth A. A comparison of complications of pregnancy and delivery in morbidly
obese and non-obese women. J Obstet Gynaecol. 2006;26:527–530.
31. Saravanakumar K, Rao SG, Cooper GM. The challenges of obesity and obstetric anaesthesia. Curr
Opin Obstet Gynecol. 2006;18:631–635.
32. Hood DD, Dewan DM. Anesthetic and obstetric outcome in morbidly obese parturients.
Anesthesiology. 1993;79:1210–1218.
33. Adams JP, Murphy PG. Obesity in anaesthesia and intensive care. Br J Anaesth. 2000;85:91–108.
34. Endler GC, Mariona FG, Sokol RJ, et al. Anesthesia-related maternal mortality in Michigan: 1972 to
1984. Am J Obstet Gynecol. 1988;159:187–193.
35. American College of Obstetricians and Gynecologists. ACOG Committee Opinion No. 315: obesity in
pregnancy. Obstet Gynecol. 2005;106:671–675.
36. Brodsky JB, Lemmens HJ, Brock-Utne JG, et al. Anesthetic considerations for bariatric surgery:
proper positioning is important for laryngoscopy. Anesth Analg. 2003;96:1841–1842.
37. Hawkins JL. Anesthesia-related maternal mortality. Clin Obstet Gynecol. 2003;46:679–687.
38. Awan R, Nolan JP, Cook TM. Use of a ProSeal laryngeal mask airway for airway maintenance during
emergency caesarean section after failed tracheal intubation. Br J Anaesth. 2004;92:144–146.
39. Morgan PJ, Pittini R, Regehr G, et al. Evaluating teamwork in a simulated obstetric environment.
Anesthesiology. 2007;106:907–915.
40. American College of Obstetricians and Gynecologists. ACOG Committee Opinion No. 104:
anesthesia for emergency deliveries. Int J Gynaecol Obstet. 1992;39:148.
41. Gaiser RR, McGonigal ET, Litts P, et al. Obstetricians’ ability to assess the airway. Obstet Gynecol.
1999;93:648–652.
42. Wong CA. Epidural and spinal analgesia/anesthesia for labor and vaginal delivery. In: Chestnut DH,
ed. Chestnut’s Obstetric Anesthesia Principles and Practice. 4th ed. Philadelphia, PA: Mosby Elsevier;
2009:429.
43. Bloom SL, Spong CY, Weiner SJ, et al. Complications of anesthesia for cesarean delivery. Obstet
Gynecol. 2005;106:281–287.
44. Dadarkar P, Philip J, Weidner C, et al. Spinal anesthesia for cesarean section following inadequate
labor epidural analgesia: a retrospective audit. Int J Obstet Anesth. 2004;13:239–243.
45. Portnoy D, Vadhera RB. Mechanisms and management of an incomplete epidural block for cesarean
section. Anesthesiol Clin North America. 2003;21:39–57.
46. Wheeler M, Ovassapian A. Fiberoptic endoscopy-aided techniques. In: Hagberg C, ed. Benumof’s
Airway Management. 2nd ed. Philadelphia, PA: Mosby Elsevier; 2007:399–438.
47. Ovassapian A, Krejcie TC, Yelich SJ, et al. Awake fibreoptic intubation in the patient at high risk of
aspiration. Br J Anaesth. 1989;62:13–16.
48. Trevisan P. Fibre-optic awake intubation for caesarean section in a parturient with predicted difficult
airway. Minerva Anesthesiol. 2002;68:775–781.
49. Doyle DJ. Awake intubation using the Glidescope video laryngoscope: initial experience in four cases.
Can J Anaesth. 2004;51:520–521.
50. Jakushenko N, Kopeika U, Nagobade D, et al. Comparison of awake endotracheal intubation with
GlideScope videolaryngoscope and fibreoptic bronchoscope in patients with difficult airway. Eur J
Anaesth. 2010;27:264.
51. Suresh MS, Wali A, Crosby ET. Difficult and failed intubation: strategies, prevention and management
of airway-related catastrophes in obstetrical patients. In: Suresh MS, Segal BS, Preston RL, et al., eds.
Shnider and Levison’s Anesthesia for Obstetrics. 5th ed. Philadelphia, PA: Lippincott Williams &
Wilkins; 2013:363–403.
52. Kilicaslan A, Topal A, Tavlan A, et al. Effectiveness of the C-MAC video laryngoscope in the
management of unexpected failed intubations. Braz J Anesthesiol. 2014;64:62–65.
53. Dhonneur G, Ndoko S, Amathieu R, et al. Tracheal intubation using the Airtraq in morbid obese
patients undergoing emergency cesarean delivery. Anesthesiology. 2007;106:629–630.
54. Han TH, Brimacombe J, Lee EJ, et al. The laryngeal mask airway is effective (and probably safe) in
selected healthy parturients for elective cesarean section: a prospective study of 1067 cases. Can J
Anaesth. 2001;48:1117–1121.
55. Cook TM, Brooks TS, Van der Westhuizen J, et al. The ProSeal LMA is a useful rescue device during
failed rapid sequence intubation: two additional cases. Can J Anaesth. 2005;52:630–633.
56. Keller C, Brimacombe J, Lirk P, et al. Failed obstetric tracheal intubation and postoperative respiratory
support with the ProSeal laryngeal mask airway. Anesth Analg. 2004;98:1467–1470.
57. Sharma B, Sahai C, Sood J, et al. The ProSeal laryngeal mask airway in two failed obstetric tracheal
intubation scenarios. Int J Obstet Anesth. 2006;15:338–339.
58. Brown NI, Mack PF, Mitera DM, et al. Use of the ProSeal laryngeal mask airway in a pregnant patient
with a difficult airway during electroconvulsive therapy. Br J Anaesth. 2003;91:752–754.
59. González GG, Marenco de la Fuente ML, Cornejo MB. Fastrach mask to resolve a difficult airway
during emergency cesarean section [in Spanish]. Rev Esp Anestesiol Reanim. 2005;52:56–57.
60. Minville V, N’guyen L, Coustet B, et al. Difficult airway in obstetric using Ilma-Fastrach. Anesth
Analg. 2004;99:1873.
61. Wissler RN. The esophageal-tracheal combitube. Anesthesiol Rev. 1993;20:147–152.
62. Dorges V, Ocker H, Wenzel V, et al. The laryngeal tube: a new simple airway device. Anesth Analg.
2000;90:1220–1222.
63. Zand F, Amini A. Use of the laryngeal tube-S for airway management and prevention of aspiration
after a failed tracheal intubation in a parturient. Anesthesiology. 2005;102:481–483.
64. Lipman S, Cohen S, Einav S, et al. The Society for Obstetric Anesthesia and Perinatology consensus
statement on the management of cardiac arrest in pregnancy. Anesth Analg. 2014;118:1003–1016.
65. Wong DT, Prabhu AJ, Coloma M, et al. What is the minimum training required for successful
cricothyroidotomy?: a study in mannequins. Anesthesiology. 2003;98:349–353.
66. Vadodaria BS, Gandhi SD, McIndoe AK. Comparison of four different emergency airway access
equipment sets on a human patient simulator. Anaesthesia. 2004;59:73–79.
67. DeLaurier GA, Hawkins ML, Treat RC, et al. Acute airway management: role of cricothyroidotomy.
Am Surg. 1990;56: 12–15.
68. Brofeldt BT, Panacek EA, Richards JR. An easy cricothyrotomy approach: the rapid four-step
technique. Acad Emerg Med. 1996;3:1060–1063.
69. Morris A, Lockey D, Coats T. Fat necks: modification of a standard surgical airway protocol in the
pre-hospital environmental. Resuscitation. 1997;35:253–254.
70. Gambling DR, Shay DC. The mother of all breathing problems. J Clin Anesth. 2003;15:491–494.
71. Baker DP, Day R, Salas E. Teamwork as an essential component of high-reliability organizations.
Health Serv Res. 2006;41:1576–1598.
Anesthesia for Multiple Gestation and Breech
Presentation
Carolyn F. Weiniger
ANESTHESIA FOR MULTIPLE GESTATION
I. Introduction
II. National Guidelines
A. Practice guidelines for obstetric anesthesia: an updated report
by the American Society of Anesthesiologists Task Force on
Obstetric Anesthesia
B. American College of Obstetricians and Gynecologists 2014
Practice Bulletin 144: multiple gestation—complicated twin,
triplet, and high-order multifetal pregnancy
C. American College of Obstetricians and Gynecologists 2013
Committee Opinion 560: medically indicated late-preterm and
early-term deliveries
D. American College of Obstetricians and Gynecologists 2013
Committee Opinion 573: magnesium sulfate use in obstetrics
III. Maternal adaptation to multiple gestation pregnancy
A. Cardiovascular system
B. Respiratory system
C. Central nervous system
D. Hematologic changes
IV. Obstetric conditions and concerns for multiple gestation pregnancy
A. Preeclampsia
B. Maternal hemorrhage
C. Fetal surgery
D. Maternal morbidity
E. Other concerns
V. Timing of delivery for multiple gestation pregnancy
A. Overview
B. Medically indicated delivery timing
C. Preventing multiple gestation preterm birth
VI. Preterm birth in multiple gestation pregnancy
A. Predicting preterm birth
B. Interventions to prevent preterm birth
C. Pharmacologic therapies to prevent preterm birth
D. Anesthesia for multiple gestation preterm birth
VII. Delivery route for multiple gestation pregnancy
A. Overview
B. Delivery route
C. Cephalic-cephalic presenting twins
D. Cephalic-noncephalic presenting twins
VIII. Anesthesia for vaginal delivery in multiple gestation pregnancy
A. Labor patterns
B. Unplanned operative delivery
IX. Anesthesia for cesarean delivery in multiple gestation pregnancy
A. Cesarean delivery
B. Unplanned cesarean delivery
X. Pharmacologic therapies for multiple gestation pregnancy
A. Corticosteroids
B. Magnesium sulfate
XI. Costs of multiple gestation pregnancy
ANESTHESIA FOR BREECH PRESENTATION
I. Introduction
II. American College of Obstetricians and Gynecologists Committee
Opinion 2006: breech presentation management
III. Historical perspective for breech presentation management
IV. Demographics for breech presentation
A. Incidence
B. Risk factors
V. Obstetric management for breech presentation
A. Historical perspective
B. Vaginal breech delivery
C. Anesthesia for vaginal breech delivery
D. Uterine relaxation for vaginal breech delivery
VI. Complications of vaginal breech delivery
A. Preterm breech delivery
B. Umbilical cord prolapse
C. Fetal head entrapment
D. Anesthesia for fetal head entrapment
E. Emergent cesarean delivery (Zavanelli maneuver)
F. Emergent anesthesia for breech cesarean delivery
VII. Cesarean delivery for breech presentation
A. Overview
B. Anesthesia for planned breech cesarean delivery
C. Risks
D. Anesthesia for urgent cesarean delivery
VIII. External cephalic version for breech presentation
A. Overview
B. Maternal and fetal indications for external cephalic version
C. Utilization of external cephalic version
D. Success of external cephalic version
E. Risks of external cephalic version
IX. Labor following external cephalic version
A. Successful vaginal delivery
B. Neonatal outcomes following external cephalic version
C. Labor augmentation
X. Anesthesia for external cephalic version in breech presentation
A. Overview
B. Anesthesia and external cephalic version success
C. Conversion of neuraxial technique for subsequent use
D. Emergency preparations
XI. Costs for external cephalic version
XII. Neonatal resuscitation in breech presentation delivery
KEYPOINTS
1. Multiple gestation pregnancies have become considerably more frequent
in past decades, although the twin pregnancy rate appears to have peaked,
and non-twin multiple gestations are decreasing. Multiple gestation
delivery is high risk, associated with cesarean delivery (CD), preterm
birth, and an increased incidence of concurrent medical and obstetric
conditions, such as preeclampsia.
2. Postpartum hemorrhage should be anticipated due to the distended uterus
in multiple gestation pregnancy. Protocols can guide management of this
potentially life-threatening disaster.
3. The majority of twin pregnancies will deliver before 37 weeks. Multiple
gestation is associated with an increased incidence of fetal
malpresentation and congenital anomalies. These factors may impact
delivery route choice.
4. Perinatal mortality is significantly increased following vaginal breech
delivery, and CD is the prevalent delivery mode for breech presentation.
5. Women should be offered external cephalic version (ECV) if possible,
and anesthesia may increase the success of ECV, in turn enabling vaginal
delivery.
A. Cardiovascular system
Many of the cardiovascular changes associated with singleton
pregnancy have been described in the past decade using
echocardiography. Cardiac output increases with gestational age
along with stroke volume; these changes are more dramatic than
with singleton pregnancy. Cardiac output is 20% greater—a
consequence of exaggerated responses of stroke volume and heart
rate.11 Preload is further increased above singleton pregnancy
after 20 weeks. Aortocaval compression, seen from 20 weeks in
singleton pregnancy, is exaggerated in twin pregnancy and may
present earlier.12,13 Preeclampsia is more common in multiple
gestations pregnancies.11,14 Gestational hypertension presenting
during twin pregnancy progresses more frequently to
preeclampsia than in singleton pregnancy.15
B. Respiratory system
The greater abdominal size with multiple gestation pregnancies
compounds respiratory changes and may cause dyspnea.16
However, respiratory function does not appear to differ markedly
compared to singleton pregnancies.17 Nevertheless, the enlarged
abdomen and elevated serum progesterone levels may impact
respiration in multiple gestation pregnancy.18
C. Central nervous system
The mechanism for enhanced effect of spinal drug in multiple
gestation pregnancy may be related to aortocaval compression.12
Spinal anesthesia drug spread is greater compared to singleton
pregnancy and is reportedly two dermatomes higher. Another
explanation relates to higher cerebrospinal fluid progesterone
levels that contribute to a neural sensitizing effect with lower
doses achieving a denser block.19 Hemodynamic instability after
spinal anesthesia is not greater in multiple gestation pregnancies,
as expressed by vasopressor requirements during CD.20 Spinal
anesthesia has been reported to be safe in triplet21 and quadruplet
pregnancies.22
D. Hematologic changes
Blood volumes increase more in twin pregnancy compared with
singletons.23 Morbidity and mortality are increased in multiple
gestation pregnancy and in an advanced age population.24 Aside
from thrombocytopenia associated with HELLP syndrome,
multiple gestation pregnancies frequently feature gestational
thrombocytopenia. Morikawa et al.25 reported a count below
100,000 in 43% of triplet pregnancies compared with 4.3% for
twin pregnancy. Triplet pregnancies are associated with further
increased risk for HELLP and acute fatty liver disease compared
with twin pregnancies. Regarding platelet counts that may impact
neuraxial anesthesia, 3.2% of women carrying triplets had platelet
counts <70,000 per mcL,26 and thrombocytopenia frequently
preceded the diagnosis of preeclampsia.
IV. Obstetric conditions and concerns for multiple gestation
pregnancy
Complications of multiple gestation pregnancy are listed in Table
15.2.
A. Preeclampsia
Hypertension is more frequent in twin pregnancy than singletons.
In a perinatal database case-control study from 1982 to 1987, the
likelihood of hypertension was 2.5 times higher among 1,253
twin versus 5,119 singleton pregnancies (odds ratio [OR], 2.5;
95% confidence interval [CI], 2.1 to 3.1).6 Preeclampsia may be
more prevalent in multiple gestation pregnancies. Foo et al.15
reported that multiple gestation pregnancies with de novo
hypertensive disorders were more likely to progress to
preeclampsia than singletons (24 per 70 [34%] vs. 279 per 1,881
[15%], P < .001). A similar finding was reported for progression
of chronic hypertension to preeclampsia in twins (53%) versus
singletons (18%), P < .01. The initial hypertensive-related
disorder diagnosed in twin pregnancies was more likely to be
preeclampsia rather than gestational hypertension.
B. Maternal hemorrhage
1. Multiple gestation pregnancy has a greater blood loss (>900
mL)—similar to that seen in CD and double that seen
following singleton delivery.23 Placental abruption is three
times more likely among twin pregnancies compared with
singletons (OR, 3.0; 95% CI, 1.9 to 4.7) and the frequency is
more apparent after adjustment for maternal age and weight
gain (OR, 3.7; 95% CI, 2.6 to 3.5).6
2. The rate of postpartum hemorrhage (PPH) is doubled in
multiple gestation pregnancies and occurs in approximately
9% of deliveries, in part due to the increased abruption rate.27
The distended uterus may also become atonic. Independent
risk factors for PPH in twin pregnancies were reported in a 9-
year Japanese cohort. When the gestational age was >41
weeks, the risk of PPH was over 8 times more likely
compared to a singleton pregnancy. Hypertensive disorders
were over 5 times more likely to be associated with PPH.28
3. PPH protocols can guide management of this potentially life-
threatening event. PPH should be anticipated in multiple
gestation deliveries.29 Airway considerations, readily
available equipment (e.g., intravenous [IV] access, rapid
infusers, warming blankets), massive transfusion protocols,
uterotonics (e.g., oxytocin, Methergine, misoprostol, and
Hemabate), and regular protocol drills are all important.30,31
REFERENCES
1. Martin JA, Hamilton BE, Osterman MJ. Three decades of twin births in the United States, 1980-2009.
NCHS Data Brief. 2012:1–8.
2. Lee YM. Delivery of twins. Semin Perinatol. 2012;36:195–200.
3. Martin JA, Hamilton BE, Sutton PD, et al. Births: final data for 2002. Natl Vital Stat Rep. 2003;52:1–
113.
4. Martin JA, Hamilton BE, Sutton PD, et al. Births: final data for 2005. Natl Vital Stat Rep. 2007;56:1–
103.
5. Martin JA, Hamilton BE, Osterman MJ, et al. Births: final data for 2012. Natl Vital Stat Rep.
2013;62:1–68.
6. Spellacy WN, Handler A, Ferre CD. A case-control study of 1253 twin pregnancies from a 1982-1987
perinatal data base. Obstet Gynecol. 1990;75:168–171.
7. American Society of Anesthesiologists Task Force on Obstetric Anesthesia. Practice guidelines for
obstetric anesthesia: an updated report by the American Society of Anesthesiologists Task Force on
Obstetric Anesthesia. Anesthesiology. 2007;106:843–863.
8. American College of Obstetricians and Gynecologists, Society for Maternal-Fetal Medicine. ACOG
Practice Bulletin No. 144: multifetal gestations: twin, triplet, and higher-order multifetal pregnancies.
Obstet Gynecol. 2014;123:1118–1132.
9. American College of Obstetricians and Gynecologists. ACOG Committee Opinion No. 560: medically
indicated late-preterm and early-term deliveries. Obstet Gynecol. 2013;121:908–910.
10. American College of Obstetricians and Gynecologists Committee on Obstetric Practice. ACOG
Committee Opinion No. 573: magnesium sulfate use in obstetrics. Obstet Gynecol. 2013;122:727–
728.
11. Kametas NA, McAuliffe F, Krampl E, et al. Maternal cardiac function in twin pregnancy. Obstet
Gynecol. 2003;102:806–815.
12. Barclay DL, Renegar OJ, Nelson EW Jr. The influence of inferior vena cava compression on the level
of spinal anesthesia. Am J Obstet Gynecol. 1968;101:792–800.
13. Kim YI, Chandra P, Marx GF. Successful management of severe aortocaval compression in twin
pregnancy. Obstet Gynecol. 1975;46:362–364.
14. Coonrod DV, Hickok DE, Zhu K, et al. Risk factors for preeclampsia in twin pregnancies: a
population-based cohort study. Obstet Gynecol. 1995;85:645–650.
15. Foo JY, Mangos GJ, Brown MA. Characteristics of hypertensive disorders in twin versus singleton
pregnancies. Pregnancy Hypertens. 2013;3:3–9.
16. Milne JA, Howie AD, Pack AI. Dyspnoea during normal pregnancy. BJOG. 1978;85:260–263.
17. McAuliffe F, Kametas N, Costello J, et al. Respiratory function in singleton and twin pregnancy.
BJOG. 2002;109:765–769.
18. Craft JB, Levinson G, Shnider SM. Anaesthetic considerations in caesarean section for quadruplets.
Can Anaesth Soc J. 1978;25:236–239.
19. Hirabayashi Y, Shimizu R, Saitoh K, et al. Cerebrospinal fluid progesterone in pregnant women. Br J
Anaesth. 1995;75:683–687.
20. Ngan Kee WD, Khaw KS, Ng FF, et al. A prospective comparison of vasopressor requirement and
hemodynamic changes during spinal anesthesia for cesarean delivery in patients with multiple
gestation versus singleton pregnancy. Anesth Analg. 2007;104:407–411.
21. Marino T, Goudas LC, Steinbok V, et al. The anesthetic management of triplet cesarean delivery: a
retrospective case series of maternal outcomes. Anesth Analg. 2001;93:991–995.
22. Elliott JP, Radin TG. Quadruplet pregnancy: contemporary management and outcome. Obstet
Gynecol. 1992;80:421–424.
23. Pritchard JA. Changes in the blood volume during pregnancy and delivery. Anesthesiology.
1965;26:393–399.
24. Mhyre JM. Maternal mortality. Curr Opin Anaesthesiol. 2012;25:277–285.
25. Morikawa M, Yamada T, Kataoka S, et al. Changes in antithrombin activity and platelet counts in the
late stage of twin and triplet pregnancies. Semin Thromb Hemost. 2005;31:290–296.
26. Al-Kouatly HB, Chasen ST, Kalish RB, et al. Causes of thrombocytopenia in triplet gestations. Am J
Obstet Gynecol. 2003;189:177–180.
27. Ananth CV, Demissie K, Smulian JC, et al. Placenta previa in singleton and twin births in the United
States, 1989 through 1998: a comparison of risk factor profiles and associated conditions. Am J Obstet
Gynecol. 2003;188:275–281.
28. Suzuki S, Hiraizumi Y, Miyake H. Risk factors for postpartum hemorrhage requiring transfusion in
cesarean deliveries for Japanese twins: comparison with those for singletons. Arch Gynecol Obstet.
2012;286:1363–1367.
29. Shields LE, Wiesner S, Fulton J, et al. Comprehensive maternal hemorrhage protocols reduce the use
of blood products and improve patient safety. Am J Obstet Gynecol. 2015;212:272–280.
30. Einerson BD, Miller ES, Grobman WA. Does a postpartum hemorrhage patient safety program result
in sustained changes in management and outcomes? Am J Obstet Gynecol. 2015;212:140–144.
31. Daniels K, Clark A, Lipman S, et al. Multidisciplinary simulation drills improve efficiency of
emergency medication retrieval. Obstet Gynecol. 2014;123(suppl 1):143s–144s.
32. D’Alton ME, Simpson LL. Syndromes in twins. Semin Perinatol. 1995;19:375–386.
33. Chang YL, Chao AS, Chang SD, et al. Mirror syndrome after fetoscopic laser therapy for twin-twin
transfusion syndrome due to transient donor hydrops that resolved before delivery. A case report. J
Reprod Med. 2014;59:90–92.
34. Tayler E, DeSimone C. Anesthetic management of maternal Mirror syndrome. Int J Obstet Anesth.
2014;23:386–389.
35. Baud D, Windrim R, Keunen J, et al. Fetoscopic laser therapy for twin-twin transfusion syndrome
before 17 and after 26 weeks’ gestation. Am J Obstet Gynecol. 2013;208:197.e1–e7.
36. Wohlmuth C, Tulzer G, Arzt W, et al. Maternal aspects of fetal cardiac intervention. Ultrasound
Obstet Gynecol. 2014;44:532–537.
37. Haas J, Hourvitz A, Dor J, et al. Perinatal outcome of twin pregnancies after early transvaginal
multifetal pregnancy reduction. Fertil Steril. 2014;101:1344–1348.
38. Hogan MC, Foreman KJ, Naghavi M, et al. Maternal mortality for 181 countries, 1980-2008: a
systematic analysis of progress towards Millennium Development Goal 5. Lancet. 2010;375:1609–
1623.
39. Lang CT, King JC. Maternal mortality in the United States. Best Pract Res Clin Obstet Gynaecol.
2008;22:517–531.
40. Martin JA, Hamilton BE, Sutton PD, et al. Births: final data for 2008. Natl Vital Stat Rep. 2010;59:1,
3–71.
41. Dodd JM, Crowther CA, Haslam RR, et al. Elective birth at 37 weeks of gestation versus standard care
for women with an uncomplicated twin pregnancy at term: the twins timing of birth randomised trial.
BJOG. 2012;119:964–973.
42. Barrett JF. Twin delivery: method, timing and conduct. Best Pract Res Clin Obstet Gynaecol.
2014;28:327–338.
43. Desai N, Lewis D, Sunday S, et al. Current antenatal management of monoamniotic twins: a survey of
maternal-fetal medicine specialists. J Matern Fetal Neonatal Med. 2012;25:1913–1916.
44. Durnwald CP, Momirova V, Rouse DJ, et al. Second trimester cervical length and risk of preterm birth
in women with twin gestations treated with 17-α hydroxyprogesterone caproate. J Matern Fetal
Neonatal Med. 2010;23:1360–1364.
45. Norman JE, Mackenzie F, Owen P, et al. Progesterone for the prevention of preterm birth in twin
pregnancy (STOPPIT): a randomised, double-blind, placebo-controlled study and meta-analysis.
Lancet. 2009;373:2034–2040.
46. Berghella V, Odibo AO, To MS, et al. Cerclage for short cervix on ultrasonography: meta-analysis of
trials using individual patient-level data. Obstet Gynecol. 2005;106:181–189.
47. American College of Obstetricians and Gynecologists, Committee on Practice Bulletins—Obstetrics.
ACOG Practice Bulletin No. 127: management of preterm labor. Obstet Gynecol. 2012;119:1308–
1317.
48. Vogel JP, Nardin JM, Dowswell T, et al. Combination of tocolytic agents for inhibiting preterm labour.
Cochrane Database Syst Rev. 2014;(7):CD006169.
49. Marks RJ, De Chazal RC. Ritodrine-induced pulmonary oedema in labour. Successful management
using epidural anaesthesia. Anaesthesia. 1984;39:1012–1014.
50. Bain ES, Middleton PF, Yelland LN, et al. Maternal adverse effects with different loading infusion
rates of antenatal magnesium sulphate for preterm fetal neuroprotection: the IRIS randomised trial.
BJOG. 2014;121:595–603.
51. Carvalho B, Saxena A, Butwick A, et al. Vaginal twin delivery: a survey and review of location,
anesthesia coverage and interventions. Int J Obstet Anesth. 2008;17:212–216.
52. Conde-Agudelo A, Belizán JM, Lindmark G. Maternal morbidity and mortality associated with
multiple gestations. Obstet Gynecol. 2000;95:899–904.
53. Chasen ST, Spiro SJ, Kalish RB, et al. Changes in fetal presentation in twin pregnancies. J Matern
Fetal Neonatal Med. 2005;17:45–48.
54. Yang Q, Wen SW, Chen Y, et al. Occurrence and clinical predictors of operative delivery for the vertex
second twin after normal vaginal delivery of the first twin. Am J Obstet Gynecol. 2005;192:178–184.
55. Rossi AC, Mullin PM, Chmait RH. Neonatal outcomes of twins according to birth order, presentation
and mode of delivery: a systematic review and meta-analysis. BJOG. 2011;118:523–532.
56. Wen SW, Fung KF, Oppenheimer L, et al. Occurrence and predictors of cesarean delivery for the
second twin after vaginal delivery of the first twin. Obstet Gynecol. 2004;103:413–419.
57. Barrett JF, Hannah ME, Hutton EK, et al. A randomized trial of planned cesarean or vaginal delivery
for twin pregnancy. N Engl J Med. 2013;369:1295–1305.
58. Leftwich HK, Zaki MN, Wilkins I, et al. Labor patterns in twin gestations. Am J Obstet Gynecol.
2013;209:254.e1–e5.
59. Pallasmaa N, Alanen A, Ekblad U, et al. Variation in cesarean section rates is not related to maternal
and neonatal outcomes. Acta Obstet Gynecol Scand. 2013;92:1168–1174.
60. Nicholls K, Ayers S. Childbirth-related post-traumatic stress disorder in couples: a qualitative study.
Br J Health Psychol. 2007;12:491–509.
61. Wanderer JP, Leffert LR, Mhyre JM, et al. Epidemiology of obstetric-related ICU admissions in
Maryland: 1999-2008. Crit Care Med. 2013;41:1844–1852.
62. Kuklina EV, Meikle SF, Jamieson DJ, et al. Severe obstetric morbidity in the United States: 1998-
2005. Obstet Gynecol. 2009;113:293–299.
63. Kathirgamanathan A, Douglas MJ, Tyler J, et al. Speed of spinal vs general anaesthesia for category-1
caesarean section: a simulation and clinical observation-based study. Anaesthesia. 2013;68:753–759.
64. Bauer ME, Kountanis JA, Tsen LC, et al. Risk factors for failed conversion of labor epidural analgesia
to cesarean delivery anesthesia: a systematic review and meta-analysis of observational trials. Int J
Obstet Anesth. 2012;21:294–309.
65. Hawkins JL, Chang J, Palmer SK, et al. Anesthesia-related maternal mortality in the United States:
1979-2002. Obstet Gynecol. 2011;117:69–74.
66. Leveno KJ, Quirk JG, Whalley PJ, et al. Fetal lung maturation in twin gestation. Am J Obstet Gynecol.
1984;148:405–411.
67. Fruscalzo A, Londero AP, Salvador S, et al. New and old predictive factors for breech presentation:
our experience in 14 433 singleton pregnancies and a literature review. J Matern Fetal Neonatal Med.
2014;27:167–172.
68. Hill LM. Prevalence of breech presentation by gestational age. Am J Perinatol. 1990;7:92–93.
69. American College of Obstetricians and Gynecologists Committee on Obstetric Practice. ACOG
Committee Opinion No. 340. Mode of term singleton breech delivery. Obstet Gynecol. 2006;108:235–
237.
70. Siegel IA, McNally HB. Primary breech presentation and external cephalic version; the management
of 308 primary breech presentations. South Med J. 1951;44:942–950.
71. Hannah ME, Hannah WJ, Hewson SA, et al. Planned caesarean section versus planned vaginal birth
for breech presentation at term: a randomised multicentre trial. Lancet. 2000;356:1375–1383.
72. Committee on Obstetric Practice. ACOG Committee Opinion: Number 265, December 2001. Mode of
term single breech delivery. Obstet Gynecol. 2001;98:1189–1190.
73. van Roosmalen J, Meguid T. The dilemma of vaginal breech delivery worldwide. Lancet.
2014;383:1863–1864.
74. Azria E, Le Meaux JP, Khoshnood B, et al. Factors associated with adverse perinatal outcomes for
term breech fetuses with planned vaginal delivery. Am J Obstet Gynecol. 2012;207:285.e1–e9.
75. Eller DP, VanDorsten JP. Route of delivery for the breech presentation: a conundrum. Am J Obstet
Gynecol. 1995;173:393–398.
76. Beilin Y, Bernstein HH. Fetal malpresentation and multiple birth. In: MC Norris, ed. Obstetric
Anesthesia. 2nd ed. Philadelphia, PA: Lippincott Williams & Wilkins; 1999:666–673.
77. George RT, Singh N, Yentis SM. External cephalic version—the bad, the good and the what now? Int
J Obstet Anesth. 2014;23:4–7.
78. Hall JE, Kohl S. Breech presentation: a study of 1,456 cases. Am J Obstet Gynecol. 1956;72:977–990.
79. Bragg F, Cromwell DA, Edozien LC, et al. Variation in rates of caesarean section among English NHS
trusts after accounting for maternal and clinical risk: cross sectional study. Br Med J. 2010;341:c5065.
80. Caughey AB, Cahill AG, Guise JM, et al. Safe prevention of the primary cesarean delivery. Am J
Obstet Gynecol. 2014;210:179–193.
81. Freeth D, MacVine JS. The value of external cephalic version under anaesthesia. Br Med J.
1951;2:881–884.
82. Crawford JS. Continuous lumbar epidural analgesia for labour and delivery. Br Med J. 1979;1:1560–
1561.
83. Breeson AJ, Kovacs GT, Pickles BG, et al. Extradural analgesia—the preferred method of analgesia
for vaginal breech delivery. Br J Anaesth. 1978;50:1227–1230.
84. Darby S, Hunter DJ. Extradural analgesia in labour when the breech presents. BJOG. 1976;83:35–38.
85. Cheng M, Hannah M. Breech delivery at term: a critical review of the literature. Obstet Gynecol.
1993;82:605–618.
86. Ginosar Y, Weiniger C, Elchalal U, et al. Emergency cesarean delivery for umbilical cord prolapse: the
head-down, knee-chest prone position for spinal anesthesia. Can J Anaesth. 2008;55:612–615.
87. Vinatier D, Dufour P, Bérard J. Utilization of intravenous nitroglycerin for obstetrical emergencies. Int
J Gynaecol Obstet. 1996;55:129–134.
88. Sandberg EC. The Zavanelli maneuver: 12 years of recorded experience. Obstet Gynecol.
1999;93:312–317.
89. Flamm BL, Newman LA, Thomas SJ, et al. Vaginal birth after cesarean delivery: results of a 5-year
multicenter collaborative study. Obstet Gynecol. 1990;76:750–754.
90. Weiner CP. Vaginal breech delivery in the 1990s. Clin Obstet Gynecol. 1992;35:559–569.
91. Royal College of Obstetricians and Gynaecologists. External Cephalic Version and Reducing the
Incidence of Breech Presentation (Guideline No. 20a). London, United Kingdom: Royal College of
Obstetricians and Gynaecologists; 2006.
92. Burgos J, Cobos P, Rodríguez L, et al. Is external cephalic version at term contraindicated in previous
caesarean section? A prospective comparative cohort study. BJOG. 2014;121:230–235.
93. Rosman AN, Guijt A, Vlemmix F, et al. Contraindications for external cephalic version in breech
position at term: a systematic review. Acta Obstet Gynecol Scand. 2013;92:137–142.
94. Vlemmix F, Kuitert M, Bais J, et al. Patient’s willingness to opt for external cephalic version. J
Psychosom Obstet Gynaecol. 2013;34:15–21.
95. Say R, Thomson R, Robson S, et al. A qualitative interview study exploring pregnant women’s and
health professionals’ attitudes to external cephalic version. BMC Pregnancy and Childbirth.
2013;13:4.
96. Maier B, Georgoulopoulos A, Zajc M, et al. Fetal outcome for infants in breech by method of
delivery: experiences with a stand-by service system of senior obstetricians and women’s choices of
mode of delivery. J Perinat Med. 2011;39:385–390.
97. Yogev Y, Horowitz E, Ben-Haroush A, et al. Changing attitudes toward mode of delivery and external
cephalic version in breech presentations. Int J Gynaecol Obstet. 2002;79:221–224.
98. Rosman AN, Vlemmix F, Beuckens A, et al. Facilitators and barriers to external cephalic version for
breech presentation at term among health care providers in the Netherlands: a quantitative analysis.
Midwifery. 2014;30:e145–e150.
99. Collaris RJ, Oei SG. External cephalic version: a safe procedure? A systematic review of version-
related risks. Acta Obstet Gynecol Scand. 2004;83:511–518.
100. Gottvall T, Ginstman C. External cephalic version of non-cephalic presentation; is it worthwhile? Acta
Obstet Gynecol Scand. 2011;90:1443–1445.
101. De Hundt M, Vlemmix F, Kok M, et al. External validation of a prediction model for successful
external cephalic version. Am J Perinatol. 2012;29:231–236.
102. Burgos J, Cobos P, Rodriguez L, et al. Clinical score for the outcome of external cephalic version: a
two-phase prospective study. Aust N Z J Obstet Gynaecol. 2012;52:59–61.
103. Cluver C, Gyte GM, Sinclair M, et al. Interventions for helping to turn term breech babies to head first
presentation when using external cephalic version. Cochrane Database Syst Rev. 2015;(2):CD000184.
104. Zhang J, Bowes WA Jr, Fortney JA. Efficacy of external cephalic version: a review. Obstet Gynecol.
1993;82:306–312.
105. Clock C, Kurtzman J, White J, et al. Cesarean risk after successful external cephalic version: a
matched, retrospective analysis. J Perinatol. 2009;29:96–100.
106. de Hundt M, Velzel J, de Groot CJ, et al. Mode of delivery after successful external cephalic version: a
systematic review and meta-analysis. Obstet Gynecol. 2014;123:1327–1334.
107. Policiano C, Costa A, Valentim-Lourenço A, et al. Route of delivery following successful external
cephalic version. Int J Gynaecol Obstet. 2014;126:272–274.
108. Jain S, Mulligama C, Tagwira V, et al. Labour outcome of women with successful external cephalic
version: a prospective study. J Obstet Gynaecol. 2010;30:13–16.
109. Ben-Haroush A, Perri T, Bar J, et al. Mode of delivery following successful external cephalic version.
Am J Perinatol. 2002;19:355–360.
110. Balayla J, Dahdouh EM, Villeneuve S, et al. Obstetrical and neonatal outcomes following unsuccessful
external cephalic version: a stratified analysis amongst failures, successes, and controls. J Matern
Fetal Neonatal Med. 2015;28:605–610.
111. Suen SS, Khaw KS, Law LW, et al. The force applied to successfully turn a foetus during reattempts of
external cephalic version is substantially reduced when performed under spinal analgesia. J Matern
Fetal Neonatal Med. 2012;25:719–722.
112. Sultan P, Carvalho B. Neuraxial blockade for external cephalic version: a systematic review. Int J
Obstet Anesth. 2011;20:299–306.
113. Weiniger CF, Ginosar Y, Elchalal U, et al. Randomized controlled trial of external cephalic version in
term multiparae with or without spinal analgesia. Br J Anaesth. 2010;104:613–618.
114. Weiniger CF, Ginosar Y, Elchalal U, et al. External cephalic version for breech presentation with or
without spinal analgesia in nulliparous women at term: a randomized controlled trial. Obstet Gynecol.
2007;110:1343–1350.
115. Mancuso KM, Yancey MK, Murphy JA, et al. Epidural analgesia for cephalic version: a randomized
trial. Obstet Gynecol. 2000;95:648–651.
116. Schorr SJ, Speights SE, Ross EL, et al. A randomized trial of epidural anesthesia to improve external
cephalic version success. Am J Obstet Gynecol. 1997;177:1133–1137.
117. Sullivan JT, Grobman WA, Bauchat JR, et al. A randomized controlled trial of the effect of combined
spinal-epidural analgesia on the success of external cephalic version for breech presentation. Int J
Obstet Anesth. 2009;18:328–334.
118. Lavoie A, Guay J. Anesthetic dose neuraxial blockade increases the success rate of external fetal
version: a meta-analysis. Can J Anaesth. 2010;57:408–414.
119. Carvalho B, Tan JM, Macario A, et al. Brief report: a cost analysis of neuraxial anesthesia to facilitate
external cephalic version for breech fetal presentation. Anesth Analg. 2013;117:155–159.
120. Adams EK, Mauldin PD, Mauldin JG, et al. Determining cost savings from attempted cephalic version
in an inner city delivering population. Health Care Manag Sci. 2000;3:185–192.
Obstetric Emergencies
Michael Frölich and Brenda A. Bucklin
I. Nonreassuring fetal status
A. Pathophysiology
B. Diagnosis
C. Treatment
II. Peripartum bleeding
A. Management of obstetric hemorrhage
B. Antepartum hemorrhage
C. Postpartum hemorrhage
III. Intrapartum emergencies
A. Preterm labor and delivery
B. Shoulder dystocia
C. Umbilical cord compression/prolapse
D. Amniotic fluid embolism
KEYPOINTS
1. Management of the pregnant patient includes the assessment and
screening of pregnant patients, perioperative management, and
postoperative care as appropriate. When managing the obstetric patient
both maternal and fetal well-being must be considered.
2. Intrauterine asphyxia is an uncommon cause of newborn depression.
3. A neonate who has experienced hypoxia proximate to delivery severe
enough to result in hypoxic encephalopathy will demonstrate other signs
of hypoxic damage, such as a profound metabolic or mixed acidemia (pH
<7.00), a low Apgar score of 0 to 3 for more than 5 minutes, and
evidence of neonatal neurologic sequelae.
4. Obstetric emergencies contribute significantly to maternal morbidity and
mortality.
5. Recent developments in the management of medical emergencies
emphasize the value of a protocol-based team approach and regular
simulation-based rehearsal of management algorithms.
6. Obstetric blood loss is often underestimated. However, visual aids can be
used to objectively and subjectively improve estimation of blood loss.
7. “Never treat ‘postpartum hemorrhage’ without simultaneously pursuing
an actual clinical diagnosis” because postpartum hemorrhage is a
“clinical sign of an underlying condition that is amenable to diagnosis.”1
A. Pathophysiology
Epidemiologic studies have demonstrated that <20% of cases of
neonatal encephalopathy meet nonstringent criteria for
intrapartum hypoxia with another 10% demonstrating an
association with intrapartum hypoxia. These studies concluded
that approximately 70% of cases of neonatal encephalopathy
result from events arising before the onset of labor. Although it is
difficult to distinguish between birth asphyxia, newborn asphyxia,
and hypoxic-ischemic encephalopathy of the newborn, the overall
incidence of neonatal encephalopathy attributable to intrapartum
hypoxia in the absence of any other preconception or antepartum
abnormalities is estimated to be approximately 1.6 per 10,000.7
The ACOG has suggested abandoning the term birth asphyxia.2
1. Causes of fetal asphyxia. There are several mechanisms by
which a fetus can experience intrapartum asphyxia (see Table
16.1).
2. Presence of meconium
Meconium staining has often been associated with neonatal
depression. In a recent prospective cohort study, the presence
of meconium was associated with higher risk of the composite
morbidity.15 However, in most cases, the finding is not of
serious importance because it is not necessarily indicative of
intrauterine asphyxia. Historically, the presence of meconium-
stained amniotic fluid during labor was a potentially ominous
sign, but controversy exists over its relative impact on fetal
status. Data indicate that timing and quantity of meconium
passage are critical variables for assessing the significance of
this occurrence on fetal well-being. Presumably, these two
aspects of meconium passage correlate with the duration and
severity of the intrauterine insult. If meconium staining is
present in combination with an abnormal FHR tracing or
another risk factor (e.g., intrauterine growth restriction
[IUGR], postdates), there is an increased likelihood of
neonatal depression. More important, aspiration of
meconium-stained fluid can lead to serious neonatal
morbidity and mortality.
4. Doppler ultrasound
Doppler ultrasound is used to assess blood flow in the fetal
arterial and venous systems. It is as an important tool used in
antenatal surveillance for the management and timing of
delivery in pregnancies associated with IUGR. Evaluation of
the umbilical arteries provides information on placental
resistance. Placental resistance is elevated in uteroplacental
insufficiency. This leads to a reduction in forward umbilical
artery flow and is reflected by a relative reduction in diastolic
as compared to systolic velocities. Absent or reversed end
diastolic flow in the umbilical artery is known to be
associated with an 80-fold increased risk of perinatal
mortality.16 Neonatal intracranial volume and cerebral cortical
gray matter, as measured by magnetic resonance imaging
(MRI), were also reduced in infants born from pregnancies
complicated by fetal growth restriction and abnormal
umbilical artery Doppler studies compared with matched
infants of the same gestational age but with appropriate
intrauterine growth.17
The measurement of blood flow velocities in umbilical
and fetal cerebral arteries is a valuable tool in antepartum
surveillance.
C. Treatment
1. Obstetric management
Type II and type III FHR patterns warrant active medical
intervention. When there is an unresolved, prolonged severe
bradycardia (class III FHR pattern), emergency cesarean
delivery (CD) will be necessary to minimize fetal risk. The
management of a class II FHR pattern will depend on the
presence or absence of moderate variability or accelerations,
the response of the FHR to contractions and the progress and
stage of labor. With a category II FHR pattern, several
maneuvers can be utilized to resuscitate the fetus in utero (see
Table 16.2). Fetal O2 content and saturation can be improved
by administering supplemental O2 and by placing the mother
in the left lateral position to reduce compression of the vena
cava. Uterine stimulants (i.e., oxytocin infusion) should also
be discontinued. Tocolysis should be considered to achieve
uterine relaxation if the mother is hemodynamically stable
and there is no evidence of abruptio placentae. Terbutaline,
0.25 mg administered subcutaneously (SQ) or intravenously
(IV) has been found to be superior to placebo and to
magnesium sulfate in relieving decelerations. Sublingual
nitroglycerin spray can achieve more rapid uterine relaxation,
but its use may cause hypotension and maternal headache. IV
hydration should be considered if the volume status is in
question. Maternal hypotension resulting from neuraxial
anesthesia should also be corrected. A vaginal examination
should be performed to rule out rapid cervical change and
fetal descent as well as cord prolapse. If there is evidence of
cord compression by the presence of repetitive, moderate to
severe variable decelerations, amnioinfusion should be
considered. If the FHR abnormalities do not resolve with the
earlier mentioned measures, CD or operative vaginal delivery
is indicated. Maternal fever can lead to increased fetal oxygen
consumption and is associated with increased risk of poorer
neonatal outcome. Treatment with acetaminophen and cooling
measures should be instituted.
2. Anesthetic management
Anesthesia for CD of a distressed fetus must provide proper
operating conditions for the obstetrician within a short time
period without compromising maternal or fetal safety. The
obstetrician determines the timing and speed with which
the procedure must be performed. The choice of general,
spinal, or extension of an existing epidural anesthetic
depends on the clinical circumstances. Although general
anesthesia (GA) may be indicated in some cases of FHR
abnormality, the severity of the abnormality should be
considered before incurring risk associated with GA.18
a. Interpretation of the 30-minute rule
Clinicians have attempted to provide guidance on the time
frame when an emergent CD should be initiated. In 1982,
ACOG proposed a 15-minute interval between the time of
decision for a CD to its start. These guidelines, however,
were revised in 1989, and the 15-minute recommendation
was extended to 30 minutes.19
A 30-minute interval is reported to be adequate for
most CD. The Optimal Goals for Anesthesia Care in
Obstetrics emphasize the “Availability of anesthesia and
surgical personnel to permit the start of a cesarean
delivery within 30 minutes of the decision to perform the
procedure”20; in cases of vaginal birth after cesarean
(VBAC), appropriate facilities and personnel, including
obstetric anesthesia, nursing personnel, and a physician
capable of monitoring labor and performing CD,
immediately available during active labor to perform
emergency CD. “The definition of immediate availability
of personnel and facilities remains a local decision, based
on each institution’s available resources and geographic
location.”20 Although there are early reports of evidence
for increased risk of fetal loss if the “alarm to surgery”
interval exceeds 20 minutes21 and another report
suggested that permanent fetal CNS damage may begin
after approximately 10 minutes of fetal anoxia,22 most
cases of fetal asphyxia are not the result of fetal anoxia. In
cases where complete fetal anoxia is suspected, delivery
should occur as soon as possible. Because these
emergencies may occur at any time, the anesthesia
provider must always be prepared for emergent operative
delivery.
CLINICAL PEARL “Availability of anesthesia and surgical
personnel to permit the start of a cesarean delivery within 30
minutes of the decision to perform the procedure”; in cases of
VBAC, appropriate facilities and personnel, including obstetric
anesthesia, nursing personnel, and a physician capable of
monitoring labor and performing CD, immediately available during
active labor to perform emergency CD. “The definition of
immediate availability of personnel and facilities remains a local
decision, based on each institution’s available resources and
geographic location.”20
b. Anesthetic choice
Every woman admitted to labor and delivery has the
potential to develop an obstetric emergency as a result of
nonreassuring fetal status. When choosing an anesthetic,
the anesthesia provider must communicate with the
obstetrician to determine the urgency of the situation. “A
communication system should be in place to encourage
early and ongoing contact between obstetric providers,
anesthesiologists, and other members of the
multidisciplinary team.”23
Because most anesthesiologists and other anesthesia
providers are not experts in FHR monitoring, it is up to
the obstetrician to determine the severity of the FHR
abnormality. Emergency CD is performed when the
situation is life threatening for either the mother or fetus.
In cases of abnormal FHR patterns, emergency CD is
indicated for prolonged fetal bradycardia, late
decelerations without FHR variability, and cord prolapse
with bradycardia.
“The decision to use a particular anesthetic
technique for cesarean delivery should be
individualized, based on several factors. These include
anesthetic, obstetric, or fetal risk factors (e.g., elective vs.
emergency), the preferences of the patient, and the
judgment of the anesthesiologist. Neuraxial techniques
are preferred to GA for most cesarean deliveries. An
indwelling epidural catheter may provide equivalent onset
of anesthesia compared to initiation of spinal anesthesia
for urgent cesarean delivery. . . . However, GA may be
the most appropriate choice in some circumstances
(e.g., profound fetal bradycardia, ruptured uterus,
severe hemorrhage, severe placental abruption)”23 but
the risk of a difficult airway (and availability of
backup and advanced airway equipment) should
always be considered.
c. Diagnosis
The diagnosis of placenta previa has shifted from clinical
examination of the dilated cervix to sonographic
assessment of the closed internal os, resulting in
confusing terminology. Placenta previa is classified
according to the extent of encroachment of the placenta
(see Fig. 16.2):
(1) A low-lying placenta encroaches the lower segment
of the uterus but does not infringe on the cervical os.
(2) Marginal placenta touches but does not cover the top
of the cervix.
(3) Partial placenta previa partially covers the cervix.
(4) Complete placenta previa covers the top of the
cervix entirely.
Placenta previa often presents as painless bleeding in
the late second or early third trimester. All vaginal
bleeding in the third trimester should be considered to
be placenta previa until proven otherwise. Some
cases will not bleed until labor begins, but the
majority of cases are diagnosed during routine
ultrasonography in asymptomatic women, usually
during the second trimester. Transvaginal
ultrasonography appears to be superior to
transabdominal ultrasonography for diagnosis.
d. Obstetric management
Obstetric management depends on the degree of blood
loss as well as fetal maturity and status. In such cases, the
fetus is at risk for compromise because of placental
separation resulting in uteroplacental insufficiency or
premature delivery. CD is recommended if the placenta
reaches the cervical margin at time of delivery, and this
entity may be grouped with placenta previa.60 In these
patients, bleeding may stop spontaneously or be sudden
and severe. If the bleeding is ongoing, an urgent CD is
indicated. However, if bleeding has abated and the fetus is
preterm, obstetric management is “expectant” (i.e., bed
rest). There are reports of tocolytic administration in
women who have a preterm fetus and are contracting with
placenta previa. Judicious tocolytic administration is
reasonable if both mother and fetus are stable.61 Steroids
should be administered in women between 24 and 34
weeks of gestation, generally at the time of admission for
bleeding, to promote fetal lung maturation. In women
who have a history of CD or prior uterine surgery,
detailed ultrasonography should be performed to exclude
placenta accreta. As the fetus approaches “term,” fetal
maturity will be assessed (usually be amniocentesis) and
an elective CD will be performed when fetal lung
maturity is confirmed.
e. Anesthetic management
Anesthetic management is dependent on the urgency of
the situation as well as maternal hemodynamic (i.e.,
degree and rate of hemorrhage) status. If there is rapid
or massive hemorrhage, then GA is the most expedient
way to deliver the fetus and stabilize the mother. It may
not be possible to fully resuscitate the mother prior to
delivery because bleeding will continue until after
delivery of both the fetus and placenta (unless there is no
evidence of placenta accreta). The importance of adequate
IV access cannot be overemphasized in patients with
active bleeding. Flow through an IV cannula is directly
proportional to the fourth power of the radius and
inversely proportional to length. Consequently, one or
more short, large bore peripheral IV catheters are often
preferable to central venous access with longer catheters
(i.e., double-or triple-lumen catheters). An arterial line
can be extremely helpful during a hemorrhagic
emergency, both for beat-to-beat monitoring of BP and for
obtaining frequent laboratory tests.
The ASA Task Force on Obstetric Anesthesia and
the ACOG recommend that all facilities providing
obstetric care be prepared to manage hemorrhagic
emergencies.23 Hand-inflated pressure bags, an automatic
rapid infusion system, a fluid warmer, and a forced-air
warming device are recommended. Because all hospitals
have different capabilities, it is paramount to have
knowledge about blood bank resources for managing
hemorrhage. In cases where the bleeding has stopped
spontaneously, neuraxial anesthesia can be used after
careful assessment of maternal volume status (HR, BP,
urine output). Neuraxial anesthesia may decrease the
estimated blood loss.62 However, hypovolemia is a
relative contraindication to neuraxial anesthesia.
2. Vasa previa
Vasa previa, if undiagnosed, is associated with perinatal
mortality rates of approximately 60% because rupture of
fetal vessels can lead to exsanguination of the fetus and is
often disastrous for fetal well-being.63
a. Definition
Vasa previa occurs when the fetal vessels cross the
membranes ahead of the fetal presenting part.
Consequently, fetal vessels are unprotected by either the
placenta or umbilical cord, resulting in shearing of the
fetal vessels if rupture of membranes occurs.
b. Epidemiology
Vasa previa is a rare obstetric complication with an
estimated incidence of approximately 1 in 2,500
deliveries. Vasa previa is most commonly diagnosed when
vaginal bleeding and fetal bradycardia, or fetal death
accompany rupture of the membranes. Although vasa
previa does not endanger the mother’s life, achieving
optimal fetal outcomes depends on prenatal diagnosis and
appropriate management at the time of delivery.63
Advances in ultrasonography have made it possible to
diagnose vasa previa with reasonable accuracy. Such
diagnostic capabilities allow for proper delivery planning
and management.
c. Obstetric management
The primary obstetric treatment goal is fetal survival. A
ruptured vasa previa is a true obstetric emergency
requiring immediate CD. If diagnosed in early pregnancy,
these patients can be managed in an antenatal unit with
bed rest and frequent monitoring of fetal status. Elective
CD is then performed when fetal maturity has improved
(approximately 34 weeks estimated gestational age
[EGA]).
d. Anesthetic management
Anesthetic management is dependent on the urgency of
the situation. In cases of ruptured vasa previa, GA is often
required.
3. Abruptio placentae
a. Definition
Abruptio placentae (placental abruption) is defined as
premature separation of a normally implanted placenta
from the decidua basalis after 20 weeks of gestation and
prior to birth. During a normal delivery, maternal blood
loss is limited by constriction of the spiral arteries
following delivery of the placenta. In acute cases of
abruption, bleeding results from exposure of decidual
vessels because the uterus is unable to selectively
constrict the area of abruption. The separation can be a
complete or partial and associated with maternal
hemorrhage. The hemorrhage may be concealed or
present as vaginal bleeding. Loss of placental-uterine
surface area reduces gas exchange and can result in a
distressed fetus and/or fetal asphyxia. Many cases involve
a premature infant and at least half occur prior to the onset
of labor.
b. Epidemiology
Placental abruption occurs in approximately 1% of
pregnancies worldwide with a fetal mortality rate of up to
20% to 40% depending on the degree of separation.
Placental abruption can contribute to maternal mortality64
and may be implicated in up to 10% of preterm births.65
Perinatal mortality is estimated to be 12% among
pregnancies complicated by abruption.66 Risk factors
include (1) advanced maternal age, (2) multiparity, (3)
hypertension, (4) smoking, (5) trauma, (6) premature
rupture of membranes, (7) trauma, and (8) cocaine
abuse.67 Uterotonic drugs do not increase the risk of
abruptio placentae.68
c. Obstetric considerations
(1) Signs and symptoms
The classically described symptoms of placental
abruption are vaginal bleeding and abdominal pain.
Typically, there is uterine hypertonus associated with
frequent uterine contractions. The uterus is frequently
tender and tense to palpation. Breakthrough pain in a
patient with an otherwise effective labor epidural
may indicate acute abruption. Decidual bleeding with
subsequent hematoma formation leads to progressive
placental separation. Blood may also force its way
through the uterine wall into the serosa, a condition
known as Couvelaire uterus. Up to 90% of abruptions
are mild to moderate without fetal compromise,
maternal hypotension, or coagulopathy. However,
vaginal blood loss is often underestimated and
misleading. In some cases, as much as 3,000 mL of
blood can be sequestered behind the placenta (i.e.,
“concealed bleeding”) without evidence of vaginal
bleeding. Consequently, the amount of vaginal
bleeding can grossly underestimate the volume
status because of the concealed retroplacental
hematoma formation. The primary risk to the
mother is hypovolemic shock due to acute blood loss.
4. Uterine rupture
Uterine rupture is a potentially catastrophic complication for
mother and fetus. Even though it occurs most commonly in
patients with prior uterine scars (e.g., CD, myomectomy), the
overall risk is low.
a. Definition
Uterine rupture refers to separation of a uterine scar that is
clinically apparent and results in fetal distress and
maternal hemorrhage requiring emergency CD or
postpartum laparotomy. Uterine rupture has also been
reported in women with an unscarred uterus; it is
sometimes related to trauma.70
b. Epidemiology
Uterine rupture is a potentially catastrophic event in
which the integrity of the myometrial wall is breached.
However, in most patients with a prior uterine scar,
the risk of rupture is <1%. Defects may occur
antepartum, intrapartum, or postpartum. The most
common type of defect is uterine scar separation or
dehiscence. Most of these cases are asymptomatic and do
not result in maternal or fetal morbidity. However, actual
uterine rupture is a defect of the uterine wall that is
profound enough to cause fetal compromise and/or
maternal hemorrhage resulting in CD or laparotomy.
Conditions associated with uterine rupture include
(1) separation of a prior uterine scar (e.g., CD,
myomectomy), (2) rapid labor, (3) prolonged labor
associated with oxytocin infusion, (4) traumatic
rupture, (5) weakened uterine musculature (e.g., grand
multiparity, polyhydramnios, connective tissue
disorders), (6) excessive fundal pressure, and (7)
forceps delivery. However, the single most important risk
factor is prior uterine surgery. Rupture of a classical
uterine incision scar (i.e., vertical incision) is associated
with the greatest morbidity and mortality. Compared to a
low transverse uterine scar, the vertical scar involves a
more vascular area and is often the site of placental
implantation. A retrospective analysis of more than
20,000 deliveries in women with one prior CD
determined that the risk of uterine rupture was (1) 1.6 per
1,000 in nonlaboring women; (2) 5.2 per 1,000 in women
with spontaneous labor; (3) 7.7 per 1,000 in women
undergoing induction of labor; and (4) 24.5 per 1,000
among women undergoing a prostaglandin induction.71
Risk of rupture increases in patients undergoing an
induction of labor, particularly in the presence of an
unfavorable cervix or high fetal station. This practice is
discouraged by the ACOG.72 Maternal mortality is
increased in patients without a prior uterine scar or if
the rupture is traumatic.
c. Diagnosis
Although the diagnosis can be difficult because of
variable presentation, the most consistent clinical feature
of uterine rupture is fetal bradycardia in approximately
70% of patients.73,74 Other signs and symptoms may
include vaginal bleeding, severe abdominal pain, shoulder
pain, and/or hypotension. If rupture is diagnosed
intrapartum, prompt management is essential, and if there
is hypovolemia, fluid resuscitation should begin before
inducing GA and laparotomy. In more severe cases, the
fetus will already be expelled from the uterus into the
abdomen. This is a situation in which the fetus rarely
survives. Serious maternal and fetal morbidity and
mortality occur in 10% to 25% of cases of uterine
rupture.75 Maternal morbidity is associated with gravid
hysterectomy and a high rate of blood loss and
transfusion.
d. Obstetric management
Because most cases of uterine rupture are associated with
trial of labor after cesarean (TOLAC) attempts, this has
been a controversial area of obstetric practice. Despite an
increasing CD rate, the TOLAC rate has declined
substantially. The most recent ACOG Practice Bulletin on
TOLAC states, “A trial of labor after previous cesarean
delivery should be undertaken at facilities capable of
emergency deliveries.”76 The recommendations further
indicated that “TOLAC is not contraindicated for women
with previous cesarean delivery with an unknown uterine
scar type unless there is a high clinical suspicion of a
previous classical uterine incision.” The ASA Optimal
Goals are also consistent and state that “in cases of
TOLAC, appropriate facilities and personnel, including
obstetric anesthesia, nursing personnel, and a physician
capable of monitoring labor and performing CD,
immediately available during active labor to perform
emergency CD. The definition of immediate availability
of personnel and facilities remains a local decision, based
on each institution’s available resources and geographic
location.”77
When uterine rupture is an incidental finding after
vaginal delivery, the obstetrician must carefully monitor
the patient for concealed bleeding. Close observation is
appropriate in this circumstance. However, when uterine
rupture occurs during labor, explorative laparotomy and
hysterectomy may be required to manage bleeding. The
treatment must be individualized based on maternal and
fetal considerations.
e. Anesthetic considerations
In patients attempting TOLAC, the major signs of
intrapartum rupture are a change in uterine tone or
contraction pattern and FHR abnormalities. Epidural
analgesia is not contraindicated in patients attempting
TOLAC because epidural analgesia with low
concentrations of local anesthetic will not mask the
pain of uterine rupture. Success rates for TOLAC are
similar to women who do or do not receive epidural
analgesia.76 In these cases, the lowest concentration
possible of local anesthetic with opioid should be used to
ensure adequate analgesia. If the trial of labor is
unsuccessful and both mother and fetus are stable, the
epidural block may be extended and used for CD.
However, in a true emergency where the uterine scar is
ruptured and the fetus is distressed or there is maternal
cardiovascular instability, GA for emergent CD is
indicated. In such cases, invasive hemodynamic
monitoring, volume resuscitation, and transfusion may be
necessary. Hemorrhage may be uncontrollable and
hysterectomy is necessary.
C. Postpartum hemorrhage
PPH is often defined as the loss of more than 500 mL of blood
after vaginal delivery or 1,000 mL after CD within the first 24
hours following delivery. Severe hemorrhage is the primary cause
of maternal death worldwide78 with >50% of these deaths
occurring within 24 hours of delivery. In the United States, it is an
important contributor to maternal morbidity and mortality.79,80
Morbidity results in coagulopathy, shock, adult respiratory
distress syndrome, loss of fertility, and pituitary necrosis.46
Recently, Clark and Hankins1 stated to “ never treat ‘postpartum
hemorrhage’ without simultaneously pursuing an actual clinical
diagnosis” because PPH is a “clinical sign of an underlying
condition that is amenable to diagnosis.”
1. Uterine atony
a. Epidemiology
Uterine atony is the most common cause of PPH.79 It
complicates up to 10% of deliveries and is the most
common cause of serious obstetric hemorrhage. Although
rates of PPH from uterine atony are increasing, this is not
explained by changes in rates of CD, TOLAC, maternal
age, multiple births, hypertension, or diabetes mellitus.79
Uterine atony manifests as a noncontracted uterus with
severe bleeding. Risk factors include (1) rapid or
protracted delivery, (2) tocolysis, (3) overdistention of the
uterus (e.g., multiple gestations, macrosomia,
polyhydramnios), (4) high parity, (5) prolonged oxytocin
infusion (i.e., either induced labor or augmented labor),
(6) chorioamnionitis, (7) retained placenta, (8) operative
assisted vaginal delivery, (9) GA with volatile agents, and
(10) previous or current gestational trophoblastic disease,
(11) hypertensive disease, (12) diabetes, and (13)
advanced maternal age.
b. Medical management
The initial treatment of uterine atony is medical
management, including the active management of the
third stage of labor (i.e., oxytocin administration and
uterine massage). The ACOG recommends prophylactic
uterotonic administration to stimulate uterine contraction
and prevent uterine atony.34
(1) These uterotonic agents include:
(a) Oxytocin. Oxytocin is an effective first-line
treatment for PPH. Oxytocin is usually
administered by rapid IV infusion with 20 to 40
U added per 1 L normal saline (NS) or Ringer’s
lactate (LR) solution.81 Many practitioners avoid
bolus dosing because oxytocin is a systemic
vasodilator and may aggravate hypotension,
especially during severe hemorrhage.
(b) Methylergonovine. When oxytocin fails,
prostaglandins and ergot alkaloids are
considered second-and third-line treatments.
They enhance uterine contractility and cause
vasoconstriction. Methylergonovine
(Methergine) is an ergot alkaloid that causes
generalized smooth muscle contraction in which
the upper and lower segments of the uterus
contract simultaneously. Because ergot alkaloid
agents increase BP, they are relatively
contraindicated in women with preeclampsia or
hypertension. Other relative contraindications
include peripheral vascular disease and coronary
artery disease. Methergine should not be
administered IV, but intramuscularly (IM) at a
dose of 0.2 mg. Other adverse effects include
nausea, vomiting, and in rare cases, pulmonary
edema. Recent evidence suggests that the risk of
myocardial ischemia is negligible.82
(c) 15-Methyl prostaglandin F2α. The
prostaglandin most commonly used is 15-methyl
prostaglandin F2α, or carboprost (Hemabate). It
can be administered intramyometrially or IM in
a dose of 0.25 mg; this dose can be repeated
every 15 minutes for a total dose of up to 2 mg.
Carboprost has been proved to control
hemorrhage in up to 87% of patients and may
control hemorrhage when other medical
treatments fail.83 Despite the success of this
medication, it should be used cautiously in
asthmatic patients. It is known to cause
abnormal ventilation–perfusion ratios, increased
pulmonary shunt fraction, and hypoxemia.
Relative contraindications include hepatic,
cardiac, and renal disease. It is also known to
produce tachycardia, diarrhea, and fever.
(d) Misoprostol (PGE1, Cytotec). Prostaglandins
have been used for PPH when other measures
fail.84 Misoprostol, is an inexpensive
prostaglandin E1 analog and has been suggested
as an alternative for routine management of the
third stage of labor. When administered rectally
(800 to 1,000 μg), misoprostol has been effective
in treating PPH when other medical treatments
fail. There are few, if any, side effects.
Misoprostol does not require refrigeration for
storage, thus making it attractive for use in
underdeveloped countries.
c. Invasive therapy
Conservative therapy sometimes fails to treat uterine
atony.
(1) Uterine atony unresponsive to medical therapy is
treated with tamponade techniques (e.g., Sengstaken-
Blakemore tube, Bakri balloon) or surgical
procedures (e.g., uterine curettage, uterine artery
ligation, B-Lynch suture, hypogastric artery
ligation, hysterectomy),46 whereas fluid
resuscitation is initiated as necessary to prevent
obstetric shock. Invasive management is aimed at
preventing uterine atony by compressive methods
(i.e., uterus hemostatic suture by B-Lynch or uterine
Z-suture) or achieving selective uterine
devascularization (e.g., vascular ligation of uterine
arteries, ovarian arteries, and hypogastric arteries;
and/or selective transarterial embolization of uterine
arteries) to prevent hemorrhage.
(2) In some cases, arterial embolization is used to
reduce hemorrhage. It is a procedure that is available
in a number of centers that are staffed with
appropriately trained interventional radiologists. The
catheter is introduced via the femoral artery and is
advanced above the bifurcation of the aorta where the
bleeding point is identified by contrast injection. The
feeder artery is catheterized and embolized with
absorbable gelatin sponge, which is usually resorbed
in about 10 days. However, “angiographic
embolization is not meant to be used for acute,
massive postpartum hemorrhage.”1
(3) Hysterectomy is the last resort in the management
of PPH due to uterine causes. In most instances, a
subtotal hysterectomy, which is rapid, simple, safe,
and associated with less blood loss, is effective. Total
hysterectomy is necessary in cases where bleeding
occurs in the lower segment such as placenta previa
with accreta and tears into the lower segment.
Hysterectomy is reserved when all other treatments
available have been exhausted, bleeding continues,
and when further delay may compromise a patient
who is in severe shock. It is also the definitive
treatment in cases of coagulopathy when no
replacement blood products are available.19
2. Retained placenta
The third stage of labor is delivery of the placenta. The
retroplacental myometrium must contract to allow the
placenta to shear away from its bed and be expelled.
a. Epidemiology
Retained placenta complicates approximately 1% of
deliveries worldwide and is a significant cause of
maternal mortality and morbidity. In the developing
world, the associated mortality approaches 10% from
resulting PPH. However, not all cases of retained placenta
result in PPH. If the uterus contracts despite retained
fragments, little or no hemorrhage occurs. Risk factors
include previous history of retained placenta, previous
injury to uterus, preterm delivery, induced labor, and
multiparity.
b. Obstetric management
Oxytocics cause myometrial contraction, which generates
a shearing force that detaches the placenta from the
uterine wall. However, after placental detachment,
cervical constriction may trap the placenta within the
uterus. Manual removal of the placenta is standard
treatment. After removal, the placenta should be inspected
to ensure that there are no other retained fragments. In
addition, uterine tone should be assessed for evidence of
uterine atony and ongoing hemorrhage. Uterine curettage
will be necessary in some cases.
c. Anesthetic management
(1) Analgesia or anesthesia is often necessary in cases of
retained placenta because the obstetrician must
explore a uterus that is often partially contracted. In
some patients who have not received a neuraxial
block prior to delivery, IV analgesia with ketamine
(0.1 mg per kg), benzodiazepines, and judicious
administration of opioids (e.g., fentanyl) may be
adequate. However, it is essential to prevent
excessive sedation and loss of the maternal airway.
(2) If sedation is inadequate, the choice of anesthetic
technique depends largely on the degree of
hemorrhage. Spinal anesthesia is a reasonable choice
for patients who are hemodynamically stable with
little evidence of hemorrhage. However, in patients
with profound hemorrhage, GA is a better choice
because of the risk of hypotension resulting from
sympathetic blockade in a patient with hypovolemia
who has just received spinal anesthesia. In cases
where there is an indwelling neuraxial catheter and
the patient is hemodynamically stable, the addition of
more concentrated local anesthetic (e.g., 3% 2-
chloroprocaine) may be necessary to achieve
adequate anesthesia for uterine exploration.
(3) Obstetricians may request uterine relaxation for
removal of retained placenta. In the past, GA with
volatile anesthetic provided such relaxation. More
recently, reports consistently suggest that modest
incremental doses of IV nitroglycerin (25 to 50 µg)
are effective in producing adequate uterine relaxation
without major side effects. Nitroglycerin spray has
also been administered sublingually as an effective
uterine relaxant.
3. Placenta accreta
Placenta accreta is a potentially life-threatening pregnancy-
related complication. Although the incidence of placenta
accreta has increased over the past 20 to 30 years,85,86
estimates suggest that the complication occurs between 1 and
90 per 10,000 deliveries and is dependent on the frequency of
CD.87 A recent retrospective cohort study using data from the
Canadian Institute for Health Information of more than
570,000 deliveries from 2009 to 2010 determined that there
was a strong association between placenta accreta and PPH
with hysterectomy.88
a. Definition
Placenta accreta refers to a placenta that is abnormally
attached to the uterus. When the placenta invades the
myometrium, the term placenta increta is used, whereas
placenta percreta refers to a placenta that has invaded
through the myometrium and serosa, sometimes into
adjacent organs, such as the bladder (see Fig. 16.2). The
term placenta accreta is often used interchangeably as a
general term to describe all of these conditions. These
conditions can all cause maternal hemorrhage.
b. Epidemiology
Prior CD and presence of placenta previa in the current
pregnancy are important risk factors. In a multicenter
study of more than 30,000 patients who underwent CD
without labor, the risk of placenta accreta was
approximately 0.2%, 0.3%, 0.6%, 2.1%, and 7.7% for
women experiencing their first through sixth CD,
respectively. In patients with placenta previa, the risk of
accreta was 3%, 11%, 40%, 61%, and 67% for those
undergoing their first through their fifth or greater CD,
respectively.59 Other risk factors include (1) placenta
previa with or without prior uterine surgery, (2) prior
myomectomy, (3) prior CD, (4) Asherman syndrome, (5)
submucous leiomyomata, and (6) maternal age older than
35 years. Placenta accreta accounts for up to 50% of
all cesarean hysterectomies, most of which are
unplanned.
c. Diagnosis
There should be a high degree of clinical suspicion in
women with placenta previa or a history of CD or other
uterine surgery. Vigilance is particularly indicated when
the placenta is anterior and overlies the cesarean scar.
Ultrasonography is the primary imaging modality for
diagnosing accreta and is readily available in most
centers. Color Doppler techniques may also be helpful in
the diagnosis.34 Although most studies have suggested
reasonable diagnostic accuracy of MRI for placenta
accreta, it appears that MRI is no more sensitive than
ultrasonography for diagnosing placenta accreta.89
However, MRI may be more useful in the diagnosis of
cases when the placenta is situated posteriorly.
d. Obstetric management
(1) The ACOG suggests that the following measures
be taken when there is a strong suspicion of
placenta accreta before delivery:34
(a) The patient should be counseled about the
likelihood of hysterectomy and blood
transfusion.
(b) Blood products and clotting factors should be
available.
(c) Cell saver technology should be considered if
available.
(d) The appropriate location and timing for delivery
should be considered to allow access to adequate
surgical personnel and equipment.
(e) A preoperative anesthesia assessment should be
obtained.
(2) The area and depth of the abnormal attachment will
determine the obstetric treatment (i.e., curettage,
wedge resection, medical management, or
hysterectomy). When there is a small focal area of
abnormal attachment, conservative treatment may be
effective. However, in many cases, abdominal
hysterectomy is the definitive treatment approach.
e. Anesthetic management
(1) The role of experienced anesthesiology personnel
who are skilled in obstetric anesthesia cannot be
overemphasized, and they should be involved in
preoperative assessment of the patient.
(2) In cases of unexpected placenta accreta during
elective CD, GA is often necessary to protect the
patient’s airway. In all cases of placenta accreta, the
anesthesia provider should be prepared for massive
hemorrhage with adequate IV access and large
amounts of crystalloid, colloid, and blood products.
(3) In patients who are hemodynamically stable and
euvolemic, but are at risk for placenta accreta and
hysterectomy, a continuous catheter neuraxial
technique has been shown to be a reasonable
option.90,91 However, the neuraxial technique may
fail because (1) extended operative time predisposes
the patient to restlessness and fatigue, (2) surgical
retraction exceeds that required by CD alone, and (3)
a quiet operative field is necessary for dissection.90
When patients are candidates for neuraxial
anesthesia, they should be counseled about the risks
and benefits of such techniques, specifically that the
neuraxial technique might be inadequate and require
conversion to GA. When an epidural is in place but
GA becomes necessary, the epidural anesthetic
reduces volatile anesthetic requirements and can be
used for postoperative analgesia.
f. Interventional radiology techniques
Although controversial, preoperative placement of
internal iliac artery balloon catheters and ureteral stents
may be helpful to control hemorrhage during CD and
subsequent hysterectomy. In the largest of studies,
placement of balloon catheters preoperatively reduced (1)
the mean estimated blood loss, (2) the number of cases
with blood loss more than 2.5 L, and (3) the number of
massive transfusions.92 However, there can be
complications related to placement (e.g., fetal
bradycardia) and postoperatively (e.g., lower extremity
ischemia) (see Table 16.7). The Society of Maternal-Fetal
Medicine recommends prophylactic intra-arterial balloon
catheters for women who (1) have a desire to preserve
fertility, (2) decline blood products, and (3) who have
unresectable placenta percreta.93 In cases where
embolization techniques are unsuccessful, hysterectomy
will be necessary and the anesthesia provider should be
prepared for significant hemorrhage and transfusion
requirements.
4. Uterine inversion
a. Definition
Uterine inversion is a “turning inside out” of the uterus.
Although rare, it has potentially serious consequences. It
is classified as complete if the fundus passes through the
cervix, or incomplete if it remains above this level. The
underlying causes are not completely understood94 but
risk factors include (1) overzealous fundal pressure, (2)
excessive umbilical cord traction, (3) uterine
anomalies, (4) uterine atony, and (5) placenta accreta.
b. Diagnosis
Cases of uterine inversion are often obvious because of
hemorrhage and a mass in the vagina. In the past,
obstetricians have suggested that the degree of
hemorrhage was disproportionate to the degree of shock.
However, an atonic, inverted uterus will contribute to
severe hemorrhage and shock.
c. Obstetric management
Management of uterine inversion has two important
components: the immediate treatment of hemorrhagic
shock and replacement of the uterus. Atony usually
resolves after replacement followed by medical treatment
(e.g., oxytocin infusion).
d. Anesthetic management
Resuscitation should begin immediately while attempts
are made to replace the uterus manually because
hypotension and bradycardia may occur. In most cases,
neuraxial analgesia does not provide adequate relaxation
but may be helpful in providing analgesia. Reports
suggest that nitroglycerin may obviate the need for GA,
although GA with a volatile halogenated agent may be
necessary for uterine relaxation. Other anecdotal reports
suggest terbutaline as well as magnesium sulfate tocolytic
therapies. Severe cases require laparotomy.
REFERENCES
1. Clark SL, Hankins GD. Preventing maternal death: 10 clinical diamonds. Obstet Gynecol.
2012;119:360–364.
2. American College of Obstetricians and Gynecologists. ACOG Committee Opinion No. 326:
inappropriate use of the terms fetal distress and birth asphyxia. Obstet Gynecol. 2005;106:1469–1470.
3. Bax M, Nelson KB. Birth asphyxia: a statement. Dev Med Child Neurol. 1993;35:1022–1024.
4. American College of Obstetricians and Gynecologists Task Force on Neonatal Encephalopathy and
Cerebral Palsy, American Academy of Pediatrics. Neonatal Encephalopathy and Cerebral Palsy:
Defining the Pathogenesis and Pathophysiology. Washington, DC: American College of Obstetricians
and Gynecologists; 2003.
5. Herrera-Marschitz M, Neira-Pena T, Rojas-Mancilla E, et al. Perinatal asphyxia: CNS development
and deficits with delayed onset. Front Nuerosci. 2014;8:47.
6. MacLennan A. A template for defining a causal relation between acute intrapartum events and cerebral
palsy: international consensus statement. BMJ. 1999;319:1054–1059.
7. American College of Obstetricians and Gynecologists. Neonatal encephalopathy and cerebral palsy:
executive summary. Obstet Gynecol. 2004;103:780–781.
8. American College of Obstetricians and Gynecologists. ACOG Committee Opinion No. 348: umbilical
cord blood gas and acid-base analysis. Obstet Gynecol. 2006;108:1319–1322.
9. Dawes GS. Sudden death in babies: physiology of the fetus and newborn. Am J Cardiol. 1968;22:469–
478.
10. Fetal physiology and cell biology. In: American College of Obstetricians and Gynecologists, American
Academy of Pediatrics, eds. Neonatal Encephalopathy and Neurologic Outcome. 2nd ed. Washington,
DC: American College of Obstetricians and Gynecologists; 2014:21–36.
11. Badawi N, Kurinczuk JJ, Keogh JM, et al. Antepartum risk factors for newborn encephalopathy: the
Western Australian case-control study. BMJ. 1998;317:1549–1553.
12. Leveno KJ, Cunningham FG, Nelson S, et al. A prospective comparison of selective and universal
electronic fetal monitoring in 34,995 pregnancies. N Engl J Med. 1986;315:615–619.
13. 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.
14. Clark SL, Nageotte MP, Garite TJ, et al. Intrapartum management of category II fetal heart rate
tracings: towards standardization of care. Am J Obstet Gynecol. 2013;209:89–97.
15. Graham EM, Adami RR, McKenney SL, et al. Diagnostic accuracy of fetal heart rate monitoring in
the identification of neonatal encephalopathy. Obstet Gynecol. 2014;124:507–513.
16. Alfirevic Z, Neilson JP. Doppler ultrasonography in high-risk pregnancies: systematic review with
meta-analysis. Am J Obstet Gynecol. 1995;172:1379–1387.
17. Tolsa CB, Zimine S, Warfield SK, et al. Early alteration of structural and functional brain development
in premature infants born with intrauterine growth restriction. Pediatr Res. 2004;56:132–138.
18. American College of Obstetricians and Gynecologists. ACOG Practice Bulletin No. 36: obstetric
analgesia and anesthesia. Int J Gynaecol Obstet. 2002;78:321–335.
19. American College of Obstetricians and Gynecologists. Standards of Obstetric-Gynecologic Services.
7th ed. Washington, DC: American College of Obstetricians and Gynecologists; 1989:39.
20. American College of Obstetricians and Gynecologists. ACOG Committee Opinion No. 433: optimal
goals for anesthesia care in obstetrics. Obstet Gynecol. 2009;113:1197–1199.
21. Korhonen J, Kariniemi V. Emergency cesarean section: the effect of delay on umbilical arterial gas
balance and Apgar scores. Acta Obstet Gynecol Scand. 1994;73:782–786.
22. Myers RE. Two patterns of perinatal brain damage and their conditions of occurrence. Am J Obstet
Gynecol. 1972;112:246–276.
23. American Society of Anesthesiologists Task Force on Obstetric Anesthesia. Practice guidelines for
obstetric anesthesia: an updated report by the American Society of Anesthesiologists Task Force on
Obstetric Anesthesia. Anesthesiology. 2007;106:843–863.
24. Centers for Disease Control and Prevention. Pregnancy mortality surveillance system.
http://www.cdc.gov/reproductivehealth/maternalinfanthealth/pmss.html. Accessed March 22, 2015.
25. Hawkins JL, Koonin LM, Palmer SK, et al. Anesthesia-related deaths during obstetric delivery in the
United States, 1979–1990. Anesthesiology. 1997;86:277–284.
26. D’Angelo R, Smiley RM, Riley ET, et al. Serious complications related to obstetric anesthesia: the
serious complication repository project of the Society for Obstetric Anesthesia and Perinatology.
Anesthesiology. 2014;120:1505–1512.
27. Frölich MA, Banks C, Brooks A, et al. Why do pregnant women die? A review of maternal deaths
from 1990 to 2010 at the University of Alabama at Birmingham. Anesth Analg. 2014;119:1135–1139.
28. Shields LE, Smalarz K, Reffigee L, et al. Comprehensive maternal hemorrhage protocols improve
patient safety and reduce utilization of blood products. Am J Obstet Gynecol. 2011;205:368.e1–
368.e8.
29. Reiss RF. Hemostatic defects in massive transfusion: rapid diagnosis and management. Am J Crit
Care. 2000;9:158–165.
30. The Joint Commission. Preventing maternal death. Sentinel Event Alert. 2010;44:1–4.
31. Shields LE, Wiesner S, Fulton J, et al. Comprehensive maternal hemorrhage protocols reduce the use
of blood products and improve patient safety. Am J Obstet Gynecol. 2015;212:272–280.
32. Scavone BM, Tung A. The transfusion dilemma: more, less, or more organized? Anesthesiology.
2014;121:439–441.
33. Zuckerwise LC, Pettker CM, Illuzzi J, et al. Use of a novel visual aid to improve estimation of
obstetric blood loss. Obstet Gynecol. 2014;123:982–986.
34. American College of Obstetricians and Gynecologists. ACOG Practice Bulletin: Clinical Management
Guidelines for Obstetrician-Gynecologists No. 76: postpartum hemorrhage. Obstet Gynecol.
2006;108:1039–1047.
35. Hébert PC, Wells G, Blajchman MA, et al. A multicenter, randomized, controlled clinical trial of
transfusion requirements in critical care. Transfusion requirements in critical care investigators,
Canadian Critical Care Trials Group. N Engl J Med. 1999;340:409–417.
36. Karpati PCJ, Rossignol M, Pirot M, et al. High incidence of myocardial ischemia during postpartum
hemorrhage. Anesthesiology. 2004;100:30–36.
37. Matot I, Einav S, Goodman S, et al. A survey of physicians’ attitudes toward blood transfusion in
patients undergoing cesarean section. Am J Obstet Gynecol. 2004;190:462–467.
38. American Society of Anesthesiologists Task Force on Perioperative Blood Management. Practice
guidelines for perioperative blood management: an updated report by the American Society of
Anesthesiologists Task Force on Perioperative Blood Management. Anesthesiology. 2015;122:241–
275.
39. Vanderlinde ES, Heal JM, Blumberg N. Autologous transfusion. BMJ. 2002;324:772–775.
40. Goodnough LT, Brecher ME, Kanter MH, et al. Transfusion medicine. Second of two parts—blood
conservation. N Engl J Med. 1999;340:525–533.
41. Yamada T, Mori H, Ueki M. Autologous blood transfusion in patients with placenta previa. Acta
Obstet Gynecol Scand. 2005;84:255–259.
42. Allam J, Cox M, Yentis SM. Cell salvage in obstetrics. Int J Obstet Anesth. 2008;17:37–45.
43. Petroianu GA, Altmannsberger SH, Maleck WH, et al. Meconium and amniotic fluid embolism:
effects on coagulation in pregnant mini-pigs. Crit Care Med. 1999;27:348–355.
44. Waters JH, Biscotti C, Potter PS, et al. Amniotic fluid removal during cell salvage in the cesarean
section patient. Anesthesiology. 2000;92:1531–1536.
45. Grange CS, Douglas MJ, Adams TJ, et al. The use of acute hemodilution in parturients undergoing
cesarean section. Am J Obstet Gynecol. 1998;178:156–160.
46. Malone DL, Hess JR, Fingerhut A. Massive transfusion practices around the globe and a suggestion
for a common massive transfusion protocol. J Trauma. 2006;60:S91–S96.
47. Mhyre JM, D’Oria R, Hameed AB, et al. The maternal early warning criteria: a proposal from the
national partnership for maternal safety. Obstet Gynecol. 2014;124:782–786.
48. Kacmar RM, Mhyre JM, Scavone BM, et al. The use of postpartum hemorrhage protocols in United
States academic obstetric anesthesia units. Anesth Analg. 2014;119:906–910.
49. Karlsson O, Jeppsson A, Hellgren M. Major obstetric haemorrhage: monitoring with
thromboelastography, laboratory analyses or both? Int J Obstet Anesth. 2014;23:10–17.
50. Sharma SK, Philip J, Whitten CW, et al. Assessment of changes in coagulation in parturients with
preeclampsia using thromboelastography. Anesthesiology. 1999;90:385–390.
51. Sharma SK, Philip J, Wiley J. Thromboelastographic changes in healthy parturients and postpartum
women. Anesth Analg. 1997;85:94–98.
52. Cortet M, Deneux-Tharaux C, Dupont C, et al. Association between fibrinogen level and severity of
postpartum haemorrhage: secondary analysis of a prospective trial. Br J Anaesthesia. 2012;108:984–
989.
53. Theusinger OM, Baulig W, Seifert B, et al. Relative concentrations of haemostatic factors and
cytokines in solvent/detergent-treated and fresh-frozen plasma. Br J Anaesth. 2011;106:505–511.
54. Caudill JSC, Nichols WL, Plumhoff EA, et al. Comparison of coagulation factor XIII content and
concentration in cryoprecipitate and fresh-frozen plasma. Transfusion. 2009;49:765–770.
55. Levy JH, Szlam F, Tanaka KA, et al. Fibrinogen and hemostasis: a primary hemostatic target for the
management of acquired bleeding. Anesth Analg. 2012;114:261–274.
56. Lewis NR, Brunker P, Lemire SJ, et al. Failure of recombinant factor VIIa to correct the coagulopathy
in a case of severe postpartum hemorrhage. Transfusion. 2009;49:689–695.
57. Sentilhes L, Lasocki S, Ducloy-Bouthors AS, et al. Tranexamic acid for the prevention and treatment
of postpartum haemorrhage. Br J Anaesth. 2015;114:576–587.
58. Iyasu S, Saftlas AK, Rowley DL, et al. The epidemiology of placenta previa in the United States, 1979
through 1987. Am J Obstet Gynecol. 1993;168:1424–1429.
59. Silver RM, Landon MB, Rouse DJ, et al. Maternal morbidity associated with multiple repeat cesarean
deliveries. Obstet Gynecol. 2006;107:1226–1232.
60. Dashe JS. Toward consistent terminology of placental location. Semin Perinatol. 2013;37:375–379.
61. Sharma A, Suri V, Gupta I. Tocolytic therapy in conservative management of symptomatic placenta
previa. Int J Gynaecol Obstet. 2004;84:109–113.
62. Bonner SM, Haynes SR, Ryall D. The anaesthetic management of caesarean section for placenta
praevia: a questionnaire survey. Anaesthesia. 1995;50:992–994.
63. Oyelese Y, Catanzarite V, Prefumo F, et al. Vasa previa: the impact of prenatal diagnosis on outcomes.
Obstet Gynecol. 2004;103:937–942.
64. Sheiner E, Shoham-Vardi I, Hallak M, et al. Placental abruption in term pregnancies: clinical
significance and obstetric risk factors. J Matern Fetal Neonatal Med. 2003;13:45–49.
65. Ananth CV, Oyelese Y, Yeo L, et al. Placental abruption in the United States, 1979 through 2001:
temporal trends and potential determinants. Am J Obstet Gynecol. 2005;192:191–198.
66. Ananth CV, Wilcox AJ. Placental abruption and perinatal mortality in the United States. Am J
Epidemiol. 2001;153: 332–337.
67. Tikkanen M, Nuutila M, Hiilesmaa V, et al. Clinical presentation and risk factors of placental
abruption. Acta Obstet Gynecol Scand. 2006;85:700–705.
68. Morikawa M, Cho K, Yamada T, et al. Do uterotonic drugs increase risk of abruptio placentae and
eclampsia? Arch Gynecol Obstet. 2014;289:987–991.
69. Kayani SI, Walkinshaw SA, Preston C. Pregnancy outcome in severe placental abruption. BJOG.
2003;110:679–683.
70. Walsh CA, Baxi LV. Rupture of the primigravid uterus: a review of the literature. Obstet Gynecol Surv.
2007;62:327–334.
71. Lydon-Rochelle M, Holt VL, Easterling TR, et al. Risk of uterine rupture during labor among women
with a prior cesarean delivery. N Engl J Med. 2001;345:3–8.
72. American College of Obstetricians and Gynecologists. ACOG Committee Opinion No. 342: induction
of labor for vaginal birth after cesarean delivery. Obstet Gynecol. 2006;108:465–468.
73. Eden RD, Parker RT, Gall SA. Rupture of the pregnant uterus: a 53-year review. Obstet Gynecol.
1986;68:671–674.
74. Scavone B. Antepartum and postpartum hemorrhage. In: Chestnut DH, Wong CA, Tsen LC, et al, eds.
Chestnut’s Obstetric Anesthesia: Principles and Practice. 5th ed. Philadelphia, Pa: Elsevier Saunders;
2014:881–914.
75. Stalnaker BL, Maher JE, Kleinman GE, et al. Characteristics of successful claims for payment by the
Florida Neurologic Injury Compensation Association Fund. Am J Obstet Gynecol. 1997;177:268–271.
76. American College of Obstetricians and Gynecologists. ACOG Practice Bulletin No. 115: vaginal birth
after previous cesarean delivery. Obstet Gynecol. 2010;116:450–463.
77. American Society of Anesthesiologists. Optimal goals for anesthesia care in obstetrics.
http://www.asahq.org/publications AndServices/standards/24.html. Accessed April 7, 2015.
78. World Health Organization. World Health Organization recommendations for the prevention and
treatment of postpartum haemorrhage.
http://apps.who.int/iris/bitstream/10665/75411/1/9789241548502_eng.pdf. Accessed March 22, 2015.
79. Callaghan WM, Kuklina EV, Berg CJ. Trends in postpartum hemorrhage: United States, 1994–2006.
Am J Obstet Gynecol. 2010;202:353.e351–353.e356.
80. Mhyre JM. Maternal mortality. Curr Opin Anaesthesiol. 2012;25:277–285.
81. Tita ATN, Szychowski JM, Rouse DJ, et al. Higher-dose oxytocin and hemorrhage after vaginal
delivery: a randomized controlled trial. Obstet Gynecol. 2012;119:293–300.
82. Bateman BT, Huybrechts KF, Hernandez-Diaz S, et al. Methylergonovine maleate and the risk of
myocardial ischemia and infarction. Am J Obstet Gynecol. 2013;209:459.e1–459.e13.
83. Oleen MA, Mariano JP. Controlling refractory atonic postpartum hemorrhage with Hemabate sterile
solution. Am J Obstet Gynecol. 1990;162:205–208.
84. Gülmezoglu AM, Forna F, Villar J, et al. Prostaglandins for preventing postpartum haemorrhage.
Cochrane Database Syst Rev. 2007;3:CD000494.
85. Timor-Tritsch IE, Monteagudo A. Unforeseen consequences of the increasing rate of cesarean
deliveries: early placenta accreta and cesarean scar pregnancy. A review. Am J Obstet Gynecol.
2012;207:14–29.
86. Wu S, Kocherginsky M, Hibbard JU. Abnormal placentation: twenty-year analysis. Am J Obstet
Gynecol. 2005;192: 1458–1461.
87. Gielchinsky Y, Rojansky N, Fasouliotis SJ, et al. Placenta accreta—summary of 10 years: a survey of
310 cases. Placenta. 2002;23:210–214.
88. Mehrabadi A, Hutcheon JA, Liu S, et al. Contribution of placenta accreta to the incidence of
postpartum hemorrhage and severe postpartum hemorrhage. Obstet Gynecol. 2015;125:814–821.
89. Palacios Jaraquemada JM, Bruno CH. Magnetic resonance imaging in 300 cases of placenta accreta:
surgical correlation of new findings. Acta Obstet Gynecol Scand. 2005;84:716–724.
90. Chestnut DH, Redick LF. Continuous epidural anesthesia for elective cesarean hysterectomy. South
Med J. 1985;78: 1168–1169.
91. Chestnut DH, Dewan DM, Redick LF, et al. Anesthetic management for obstetric hysterectomy: a
multi-institutional study. Anesthesiology. 1989;70:607–610.
92. Ballas J, Hull AD, Saenz C, et al. Preoperative intravascular balloon catheters and surgical outcomes
in pregnancies complicated by placenta accreta: a management paradox. Am J Obstet Gynecol.
2012;207:216.e211–216.e215.
93. Belfort MA. Placenta accreta. Am J Obstet Gynecol. 2010;203:430–439.
94. Wendel PJ, Cox SM. Emergent obstetric management of uterine inversion. Obstet Gynecol Clin North
Am. 1995;22: 261–274.
95. Goldenberg RL, Culhane JF, Iams JD, et al. Epidemiology and causes of preterm birth. Lancet.
2008;371:75–84.
96. Eichenwald EC, Stark AR. Management and outcomes of very low birth weight. N Engl J Med.
2008;358:1700–1711.
97. Patel P, Sun L. Update on neonatal anesthetic neurotoxicity: insight into molecular mechanisms and
relevance to humans. Anesthesiology. 2009;110:703–708.
98. Palanisamy A. Maternal anesthesia and fetal neurodevelopment. Int J Obstet Anesth. 2012;21:152–
162.
99. Yamasato K, Burlingame J, Kaneshiro B. Hemodynamic effects of nifedipine tocolysis. J Obstet
Gynaecol Res. 2015;41:17–22.
100. van Vliet EO, Boormans EM, de Lange TS, et al. Preterm labor: current pharmacotherapy options for
tocolysis. Expert Opin Pharmacother. 2014;15:787–797.
101. Loe SM, Sanchez-Ramos L, Kaunitz AM. Assessing the neonatal safety of indomethacin tocolysis: a
systematic review with meta-analysis. Obstet Gynecol. 2005;106:173–179.
102. Chau AC, Gabert HA, Miller JM Jr. A prospective comparison of terbutaline and magnesium for
tocolysis. Obstet Gynecol. 1992;80:847–851.
103. Hollander DI, Nagey DA, Pupkin MJ. Magnesium sulfate and ritodrine hydrochloride: a randomized
comparison. Am J Obstet Gynecol. 1987;156:631–637.
104. DeVore JS, Asrani R. Magnesium sulfate prevents succinylcholine-induced fasciculations in toxemic
parturients. Anesthesiology. 1980;52:76–77.
105. Neilson JP, West HM, Dowswell T. Betamimetics for inhibiting preterm labour. Cochrane Database
Syst Rev. 2014;2: CD004352.
106. Flenady V, Reinebrant HE, Liley HG, et al. Oxytocin receptor antagonists for inhibiting preterm
labour. Cochrane Database Syst Rev. 2014;6:CD004452.
107. Beall MH, Spong C, McKay J, et al. Objective definition of shoulder dystocia: a prospective
evaluation. Am J Obstet Gynecol. 1998;179:934–937.
108. Rouse DJ, Owen J, Goldenberg RL, et al. The effectiveness and costs of elective cesarean delivery for
fetal macrosomia diagnosed by ultrasound. JAMA. 1996;276:1480–1486.
109. Sandberg EC. The Zavanelli maneuver: 12 years of recorded experience. Obstet Gynecol.
1999;93:312–317.
110. Gibbons C, O’Herlihy C, Murphy JF. Umbilical cord prolapse—changing patterns and improved
outcomes: a retrospective cohort study. BJOG. 2014;121:1705–1708.
111. Katz Z, Shoham Z, Lancet M, et al. Management of labor with umbilical cord prolapse: a 5-year study.
Obstet Gynecol. 1988;72:278–281.
112. McDonnell NJ, Percival V, Paech MJ. Amniotic fluid embolism: a leading cause of maternal death yet
still a medical conundrum. Int J Obstet Anesth. 2013;22:329–336.
113. Abenhaim HA, Azoulay L, Kramer MS, et al. Incidence and risk factors of amniotic fluid embolisms:
a population-based study on 3 million births in the United States. Am J Obstet Gynecol.
2008;199:49.e1–49.e8.
114. Knight M, Tuffnell D, Brocklehurst P, et al. Incidence and risk factors for amniotic-fluid embolism.
Obstet Gynecol. 2010;115:910–917.
115. Kramer MS, Rouleau J, Baskett TF, et al. Amniotic-fluid embolism and medical induction of labour: a
retrospective, population-based cohort study. Lancet. 2006;368:1444–1448.
116. Roberts CL, Algert CS, Knight M, et al. Amniotic fluid embolism in an Australian population-based
cohort. BJOG. 2010;117:1417–1421.
117. Knight M, Berg C, Brocklehurst P, et al. Amniotic fluid embolism incidence, risk factors and
outcomes: a review and recommendations. BMC Pregnancy Childbirth. 2012;12:7.
118. Dean LS, Rogers RP III, Harley RA, et al. Case scenario: amniotic fluid embolism. Anesthesiology.
2012;116:186–192.
119. Clark SL. Amniotic fluid embolism. Obstet Gynecol. 2014;123:337–348.
120. Vanden Hoek TL, Morrison LJ, Shuster M, et al. Part 12: cardiac arrest in special situations: 2010
American Heart Association Guidelines for Cardiopulmonary Resuscitation and Emergency
Cardiovascular Care. Circulation. 2010;122:S829–S861.
121. Farinelli CK, Hameed AB. Cardiopulmonary resuscitation in pregnancy. Cardiol Clin. 2012;30:453–
461.
122. Paterson-Brown S, Bamber J. Prevention and treatment of haemorrhage. In: Knight M, Kenyon S,
Brocklehurst P, et al, eds. Saving Lives, Improving Mothers’ Care : Lessons Learned to Inform Future
Maternity Care from the UK and Ireland Confidential Enquiries into Maternal Deaths and Morbidity
2009-2012. Oxford, United Kingdom: University of Oxford; 2014:45–55.
123. Conde-Agudelo A, Romero R. Amniotic fluid embolism: an evidence-based review. Am J Obstet
Gynecol. 2009;201:445.e1–455.e13.
124. Girard T, Mörtl M, Schlembach D. New approaches to obstetric hemorrhage: the postpartum
hemorrhage consensus algorithm. Curr Opin Anaesthesiol. 2014;27:267–274.
125. Leighton BL, Wall MH, Lockhart EM, et al. Use of recombinant factor VIIa in patients with amniotic
fluid embolism: a systematic review of cases. Anesthesiology. 2011;115:1201–1208.
126. Morgan M. Amniotic fluid embolism. Anaesthesia. 1979;34:20–32.
127. Kramer MS, Rouleau J, Liu S, et al. Amniotic fluid embolism: incidence, risk factors, and impact on
perinatal outcome. BJOG. 2012;119:874–879.
Newborn Resuscitation
Richard A. Month
I. Neonatal adaptations to extrauterine life
A. Fetal cardiovascular and pulmonary physiology
B. Normal peripartum transition to extrauterine life
C. Prolonged hypoxemia/acidosis and failure of transition
II. Anticipating the depressed newborn
A. Antepartum evaluation
B. Intrapartum evaluation
III. Evaluating the neonate
A. Apgar score
B. Umbilical cord blood gas measurements
IV. Resuscitation of the neonate
A. Preparation for resuscitation
B. The resuscitation algorithm
C. Initial resuscitation
D. Maintain normothermia
E. Assisted ventilation
F. Considerations for establishing ventilation
G. Oxygen administration
H. Chest compressions
I. Umbilical vein catheterization
J. Medications
K. Discontinuation of resuscitative efforts
V. Special resuscitation circumstances
A. Meconium-stained amniotic fluid
B. Premature infants
C. Opioid-induced respiratory depression
D. Magnesium toxicity
KEYPOINTS
1. The transition from intrauterine to extrauterine life requires a reduction in
neonatal pulmonary vascular resistance and an increase in systemic
vascular resistance that accompanies the onset of breathing.
2. Prolonged acidosis and hypoxia after delivery can prevent this transition.
3. Most cases of neonatal depression can be predicted by antepartum and
intrapartum fetal assessment.
4. The Apgar score gives practitioners a guide to the need for resuscitation.
It is not a good predictor of poor neurologic outcome following delivery.
5. The primary responsibility of the anesthesia provider is to the mother. A
second individual skilled in neonatal resuscitation should be immediately
available to resuscitate the neonate when possible.
6. Assisted ventilation is indicated 30 seconds after delivery in the neonate
who has ineffective breathing following initial resuscitative measures.
7. The administration of 100% oxygen during assisted ventilation is no
longer routinely recommended. However, the optimal inspired oxygen
concentration has not been established.
8. Hypovolemia is rarely a cause of neonatal cardiopulmonary depression.
The administration of large volumes of fluid to a normovolemic neonate
is associated with significant adverse effects.
9. Intrapartum oropharyngeal and tracheal suctioning of a vigorous neonate
born through meconium-stained fluid is not recommended.
10. Initial treatment of the neonate with opioid-induced respiratory
depression is with assisted ventilation, not naloxone administration.
A SUCCESSFUL TRANSITION FROM THE INTRAUTERINE TO EXTRAUTERINE
ENVIRONMENT requires numerous complex physiologic changes. Approximately
10% of newborns will require assistance with ventilation at birth, and 1% will
need extensive resuscitative measures.1 Worldwide, an estimated 5 to 10 million
interventions will be required annually; it is imperative that all providers
working in labor and delivery suites understand the neonatal adaptions to
extrauterine life, recognize the need for resuscitation, and have the expertise to
respond appropriately.
I. Neonatal adaptations to extrauterine life
A. Fetal cardiovascular and pulmonary physiology
1. Unlike the adult, where pulmonary and systemic circulations
operate in parallel, the fetal circulation operates in series
(see Fig. 17.1).2 Oxygenated blood from the placenta returns
via the umbilical vein and is shunted through the ductus
venosus into the inferior vena cava (IVC). Upon return to the
heart, nearly 40% of IVC flow is directed across the foramen
ovale and through the left heart and ascending aorta in order
to perfuse the fetal heart and brain with maximally
oxygenated blood.3 Deoxygenated blood from the head and
upper extremities enters the heart through the superior vena
cava (SVC). This blood is preferentially directed into the right
ventricle where it mixes with the remainder of the maximally
oxygenated IVC return. High pulmonary vascular resistance
(PVR) ensures that 90% of right ventricular outflow shunts
across the ductus arteriosus to the descending aorta to perfuse
the abdomen, pelvis, and lower extremities.4 This creates a
substantial right-to-left cardiac shunt. Forty percent of
cardiac output flows through the umbilical arteries to the
placenta; due to the large total cross sectional area of the
placenta, it has low resistance and contributes little to fetal
systemic vascular resistance (SVR).
2. Fetal lung development takes place throughout pregnancy,
and lung maturation continues well into the neonatal period.
The pulmonary transition that occurs in the fetus is initiated
before labor begins. Cortisol production increases in the third
trimester to begin multisystem organ preparation for the
newborn transition, including preparation for pulmonary
maturation. In addition, surfactant production increases to
reduce alveolar surface tension, promote alveolar expansion,
foster gas exchange, and stimulate fetal lung development.5
B. Normal peripartum transition to extrauterine life
1. Delivery leads to rapid and profound changes in neonatal
hemodynamics and pulmonary mechanics. During
development, the fetal airways contain approximately 30 mL
per kg of fluid. This fluid, an ultrafiltrate of fetal plasma,
begins to reabsorb during labor.3 During vaginal delivery,
compression of the thorax expels further fluid from the mouth
and upper airways.2 With the first breaths, the lungs fill with
air, surfactant is released, and oxygenation increases
dramatically.6 This increase in oxygen tension and blood flow
increases the release of nitric oxide in the pulmonary
vasculature resulting in pulmonary vasodilation and a
substantial decrease in PVR.7 Simultaneously, clamping of the
umbilical cord removes the low-resistance placenta from the
systemic circulation increasing SVR. Functional closure of
the foramen ovale occurs rapidly after birth as left atrial
pressures exceed right atrial pressures.
2. The right-to-left shunt across the foramen ovale and ductus
arteriosus is substantially reduced within the first few minutes
of life.
C. Initial resuscitation
The general condition of the neonate is quickly assessed with four
screening questions addressed immediately following delivery
(see Fig. 17.3): (1) Is the infant full-term? (2) Is the amniotic fluid
clear? (3) Is the newborn breathing or crying? (4) Does the infant
have good muscle tone? Routine care is provided for full-term
newborns with clear amniotic fluid, good muscle tone, and
breathing without distress. Neonates with difficulty making the
transition to extrauterine life should be warmed, gently
stimulated, and placed in the “sniffing position” to open their
airways. Suctioning of the pharynx and nose should be brief and
gentle because prolonged or vigorous suctioning may result in
breath holding, laryngospasm, or bradycardia.
D. Maintain normothermia
Minimizing heat loss is an integral part of neonatal resuscitation.
All infants have an unstable thermal regulatory system and this is
enhanced in depressed or asphyxiated neonates. The large surface
area to body mass ratio of the neonate facilitates rapid heat loss
by conduction, convection, evaporation, and radiation. The term
neonate’s primary defense against hypothermia is nonshivering
thermogenesis through catecholamine-mediated metabolism of
brown fat. This, in turn, leads to an increase in oxygen
consumption, calorie utilization, and metabolic rate.26 The
resulting hypoxemia, hypercarbia, and hypoglycemia promote
persistence of the fetal circulation and hinder resuscitation.
E. Assisted ventilation
Spontaneously breathing infants should make their first
respiratory effort seconds after delivery of the thorax, generating
a negative intrathoracic pressure of 60 to 100 cm H2O and
inspiring a tidal volume of approximately 80 mL. The majority of
this first breath is retained within the lungs as part of the
functional residual capacity.27 Infants who fail to initiate
respirations with gentle stimulation, warming, and airway
suctioning require further intervention to ensure adequate
ventilation.
1. Assisted ventilation is required in about 3% to 5% of all
newborn infants. Positive pressure ventilation is indicated in
neonates who remain apneic, have ineffective or gasping
ventilation, or a heart rate less than 100 bpm more than 30
seconds after delivery (see Fig. 17.3). Assisted ventilation can
be initiated via a bag valve mask device, a laryngeal mask
airway (LMA), or an endotracheal tube. Ventilation is usually
first established with a bag valve mask device followed by
endotracheal intubation in the absence of clinical
improvement.
2. Common indications for endotracheal intubation include
tracheal suctioning for meconium, ineffective or prolonged
bag valve mask ventilation, or endotracheal administration of
medications (although this practice is now strongly
discouraged). For ideal intubating conditions, the infant’s
head is placed in a neutral “sniffing position” (see Fig. 17.4).
A small straight blade, such as a Miller 0 or 1, provides the
best visualization of the neonatal larynx given its unique
anatomic characteristics. Specifically, the newborn larynx is
more anterior than the adult and at the level of the third
cervical vertebra rather than the sixth. A small air leak with
positive pressure ventilation indicates the uncuffed
endotracheal tube is appropriately sized (see Table 17.7). An
oversized endotracheal tube may cause subglottic stenosis,
whereas an undersized endotracheal tube can impede
adequate ventilation and may become easily occluded. The
endotracheal tube is inserted 2 cm past the vocal cords, and
tracheal placement is confirmed via detection of end tidal
CO2, bilateral breath sounds, and symmetric chest rise.
3. Both bag valve mask and endotracheal ventilation are known
to produce significant morbidity. Prolonged bag valve mask
ventilation can result in distention of the stomach and/or
ocular or facial abrasions from pressure applied to the mask.
On the other hand, direct laryngoscopy can be accompanied
by a significant hypertensive response contributing to cerebral
hemorrhage. In addition, intubation can be challenging in
neonates. A recent survey found that intubation by third-year
pediatric residents was successful only 62% of the time on the
first or second attempt.28
4. Given some of the limitations of bag valve mask and
endotracheal ventilation, some advocate for the use of LMAs
in neonatal resuscitation.29–31 LMAs have been used
successfully in resuscitation of both full-term and preterm
infants and may have significant advantages over
endotracheal intubation, including ease of use and decreased
hemodynamic response. Both novice and experienced
providers have had success with LMA use in the neonate. One
study reported the successful resuscitation of 20 neonates with
the LMA as a conduit for positive pressure ventilation by
novice providers.29 Another trial found no significant
difference between the LMA and endotracheal intubation
during resuscitation of infants by experienced providers after
CD.31 Although endotracheal intubation is still preferred in
situations requiring ventilation with high peak airway
pressures, tracheal suctioning, or administration of
endotracheal medications, the LMA can be lifesaving in
neonates with difficult airways, specifically in conditions with
a congenital hypoplastic mandible, such as Pierre-Robin and
Treacher-Collins syndromes.
H. Chest compressions
Regardless of the fraction of inspired oxygen used during initial
resuscitative efforts, if the heart rate remains less than 60 bpm
after 30 seconds of positive pressure ventilation, then chest
compressions are indicated (see Fig. 17.3). Neonatal cardiac
arrest occurs in less than 0.1% of term deliveries and is most
commonly associated with respiratory failure because hypoxemia
and tissue acidosis lead to bradycardia, decreased cardiac
contractility, and eventually cardiac arrest.
1. Chest compressions should be initiated at a ratio of 3:1 with
90 compressions and 30 breaths per minute, by one of two
compressive techniques (see Fig. 17.5). With the “thumb
compression technique,” both thumbs are positioned at the
lower third of the sternum and the fingers encircle the chest
and support the back; this method generates higher peak
systolic and coronary perfusion pressures and is generally
recommended over the “two-finger technique.”43 The “two-
finger technique,” with the tips of the middle and index
fingers positioned perpendicular to the chest and a second
hand supporting the back, facilitates simultaneous
interventions on the neonate and may be preferred in
situations where access to the umbilical vessels is necessary.
Regardless of the technique, the sternum is displaced one-
third the anterior–posterior chest diameter with each
compression. An improvement in the neonate’s oxygenation
and/or color and the presence of a palpable pulse confirms the
adequacy of cardiac output. Chest compressions can be
paused every 2 minutes for evaluation of the endogenous
heart rate.
2. Discontinuation of chest compressions is indicated when the
heart rate is greater than 80 bpm and the pulse remains
palpable.
I. Umbilical vein catheterization
Cannulation of the umbilical vessels allows for both prompt
administration of resuscitation drugs and hemodynamic
monitoring. In addition, they provide a reliable conduit for the
administration of epinephrine and/or fluids. The umbilical artery
can be cannulated when frequent assessment of systemic blood
pressure or arterial blood gases is necessary.
1. A 3.5 or 5 French catheter is sterilely inserted into the vein at
the stump of the umbilical cord. The catheter is advanced 2 to
3 cm, at which point aspiration of blood should be possible.
The catheter must remain infrahepatic because hepatic
infusion of drugs or hypertonic solutions can cause hepatic
necrosis or portal vein thrombosis.
2. The tip of the umbilical artery catheter should be positioned
above the bifurcation of the aorta and below the celiac, renal,
and mesenteric arteries. In a term infant, advancement of the
catheter between 9 and 12 cm generally allows for optimal
positioning. Radiographic verification of the tip position is
indicated if the catheter is to remain in place for an extended
period.
J. Medications
1. Epinephrine
In approximately 1 in 2,000 deliveries where the heart rate
remains less than 60 bpm despite adequate ventilation and 30
seconds of chest compressions, epinephrine is the vasopressor
of choice (see Fig. 17.3). Administration of epinephrine leads
to increased myocardial blood flow from α-adrenergic-
mediated vasoconstriction and a subsequent increase in
coronary perfusion pressure.
a. Intravascular epinephrine is given at doses of 0.01 to 0.03
mg per kg and can be repeated every 3 to 5 minutes until
the heart rate is greater than 60 bpm (see Table 17.8).
Adequate ventilation must be established prior to its
administration because the α-and β-adrenergic activity of
epinephrine increases oxygen consumption and can lead
to myocardial damage in the setting of hypoxemia.
3. Sodium bicarbonate
Although acidemia impairs myocardial performance and may
attenuate the hemodynamic response to catecholamines, the
benefit of sodium bicarbonate infusions has been refuted by
several studies in adults and neonates.49
a. Among 19 retrospective adult studies examining mortality
rates and other outcomes, none demonstrated benefit: 11
showed no difference in outcomes, and 8 suggested a
deleterious effect of sodium bicarbonate administration
during cardiopulmonary resuscitation.50 One randomized
controlled trial of sodium bicarbonate use in neonatal
resuscitation demonstrated no difference in mortality rates
or neurologic outcomes.51 Recent AHA guidelines
highlight the adverse neurologic and cardiac effects linked
to bicarbonate administration.52
b. Exogenously administered sodium bicarbonate is readily
converted into carbon dioxide. It diffuses into myocardial
and cerebral cells and paradoxically contributes to
intracellular acidosis and reduced cellular function. The
extracellular alkalosis created by sodium bicarbonate
shifts the oxyhemoglobin saturation curve, further
worsening the intracellular acidosis by impeding oxygen
release to the tissues. Sodium bicarbonate infusions also
hinder cerebral and coronary perfusion by reducing SVR.
The strong relationship between intracranial hemorrhage
and rapid infusions of hyperosmolar sodium bicarbonate
further argue against its routine use. The administration of
sodium bicarbonate is no longer recommended during
initial neonatal resuscitation.
B. Premature infants
Although the survival rate of premature infants has steadily
increased over the past few decades,66,67 long-term morbidity and
mortality is still common in neonates less than 28 weeks’
gestational age.68 Appropriate resuscitation of this subset of
newborns requires an understanding of their unique physiologic
limitations. Although premature infants are at higher risk for
problems with multiple organ systems, the pulmonary and
cerebral systems deserve special consideration.
1. Fetal lung development during the third trimester is
characterized by the saccular development of terminal
bronchioles and the formation of surfactant by type II
pneumocytes. The reduction in pulmonary surface area and
surfactant production characteristic of immature lungs
contributes to difficulty in ventilation and increases
vulnerability to barotrauma during positive pressure
ventilation. In addition, the antioxidant defense system
develops late in gestation, and premature neonates may be
more susceptible to the adverse effects of excess oxygen. In
one study, preterm infants resuscitated with 30% oxygen had
less oxidative stress, inflammation, and chronic lung disease
compared to those resuscitated with 100% oxygen.69
However, room air appears to be insufficient for the
resuscitation of most neonates born at less than 28 weeks’
gestation.70–72 In another study, attempts to titrate oxygen
concentrations in preterm infants initially receiving room air
left several babies with uncertain or dangerously low levels of
oxygenation at 9 minutes of life.72 In an attempt to ensure
oxygenation while avoiding the complications of hyperoxia,
neonates <34 weeks should initially be resuscitated with 30%
oxygen. If oxygen saturation (SaO2) is less than 70% at 5
minutes of age or the heart rate is not increasing satisfactorily,
additional oxygen should be given.73
2. The cerebral architecture is often insufficiently developed in
the premature infant. Although the survival rate of infants
weighing less than 1,500 g is approximately 85%,74 a large
number of these infants display neurologic deficits later in
life. From 5% to 10% of very low birth weight infants
develop cerebral palsy and 25% to 50% exhibit behavioral
and cognitive deficits.75,76 The fragility of the immature
subependymal germinal matrix predisposes preterm neonates
to intraventricular hemorrhage (IVH), which is an early
marker for brain injury. Antenatal interventions, such as
corticosteroids and magnesium sulfate,77–79 and postnatal
management strategies can decrease the risk of IVH and
improve neurologic outcome in the premature infant.
Establishing adequate ventilation helps maintain the integrity
of the cerebral vasculature. Hypoxemia and hypercarbia have
been linked to disruption of cerebrovascular autoregulation
and the development of pressure-passive cerebral
circulation.76 In addition, the rapid infusion of volume
expanders and hypertonic solutions, such as sodium
bicarbonate during neonatal resuscitation has been shown to
increase the risk of IVH and thus neurologic injury.76
3. Hypothermia can also cause significant morbidity and
mortality in the premature infant. Preterm neonates are prone
to thermoregulatory problems. Thin skin and a large body
surface area contribute to rapid heat loss, whereas inadequate
brown fat stores limit nonshivering thermogenesis. Cold stress
in this population leads to increased oxygen consumption,
hypoglycemia, and metabolic acidosis, which hinders
resuscitative efforts. Additional warming techniques,
including wrapping in polyethylene bags and radiant warmers,
are recommended in preterm, low birth weight infants to
prevent hypothermia.80
D. Magnesium toxicity
Hypermagnesemia may be encountered in neonates born to
mothers treated with large doses of magnesium for preeclampsia
and eclampsia. Magnesium easily crosses the placenta and can
influence the neuromuscular and cardiac systems of newborns.
Specifically, hypermagnesemic infants are generally flushed,
hypotensive, hypotonic, and peripherally vasodilated. In rare
circumstances, hypermagnesemic infants require intubation and
mechanical ventilation because of poor respiratory effort. In the
presence of adequate renal function, magnesium levels normalize
over 24 to 48 hours. If severe hypotension is present, calcium can
be used as an antidote. The blood pressure of hypermagnesemic
neonates should increase with a bolus of 100 to 200 mg per kg of
calcium gluconate given over 5 minutes.
REFERENCES
1. American Heart Association. 2005 American Heart Association (AHA) guidelines for
cardiopulmonary resuscitation (CPR) and emergency cardiovascular care (ECC) of pediatric and
neonatal patients: neonatal resuscitation guidelines. Pediatrics. 2006;117:e1029–e1038.
2. Ostheimer GW. Anaesthetists’ role in neonatal resuscitation and care of the newborn. Can J Anaesth.
1993;40:R50–R62.
3. Rudolph AM, Heyman MA. Fetal and neonatal circulation and respiration. Annu Rev Physiol.
1974;36:187–207.
4. Wimmer JE Jr. Neonatal resuscitation. Pediatr Rev. 1994;15:255–265.
5. Swanson JR, Sinkin RA. Transition from fetus to newborn. Pediatr Clin North Am. 2015;62:329–343.
6. Lawson EE, Birdwell RL, Huang PS, et al. Augmentation of pulmonary surfactant secretion by lung
expansion at birth. Pediatr Res. 1979;13:611–614.
7. Lakshminrusimha S, Steinhorn RH. Pulmonary vascular biology during neonatal transition. Clin
Perinatol. 1999;26: 601–619.
8. Dawes G. Foetal and Neonatal Physiology: A Comparative Study of the Changes at Birth. Chicago,
IL: Year Book Medical; 1968:141–157.
9. Posen R, Friedlich P, Chan L, et al. Relationship between fetal monitoring and resuscitative needs:
fetal distress versus routine cesarean deliveries. J Perinatol. 2000;20:101–104.
10. Ray M, Freeman R, Pine S, et al. Clinical experience with the oxytocin challenge test. Am J Obstet
Gynecol. 1972;114:1–9.
11. Lagrew DC Jr. The contraction stress test. Clin Obstet Gynecol. 1995;38:11–25.
12. Manning FA, Platt LD, Sipos L. Antepartum fetal evaluation: development of a fetal biophysical
profile. Am J Obstet Gynecol. 1980;136:787–795.
13. Freeman RK, Anderson G, Dorchester W. A prospective multi-institutional study of antepartum fetal
heart rate monitoring. I. Risk of perinatal mortality and morbidity according to antepartum fetal heart
rate test results. Am J Obstet Gynecol. 1982;143:771–777.
14. Nageotte MP, Towers CV, Asrat T, et al. Perinatal outcome with the modified biophysical profile. Am J
Obstet Gynecol. 1994;170:1672–1676.
15. Vintzileos AM, Knuppel RA. Multiple parameter biophysical testing in the prediction of fetal acid-
base status. Clin Perinatol. 1994;21:823–848.
16. Schifrin BS, Dame L. Fetal heart rate patterns. Prediction of Apgar score. JAMA. 1972;219:1322–
1325.
17. 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.
18. Saling E. A new method for examination of the child during labor. Introduction, technic and principles
[in German]. Arch Gynakol. 1962;197:108–122.
19. Skupski DW, Rosenberg CR, Eglinton GS. Intrapartum fetal stimulation tests: a meta-analysis. Obstet
Gynecol. 2002;99:129–134.
20. Apgar V. A proposal for a new method of evaluation of the newborn infant. Curr Res Anesth Analg.
1953;32:260–267.
21. Casey BM, McIntire DD, Leveno KJ. The continuing value of the Apgar score for the assessment of
newborn infants. N Engl J Med. 2001;344:467–471.
22. Moster D, Lie RT, Irgens LM, et al. The association of Apgar score with subsequent death and cerebral
palsy: a population-based study in term infants. J Pediatr. 2001;138:798–803.
23. O’Donnell CP, Kamlin CO, Davis PG, et al. Interobserver variability of the 5-minute Apgar score. J
Pediatr. 2006;149: 486–489.
24. American Academy of Pediatrics, Committee on Fetus and Newborn, American College of
Obstetricians and Gynecologists, Committee on Obstetric Practice. The Apgar score. Pediatrics.
2006;117:1444–1447.
25. American College of Obstetricians and Gynecologists Committee on Obstetric Practice. ACOG
Committee Opinion No. 433: optimal goals for anesthesia care in obstetrics. Obstet Gynecol.
2009;113:1197–1199.
26. Adamson SK Jr, Gandy GM, James LS. The influence of thermal factors upon oxygen consumption of
the newborn human infant. J Pediatr. 1965;66:495–508.
27. Karlberg P. The breaths of life. In: Gluck L, ed. Modern Perinatal Medicine. Chicago, IL: Year Book
Medical; 1974:391–408.
28. Falck AJ, Escobedo MB, Baillargeon JG, et al. Proficiency of pediatric residents in performing
neonatal endotracheal intubation. Pediatrics. 2003;112:1242–1247.
29. Paterson SJ, Byrne PJ, Molesky MG, et al. Neonatal resuscitation using the laryngeal mask airway.
Anesthesiology. 1994;80:1248–1253.
30. Trevisanuto D, Micaglio M, Ferrarese P, et al. The laryngeal mask airway: potential applications in
neonates. Arch Dis Child Fetal Neonatal Ed. 2004;89:F485–F489.
31. Esmail N, Saleh M, Ali A. Laryngeal mask airway versus endotracheal intubation for Apgar score
improvement in neonatal resuscitation. Eg J Anaesth. 2002;18:115–121.
32. Probyn ME, Hooper SB, Dargaville PA, et al. Positive end expiratory pressure during resuscitation of
premature lambs rapidly improves blood gases without adversely affecting arterial pressure. Pediatr
Res. 2004;56:198–204.
33. Ramji S, Ahuja S, Thirupuram S, et al. Resuscitation of asphyxic newborn infants with room air or
100% oxygen. Pediatr Res. 1993;34:809–812.
34. Saugstad OD, Rootwelt T, Aalen O. Resuscitation of asphyxiated newborn infants with room air or
oxygen: an international controlled trial: the Resair 2 study. Pediatrics. 1998;102:e1.
35. Saugstad OD, Ramji S, Irani SF, et al. Resuscitation of newborn infants with 21% or 100% oxygen:
follow-up at 18 to 24 months. Pediatrics. 2003;112:296–300.
36. Saugstad OD, Ramji S, Vento M. Resuscitation of depressed newborn infants with ambient air or pure
oxygen: a meta-analysis. Biol Neonate. 2005;87:27–34.
37. Ramji S, Saugstad OD. Use of 100% oxygen or room air in neonatal resuscitation. Neoreviews.
2005;6:e172–e196.
38. Mendoza-Paredes A, Liu H, Schears G, et al. Resuscitation with 100%, compared with 21%, oxygen
following brief, repeated periods of apnea can protect vulnerable neonatal brain regions from
apoptotic injury. Resuscitation. 2008;76:261–270.
39. House JT, Schultetus RR, Gravenstein N. Continuous neonatal evaluation in the delivery room by
pulse oximetry. J Clin Monit. 1987;3:96–100.
40. Vento M, Asensi M, Sastre J, et al. Resuscitation with room air instead of 100% oxygen prevents
oxidative stress in moderately asphyxiated term neonates. Pediatrics. 2001;107:642–647.
41. Ten VS, Matsiukevich D. Room air or 100% oxygen for resuscitation of infants with perinatal
depression. Curr Opin Pediatr. 2009;21:188–193.
42. Niijima S, Shortland DB, Levene MI, et al. Transient hyperoxia and cerebral blood flow velocity in
infants born prematurely and at full term. Arch Dis Child. 1988;63:1126–1130.
43. Menegazzi JJ, Auble TE, Nicklas KA, et al. Two-thumb versus two-finger chest compression during
CRP in a swine infant model of cardiac arrest. Ann Emerg Med. 1993;22:240–243.
44. Barber CA, Wyckoff MH. Use and efficacy of endotracheal versus intravenous epinephrine during
neonatal cardiopulmonary resuscitation in the delivery room. Pediatrics. 2006;118:1028–1034.
45. Ralston SH, Voorhees WD, Babbs CF. Intrapulmonary epinephrine during prolonged cardiopulmonary
resuscitation: improved regional blood flow and resuscitation in dogs. Ann Emerg Med. 1984;13:79–
86.
46. Kleinman ME, Oh W, Stonestreet BS. Comparison of intravenous and endotracheal epinephrine during
cardiopulmonary resuscitation in newborn piglets. Crit Care Med. 1999;27:2748–2754.
47. Wyckoff MH, Perlman JM, Laptook AR. Use of volume expansion during delivery room resuscitation
in near-term and term infants. Pediatrics. 2005;115:950–955.
48. Oca MJ, Nelson M, Donn SM. Randomized trial of normal saline versus 5% albumin for the treatment
of neonatal hypotension. J Perinatol. 2003;23:473–476.
49. Aschner JL, Poland RL. Sodium bicarbonate: basically useless therapy. Pediatrics. 2008;122:831–835.
50. Levy MM. An evidence-based evaluation of the use of sodium bicarbonate during cardiopulmonary
resuscitation. Crit Care Clin. 1998;14:457–483.
51. Lokesh L, Kumar P, Murki S, et al. A randomized controlled trial of sodium bicarbonate in neonatal
resuscitation-effect on immediate outcome. Resuscitation. 2004;60:219–223.
52. Emergency Cardiovascular Care Committee. Subcommittees and Task Forces of the American Heart
Association. 2005 American Heart Association guidelines for cardiopulmonary resuscitation and
emergency cardiovascular care. Circulation. 2005;112:1–203.
53. Harrington DJ, Redman CW, Moulden M, et al. The long-term outcome in surviving infants with
Apgar zero at 10 minutes: a systematic review of the literature and hospital-based cohort. Am J Obstet
Gynecol. 2007;196:463.e1–463.e5.
54. Wiswell TE, Bent RC. Meconium staining and the meconium aspiration syndrome. Unresolved issues.
Pediatr Clin North Am. 1993;40:955–981.
55. Wiswell TE, Tuggle JM, Turner BS. Meconium aspiration syndrome: have we made a difference?
Pediatrics. 1990;85:715–721.
56. Gregory GA, Gooding CA, Phibbs RH, et al. Meconium aspiration in infants—a prospective study. J
Pediatr. 1974;85:848–852.
57. Carson BS, Losey RW, Bowes WA Jr, et al. Combined obstetric and pediatric approach to prevent
meconium aspiration syndrome. Am J Obstet Gynecol. 1976;126:712–715.
58. Jovanovic R, Nguyen HT. Experimental meconium aspiration in guinea pigs. Obstet Gynecol.
1989;73:652–656.
59. Cornish JD, Dreyer GL, Snyder GE, et al. Failure of acute perinatal asphyxia or meconium aspiration
to produce persistent pulmonary hypertension in a neonatal baboon model. Am J Obstet Gynecol.
1994;171:43–49.
60. Ghidini A, Spong CY. Severe meconium aspiration syndrome is not caused by aspiration of
meconium. Am J Obstet Gynecol. 2001;185:931–938.
61. Vain NE, Szyld EG, Prudent LM, et al. Oropharyngeal and nasopharyngeal suctioning of meconium-
stained neonates before delivery of their shoulders: multicentre, randomised controlled trial. Lancet.
2004;364:597–602.
62. Wiswell TE, Gannon CM, Jacob J, et al. Delivery room management of the apparently vigorous
meconium-stained neonate: results of the multicenter, international collaborative trial. Pediatrics.
2000;105:1–7.
63. Linder N, Aranda JV, Tsur M, et al. Need for endotracheal intubation and suction in meconium-stained
neonates. J Pediatr. 1988;112:613–615.
64. Falciglia HS, Henderschott C, Potter P, et al. Does DeLee suction at the perineum prevent meconium
aspiration syndrome? Am J Obstet Gynecol. 1992;167:1243–1249.
65. Velaphi S, Vidyasagar D. The pros and cons of suctioning at the perineum (intrapartum) and post-
delivery with and without meconium. Semin Fetal Neonatal Med. 2008;13:375–382.
66. Markestad T, Kaaresen PI, Rønnestad A, et al. Early death, morbidity, and need of treatment among
extremely premature infants. Pediatrics. 2005;115:1289–1298.
67. Fanaroff AA, Stoll BJ, Wright LL, et al. Trends in neonatal morbidity and mortality for very low
birthweight infants. Am J Obstet Gynecol. 2007;196:147.e1–147.e8.
68. Landmann E, Misselwitz B, Steiss JO, et al. Mortality and morbidity of neonates born at <26 weeks of
gestation (1998–2003). A population-based study. J Perinat Med. 2008;36:168–174.
69. Vento M, Moro M, Escrig R, et al. Preterm resuscitation with low oxygen causes less oxidative stress,
inflammation, and chronic lung disease. Pediatrics. 2009;124:e439–e449.
70. Wang CL, Anderson C, Leone TA, et al. Resuscitation of preterm neonates by using room air or 100%
oxygen. Pediatrics. 2008;121:1083–1089.
71. Escrig R, Arruza L, Izquierdo I, et al. Achievement of targeted saturation values in extremely low
gestational age neonates resuscitated with low or high oxygen concentrations: a prospective,
randomized trial. Pediatrics. 2008;121:875–881.
72. Dawson JA, Kamlin CO, Wong C, et al. Oxygen saturation and heart rate during delivery room
resuscitation of infants <30 weeks’ gestation with air or 100% oxygen. Arch Dis Child Fetal Neonatal
Ed. 2009;94:F87–F91.
73. Vento M, Saugstad OD. Resuscitation of the term and preterm infant. Semin Fetal Neonatal Med.
2010;15:216–222.
74. Hamilton BE, Miniño AM, Martin JA, et al. Annual summary of vital statistics: 2005. Pediatrics.
2007;119:345–360.
75. Wolke D, Meyer R. Cognitive status, language attainment, and prereading skills of 6-year-old very
preterm children and their peers: the Bavarian longitudinal study. Dev Med Child Neurol. 1999;41:94–
109.
76. Volpe JJ. Neurology of the Newborn. 5th ed. Philadelphia, PA: Saunders Elsevier; 2008.
77. Moïse AA, Wearden ME, Kozinetz CA, et al. Antenatal steroids are associated with less need for
blood pressure support in extremely premature infants. Pediatrics. 1995;95:845–850.
78. Nelson KB, Grether JK. Can magnesium sulfate reduce the risk of cerebral palsy in very low
birthweight infants? Pediatrics. 1995;95:263–269.
79. Hirtz DG, Nelson K. Magnesium sulfate and cerebral palsy in premature infants. Curr Opin Pediatr.
1998;10:131–137.
80. Watkinson M. Temperature control of premature infants in the delivery room. Clin Perinatol.
2006;33:43–53.
81. American Academy of Pediatrics. Committee on drugs. Naloxone use in newborns. Pediatrics.
1980;65:667–669.
82. Herschel M, Khoshnood B, Lass NA. Role of naloxone in newborn resuscitation. Pediatrics.
2000;106:831–834.
Postpartum Issues
Postcesarean Analgesia
Richard N.Wissler
I. Introduction
II. Multimodal therapy
A. Goals
B. Components of multimodal therapy
III. Medications: oral, systemic, neuraxial, and regional administration
A. Opioids
B. Local anesthetics
C. Nonsteroidal anti-inflammatory drugs
D. Acetaminophen
E. Other medications
F. Medications in breast milk
IV. Summary
KEYPOINTS
1. The use of multimodal analgesia and differing routes of administration
will increase overall postcesarean delivery (post-CD) analgesia and
reduce the incidence of unwanted side effects.
2. Opioids are the mainstay of post-CD analgesia. Intravenous patient-
controlled analgesia (PCA) opioid administration provides superior pain
relief compared to intermittent caregiver administration and may have
analgesic effectiveness comparable to neuraxial opioid administration.
3. Neuraxial opioids are the gold standard in providing post-CD analgesia
by decreasing overall opioid consumption, improving ambulation,
allowing earlier return of bowel function, and improving reductions in
breast milk levels of opioids compared to systemically administered
opioids.
4. Neuraxial opioids are associated with nausea, vomiting, urinary retention,
respiratory depression, and pruritus.
5. Continuous wound infiltration with local anesthetic using catheter
techniques may offer significant post-CD analgesia. The use of
transversus abdominis blockade does not offer significant analgesic
advantage when added to neuraxial opioid administration.
6. Nonsteroidal anti-inflammatory drugs, when administered orally or
intravenously and when added to local anesthetics used in neuraxial
blockade, are useful analgesic adjuncts to opioids. Acetaminophen may
offer significant analgesia and is safe for mother and child.
7. Predicting the effects of maternally administered medications that appear
in breast milk is complex. Consulting the published guidelines written by
professional organizations is recommended
I. Introduction
Pain relief after cesarean delivery (CD) has many of the same clinical
considerations as analgesia following other forms of abdominal
surgery. Additional concerns in the post-CD population include the
goal of minimizing maternal sedation to facilitate interactions with the
newborn, family, and friends; transfer of analgesic medications in
breast milk; and reducing time to discharge to home. CD is a very
common surgical procedure, and anesthesiologists should be familiar
with current post-CD analgesic techniques. More than 1.28 million
CD were performed in the United States in 2013, representing
32.7% of all births.1 As seen in the subsequent text, the anesthetic
method used for CD often influences the choices for postcesarean
analgesia. Both scheduled and unplanned CD have similar acute
postoperative pain scores and analgesic requirements.2
Acute postoperative or nociceptive pain following CD is a
combination of visceral pain from the uterus and somatic pain
from the abdominal wall. Recent research has focused on the
subsequent development of chronic neuropathic pain at the abdominal
incision site. Retrospective and prospective studies of chronic pain
after CD report an incidence range of 1% to 18%.3 At this time,
modifications of post-CD analgesia to decrease the incidence of
chronic incisional pain are investigational only. This chapter focuses
on current analgesic strategies for acute nociceptive post-CD pain.
However, some data suggest that effective treatment of acute
postoperative pain may minimize the development of chronic pain in
these patients.4
Effective post-CD analgesia is an attainable goal recommended
by several national professional organizations.5,6 One prospective
written survey of pregnant patients before delivery demonstrates that
pain during and after CD are the two most concerning anesthesia
outcomes.7 A prospective study of healthy parturients shows that
preoperative questions about anxiety, expected postoperative pain,
and expected needs of postoperative analgesic medications are
moderately predictive of post-CD analgesic requirements.8 However,
the practical application of preoperative questionnaires for routine
clinical practice is not clear.
II. Multimodal therapy
A. Goals. Utilizing several medications and/or routes of
administration that complement each other.5,9 The goals are:
1. Increasing the effectiveness of analgesia
2. Decreasing medication side effects by decreasing doses of
individual medications
B. Components of multimodal therapy. Examples of multimodal
therapy for post-CD analgesia include different combinations of
the following:
1. Systemic opioids
2. Systemic nonsteroidal anti-inflammatory drugs (NSAIDs)
3. Epidural medications
a. Opioids
b. Adjuncts
c. Local anesthetics
4. Intrathecal (IT) medications
a. Opioids
b. Adjuncts
5. Direct wound site treatments
a. Local anesthetics
b. NSAIDs
3. Epidural opioids
a. Specific medications studied include morphine,
meperidine, hydromorphone, fentanyl, sufentanil,
oxymorphone, methadone, nalbuphine, butorphanol,
buprenorphine, and diamorphine (not available in the
United States).
b. The rationale for neuraxial (i.e., epidural or spinal)
administration of opioids is to improve the analgesic
efficacy while minimizing the side effects of the opioids
when administered by other routes. These side effects
include sedation, respiratory depression, nausea and
vomiting, itching, and urinary retention. However, all
these side effects can occur with either systemic or
neuraxial opioids.
c. A single dose of epidural morphine:
(1) Provides better post-CD analgesia than a saline
placebo17 or IM opioids12,18
(2) Has similar analgesic efficacy and patient
satisfaction compared to IV PCA morphine12,18
(3) Is associated with more pruritus than IV PCA or
IM morphine
(4) May be less effective than if given by continuous
infusion19
d. Studies of PCEA in patients receiving post-CD analgesia
with fentanyl, sufentanil, or hydromorphone demonstrate:
(1) Better pain relief, less medication administration, and
more pruritus compared to IV PCA opioids20,21
(2) No benefit of continuous basal infusion PCEA versus
demand-only PCEA22,23
e. A single dose of epidural fentanyl should be diluted to an
injection volume of at least 10 mL for the best effect.24
However, injection volume variations with single doses of
epidural morphine do not have a significant effect on
post-CD analgesic efficacy.25
f. Extended-release epidural morphine
(1) It was approved by the U.S. Food and Drug
Administration (FDA) in 2004 for postoperative
analgesia, with the trade name of DepoDur.26
(2) Two prospective randomized studies have compared
DepoDur® to conventional morphine for post-CD
epidural analgesia.27,28 The patients who received
DepoDur had improved pain scores at rest and with
movement as well as improved functional ability. The
differences between DepoDur and conventional
morphine were restricted to the 24 to 48 hour
postoperative time frame, but the magnitude of
those differences was clinically modest.
(3) Although it is approved for clinical use, it is currently
not being produced by the pharmaceutical company,
Pacira, who currently own the license. They are using
the carrier, DepoFoam to manufacture the long acting
local anesthetic preparation, Exparel.
g. Table 18.2 lists the recommended single doses of
conventional epidural opioids, based on prospective
randomized studies.
h. Adjuncts and epidural opioids
(1) Clonidine (75 to 150 µg) improves post-CD
analgesia when added to single epidural doses of
fentanyl or morphine.29,30 However, there is an FDA
black box warning for clonidine. “Duraclon®
(epidural clonidine) is not recommended for
obstetric, postpartum, or perioperative pain
management . . . ” Use of clonidine alone does not
offer significant analgesic improvement over
neuraxial opioid use, and the associated side effects
of frequent sedation and hypotension mitigate against
its routine use.
(2) Epinephrine has demonstrated inconsistent effects in
improving analgesia and reducing side effects when
added to single doses of epidural opioids.
(3) The use of N-methyl-D-aspartate (NMDA)
antagonists (e.g., ketamine, magnesium), although
safe and effective in producing analgesia in a very
limited number of studies, should be considered
experimental at present.
4. Intrathecal Opioids
a. Specific medications studied include morphine,
meperidine, fentanyl, sufentanil, nalbuphine, butorphanol,
and buprenorphine.
b. In contrast to systemic and epidural routes of
administration, IT administration of opioids does not
lead to clinically significant maternal serum
concentrations of these agents.31,32
c. In most circumstances, IT opioid administration implies a
“single-shot” opioid delivery as part of a spinal or
combined spinal-epidural (CSE) anesthetic. A number of
IT local anesthetics have been combined successfully with
IT opioids including lidocaine, bupivacaine, tetracaine,
mepivacaine, and ropivacaine.
d. Direct comparisons of the IT and epidural routes of
administration for single-dose morphine show similar
efficacy for post-CD analgesia.31,32 However, the
recommended dose is reduced by approximately 20-fold
in the IT space.
e. In many instances, pruritus is more prevalent and intense
with IT opioids, compared with either epidural or
systemic administration.
f. Table 18.3 lists the recommended single doses of IT
opioids based on the best available evidence.
B. Local anesthetics
1. Epidural
a. Local anesthetics, alone or in combination with opioids,
may be infused by the lumbar epidural route for post-CD
analgesia. The infusion strategy may be continuous or by
PCEA, analogous to postoperative analgesia regimens for
other lower abdominal surgeries.5
b. Adding a local anesthetic to an epidural opioid infusion
may have a dose-sparing effect,42 but it can result in
lower extremity motor block and delayed ambulation in
women after CD.43
2. Iliohypogastric and ilioinguinal peripheral nerve blocks
a. These two peripheral nerves originate from the L1 spinal
nerve root and innervate the abdominal wall region that
corresponds to a lower transverse cesarean incision. The
blocks need to be performed bilaterally.
b. The existing literature with single-shot nerve blocks
presents conflicting results about the effectiveness of
perioperative placement of these blocks in decreasing
subsequent supplemental opioid use after low transverse
CD.44–46
c. A case series of ultrasound-guided continuous ilioinguinal
and iliohypogastric nerve blocks in post-CD patients
describes apparent opioid-sparing in the postoperative
period.47
3. Transversus abdominis plane blocks
a. A transversus abdominis plane (TAP) block is a lower
abdominal wall block accomplished by placing local
anesthetic medications in the fascial plane between the
internal oblique and the transversus abdominis muscles. It
must be performed bilaterally and uses approximately 20
mL of local anesthetic per side. Although initially
described using surface anatomy landmarks, it is now
placed with real-time ultrasound guidance.
b. When used after spinal anesthesia for CD, the TAP block
adds value only when IT morphine is omitted from a
multimodal analgesic regimen.48,49 A prospective direct
comparison study in post-CD patients demonstrates that
IT morphine is a more effective analgesic technique
compared to a TAP block, but with a higher side effect
profile.50 A case series in this patient population describes
a continuous bilateral TAP block technique using 20
gauge epidural catheters placed with ultrasound
guidance.51
c. The role of TAP blocks in for post-CD analgesia is
evolving. Their best use maybe in treating pain after
general anesthesia or as rescue treatment after spinal
anesthesia52 when neuraxial opioids have failed or are not
indicated. Adding 75 µg of clonidine to each side of a
post-CD local anesthetic TAP block did not improve its
performance in a multimodal analgesic regimen that
included IT morphine.53
4. Surgical wound infiltration
a. Single-injection direct surgical wound infiltration with 20
mL of bupivacaine 0.5% at wound closure does not
significantly change postoperative supplemental analgesic
use after low transverse CD54,55; however, injection of
local anesthetic at the fascial level during wound closure
decreases opioid requirements during the first 12 hours of
recovery.56
b. Postoperative wound infusion, using a catheter
inserted at the time of surgery, is an effective post-CD
analgesic technique.57,58 Direct comparisons show
postoperative wound infusions with local anesthetics are
more effective than epidural morphine and similar to
systemic diclofenac.58,59 Catheter infusions are most
effective when located deep to the fascia at the time of
wound closure60 and should be placed using ultrasound
guidance to improve success.
c. Postoperative wound infusions with local anesthesia affect
specific inflammatory mediators measured in wound
exudates, suggesting an interaction with the wound
healing process.61
d. It remains to be seen if infiltration of the wound with
Exparel®, the long acting local anesthetic product,
improves post-CD analgesia, especially on day 2
postoperatively
E. Other medications
1. Ketamine
CD patients who received IV ketamine in subhypnotic doses
during either spinal or general anesthesia had only transient
small decreases in postoperative pain or none at all.67–69 One
study described a modest decrease in pain scores 2 weeks
after surgery in the ketamine group.69
2. Gabapentin
Perioperative oral dosing with gabapentin does not produce
clinically meaningful post-CD analgesia. In addition, it is
associated with increased sedation.70,71
3. Magnesium when administered intravenously demonstrates
minimal reduction in pain scores and modest reduction in
opioid use in non-pregnant women after surgery but has not
been evaluated in women after CD.72
IV. Summary
A. Multimodal therapy is the current and future direction for post-
CD analgesia.
B. Opioids have been, and continue to be, the dominant medications
for pain relief after CD despite concerns about patient safety.
C. IM route of administration should be considered only as a last
resort.
D. NSAIDs have a widespread role in post-CD analgesia, frequently
in addition to opioids.
E. Acetaminophen’s use as a postoperative medication continues to
evolve, particularly in reference to IV versus other routes of
administration. The total dose in different formulations needs to
be appreciated to minimize the risk of toxicity.
F. Patient-controlled catheter infusions are promising techniques and
can be used for postoperative wound infiltration with a local
anesthetic and/or NSAID.
G. Analgesic medications are mostly compatible with breastfeeding.
As discussed earlier, some concerns have been expressed
concerning the administration of meperidine, codeine, or
oxycodone to patients who are breastfeeding, based on different
mechanisms of drug metabolism.
H. In general, postoperative pain management is an opportunity for
anesthesiologists to collaborate with obstetricians and obstetric
nurses. We have expertise to share that will improve the
postoperative experience for many patients after CD. New
initiatives for post-CD analgesia are unlikely to succeed without
the prior knowledge and support of our clinical obstetric and
nursing colleagues. Also, in the current regulatory climate,
hospitals will appreciate assistance in improving patient
satisfaction scores.
REFERENCES
1. Martin JA, Hamilton BE, Osterman MJ, et al. Births: final data for 2013. Natl Vital Stat Rep. 2015;
64:1–65.
2. Carvalho B, Coleman L, Saxena A, et al. Analgesic requirements and postoperative recovery after
scheduled compared to unplanned cesarean delivery: a retrospective chart review. Int J Obstet Anesth.
2010;19:10–15.
3. Landau R, Bollag L, Ortner C. Chronic pain after childbirth. Int J Obstet Anesth. 2013;22:133–145.
4. Eisenach JC, Pan P, Smiley RM, et al. Resolution of pain after childbirth. Anesthesiology.
2013;118:143–151.
5. American Society of Anesthesiologists Task Force on Acute Pain Management. Practice guidelines for
acute pain management in the perioperative setting: an updated report by the American Society of
Anesthesiologists Task Force on Acute Pain Management. Anesthesiology. 2012;116:248–273.
6. American College of Obstetricians and Gynecologists. Guidelines for Perinatal Care. 7th ed.
Washington, DC: American College of Obstetricians and Gynecologists; 2012.
7. Carvalho B, Cohen SE, Lipman SS, et al. Patient preferences for anesthesia outcomes associated with
cesarean delivery. Anesth Analg. 2005;101:1182–1187.
8. Pan PH, Tonidandel AM, Aschenbrenner CA, et al. Predicting acute pain after cesarean delivery using
three simple questions. Anesthesiology. 2013;118:1170–1179.
9. Tan M, Law LS, Gan TJ. Optimizing pain management to facilitate enhanced recovery after surgery
pathways. Can J Anaesth. 2015;62:203–218.
10. Wittels B, Scott DT, Sinatra RS. Exogenous opioids in human breast milk and acute neonatal
neurobehavior: a preliminary study. Anesthesiology. 1990;73:864–869.
11. Al-Tamimi Y, Ilett KF, Paech MJ, et al. Estimation of infant dose and exposure to pethidine and
norpethidine via breast milk following patient-controlled epidural pethidine for analgesia post
cesarean delivery. Int J Obstet Anesth. 2011;20:128–134.
12. Eisenach JC, Grice SC, Dewan DM. Patient-controlled analgesia following cesarean section: a
comparison with epidural and intramuscular narcotics. Anesthesiology. 1988;68:444–448.
13. Sinatra R, Chung KS, Silverman DG, et al. An evaluation of morphine and oxymorphone administered
via patient-controlled analgesia (PCA) or PCA plus basal infusion in postcesarean-delivery patients.
Anesthesiology. 1989;71:502–507.
14. Parker RK, Holtmann B, White PF. Effects of a nighttime opioid infusion with PCA therapy on patient
comfort and analgesic requirements after abdominal hysterectomy. Anesthesiology. 1992;76:362–367.
15. Lam FY. Patient-controlled analgesia by proxy. Br J Anaesth. 1993;70:113.
16. Vicente KJ, Kada-Bekhaled K, Hillel G, et al. Programming errors contribute to death from patient-
controlled analgesia: case report and estimate of probability. Can J Anaesth. 2003;50:328–332.
17. Binsted RJ. Epidural morphine after caesarean section. Anaesth Intensive Care. 1983;11:130–134.
18. Harrison DM, Sinatra R, Morgese L, et al. Epidural narcotic and patient-controlled analgesia for post-
cesarean section pain relief. Anesthesiology. 1988;68:454–457.
19. Rauck RL, Raj PP, Knarr DC, et al. Comparison of the efficacy of epidural morphine given by
intermittent injection or continuous infusion for the management of postoperative pain. Reg Anesth.
1994;19:316–324.
20. Cooper DW, Ryall DM, Desira WR. Extradural fentanyl for postoperative analgesia: predominant
spinal or systemic action? Br J Anaesth. 1995;74:184–187.
21. Ngan Kee WD, Lam KK, Chen PP, et al. Comparison of patient-controlled epidural analgesia with
patient-controlled intravenous analgesia using pethidine or fentanyl. Anaesth Intensive Care.
1997;25:126–132.
22. Parker RK, Sawaki Y, White PF. Epidural patient-controlled analgesia: influence of bupivacaine and
hydromorphone basal infusion on pain control after cesarean delivery. Anesth Analg. 1992;75:740–
746.
23. Vercauteren MP, Coppejans HC, ten Broecke PW, et al. Epidural sufentanil for postoperative patient-
controlled analgesia (PCA) with or without background infusion: a double-blind comparison. Anesth
Analg. 1995;80:76–80.
24. Birnbach DJ, Johnson MD, Arcario T, et al. Effect of diluent volume on analgesia produced by
epidural fentanyl. Anesth Analg. 1989;68:808–810.
25. Asantila R, Eklund P, Rosenberg PH. Epidural analgesia with 4 mg of morphine following caesarean
section: effect of injected volume. Acta Anaesthesiol Scand. 1993;37:764–767.
26. U.S. Food and Drug Administration. Product label for DepoDur.
www.accessdata.fda.gov/drugsatfda_docs/label/2009/021671s020lbl.pdf. Accessed April 30, 2015.
27. Carvalho B, Riley E, Cohen SE, et al. Single-dose, sustained-release epidural morphine in the
management of postoperative pain after elective cesarean delivery: results of a multicenter randomized
controlled study. Anesth Analg. 2005;100: 1150–1158.
28. Carvalho B, Roland LM, Chu LF, et al. Single-dose, extended-release epidural morphine (DepoDur)
compared to conventional epidural morphine for post-cesarean pain. Anesth Analg. 2007;105:176–
183.
29. Eisenach JC, D’Angelo R, Taylor C, et al. An isobolographic study of epidural clonidine and fentanyl
after cesarean section. Anesth Analg. 1994;79:285–290.
30. Capogna G, Celleno D, Zangrillo A, et al. Addition of clonidine to epidural morphine enhances
postoperative analgesia after cesarean delivery. Reg Anesth. 1995;20:57–61.
31. Sarvela J, Halonen P, Soikkeli A, et al. A double-blinded, randomized comparison of intrathecal and
epidural morphine for elective cesarean delivery. Anesth Analg. 2002; 95:436–440.
32. Dualé C, Frey C, Bolandard F, et al. Epidural versus intrathecal morphine for postoperative analgesia
after caesarean section. Br J Anaesth. 2003;91:690–694.
33. Paech MJ, Pavy TJ, Orlikowski CE, et al. Postcesarean analgesia with spinal morphine, clonidine, or
their combination. Anesth Analg. 2004;98:1460–1466.
34. Lavand’homme PM, Roelants F, Waterloos H, et al. An evaluation of the postoperative
antihyperalgesic and analgesic effects of intrathecal clonidine administered during elective cesarean
delivery. Anesth Analg. 2008;107:948–955.
35. van Tuijl I, van Klei WA, van der Werff DB, et al. The effect of addition of intrathecal clonidine to
hyperbaric bupivacaine on postoperative pain and morphine requirements after cesarean section: a
randomized controlled trial. Br J Anaesth. 2006;97:365–370.
36. Chung CJ, Kim JS, Park HS, et al. The efficacy of intrathecal neostigmine, intrathecal morphine, and
their combination for post-cesarean section analgesia. Anesth Analg. 1998;87:341–346.
37. American Society of Anesthesiologists Task Force on Neuraxial Opioids, Horlocker TT, Burton AW,
et al. Practice guidelines for the prevention, detection, and management of respiratory depression
associated with neuraxial opioid administration. Anesthesiology. 2009;110:218–230.
38. Crowgey TR, Dominguez JE, Peterson-Layne C, et al. A retrospective assessment of the incidence of
respiratory depression after neuraxial morphine administration for postcesarean delivery analgesia.
Anesth Analg. 2013;117:1368–1370.
39. Dominguez JE, Habib AS. Prophylaxis and treatment of the side-effects of neuraxial morphine
analgesia following cesarean delivery. Curr Opin Anesthesiol. 2013;26:288–295.
40. Luthman JA, Kay NH, White JB. Intrathecal morphine for post cesarean section analgesia: does
naloxone reduce the incidence of pruritus? Int J Obstet Anesth. 1992;1:191–194.
41. Charuluxananan S, Kyokong O, Somboonviboon W, et al. Nalbuphine versus propofol for treatment of
intrathecal morphine-induced pruritus after cesarean delivery. Anesth Analg. 2001;93:162–165.
42. Cooper DW, Ryall DM, McHardy FE, et al. Patient-controlled extradural analgesia with bupivacaine,
fentanyl, or a mixture of both, after caesarean section. Br J Anaesth. 1996;76:611–615.
43. Cohen S, Amar D, Pantuck CB, et al. Adverse effects of epidural 0.03% bupivacaine during analgesia
after cesarean section. Anesth Analg. 1992;75:753–756.
44. Huffnagle HJ, Norris MC, Leighton BL, et al. Ilioinguinal iliohypogastric nerve blocks—before or
after cesarean delivery under spinal anesthesia? Anesth Analg. 1996;82:8–12.
45. Bell EA, Jones BP, Olufolabi AJ, et al. Iliohypogastric-ilioinguinal peripheral nerve block for post-
cesarean delivery analgesia decreases morphine use but not opioid-related side effects. Can J Anaesth.
2002;49:694–700.
46. Vallejo MC, Steen TL, Cobb BT, et al. Efficacy of the bilateral ilioinguinal-iliohypogastric block with
intrathecal morphine for postoperative cesarean delivery analgesia. Sci World J. 2012;2012:107316.
doi: 10.1100/2012/107316.
47. Gucev G, Yasui GM, Chang TY, et al. Bilateral ultrasound-guided continuous ilioinguinal-
iliohypogastric block for pain relief after cesarean delivery. Anesth Analg. 2008;106:1220–1222.
48. McDonnell JG, Curley G, Carney J, et al. The analgesic efficacy of transversus abdominis plane block
after cesarean delivery: a randomized controlled trial. Anesth Analg. 2008;106:186–191.
49. Costello JF, Moore AR, Wieczorek PM, et al. The transversus abdominis plane block, when used as
part of a multimodal regimen inclusive of intrathecal morphine, does not improve analgesia after
cesarean delivery. Reg Anesth Pain Med. 2009;34:586–589.
50. Kanazi GE, Aouad MT, Abdallah FW, et al. The analgesic efficacy of subarachnoid morphine in
comparison with ultrasound-guided transversus abdominis plane block after cesarean delivery: a
randomized controlled trial. Anesth Analg. 2010;111:475–481.
51. Bollag L, Richebe P, Ortner C, et al. Transversus abdominis plane catheters for post-cesarean delivery
analgesia: a series of five cases. Int J Obstet Anesth. 2012;21:176-180.
52. Mirza F, Carvalho B. Transversus abdominis plane blocks for rescue analgesia following cesarean
delivery: a case series. Can J Anaesth. 2013;60:299–303.
53. Bollag L, Richebe P, Siaulys M, et al. Effect of transversus abdominis plane block with and without
clonidine on post-cesarean delivery wound hyperalgesia and pain. Reg Anesth Pain Med.
2012;37:508–514.
54. Trotter TN, Hayes-Gregson P, Robinson S, et al. Wound infiltration of local anesthetic after lower
segment caesarean section. Anaesthesia. 1991;46:404–407.
55. Pavy T, Gambling D, Kliffer P, et al. Effect of preoperative skin infiltration with 0.5% bupivacaine on
postoperative pain following cesarean section under spinal anesthesia. Int J Obstet Anesth.
1994;3:199–202.
56. Niklasson B, Borjesson A, Carmnes UB, et al. Intraoperative injection of bupivacaine-adrenaline close
to the fascia reduces morphine requirements after cesarean section: a randomized controlled trial. Acta
Obstet Gynecol Scand. 2012;91: 1433–1439.
57. Ranta PO, Ala-Kokko TI, Kukkonen JE, et al. Incisional and epidural analgesia after caesarean
delivery: a prospective, placebo-controlled, randomised clinical study. Int J Obstet Anesth.
2006;15:189–194.
58. O’Neill P, Duarte F, Ribeiro I, et al. Ropivacaine continuous wound infusion versus epidural morphine
for postoperative analgesia after cesarean delivery: a randomized controlled trial. Anesth Analg.
2012;114:179–185.
59. Zohar E, Shapiro A, Eidinov A, et al. Postcesarean analgesia: the efficacy of bupivacaine wound
installation with and without supplemental diclofenac. J Clin Anesth. 2006;18:415–421.
60. Rackelboom T, Le Strat S, Silvera S, et al. Improving continuous wound infusion effectiveness for
postoperative analgesia after cesarean delivery: a randomized controlled trial. Obstet Gynecol.
2010;116:893–900.
61. Carvalho B, Clark DJ, Yeomans DC, et al. Continuous subcutaneous instillation of bupivacaine
compared to saline reduces interleukin 10 and increases substance P in surgical wounds after cesarean
delivery. Anesth Analg. 2010;111:1452–1459.
62. Nauta M, Landsmeer MLA, Koren G. Codeine-acetaminophen versus nonsteroidal anti-inflammatory
drugs in the treatment of post-abdominal surgery pain: a systemic review of randomized trials. Am J
Surg. 2009;198:256–261.
63. Lavand’homme PM, Roelants F, Waterloos H, et al. Postoperative analgesic effects of continuous
wound infiltration with diclofenac after elective cesarean delivery. Anesthesiology. 2007;106:1220–
1225.
64. Carvalho B, Lemmens HJ, Ting V, et al. Postoperative subcutaneous instillation of low-dose ketorolac
but not hydromorphone reduces wound exudate concentrations of interleukin-6 and interleukin-10 and
improves analgesia following cesarean delivery. J Pain. 2013;14:48–56.
65. Davis KM, Esposito MA, Meyer BA. Oral analgesia compared with intravenous patient-controlled
analgesia for pain after cesarean delivery: a randomized controlled trial. Am J Obstet Gynecol.
2006;194:967–971.
66. Alhashemi JA, Alotaibi QA, Mashaat MS, et al. Intravenous acetaminophen vs oral ibuprofen in
combination with morphine PCIA after cesarean delivery. Can J Anaesth. 2006;53:1200–1206.
67. Menkiti ID, Desalu I, Kushimo OT. Low-dose intravenous ketamine improves postoperative analgesia
after caesarean delivery with spinal bupivacaine in African parturients. Int J Obstet Anesth.
2012;21:217–221.
68. Reza FM, Zahra F, Esmaeel F, et al. Preemptive analgesic effect of ketamine in patients undergoing
elective cesarean section. Clin J Pain. 2010;26:223–226.
69. Bauchat JR, Higgins N, Wojciechowski KG, et al. Low-dose ketamine with multimodal postcesarean
delivery analgesia: a randomized controlled trial. Int J Obstet Anesth. 2011;20:3–9.
70. Monks DT, Hoppe DW, Downey K, et al. A perioperative course of gabapentin does not produce a
clinically meaningful improvement in analgesia after cesarean delivery: a randomized controlled trial.
Anesthesiology. 2015;123:320–326. doi:10.1097/ALN.0000000000000722.
71. Short J, Downey K, Bernstein P, et al. A single preoperative dose of gabapentin does not improve
postcesarean delivery pain management: a randomized, double-blind, placebo-controlled dose-finding
trial. Anesth Analg. 2012;115:1336–1342.
72. De Oliveria GS, Castro-Alves LJ, Khan JH, et al. Perioperative systemic magnesium to minimize
postoperative pain: a meta-analysis of randomized controlled trials. Anesthesiology. 2013;119:178–
190.
73. Madadi P, Shirazi F, Walter FG, et al. Establishing causality of CNS depression in breastfed infants
following maternal codeine use. Paediatr Drugs. 2008;10:399–404.
74. VanderVaart S, Berger H, Sistonen J, et al. CYP2D6 polymorphisms and codeine analgesia in
postpartum pain management: a pilot study. Ther Drug Monit. 2011;33:425–432.
75. Lam J, Kelly L, Matok I, et al. Putative association of ABCB1 2677G>T/A with oxycodone-induced
central nervous system depression in breastfeeding mothers. Ther Drug Monit. 2013;35:466–472.
76. American Academy of Pediatrics Committee on Drugs. Transfer of drugs and other chemicals into
human milk. Pediatrics. 2001;108:776–789.
77. Montgomery A, Hale TW, Academy of Breastfeeding Medicine Protocol Committee. ABM clinical
protocol #15: analgesia and anesthesia for the breastfeeding mother. Breastfeed Med. 2006;1:271–277.
78. Lawrence RA, Lawrence RM. Breastfeeding: A Guide for the Medical Profession. 7th ed.
Philadelphia, PA: Saunders; 2010.
79. Briggs GG, Freeman RK. Drugs in Pregnancy and Lactation: A Reference Guide to Fetal and
Neonatal Risk. 10th ed. Baltimore, MD: Lippincott Williams & Wilkins; 2014.
80. Weiner CP, Buhimschi C. Drugs for Pregnant and Lactating Women. 2nd ed. Philadelphia, PA:
Saunders; 2009.
Management of Postdural Puncture Headache
David Wlody
I. Scope of the problem
II. Pathophysiology
A. Meningeal traction
B. Cerebral vasodilation
III. Risk factors for postdural puncture headache
A. Unmodifiable risk factors
B. Modifiable risk factors
IV. Diagnosis of postdural puncture headache
A. Occurrence of postdural puncture headache
B. Clinical characteristics
V. Differential diagnosis of postpartum headache
A. Migraine
B. Tension headache
C. Intracranial hemorrhage
D. Cerebral venous and sinus thrombosis
E. Neoplasm
F. Medications/substance withdrawal
G. Preeclampsia
H. Meningitis
I. Posterior reversible leukoencephalopathy syndrome
J. Benign intracranial hypertension (pseudotumor cerebri)
K. Spontaneous intracranial hypotension
L. Lactation headache
VI. Prevention and treatment of postdural puncture headache after
accidental dural puncture
A. Bed rest
B. Hydration
C. Prone position
D. Abdominal binder
E. Caffeine
F. Serotonin agonists
G. Corticosteroids/adrenocorticotropic hormone
H. Pregabalin and gabapentin
I. Acupuncture
J. Intrathecal saline
K. Intrathecal catheter
L. Epidural morphine
M. Epidural saline
N. Epidural blood patch
O. Prophylactic epidural blood patch
P. Epidural colloids
VII. Recommendations
A. Ambulation
B. Hydration
C. Analgesics
D. Pharmacologic therapy
E. Epidural blood patch
F. Prevention after known accidental dural puncture
Summary
KEYPOINTS
1. Far from being a minor complication, postdural puncture headache
(PDPH) can significantly increase the cost of hospitalization, can have an
extremely negative impact on patient satisfaction with a consequent
increase in the risk of litigation, and can lead to significant increases in
both short-and long-term maternal morbidity.
2. When evaluating patients with presumed PDPH, it is critically important
to rule out other potentially life-threatening causes of headache.
3. Numerous pharmacologic treatments for PDPH have been proposed, but
there is little evidence to support the routine use of a specific drug.
4. Although evidence supporting the practice is inconclusive, placement of
an intrathecal catheter after accidental dural puncture may decrease the
risk of developing PDPH.
5. Although not without its own complications, epidural blood patch (EBP)
represents the gold standard for treatment of established PDPH. The
routine use of prophylactic EBP and the optimal timing of EBP remain
controversial.
VII. Recommendations
PDPH can be debilitating, can cause serious morbidity, and may result
in litigation. In view of the consequences of PDPH, the
anesthesiologist should make every effort to minimize the risk of
headache by optimizing the factors that can be controlled, namely
needle size and shape. Despite all efforts, however, PDPH will
continue to occur, and anesthesia providers will continue to be called
upon to manage them. Unfortunately, despite many years of research,
the optimal treatment of PDPH is unclear. What follows then is one
suggested management approach based on a summary of the literature
and also on personal experience.
A. Ambulation. In patients who develop spinal headache,
ambulation should not be restricted because bed rest has no
demonstrated effect on the duration of spinal headache, while at
the same time increasing the risk of thromboembolism in patients
who are already at high risk for that complication. Patients should
be encouraged to ambulate as much as they can tolerate.
B. Hydration. Although forced hydration is unlikely to augment
CSF production to any significant degree, dehydration will
worsen the headache, and IV fluids should be provided to
patients who are unable to maintain adequate oral intake.
C. Analgesics. Oral analgesics should be made available; in
severe headache, opioid analgesics may be required and should be
provided.
D. Pharmacologic therapy. In patients who decline or who cannot
receive an EBP, pharmacologic therapy should be considered.
Caffeine can be given; if the IV preparation is available, one or
two doses of caffeine benzoate 500 mg should be administered.
Otherwise, 300 mg of oral caffeine can be administered every 6
hours. In many patients, headache will return after cessation of
caffeine therapy; thus, more definitive treatment will often be
necessary. Other pharmacologic therapies such as gabapentin,
cosyntropin, and serotonin agonists are of unproven utility but
may be considered in patients who are unresponsive to or
intolerant of caffeine and who remain unwilling or unable to
undergo more definitive therapy (EBP). The administration of
either intrathecal or epidural morphine might be of benefit.
E. Epidural blood patch
1. My practice is to wait at least 24 hours after the onset of
symptoms before considering a blood patch, because some
headaches may resolve by that time, and I would prefer to
avoid the possible complications of EBP in headaches that
resolve so quickly. There are exceptions: In patients with a
debilitating headache due to accidental dural puncture with a
large epidural needle, the likelihood of rapid spontaneous
resolution is small, and I will perform EBP soon after the
development of symptoms. It should be remembered,
however, that EBP performed within 24 hours of dural
puncture has a lower success rate; whether this is because
headaches treated within 24 hours are more severe, and
therefore more likely to lead to failed blood patch, or because
there is an intrinsic increased failure rate with early blood
patch, is unclear.
SUMMARY
Dural puncture is only one of many causes of postpartum headache. Patients
with PDPH have a strong postural component to the headache. The supine
position is associated with relief or improvement of headache, whereas sitting up
or standing up is associated with a worsening of the headache. The headache is
classically frontal and occipital, with radiation to the neck and shoulders, and
photophobia and/or tinnitus can occur with severe PDPH. Initial therapy should
be conservative, aimed at maintaining adequate levels of hydration and by
administering oral or IV analgesics. The administration of caffeine, cosyntropin,
and neuraxial morphine can be considered as well. For persistent or severe
headaches, an EBP will provide a definitive treatment for most women; however,
EBP can fail and may need to be repeated.
RECOMMENDED READING
Baysinger CL. Accidental dural puncture and postdural puncture headache management. Int Anesthesiol
Clin. 2014;52:18–39.
Bradbury CL, Singh SI, Badder SR, et al. Prevention of post-dural puncture headache in parturients: a
systematic review and meta-analysis. Acta Anaesthesiol Scand. 2013;57:417–430.
Sachs A, Smiley R. Post-dural puncture headache: the worst common complication in obstetric anesthesia.
Semin Perinatol. 2014;38:386–394.
Turnbull DK, Shepherd DB. Post-dural puncture headache: pathogenesis, prevention and treatment. Br J
Anaesth. 2003;91: 718–729.
REFERENCES
1. Gerritse BM, Gielen MJ. Seven months delay for epidural blood patch in post-dural puncture
headache. Eur J Anaesthesiol. 1999;16:650–651.
2. Béchard P, Perron G, Larochelle D, et al. Case report: epidural blood patch in the treatment of
abducens palsy after a dural puncture. Can J Anaesth. 2007;54:146–150.
3. Zeidan A, Farhat O, Maaliki H, et al. Does postdural puncture headache left untreated lead to subdural
hematoma? Case report and a review of the literature. Int J Obstet Anesth. 2006;15:50–58.
4. Davies JM, Posner KL, Lee LA, et al. Liability associated with obstetric anesthesia: a closed claims
analysis. Anesthesiology. 2009;110:131–139.
5. Sachs A, Smiley R. Post-dural puncture headache: the worst common complication in obstetric
anesthesia. Semin Perinatol. 2014;38:386–394.
6. Bier A. Versuche über Cocainisirung des Rückenmarkes [in German]. Dtsch Zeitschr f Chir.
1899;51:361–369.
7. Bakshi R, Mechtler LL, Kamran S, et al. MRI findings in lumbar puncture headache syndrome:
abnormal dural-meningeal and dural venous sinus enhancement. Clin Imaging. 1999;23:73–76.
8. Wu CL, Rowlingson AJ, Cohen SR, et al. Gender and post-dural puncture headache. Anesthesiology.
2006;105:613–618.
9. Stride PC, Cooper GM. Dural taps revisited: a 20-year survey from Birmingham Maternity Hospital.
Anaesthesia. 1993;48:247–255.
10. Angle P, Thompson D, Halpern S, et al. Second stage pushing correlates with headache after
unintentional dural puncture in parturients. Can J Anaesth. 1999;46:861–866.
11. Lybecker H, Møller JT, May O, et al. Incidence and prediction of postdural puncture headache: a
prospective study of 1,021 spinal anesthetics. Anesth Analg. 1990;70:389–394.
12. Santanen U, Rautoma P, Luurila H, et al. Comparison of 27-gauge (0.41-mm) Whitacre and Quincke
spinal needles with respect to post-dural puncture headache and non-dural puncture headache. Acta
Anaesthesiol Scand. 2004;48:474–479.
13. Richman JM, Joe EM, Cohen SR, et al. Bevel direction and postdural puncture headache: a meta-
analysis. Neurologist. 2006;12:224–228.
14. Hatfalvi BI. Postulated mechanisms for postdural puncture headache and review of laboratory models:
clinical experience. Reg Anesth. 1995;20:329–336.
15. Faure E, Moreno R, Thisted R. Incidence of postdural puncture headache in morbidly obese
parturients. Reg Anesth. 1994;19:361–363.
16. Peralta F, Higgins N, Lange E, et al. The relationship of body mass index with the incidence of
postdural puncture headache in parturients. Anesth Analg. 2015;121:451–456
17. Wlody DJ. Postpartum headache other than post-dural puncture headache. In: Atlee J, ed.
Complications in Anesthesia. 2nd ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2005.
18. Headache Classification Subcommittee of the International Headache Society. The international
classification of headache disorders. 2nd ed. Cephalalgia. 2004;24(suppl 1):1–150.
19. Dias MS, Sekhar LN. Intracranial hemorrhage from aneurysms and arteriovenous malformations
during pregnancy and the puerperium. Neurosurgery. 1990;27:855–865.
20. Lockhart EM, Baysinger CL. Intracranial venous thrombosis in the parturient. Anesthesiology.
2007;107:652–658.
21. Guner D, Tiftikcioglu BI, Uludag IF, et al. Dural puncture: an overlooked cause of cerebral venous
thrombosis. Acta Neurol Belg. 2015;115:53–57.
22. Altinkaya SO, Nergiz S, Küçük M, et al. Posterior reversible encephalopathy syndrome in obstetric
patients: report of three cases with literature review. Clin Exp Obstet Gynecol. 2014;41:730–733.
23. Alhilali LM, Reynolds AR, Fakhran S. A multi-disciplinary model of risk factors for fatal outcome in
posterior reversible encephalopathy syndrome. J Neurol Sci. 2014;15;347:59–65.
24. Kesler A, Kupferminc M. Idiopathic intracranial hypertension and pregnancy. Clin Obstet Gynecol.
2013;56:389–396.
25. Lussos SA, Loeffler C. Epidural blood patch improves postdural puncture headache in a patient with
benign intracranial hypertension. Reg Anesth. 1993;18:315–317.
26. Singh T, Schroeder F, Pereira A, et al. Antenatal blood patch in a pregnant woman with spontaneous
intracranial hypotension. Int J Obstet Anesth. 2009;18:165–168.
27. Roll JD, Larson TC III, Soriano MM. Cerebral angiographic findings of spontaneous intracranial
hypotension. AJNR Am J Neuroradiol. 2003;24:707–708.
28. MacGregor EA. Headache in pregnancy. Neurol Clin. 2012;30:835–866.
29. Askmark H, Lundberg PO. Lactation headache—a new form of headache? Cephalalgia. 1989;9:119–
122.
30. Sudlow C, Warlow C. Posture and fluids for preventing post-dural puncture headache. Cochrane
Database Syst Rev. 2002;(2):CD001790.
31. Dieterich M, Brandt T. Incidence of post-lumbar puncture headache is independent of daily fluid
intake. Eur Arch Psychiatry Neurol Sci. 1988;237:194–196.
32. Mosavy SH, Shafei M. Prevention of headache consequent upon dural puncture in obstetric patient.
Anaesthesia. 1975;30:807–809.
33. Sechzer PH, Abel L. Post-spinal anesthesia headache treated with caffeine: evaluation with demand
method: part I. Curr Ther Res. 1978;24:307–312.
34. Halker RB, Demaerschalk BM, Wellik KE, et al. Caffeine for the prevention and treatment of
postdural puncture headache: debunking the myth. Neurologist. 2007;13:323–327.
35. Carp H, Singh PJ, Vadhera R, et al. Effects of the serotonin-receptor agonist sumatriptan on postdural
puncture headache: report of six cases. Anesth Analg. 1994;79:180–182.
36. Connelly NR, Parker RK, Rahimi A, et al. Sumatriptan in patients with postdural puncture headache.
Headache. 2000;40:316–319.
37. Bussone G, Tullo V, d’Onofrio F, et al. Frovatriptan for the prevention of postdural puncture headache.
Cephalalgia. 2007;27:809–813.
38. Noyan Ashraf MA, Sadeghi A, Azarbakht Z, et al. Hydrocortisone in post-dural puncture headache.
Middle East J Anesthesiol. 2007;19:415–422.
39. Rucklidge MW, Yentis SM, Paech MJ. Synacthen depot for the treatment of postdural puncture
headache. Anaesthesia. 2004;59:138–141.
40. Basurto Ona X, Uriona Tuma SM, Martínez García L, et al. Drug therapy for preventing post-dural
puncture headache. Cochrane Database Syst Rev. 2013;(2):CD001792.
41. Mahoori A, Noroozinia H, Hasani E, et al. Comparing the effect of pregabalin, gabapentin, and
acetaminophen on post-dural puncture headache. Saudi J Anesth. 2014;8:374–377.
42. Dietzel J, Witstruck T, Adler S, et al. Acupuncture for treatment of therapy-resistant post-dural
puncture headache: a retrospective case series. Br J Anaesth. 2013;111:847–849.
43. Charsley MM, Abram SE. The injection of intrathecal normal saline reduces the severity of postdural
puncture headache. Reg Anesth Pain Med. 2001;26:301–305.
44. Dennehy KC, Rosaeg OP. Intrathecal catheter insertion during labour reduces the risk of post-dural
puncture headache. Can J Anaesth. 1998;45:42–45.
45. Liu N, Montefiore A, Kermarec N, et al. Prolonged placement of spinal catheters does not prevent
postdural puncture headache. Reg Anesth. 1993;18:110–113.
46. Ayad S, Demian Y, Narouze SN, et al. Subarachnoid catheter placement after wet tap for analgesia in
labor: influence on the risk of headache in obstetric patients. Reg Anesth Pain Med. 2003;28:512–515.
47. Russell IF. A prospective controlled study of continuous spinal analgesia versus repeat epidural
analgesia after accidental dural puncture in labour. Int J Obstet Anesth. 2012;21:7–16.
48. Heesen M, Klöhr S, Rossaint R, et al. Insertion of an intrathecal catheter following accidental dural
puncture: a meta-analysis. Int J Obstet Anesth. 2013;22:26–30.
49. Verstraete S, Walters MA, Devroe S, et al. Lower incidence of post-dural puncture headache with
spinal catheterization after accidental dural puncture in obstetric patients. Acta Anaesthesiol Scand.
2014;58:1233–1239.
50. Al-metwalli RR. Epidural morphine injections for prevention of post dural puncture headache.
Anaesthesia. 2008;63:847–850.
51. Shah JL. Epidural pressure during infusion of saline in the parturient. Int J Obstet Anesth. 1993;2:190–
192.
52. Sudlow C, Warlow C. Epidural blood patching for preventing and treating post-dural puncture
headache. Cochrane Database Syst Rev. 2002;(2):CD001791.
53. Safa-Tisseront V, Thormann F, Malassiné P, et al. Effectiveness of epidural blood patch in the
management of post-dural puncture headache. Anesthesiology. 2001;95:334–339.
54. Cheek TG, Banner R, Sauter J, et al. Prophylactic extradural blood patch is effective: a preliminary
communication. Br J Anaesth. 1988;61:340–342.
55. Vasdev GM, Southern PA. Postdural puncture headache: the role of prophylactic epidural blood patch.
Curr Pain Headache Rep. 2001;5:281–283.
56. Scavone BM, Wong CA, Sullivan JT, et al. Efficacy of a prophylactic epidural blood patch in
preventing post dural puncture headache in parturients after inadvertent dural puncture.
Anesthesiology. 2004;101:1422–1427.
57. Stein MH, Cohen S, Mohiuddin MA, et al. Prophylactic vs therapeutic blood patch for obstetric
patients with accidental dural puncture—a randomised controlled trial. Anaesthesia. 2014;69:320–326.
58. Scavone BM. Timing of epidural blood patch: clearing up the confusion. Anaesthesia. 2015;70:119–
121.
59. Barrios-Alarcon J, Aldrete JA, Paragas-Tapia D. Relief of post-lumbar puncture headache with
epidural dextran 40: a preliminary report. Reg Anesth. 1989;14:78–80.
60. Vassal O, Baud MC, Bolandard F, et al. Epidural injection of hydroxyethyl starch in the management
of postdural puncture headache. Int J Obstet Anesth. 2013;22:153–155.
61. Martin R, Jourdain S, Clairoux M, et al. Duration of decubitus position after epidural blood patch. Can
J Anaesth. 1994;41:23–25.
Neurologic Deficits Following Labor and Delivery
Mark I. Zakowski and Andrew Geller
I. Neurologic injury
A. Incidence
II. History and initial evaluation
A. Relevant questions during evaluation
B. The causes of postpartum neurologic injury
C. The extent of the injury
III. Basic anatomy
A. The lumbar plexus
B. The sacral plexus
C. Trauma caused by the fetal head
IV. Common obstetric neuropathies
A. The lateral femoral cutaneous nerve of the thigh
B. The femoral nerve
C. The obturator nerve
D. The sciatic nerve
V. Ischemic injury to the spinal cord
A. Blood supply to the spinal cord
B. The artery of Adamkiewicz
C. The lumbar arteries
D. Diagnosis of spinal cord ischemia
VI. Types of lesions
A. Chemical injury
B. Direct nerve trauma
VII. Diagnosis and treatment of neuropathies
A. Electromyography
B. Nerve conduction studies
C. Timing of electrophysiologic studies
VIII. Approach to patients with peripheral nerve injuries
A. Preexisting causes
B. Neurologist consultation and imaging studies
C. Plexus injury versus spinal cord injury
IX. Complications related to spinal fluid leakage
A. Postdural puncture headache
B. Characteristics of postdural puncture headache
C. Imaging studies
D. Autologous epidural blood patch
E. Complications of epidural blood patch
F. Managing an unintended postdural puncture headache
G. Prophylactic epidural blood patch
H. Intracranial hematomas
I. Seizures
J. Spontaneous intracranial hypotension
X. Infectious complications of neuraxial blocks
A. Postdural puncture meningitis
B. Epidural abscess
XI. Epidural hematoma
A. Predisposing factors
B. Symptoms
C. Diagnosis
D. Treatment
E. Catheter removal causing epidural hematoma
F. Guidelines for administering neuraxial blocks
XII. Recommendations
KEYPOINTS
1. Neuraxial anesthesia for labor and delivery is extremely safe with a
prolonged complication rate of 1:2,000 to 1:13,000. Most neurologic
injuries are a result of birth trauma and not from neuraxial anesthesia.
2. Permanent nerve injuries are extremely rare but need to be expeditiously
evaluated with a complete history and physical exam. The neurologic
complaint may need to be further evaluated with appropriate diagnostic
testing and consultations.
3. Understanding the anatomy and nerve innervations is crucial in
diagnosing nerve injuries.
4. Careful procedural technique, including sterile precautions, with diligent
monitoring of patients can help reduce neurologic complications from
neuraxial anesthesia.
VII. Diagnosis and treatment of neuropathies (see Fig. 20.5 and Table
20.5)
A. Electromyography. Electromyography (EMG) is extremely
useful in diagnosing the extent of injury to a peripheral nerve (see
Fig. 20.6).48 However, the timing of the test is critical. The
presence of abnormal spontaneous activity in a resting muscle
(fibrillation potentials; see Fig. 20.6) and increased activity
induced by the insertion of the recording needle into the muscle
(insertion activity) suggest differing duration of injury. Insertion
activity becomes noticeable on the EMG within a few days,
whereas fibrillation potentials take 2 to 4 weeks to develop (see
Fig. 20.6).48 A progressive increase in the recruitment of more
motor units, when a muscle is stimulated, is an important
finding.48 In a completely denervated muscle, no recruitment
occurs, whereas partial recruitment may occur when the nerve is
damaged slightly, causing decreased conduction velocity (see Fig.
20.6). EMG is also useful in diagnosing whether the clinical
deficit involves a nerve injury, the extent of the injury, and if
there is evidence of plexus or nerve root injury. EMG studies can
be helpful to differentiate a radiculopathy from a plexopathy;
however, almost half of the patients with a radiculopathy may
have a false-negative paraspinous EMG. EMG can optimally
detect nerve injury between 3 weeks and 6 months following the
inciting event. Fibrillations that identify radiculopathy are seen
most commonly in the paraspinous muscle 1 week after injury
and after 3 to 6 weeks in the distal limb muscles. An early EMG
showing immediate fibrillations (test performed <1 week after
injury) indicates preexisting disease.
B. Nerve conduction studies
1. Both motor and sensory functions of a nerve can be assessed
using evoked potential responses (see Fig. 20.6 and Table
20.5).48 For motor nerve function, the nerve is stimulated at
two points and the compound action potential is recorded
from one of the muscles it innervates. Conduction velocity, as
well as the size of the evoked action potential from the
muscle, is obtained by stimulating the nerve at a point
presumed to be proximal to the site of injury and a point distal
to the site of injury. An abnormality in either of the
parameters recorded may indicate an injury. Similarly, sensory
conduction velocity may be studied by stimulating the nerve
at one point and studying the action potential at another
rostral point on the nerve. When attempting to differentiate
lesions at the level of the spinal root from a peripheral nerve
injury, EMG studies of the paraspinal muscles may help
because the short posterior branch of the spinal root
innervates them, and any combined EMG abnormality of a
lower limb muscle and the paraspinal muscles usually
indicates a lesion at the root level.48–50 If only the peripheral
muscle shows an abnormal EMG pattern, it usually indicates a
peripheral nerve lesion. Paraspinal EMG alone without a
peripheral muscle EMG may lack specificity and sensitivity to
differentiate between spinal root and peripheral nerve
lesions.50,51
2. Usefulness of nerve conduction studies48,49
a. Nerve conduction studies evaluate the functional integrity
of the nerve.
b. They can help localize the lesion. It may not be possible
for the clinician to pinpoint the nerve involved when a
serious clinical sign or symptom occurs. For instance, a
foot drop may be the result of sciatic, common peroneal,
or a lumbar root involvement, and only an
electrophysiologic study will be able to differentiate
between them. This type of study will also determine the
extent of the involvement based on conduction velocity
and muscle action potentials.
c. Evoked potential measurements, in combination with
needle EMG, can delineate the extent of injury and
thereby help to determine the prognosis.
d. Such studies help to determine whether a given clinical
picture results from injury to a single abnormal nerve or
represents an underlying multiple nerve involvement.
e. In lesions where axonal loss has occurred, nerve
conduction studies reveal smaller than usual amplitude for
sensory and muscular compounded action potentials, with
near normal conduction velocities.
f. Demyelination is associated with drastically slowed
conduction velocities with mild to moderate conduction
block.
C. Timing of electrophysiologic studies
Electrophysiologic (EP) examination performed within the first 2
days of injury may provide useful information.48 Any motor
action potential recorded from the muscle indicates only partial
damage. Fibrillation potentials suggest a long-standing lesion,
which may have occurred before the anesthetic and delivery.
Studies done 4 weeks later can be useful in assessing the severity
of the lesion and progress of regeneration.52 For example, if
fibrillation potentials are recorded with no action potential from a
muscle after a month, it indicates a poor prognosis and surgical
exploration of the involved nerve may be necessary.
VIII. Approach to patients with peripheral nerve injuries
A. Preexisting causes. During the assessment of patients with
postpartum neurologic deficits, other preexisting causes of
neuropathy, such as diabetes mellitus, obesity, HIV infection, and
MS, should be considered.53 Although most clinicians are
familiar with diabetic neuropathy, neuropathy associated with
HIV is often unrecognized.54 Peripheral neuropathy is the most
frequent neurologic complication associated with HIV infection.
Polyneuropathies may also occur secondary to the neurotoxicity
of antiretroviral treatment. The anesthesiologist should document
in the preoperative evaluation all preexisting neurologic
abnormalities in patients with systemic diseases to avoid future
medicolegal problems. MS is particularly prone to relapse in the
postpartum period54 and often involves the lateral cutaneous
nerve of the thigh.
B. Neurologist consultation and imaging studies. In the initial
evaluation, the anesthesiologist should document all sensory and
motor deficits. It is important to consult a neurologist who is
familiar with obstetric nerve injuries. Appropriate investigations
may include an MRI, CT scan, and EP studies which should be
performed without delay. Depending on the type and severity of
the lesion, it may take up to 8 weeks for the neurologic lesion to
resolve completely. A repeat EP study may be required to assess
progress of the lesion. A consultation from a physiotherapist may
be valuable to decide on the best rehabilitation procedures to
prevent muscle atrophy. A splint may be required in patients with
a significant foot drop to prevent permanent deformity. If epidural
hematoma is suspected, an MRI or CT scan followed by an
immediate decompressive laminectomy within 6 to 12 hours from
the onset of symptoms offers the best chance for a return of
neurologic function.
C. Plexus injury versus spinal cord injury. Sometimes, an
extensive injury to the lumbosacral plexus may be difficult to
distinguish from an injury to the spinal cord. With spinal cord
injury, EP studies will indicate a lesion at the root level or higher.
In patients with spinal cord ischemia, the ischemic episode may
be caused by compression of the lumbar spinal artery (which may
supply the conus) at the level of the sacrum. MRI findings
suggesting ischemia may be delayed. The possible causes of
prolonged neurologic deficits in parturients are listed in Table
20.6.
CLINICAL PEARL Electromyography and nerve conduction
studies can help identify the level of neurologic deficit and its
timing.
XII. Recommendations
The description of the serious complications in this chapter might
give the impression that neuraxial anesthesia is associated with
significant dangers for the parturient and, therefore, must be avoided.
However, when practiced judiciously, neuraxial anesthesia is
associated with a much smaller incidence of overall anesthetic
complications compared to general anesthesia. Maternal
anesthesia–related mortality was 1.7 times higher with general than
with neuraxial anesthesia in the most recently reported survey of U.S.
patients, although the rate has decreased and is very low, 1 per 1
million live births.100 Obstetric analgesia and anesthesia must be
administered by properly trained individuals. A thorough
understanding of maternal and fetal physiology, attention to technical
details, use of sterile precautions, and a comprehensive vigilant
approach are critical to minimize the incidence of complications.
When a complication occurs, the patient must be informed, followed
closely, and appropriate consultations must be obtained. The care of
patients in the postanesthesia care unit is important. It cannot be
overemphasized that the anesthesiologist or health care provider must
ensure that the block has receded before the patient is discharged to
her room, and that nursing staff continue to monitor block resolution
until complete. In the setting of a compressive lesion within the spinal
canal, timely intervention is essential to ensure recovery without
residual neurologic dysfunction.
REFERENCES
1. American Society of Anesthesiologists Task Force on Obstetric Anesthesia. Practice guidelines for
obstetric anesthesia: an updated report by the American Society of Anesthesiologists Task Force on
Obstetric Anesthesia. Anesthesiology. 2007;106:843–863.
2. Osterman MJ, Martin JA. Epidural and spinal anesthesia use during labor: 27-state reporting area,
2008. Natl Vital Stat Rep. 2011;59:1–13.
3. Zakowski MI. Obstetric-related neurological complications. Int Anesthesiol Clin. 2014;52:40–60.
4. D’Angelo R, Smiley RM, Riley ET, et al. Serious complications related to obstetric anesthesia: the
Serious Complication Repository Project of the Society for Obstetric Anesthesia and Perinatology.
Anesthesiology. 2014;120:1505–1512.
5. Wong CA. Nerve injuries after neuraxial anaesthesia and their medicolegal implications. Best Pract
Res Clin Obstet Gynaecol. 2010;24:367–381.
6. Zakowski M. Postoperative neurologic complication associated with regional anesthesia in the
parturient. In: Norris M, ed. Obstetric Anesthesia. 2nd ed. Philadelphia, PA: Lippincott Williams &
Wilkins; 1999:723–748.
7. Zakowski M. Complications associated with regional anesthesia in the obstetric patient. Semin
Perinatol. 2002;26:154–168.
8. Bromage PR. Neurologic complications of regional anesthesia in obstetrics. In: Hughes SC, Levinson
G, Rosen MA, eds. Schnider and Levinson’s Anesthesia for Obstetrics. 4th ed. Philadelphia, PA:
Lippincott Williams & Wilkins; 2002:409–428.
9. Lee LA, Posner KL, Domino KB, et al. Injuries associated with regional anesthesia in the 1980s and
1990s: a closed claims analysis. Anesthesiology. 2004;101:143–152.
10. Pitkänen MT, Aromaa U, Cozanitis DA, et al. Serious complications associated with spinal and
epidural anaesthesia in Finland from 2000 to 2009. Acta Anaesthesiol Scand. 2013;57:553–564.
11. Davies JM, Posner KL, Lee LA, et al. Liability associated with obstetric anesthesia: a closed claims
analysis. Anesthesiology. 2009;110:131–139.
12. Hayes NE, Wheelahan JM, Ross A. Self-reported post-discharge symptoms following obstetric
neuraxial blockade. Int J Obstet Anesth. 2010;19:405–409.
13. Horlocker TT, Abel MD, Messick JM Jr, et al. Small risk of serious neurologic complications related
to lumbar epidural catheter placement in anesthetized patients. Anesth Analg. 2003;96:1547–1552.
14. Cook TM, Counsell D, Wildsmith JA. Major complications of central neuraxial block: report on the
Third National Audit Project of the Royal College of Anaesthetists. Br J Anaesth. 2009;102:179–190.
15. Ruppen W, Derry S, McQuay H, et al. Incidence of epidural hematoma, infection, and neurologic
injury in obstetric patients with epidural analgesia/anesthesia. Anesthesiology. 2006;105:394–399.
16. Moen V, Dahlgren N, Irestedt L. Severe neurological complications after central neuraxial blockades
in Sweden 1990-1999. Anesthesiology. 2004;101:950–959.
17. Auroy Y, Narchi P, Messiah A, et al. Serious complications related to regional anesthesia: results of a
prospective survey in France. Anesthesiology. 1997;87:479–486.
18. Scott DB, Tunstall ME. Serious complications associated with epidural/spinal blockade in obstetrics: a
two-year prospective study. Int J Obstet Anesth. 1995;4:133–139.
19. Scott DB, Hibbard BM. Serious non-fatal complications associated with extradural block in obstetric
practice. Br J Anaesth. 1990;64:537–541.
20. Usubiaga JE. Neurological complications following epidural anesthesia. Int Anesthesiol Clin.
1975;13:1–153.
21. Wong CA, Scavone BM, Dugan S, et al. Incidence of postpartum lumbosacral spine and lower
extremity nerve injuries. Obstet Gynecol. 2003;101:279–288.
22. Brull R, McCartney CJ, Chan VW, et al. Neurological complications after regional anesthesia:
contemporary estimates of risk. Anesth Analg. 2007;104:965–974.
23. Kuczkowski KM. Neurologic complication of labor analgesia: facts and fiction. Obstet Gynecol Surv.
2004;59:47–51.
24. Lee EY, Margherita AJ, Gierada DS, et al. MRI of piriformis syndrome. AJR Am J Roentgenol.
2004;183:63–64.
25. Vallejo MC, Mariano DJ, Kaul B, et al. Piriformis syndrome in a patient after cesarean section under
spinal anesthesia. Reg Anesth Pain Med. 2004;29:364–367.
26. Redick L. Maternal perinatal nerve palsies. Postgrad Obstet Gynecol. 1992;12:1–6.
27. Grant JCB. Grant’s Atlas of Anatomy, by Regions. 6th ed. Baltimore, MD: Williams & Wilkins; 1972.
28. Cole JT. Maternal obstetric paralysis. Am J Obstet Gynecol. 1946;52:372–386.
29. Graham JG. Neurological complications of pregnancy and anaesthesia. Clin Obstet Gynaecol.
1982;9:333–350.
30. Ellis H, Feldman H. Anatomy for Anaesthetists. 3rd ed. London, United Kingdom: Blackwell Science;
1977:159–331.
31. Papadopoulos EC, Kahn SN. Piriformis syndrome and low back pain: a new classification and review
of the literature. Orthop Clin North Am. 2004;35:65–71.
32. Katirji B. Peroneal neuropathy. Neurol Clin. 1999;17:567–591.
33. Ramanathan S, Chalon J, Richards M, et al. Prolonged spinal nerve involvement after epidural
anesthesia with etidocaine. Anesth Analg. 1978;57:361–364.
34. Berlit P, Klötzsch G, Röther J, et al. Spinal cord infarction: MRI and MEP findings in three cases. J
Spinal Disord. 1992; 5:212–216.
35. Biglioli P, Roberto M, Cannata A, et al. Upper and lower spinal cord blood supply: the continuity of
the anterior spinal artery and the relevance of the lumbar arteries. J Thorac Cardiovasc Surg.
2004;127:1188–1192.
36. Dimakakos P, Arkadopoulos N. Spinal cord ischaemia. Eur J Vasc Endovasc Surg. 1999;17:544–545.
37. Masson C, Leys D, Meder JF, et al. Spinal cord ischemia. J Neuroradiol. 2004;31:35–46.
38. Castro-Moure F, Kupsky W, Goshgarian HG. Pathophysiological classification of human spinal cord
ischemia. J Spinal Cord Med. 1997;20:74–87.
39. Loher TJ, Bassetti CL, Lövblad KO, et al. Diffusion-weighted MRI in acute spinal cord ischaemia.
Neuroradiology. 2003;45:557–561.
40. Stepper F, Lövblad KO. Anterior spinal artery stroke demonstrated by echo-planar DWI. Eur Radiol.
2001;11:2607–2610.
41. Alblas Cl, Bouvy WH, Lycklama à Nijeholt GJ, et al. Acute spinal-cord ischemia: evolution of MRI
findings. J Clin Neurol. 2012;8:218–223.
42. Faccenda KA, Finucane BT. Complications of regional anaesthesia. Incidence and prevention. Drug
Saf. 2001;24:413–442.
43. Hussain SA, Gullan RW, Chitnavis BP. Cauda equina syndrome: outcome and implications for
management. Br J Neurosurg. 2003;17:164–167.
44. Pong RP, Gmelch BS, Bernards CM. Does a paresthesia during spinal needle insertion indicate
intrathecal needle placement? Reg Anesth Pain Med. 2009;34:29–32.
45. Jaime F, Mandell GL, Vallejo MC, et al. Uniport soft-tip, open-ended catheters versus multiport firm-
tipped close-ended catheters for epidural labor analgesia: a quality assurance study. J Clin Anesth.
2000;12:89–93.
46. Reynolds F. Damage to the conus medullaris following spinal anaesthesia. Anaesthesia. 2001;56:238–
247.
47. Aminoff MJ. Electrophysiologic testing for the diagnosis of peripheral nerve injuries. Anesthesiology.
2004;100:1298–1303.
48. Krarup C. An update on electrophysiological studies in neuropathy. Curr Opin Neurol. 2003;16:603–
612.
49. Czyrny JJ, Lawrence J. The importance of paraspinal muscle EMG in cervical and lumbosacral
radiculopathy: review of 100 cases. Electromyogr Clin Neurophysiol. 1996;36:503–508.
50. Bojović V, Berisavac I, Rasulic L. Significance of sensory evoked potentials in determination of the
level of brachial plexus injuries [in Serbian]. Acta Chir Iugosl. 2003;50:15–22.
51. Haig AJ, Levine JW, Ruan C, et al. Describing paraspinal EMG findings: inadequacy of the single 0-
4+ score. Am J Phys Med Rehabil. 2000;79:133–137.
52. Lalive PH, Truffert A, Magistris MR. Lombosacral radiculopathy (L3-S1) and specificity of multifidus
EMG [in French]. Neurophysiol Clin. 2004;34:41–47.
53. Wulff EA, Wang AK, Simpson DM. HIV-associated peripheral neuropathy: epidemiology,
pathophysiology and treatment. Drugs. 2000;59:1251–1260.
54. Confavreux C, Hutchinson M, Hours MM, et al. Rate of pregnancy-related relapse in multiple
sclerosis. Pregnancy in Multiple Sclerosis Group. N Engl J Med. 1998;339:285–291.
55. Vallejo MC, Mandell GL, Sabo DP, et al. Postdural puncture headache: a randomized comparison of
five spinal needles in obstetric patients. Anesth Analg. 2000;91:916–920.
56. Miu M, Paech MJ, Nathan E. The relationship between body mass index and post-dural puncture
headache in obstetric patients. Int J Obstet Anesth. 2014;23:371–375.
57. Vilming ST, Kloster R. Pain location and associated symptoms in post-lumbar puncture headache.
Cephalalgia. 1998;18:697–703.
58. Loures V, Savoldelli G, Kern K, et al. Atypical headache following dural puncture in obstetrics. Int J
Obstet Anesth. 2014;23:246–252.
59. Finegold H, Mandell G, Vallejo MC, et al. Does spinal anesthesia cause hearing loss in the obstetric
population? Anesth Analg. 2002;95:198–203.
60. Yousry I, Förderreuther S, Moriggl B, et al. Cervical MR imaging in postural headache: MR signs and
pathophysiological implications. AJNR Am J Neuroradiol. 2001;22:1239–1250.
61. Gaiser R. Postdural puncture headache. Curr Opin Anaesthesiol. 2006;19:249–253.
62. Kokki M, Sjövall S, Keinänen M, et al. The influence of timing on the effectiveness of epidural blood
patches in parturients. Int J Obstet Anesth. 2013;22:303–309.
63. Schievink WI. Spontaneous spinal cerebrospinal fluid leaks and intracranial hypotension. JAMA.
2006;295:2286–2296.
64. Kalina P, Craigo P, Weingarten T. Intrathecal injection of epidural blood patch: a case report and
review of the literature. Emerg Radiol. 2004;11:56–59.
65. Leivers D. Total spinal anesthesia following early prophylactic epidural blood patch. Anesthesiology.
1990;73:1287–1289.
66. Ayad S, Demian Y, Narouze SN, et al. Subarachnoid catheter placement after wet tap for analgesia in
labor: influence on the risk of headache in obstetric patients. Reg Anesth Pain Med. 2003;28:512–515.
67. Verstraete S, Walters MA, Devroe S, et al. Lower incidence of post-dural puncture headache with
spinal catheterization after accidental dural puncture in obstetric patients. Acta Anaesthesiol Scand.
2014;58:1233–1239.
68. Scavone BM, Wong CA, Sullivan JT, et al. Efficacy of a prophylactic epidural blood patch in
preventing post dural puncture headache in parturients after inadvertent dural puncture.
Anesthesiology. 2004;101:1422–1427.
69. Zeidan A, Farhat O, Maaliki H, et al. Does postdural puncture headache left untreated lead to subdural
hematoma? Case report and review of the literature. Middle East J Anaesthesiol. 2010;20:483–492.
70. Agrawal D, Durity FA. Seizure as a manifestation of intracranial hypotension in a shunted patient.
Pediatr Neurosurg. 2006;42:165–167.
71. Warwick WI, Neal JM. Beyond spinal headache: prophylaxis and treatment of low-pressure headache
syndromes. Reg Anesth Pain Med. 2007;32:455–461.
72. Zada G, Pezeshkian P, Giannotta S. Spontaneous intracranial hypotension and immediate
improvement following epidural blood patch placement demonstrated by intracranial pressure
monitoring. Case report. J Neurosurg. 2007;106:1089–1090.
73. Diaz JH. Treatment outcomes in spontaneous intracranial hypotension: do epidural blood patches stop
the leaks? Pain Pract. 2004;4:295–302.
74. Baer ET. Post-dural puncture bacterial meningitis. Anesthesiology. 2006;105:381–393.
75. Centers for Disease Control and Prevention. Bacterial meningitis after intrapartum spinal anesthesia—
New York and Ohio, 2008–2009. MMWR Morb Mortal Wkly Rep. 2010;59:65–69.
76. Wedel DJ, Horlocker TT. Risks of regional anesthesia—infectious, septic. Reg Anesth. 1996;21(suppl
6):57–61.
77. Kasai T, Yaegashi K, Hirose M, et al. Aseptic meningitis during combined continuous spinal and
epidural analgesia. Acta Anaesthesiol Scand. 2003;47:775–776.
78. Reynolds F. Infection as a complication of neuraxial blockade. Int J Obstet Anesth. 2005;14:183–188.
79. Horlocker TT, McGregor DG, Matsushige DK, et al. Neurologic complications of 603 consecutive
continuous spinal anesthetics using macrocatheter and microcatheter techniques. Perioperative
Outcomes Group. Anesth Analg. 1997;84:1063–1070.
80. Straus SE, Thorpe KE, Holroyd-Leduc J. How do I perform a lumbar puncture and analyze the results
to diagnose bacterial meningitis? JAMA. 2006;296:2012–2022.
81. American Society of Anesthesiologists Task Force on Infectious Complications Associated with
Neuraxial Techniques. Practice advisory for the prevention, diagnosis, and management of infectious
complications associated with neuraxial techniques: a report by the American Society of
Anesthesiologists Task Force on Infectious Complications Associated with Neuraxial Techniques.
Anesthesiology. 2010;112:530–545.
82. Videira RL, Ruiz-Neto PP, Brandao Neto M. Post spinal meningitis and asepsis. Acta Anaesthesiol
Scand. 2002;46:639–646.
83. Hepner DL. Gloved and masked—will gowns be next? The role of asepsis during neuraxial
instrumentation. Anesthesiology. 2006;105:241–243.
84. Kowalski TJ, Layton KF, Berbari EF, et al. Follow-up MR imaging in patients with pyogenic spine
infections: lack of correlation with clinical features. AJNR Am J Neuroradiol. 2007;28:693–699.
85. Cohen S, Hunter CW, Sakr A, et al. Meningitis following intrathecal catheter placement after
accidental dural puncture. Int J Obstet Anesth. 2006;15:172.
86. Grewal S, Hocking G, Wildsmith JA. Epidural abscesses. Br J Anaesth. 2006;96:292–302.
87. Horlocker TT. What’s a nice patient like you doing with a complication like this? Diagnosis, prognosis
and prevention of spinal hematoma. Can J Anaesth. 2004;51:527–534.
88. Reihsaus, E, Waldbaur H, Seeling W. Spinal epidural abscess: a meta-analysis of 915 patients.
Neurosurg Rev. 2000;23: 175–204.
89. Vandermeulen EP, Van Aken H, Vermylen J. Anticoagulants and spinal-epidural anesthesia. Anesth
Analg. 1994;79: 1165–1177.
90. Sato S, Sakuragi T, Dan K. Human skin flora as a potential source of epidural abscess. Anesthesiology.
1996;85:1276–1282.
91. Sidiropoulou T, Pompeo E, Bozzao A, et al. Epidural hematoma after thoracic epidural catheter
removal in the absence of risk factors. Reg Anesth Pain Med. 2003;28:531–534.
92. Huang J, McKenna N, Babins N. Utility of thromboelastography during neuraxial blockade in the
parturient with thrombocytopenia. AANA J. 2014;82:127–130.
93. Groen RJ, Ponssen H. The spontaneous spinal epidural hematoma. A study of the etiology. J Neurol
Sci. 1990;98:121–138.
94. Groen RJ. Non-operative treatment of spontaneous spinal epidural hematomas: a review of the
literature and a comparison with operative cases. Acta Neurochir (Wien). 2004;146:103–110.
95. Horlocker TT, Heit JA. Low molecular weight heparin: biochemistry, pharmacology, perioperative
prophylaxis regimens, and guidelines for regional anesthetic management. Anesth Analg.
1997;85:874–885.
96. Horlocker TT, Wedel DJ, Rowlingson JC, et al. Regional anesthesia in the patient receiving
antithrombotic or thrombolytic therapy: American Society of Regional Anesthesia and Pain Medicine
Evidence-Based Guidelines (Third Edition). Reg Anesth Pain Med. 2010;35:64–101.
97. Douketis JD, Kinnon K, Crowther MA. Anticoagulant effect at the time of epidural catheter removal
in patients receiving twice-daily or once-daily low-molecular-weight heparin and continuous epidural
analgesia after orthopedic surgery. Thromb Haemost. 2002;88:37–40.
98. Wu CL. Regional anesthesia and anticoagulation. J Clin Anesth. 2001;13:49–58.
99. Horlocker TT, Wedel DJ, Benzon H, et al. Regional anesthesia in the anticoagulated patient: defining
the risks (the second ASRA Consensus Conference on Neuraxial Anesthesia and Anticoagulation).
Reg Anesth Pain Med. 2003;28:172–197.
100. Hawkins JL, Chang J, Palmer SK, et al. Anesthesia-related maternal mortality in the United States:
1979-2002. Obstet Gynecol. 2011;117:69–74.
Postpartum Tubal Ligation
Brenda A. Bucklin
I. American Society of Anesthesiologists Practice Guidelines for
Obstetric Anesthesia: recommendations for postpartum
sterilization
II. Postpartum anatomic and physiologic changes
A. Cardiovascular changes
B. Gastrointestinal changes
III. Timing of postpartum tubal sterilization
A. Interval versus postpartum tubal sterilization
B. Timing from the obstetrician’s perspective
IV. Surgical considerations relevant to the anesthesiologist
A. Tubal sterilization
B. The overweight or obese patient
V. Anesthetic considerations
A. Anesthetic risk
B. Preoperative assessment
C. Aspiration risk and prophylaxis
D. Breastfeeding and anesthesia
E. Which neuraxial anesthetic is best for postpartum tubal
sterilization?
F. Anesthetic choice: general
G. Anesthetic choice: local
H. Postoperative analgesia
I. Summary of anesthetic considerations
KEYPOINTS
1. Although postpartum tubal ligation is considered an “urgent” procedure1
because of the consequences of failure to sterilize, the procedure should
not be attempted when it could compromise other aspects of patient care.2
2. Timing of the procedure should be based on anesthetic and obstetric risk
factors as well as patient preferences.
3. If postpartum tubal sterilization is anticipated within 8 hours of delivery,
neuraxial anesthesia should be encouraged during labor and delivery.
4. There should be no oral intake of solid foods within 6 to 8 hours of
surgery.
5. Aspiration prophylaxis should be considered.
6. Neuraxial techniques are preferred.
7. Epidural catheters placed for labor may be more likely to fail with longer
postdelivery time intervals.
V. Anesthetic considerations
A. Anesthetic risk
1. Although the true anesthetic risk for postpartum sterilization
performed within 8 hours of delivery is unknown, neuraxial
anesthesia should be encouraged during labor and delivery.
a. Early epidemiologic records only contain data from
laparoscopic and laparotomy sterilizations performed after
the 8-hour postpartum period.18,19 These reports indicated
that complication rates were five times higher when
general anesthesia was administered compared to local
anesthetic techniques (i.e., local, epidural, or spinal).
However, these reports occurred prior to the use of pulse
oximetry and capnography and do not reflect modern
anesthetic practice.
b. Reports of anesthetic-related mortality during obstetric
delivery in the United States do not identify PPTL as a
risk for anesthetic-related complications.6
c. Similarly, there are no reports of anesthetic-related
mortality associated with tubal sterilization in the
Reports of Confidential Enquiries into Maternal Deaths.20
These detailed reports of maternal deaths are published
every 3 years in the United Kingdom and make
recommendations for improvement in patient care.
REFERENCES
1. Committee on Health Care for Underserved Women. Committee Opinion No. 530: access to
postpartum sterilization. Obstet Gynecol. 2012;120:212–215.
2. American Society of Anesthesiologists Task Force on Obstetric Anesthesia. Practice guidelines for
obstetric anesthesia: an updated report by the American Society of Anesthesiologists Task Force on
Obstetric Anesthesia. Anesthesiology. 2007;106:843–863.
3. Mosher WD, Jones J. Use of contraception in the United States: 1982-2008. Vital Health Stat 23.
2010;29:1–44.
4. Rodriguez MI, Edelman A, Wallace N, et al. Denying postpartum sterilization to women with
Emergency Medicaid does not reduce hospital charges. Contraception. 2008;78:232–236.
5. Sadaniantz A, Saint Laurent L, Parisi AF. Long-term effects of multiple pregnancies on cardiac
dimensions and systolic and diastolic function. Am J Obstet Gynecol. 1996;174:1061–1064.
6. Hawkins JL, Chang J, Palmer SK, et al. Anesthesia-related maternal mortality in the United States:
1979-2002. Obstet Gynecol. 2011;117:69–74.
7. Löfgren M, Bäckström T. Serum concentrations of progesterone and 5 alpha-pregnane-3,20-dione
during labor and early post partum. Acta Obstet Gynecol Scand. 1990;69:123–126.
8. Hawkins JL. Postpartum tubal sterilization. In: Chestnut DH, Wong CA, Tsen LC, et al., eds.
Chestnut’s Obstetric Anesthesia: Principles and Practice. 5th ed. Philadelphia, PA: Elsevier; 2014:30–
42.
9. Thurman AR, Janecek T. One-year follow-up of women with unfulfilled postpartum sterilization
requests. Obstet Gynecol. 2010;116:1071–1077.
10. Borrero S, Zite N, Potter JE, et al. Medicaid policy on sterilization—anachronistic or still relevant? N
Engl J Med. 2014;370:102–104.
11. Colorado medicaid sterilization consent form (MED-178). Colorado Web site.
https://www.colorado.gov/pacific/sites /default/files/MED-178_092713.pdf. Accessed November 6,
2014.
12. American College of Obstetricians and Gynecologists. ACOG Practice Bulletin No. 133: benefits and
risks of sterilization. Obstet Gynecol. 2013;121:392–404.
13. Peterson HB, Xia Z, Hughes JM, et al. The risk of pregnancy after tubal sterilization: findings from
the U.S. Collaborative Review of Sterilization. Am J Obstet Gynecol. 1996;174:1161–1168.
14. Kaneshiro B, Grimes DA, Lopez LM. Pain management for tubal sterilization by hysteroscopy.
Cochrane Database System Rev. 2012:(8)CD009251.
15. Jamieson DJ, Hillis SD, Duerr A, et al. Complications of interval laparoscopic tubal sterilization:
findings from the United States Collaborative Review of Sterilization. Obstet Gynecol. 2000;96:997–
1002.
16. Flegal KM, Carroll MD, Kit BK, et al. Prevalence of obesity and trends in the distribution of body
mass index among US adults, 1999-2010. JAMA. 2012;307:491–497.
17. Rodriguez MI, Edelman AB, Kapp N. Postpartum sterilization with the titanium clip: a systematic
review. Obstet Gynecol. 2011;118:143–147.
18. Peterson HB, DeStefano F, Rubin GL, et al. Deaths attributable to tubal sterilization in the United
States, 1977 to 1981. Am J Obstet Gynecol. 1983;146:131–136.
19. Peterson HB, Greenspan JR, DeStefano F, et al. Deaths associated with laparoscopic sterilization in
the United States, 1977-79. J Reprod Med. 1982;27:345–347.
20. Cantwell R, Clutton-Brock T, Cooper G, et al. Saving mothers’ lives: reviewing maternal deaths to
make motherhood safer: 2006-2008. The eighth report of the Confidential Enquiries into Maternal
Deaths in the United Kingdom. BJOG. 2011;118:1–203.
21. Quinn AC, Milne D, Columb M, et al. Failed tracheal intubation in obstetric anaesthesia: 2 yr national
case-control study in the UK. Br J Anaesth. 2013;110:74–80.
22. Endler GC, Mariona FG, Sokol RJ, et al. Anesthesia-related maternal mortality in Michigan, 1972 to
1984. Am J Obstet Gynecol. 1988;159:187–193.
23. Suelto MD, Vincent RD Jr, Larmon JE, et al. Spinal anesthesia for postpartum tubal ligation after
pregnancy complicated by preeclampsia or gestational hypertension. Reg Anesth Pain Med.
2000;25:170–173.
24. American Academy of Pediatrics. AAP reaffirms breastfeeding guidelines. http://www.aap.org/en-
us/about-the-aap/aap-press-room/Pages/AAP-Reaffirms-Breastfeeding-Guidelines.aspx. Accessed
November 6, 2014.
25. Nitsun M, Szokol JW, Saleh HJ, et al. Pharmacokinetics of midazolam, propofol, and fentanyl transfer
to human breast milk. Clin Pharmacol Ther. 2006;79:549–557.
26. American Academy of Pediatrics Committee on Drugs. Transfer of drugs and other chemicals into
human milk. Pediatrics. 2001;108:776–789.
27. U.S. Department of Health and Human Services. Analgesia and anesthesia for the breastfeeding
mother, revised 2012. http://www.guideline.gov/content.aspx?id=39271. Accessed November 6, 2014.
28. Hamilton CL, Riley ET, Cohen SE. Changes in the position of epidural catheters associated with
patient movement. Anesthesiology. 1997;86:778–784.
29. Viscomi CM, Rathmell JP. Labor epidural catheter reactivation or spinal anesthesia for delayed
postpartum tubal ligation: a cost comparison. J Clin Anesth. 1995;7:380–383.
30. Beilin Y, Bernstein HH, Zucker-Pinchoff B. The optimal distance that a multiorifice epidural catheter
should be threaded into the epidural space. Anesth Analg. 1995;81:301–304.
31. D’Angelo R, Foss ML, Livesay CH. A comparison of multiport and uniport epidural catheters in
laboring patients. Anesth Analg. 1997;84:1276–1279.
32. Mets B, Broccoli E, Brown AR. Is spinal anesthesia after failed epidural anesthesia contraindicated for
cesarean section? Anesth Analg. 1993;77:629–631.
33. Dadarkar P, Philip J, Weidner C, et al. Spinal anesthesia for cesarean section following inadequate
labor epidural analgesia: a retrospective audit. Int J Obstet Anesth. 2004;13:239–243.
34. Abouleish EI. Postpartum tubal ligation requires more bupivacaine for spinal anesthesia than does
cesarean section. Anesth Analg. 1986;65:897–900.
35. Philip J, Sharma SK, Gottumukkala VN, et al. Transient neurologic symptoms after spinal anesthesia
with lidocaine in obstetric patients. Anesth Analg. 2001;92:405–409.
36. Schneider MC, Birnbach DJ. Lidocaine neurotoxicity in the obstetric patient: is the water safe? Anesth
Analg. 2001;92:287–290.
37. Kao YJ, Turner DR. Prolongation of succinylcholine block by metoclopramide. Anesthesiology.
1989;70:905–908.
38. Woodworth GE, Sears DH, Grove TM, et al. The effect of cimetidine and ranitidine on the duration of
action of succinylcholine. Anesth Analg. 1989;68:295–297.
39. Gin T, Chan MT, Chan KL, et al. Prolonged neuromuscular block after rocuronium in postpartum
patients. Anesth Analg. 2002;94:686–689.
40. Zhou HH, Norman P, DeLima LG, et al. The minimum alveolar concentration of isoflurane in patients
undergoing bilateral tubal ligation in the postpartum period. Anesthesiology. 1995;82:1364–1368.
41. Habib AS, Muir HA, White WD, et al. Intrathecal morphine for analgesia after postpartum bilateral
tubal ligation. Anesth Analg. 2005;100:239–243.
42. Marcus RJ, Wong CA, Lehor A, et al. Postoperative epidural morphine for postpartum tubal ligation
analgesia. Anesth Analg. 2005;101:876–881.
43. Harrison MS, DiNapoli MN, Westhoff CL. Reducing postoperative pain after tubal ligation with rings
or clips: a systematic review and meta-analysis. Obstet Gynecol. 2014;124:68–75.
44. Rorarius M, Suominen P, Baer G, et al. Peripherally administered sufentanil inhibits pain perception
after postpartum tubal ligation. Pain. 1999;79:83–88.
Disease States
Hypertensive Disorders of Pregnancy
Yelena Spitzer and Yaakov Beilin
I. Differential diagnosis and definitions
A. Gestational hypertension
B. Chronic hypertension
C. Preeclampsia
D. Chronic hypertension with superimposed preeclampsia
II. Epidemiology
A. Incidence
B. Maternal mortality
C. Neonatal mortality
III. Risk factors
A. Positive predictors
B. Protective factors
IV. Etiology
A. Decreased placental perfusion
B. Outcome of placental ischemia
C. Inflammatory mediators
D. Oxidative stress
E. Vasoactive substances
F. Antioxidants
G. Lifestyle modifications
H. Genetic influences
I. Prostaglandin imbalance
V. Pathophysiology
A. Central nervous system
B. Cardiovascular system
C. Respiratory system
D. Coagulation system
E. Genitourinary/renal system
F. Hepatobiliary system
G. Fetus and placenta
VI. Obstetric management
A. Prediction and prevention of preeclampsia
B. Timing and route of delivery
C. Seizure prophylaxis
D. Blood pressure control
VII. Anesthetic considerations
A. Assessment of volume status
B. Controlling blood pressure
C. Coagulation
VIII. Mode of delivery and anesthetic technique
A. Vaginal delivery
B. Cesarean delivery
IX. Postpartum care
A. Analgesia
B. Seizure prophylaxis
C. Blood pressure control
D. Fluid balance
E. Long-term sequelae
KEYPOINTS
1. Hypertensive disorders of pregnancy complicate 6% to 8% of all
pregnancies in the United States and result in a significant risk of
maternal and fetal morbidity and mortality.
2. In 2013, the Task Force on Hypertension in Pregnancy changed the
definition of preeclampsia. Given the dynamic nature of the disease,
preeclampsia is no longer classified as mild or severe but rather as
preeclampsia and preeclampsia with severe features.
3. Preeclampsia with severe features is a major contributor to neonatal
morbidity and mortality, due to impaired uteroplacental perfusion,
placental abruption, and iatrogenic prematurity due to delivery remote
from term.
4. Delivery of the fetus is the only definitive cure and the timing of the
delivery must take into account both maternal and fetal conditions.
5. Neuraxial anesthesia is the preferred mode of analgesia for labor and
anesthesia for cesarean delivery, and the choice is based on an assessment
of the intravascular volume, blood pressure, and coagulation status.
II. Epidemiology
Preeclampsia and gestational hypertension are primarily disorders of
older nulliparous women. Because assisted reproductive technology
allows the older patient to achieve a successful pregnancy who may
also have other comorbidities such as hypertension and diabetes, the
incidence of these disorders will continue to rise.
A. Incidence. Preeclampsia affects 6% to 8% of all pregnancies, of
which 25% have preeclampsia with severe features.5
1. HELLP syndrome occurs in 10% of women with
preeclampsia with severe features.
2. The rate of preeclampsia in the United States is on the rise.
For preeclampsia overall, the rate has increased from 3.4% in
1980 to 3.8% in 2010. The increase in preeclampsia with
severe features accounts for the overall increase. In 1980,
severe preeclampsia accounted for 0.3% of the cases
compared to 1.4% in 2010.5
B. Maternal mortality. Preeclampsia and its sequelae, one of the
three leading causes of maternal mortality, account for more than
25% of all cases of maternal mortality in the United States. Most
of the deaths related to preeclampsia are due to pulmonary
complications (e.g., pulmonary edema) and intracranial
hemorrhage.
C. Neonatal mortality. Preeclampsia with severe features is a major
contributor to neonatal morbidity and mortality, due to impaired
uteroplacental perfusion, placental abruption, and iatrogenic
prematurity due to onset of preeclampsia remote from term.
III. Risk factors (see Table 22.2)
REFERENCES
1. Martin JA, Hamilton BE, Sutton PD, et al. Births: final data for 2005. Natl Vital Stat Rep. 2007;56:1–
103.
2. National High Blood Pressure Education Program. Report of the National High Blood Pressure
Education Program working group on high blood pressure in pregnancy. Am J Obstet Gynecol.
2000;183:S1–S22.
3. American College of Obstetricians and Gynecologists. Hypertension in pregnancy. Report of the
American College of Obstetricians and Gynecologists’ Task Force on Hypertension in Pregnancy.
Obstet Gynecol. 2013;122:1122–1131.
4. Matthys LA, Coppage KH, Lambers DS, et al. Delayed postpartum preeclampsia: an experience of
151 cases. Am J Obstet Gynecol. 2004;190:1464–1466.
5. Hauth JC, Ewell MG, Levine RJ, et al. Pregnancy outcomes in healthy nulliparas who developed
hypertension. Calcium for Preeclampsia Prevention Study group. Obstet Gynecol. 2000;95:24–28.
6. American College of Obstetricians and Gynecologists Committee on Obstetric Practice. ACOG
practice bulletin. Diagnosis and management of preeclampsia and eclampsia. Number 33, January
2002. American College of Obstetricians and Gynecologists. Int J Gynaecol Obstet. 2002;77:67–75.
7. Mehendale R, Hibbard J, Fazleabas A, et al. Placental angiogenesis markers sFlt-1 and PIGF: response
to cigarette smoke. Am J Obstet Gynecol. 2007;197:363.e1–363.e5.
8. Lyall F. Priming and remodeling of human placental bed spiral arteries during pregnancy—a review.
Placenta. 2005;26 (suppl A):S31–S36.
9. Shah DM. Preeclampsia: new insights. Curr Opin Nephrol Hypertens. 2007;16:213–220.
10. Hefler LA, Tempfer CB, Gregg AR. Polymorphisms within the interleukin-1 beta gene cluster and
preeclampsia. Obstet Gynecol. 2001;97:664–668.
11. Collaborative Low-dose Aspirin Study in Pregnancy Collaborative Group. CLASP: a randomized trial
of low-dose aspirin for the prevention and treatment of pre-eclampsia among 9,364 pregnant women.
Lancet. 1994;343:619–629.
12. Duley L, Henderson-Smart DJ, Meher S, et al. Antiplatelet agents for preventing preeclampsia and its
complications. Cochrane Database Syst Rev. 2007;(2):CD004659.
doi:10.1002/14651858.CD004659.pub2
13. Belfort MA, Saade GR, Grunewald C, et al. Association of cerebral perfusion with headache in
women with preeclampsia. Br J Obstet Gynaecol. 1999;106:814–821.
14. Zeeman GG, Hatab MR, Twickler DM. Increased cerebral blood flow in preeclampsia with magnetic
resonance imaging. Am J Obstet Gynecol. 2004;191:1425–1429.
15. Cotton DB, Lee W, Huhta JC, et al. Hemodynamic profile of severe pregnancy-induced hypertension.
Am J Obstet Gynecol. 1988;158:523–529.
16. Phelan JP, Yurth DA. Severe preeclampsia. I. Peripartum hemodynamic observations. Am J Obstet
Gynecol. 1982;144:17–22.
17. Pouta A, Karinen J, Vuolteenaho O, et al. Preeclampsia: the effect of intravenous fluid preload on
atrial natriuretic peptide secretion during caesarean section under spinal anaesthesia. Acta
Anaesthesiol Scand. 1996;40:1203–1209.
18. Pritchard JA, Cunningham FG, Mason RA. Coagulation changes in eclampsia: their frequency and
pathogenesis. Am J Obstet Gynecol. 1976;124:855–864.
19. Leduc L, Wheeler JM, Kirshon B, et al. Coagulation profile in severe preeclampsia. Obstet Gynecol.
1992;79:14–18.
20. Taufield PA, Ales KL, Resnick LM, et al. Hypocalciuria in preeclampsia. N Engl J Med.
1987;316:715–718.
21. Myatt L, Clifton RG, Roberts JM, et al. The utility of uterine artery Doppler velocimetry in prediction
of preeclampsia in a low-risk population. Obstet Gynecol. 2012;120:815–822.
22. Trumbo PR, Ellwood KC. Supplemental calcium and risk reduction of hypertension, pregnancy-
induced hypertension, and preeclampsia: an evidence-based review by the US Food and Drug
Administration. Nutr Rev. 2007;65:78–87.
23. Spinnato JA, Livingston JC. Prevention of preeclampsia with antioxidants: evidence from randomized
trials. Clin Obstet Gynecol. 2005;48:416–429.
24. Haddad B, Deis S, Goffinet F, et al. Maternal and perinatal outcomes during expectant management of
239 severe preeclamptic women between 24 and 33 weeks’ gestation. Am J Obstet Gynecol.
2004;190:1590–1595.
25. Lucas MJ, Leveno KJ, Cunningham FG. A comparison of magnesium sulfate with phenytoin for the
prevention of eclampsia. N Engl J Med. 1995;333:201–205.
26. American College of Obstetricians and Gynecologists. Committee Opinion No. 623: emergent therapy
for the acute-onset, severe hypertension during pregnancy and the postpartum period. Obstet Gynecol.
2015;125:521–525.
27. Belfort MA, Clark SL, Sibai B. Cerebral hemodynamics in preeclampsia: cerebral perfusion and the
rationale for an alternative to magnesium sulfate. Obstet Gynecol Surv. 2006;61:655–665.
28. Belfort MA, Tooke-Miller C, Allen JC Jr, et al. Labetalol decreases cerebral perfusion pressure
without negatively affecting cerebral blood flow in hypertensive gravidas. Hypertens Pregnancy.
2002;21:185–197.
29. Impey L. Severe hypotension and fetal distress following sublingual administration of nifedipine to a
patient with severe pregnancy induced hypertension at 33 weeks. Br J Obstet Gynaecol.
1993;100:959–961.
30. Neustein S, Dimich I, Shiang H, et al. Cardiovascular consequences of the concomitant administration
of nifedipine and magnesium sulfate in pigs. Int J Obstet Anesth. 1998;7:247–250.
31. Bolte AC, Dekker GA, van Eyck J, et al. Lack of agreement between central venous pressure and
pulmonary capillary wedge pressure in preeclampsia. Hypertens Pregnancy. 2000;19:261–271.
32. Rodgers RP, Levin J. A critical reappraisal of the bleeding time. Semin Thromb Hemost. 1990;16:1–20.
33. Beilin Y, Arnold I, Hossain S. Evaluation of the platelet function analyzer (PFA-100) vs. the
thromboelastogram (TEG) in the parturient. Int J Obstet Anesth. 2006;15:7–12.
34. Beilin Y, Zahn J, Comerford M. Safe epidural analgesia in thirty parturients with platelet counts
between 69,000 and 98,000 mm(−3). Anesth Analg. 1997;85:385–388.
35. Vandermeulen EP, Van Aken H, Vermylen J. Anticoagulants and spinal-epidural anesthesia. Anesth
Analg. 1994;79:1165–1177.
36. Abboud T, Artal R, Sarkis F, et al. Sympathoadrenal activity, maternal, fetal, and neonatal responses
after epidural anesthesia in the preeclamptic patient. Am J Obstet Gynecol. 1982;144:915–918.
37. Jouppila P, Jouppila R, Hollmén A, et al. Lumbar epidural analgesia to improve intervillous blood
flow during labor in severe preeclampsia. Obstet Gynecol. 1982;59:158–161.
38. Ramanathan J, Vaddadi AK, Arheart KL. Combined spinal and epidural anesthesia with low doses of
intrathecal bupivacaine in women with severe preeclampsia: a preliminary report. Reg Anesth Pain
Med. 2001;26:46–51.
39. Wallace DH, Leveno KJ, Cunningham FG, et al. Randomized comparison of general and regional
anesthesia for cesarean delivery in pregnancies complicated by severe preeclampsia. Obstet Gynecol.
1995;86:193–199.
40. Hood DD, Curry R. Spinal versus epidural anesthesia for cesarean section in severely preeclamptic
patients: a retrospective survey. Anesthesiology. 1999;90:1276–1282.
41. Aya AG, Vialles N, Tanoubi I, et al. Spinal anesthesia-induced hypotension: a risk comparison
between patients with severe preeclampsia and healthy women undergoing preterm cesarean delivery.
Anesth Analg. 2005;101:869–875.
42. Ramanathan J, Sibai BM, Mabie WC, et al. The use of labetalol for the attenuation of the hypertensive
response to endotracheal intubation in preeclampsia. Am J Obstet Gynecol. 1988;159:650–654.
43. Cetin A, Yurtcu N, Guvenal T, et al. The effect of glyceryl trinitrate on hypertension in women with
severe preeclampsia, HELLP syndrome, and eclampsia. Hypertens Pregnancy. 2004;23:37–46.
44. Isler CM, Barrilleaux PS, Rinehart BK, et al. Postpartum seizure prophylaxis: using clinical
parameters to guide therapy. Obstet Gynecol. 2003;101:66–69.
45. Harskamp RE, Zeeman GG. Preeclampsia: at risk for remote cardiovascular disease. Am J Med Sci.
2007;334:291–295.
46. Wenger, NK. Recognizing pregnancy-associated cardiovascular risk factors. Am J Cardiol.
2014;113:406–409.
47. Innes KE, Byers TE. First pregnancy characteristics and subsequent breast cancer risk among young
women. Int J Cancer. 2004;112:306–311.
Endocrine Disorders
Jessica Booth, Peter H. Pan, Janine Malcolm, and Erin J. Keely
I. Diabetes mellitus
A. Introduction
B. Effect of pregnancy on diabetes mellitus
C. Effect of diabetes mellitus on pregnancy
D. Management of diabetes mellitus during pregnancy
E. Management during labor and delivery
II. Thyroid disorders
A. Introduction
B. Thyroid physiology
C. Hyperthyroidism
D. Hypothyroidism
III. Pituitary disorders
A. Introduction
B. Pituitary adenomas
C. Pituitary insufficiency during pregnancy
IV. Adrenal disorders
A. Primary adrenal insufficiency
B. Cushing disease and syndrome
C. Pheochromocytoma
V. Conclusion
KEYPOINTS
1. Diabetes and thyroid disease are the most common endocrine disorders
associated with pregnancy.
2. Neonatal macrosomia and/or shoulder dystocia occur with increased
frequency in pregnancy complicated by all types of diabetes mellitus.
3. Appropriate maternal glycemic control with oral hypoglycemic agents or
insulin is necessary to reduce fetal and maternal complications when diet
modification alone is insufficient.
4. A preanesthetic evaluation of the obstetric patient with thyroid disease
should include an evaluation of current symptoms, presence of goiter,
adequacy of treatment, and cardiovascular symptoms.
5. Early diagnosis and appropriate treatment of maternal
pheochromocytoma is critical to reduce maternal and fetal mortality.
I. Diabetes mellitus
A. Introduction
The prevalence of diabetes mellitus (DM) in pregnancy is
estimated to be 6% to 7%.1 Gestational diabetes is typically
defined as glucose intolerance with onset during the second or
third trimester of pregnancy. Overt or pregestational diabetes
is diagnosed using American Diabetes Association criteria of a
screening fasting blood glucose >125 mg per dL or by
combining the results of other standard diagnostic tests
including elevated hemoglobin A1c and elevated random
blood glucose.2 Screening for gestational diabetes is typically
performed at 24 to 28 weeks’ gestation with a 1-hour diagnostic
oral glucose tolerance test. Patients with a blood glucose >135 to
140 mg per dL after the 50-g 1-hour glucose screening test will
undergo a 100-g, 3-hour diagnostic oral glucose tolerance test.
Alternatively, some national organizations recommend a one-step
2-hour 75-g test.1
A pregnancy complicated by DM requires multidisciplinary
cooperation among obstetricians, internists/endocrinologists,
anesthesia providers, nurse educators, and dietitians to facilitate a
healthy outcome for mother and child. A woman may have a
known type 1 or type 2 DM (e.g., pregestational DM) or be
identified during pregnancy with gestational diabetes mellitus
(GDM). A small percentage of women will present with new-
onset, previously unidentified type 1 or type 2 DM during
pregnancy, which may be difficult to separate from GDM until
after delivery. Although the clinical management and risks of
types 1 and 2 DM during pregnancy are similar, there are
important differences in pathogenesis and associated conditions.
1. Type 1 DM results from the destruction of insulin-secreting
cells by an immune-mediated process leading to absolute
insulin deficiency. It represents 5% to 10% of all types of
DM. Some patients will be identified as having type 1 disease
by a sudden onset of diabetic ketoacidosis (DKA). However,
others, especially those who present in adulthood, may have a
more insidious onset. These women may be misidentified as
having type 2 DM until they develop DKA under stressful
situations such as pregnancy.
2. Type 2 DM is part of the metabolic syndrome, which includes
insulin resistance, visceral adiposity, dyslipidemia,
hypertension, accelerated cardiovascular disease, and
polycystic ovary disease. The prevalence of type 2 DM in the
pregnant population is rising as the incidence of maternal
obesity and advanced maternal age increases. Individuals with
DM, identified before pregnancy, may become pregnant while
being treated with diet and exercise, oral hypoglycemic
agents, or insulin. Obesity and hypertension often
associated with DM bring additional risk to the pregnancy
that must be identified and managed. Tight glycemic
control is required for optimal management.
3. GDM is defined as any degree of glucose intolerance with
onset during pregnancy, but does not exclude the possibility
of pregestational DM. It accounts for approximately 90% of
DM during pregnancy. The purpose of screening for GDM is
to identify pregnancies at increased risk for poor perinatal
outcomes and also to identify a cohort of women at increased
lifetime risk for type 2 DM. Although controversy continues
concerning screening and treatment strategies, most expert
groups recommend universal screening for all pregnant
women. Obesity, advanced maternal age, family history, and
glucose intolerance with prior pregnancy, are risk factors for
gestational diabetes in the United States.3 Women of color are
also at increased risk. Although most women are screened for
GDM at 24 to 28 weeks’ gestation, earlier screening may be
warranted in obese women with a body mass index (BMI)
≥30, women with a prior history of GDM, or women with a
known history of impaired glucose metabolism.1
B. Effect of pregnancy on diabetes mellitus
1. Glycemic control
Three important physiologic changes in glucose
metabolism occur during pregnancy: (a) increased risk of
fasting hypoglycemia and associated hypoglycemia
unawareness, (b) insulin resistance, and (c) accelerated
starvation resulting in increased lipolysis and ketone
production. Hypoglycemia unawareness is more prevalent
due to lack of autonomic warning from reduced
catecholamine production.4 Insulin sensitivity is decreased
by approximately 50% by the third trimester; insulin dosages
of two or three times the prepregnancy dose are often needed
during the second and third trimesters in insulin dependent
women.1 There is immediate onset of a rapid return of insulin
sensitivity postpartum, necessitating a marked reduction in
insulin dose (typically to two-thirds of the dose used
prepregnancy) immediately following delivery of the
placenta. The accelerated starvation state of pregnancy
produces lower fasting glucose levels and increased ketone
production, predisposing women to ketoacidosis. Other
stressors such as hyperemesis and corticosteroid use to
promote fetal lung maturity also predispose to keotacidosis.
2. Complications of diabetes mellitus
a. Women with type 1 or type 2 DM may have significant
microvascular disease and rarely macrovascular
disease. Approximately 34% to 50% of women with type
1 and 3% of women with type 2 DM will have evidence
of retinopathy. The presence and degree of diabetic
retinopathy are associated with the duration of DM and
long term glycemic control. Most studies show some
worsening of retinopathy during pregnancy.5
b. Diabetic nephropathy represents a spectrum of disease
ranging from microalbuminuria (microalbumin, 30 to 300
mg per dL or a microalbumin-to-creatinine ratio .2 mg per
μmol) to overt nephropathy (.300 mg of protein per 24
hours) to end-stage renal failure. Nephropathy
complicates approximately 4% of diabetic pregnancies
and is associated with increased maternal and perinatal
morbidity rates. Pregnancy may worsen the degree of
proteinuria, causing progression of renal insufficiency
which aggravates hypertension. In most cases, renal
function will return to baseline values postpartum;
however, in women with severe nephropathy or serum
creatinine >1.4 mg per dL, there is risk of a permanent
decline in GFR.6 One retrospective study further
suggested that diabetic parturients with moderate-to-
severe renal dysfunction at pregnancy onset had increased
risk of requiring dialysis about 36 months earlier than
predicted by prepregnancy estimates based on linear
decline in GFR.7
4. Postpartum care
a. Insulin requirements. Following delivery of the
placenta, insulin sensitivity returns immediately.
Maternal hypoglycemia should be avoided. A reduction of
insulin dosage to two-thirds of the prepregnancy
requirement is prudent. For women with type 1 DM, the
first SQ dose of insulin should be administered before
discontinuing an IV infusion due to the short half-life of
IV insulin. For women on oral agents or diet before
pregnancy, insulin should be used only if the patient
clearly has postpartum hyperglycemia.
b. Neonatal hypoglycemia must be anticipated and local
protocols established. In general, infants born to these
mothers should have frequent capillary glucose
measurements and, if the blood glucose level is <2 μmol
per L (<36 mg per dL), early oral feeding or the use of IV
dextrose should be implemented. Other neonatal
complications include (a) hypocalcemia, (b) jaundice, (c)
polycythemia, (d) septal hypertrophy, (e) sacral agenesis,
and (f) respiratory distress syndrome.
C. Hyperthyroidism
1. Definition and pathophysiology
Hyperthyroidism is defined by excessive thyroid hormone
production. It can be subclinical (suppressed TSH, normal
T4 and T3) or overt (suppressed TSH and elevated T4 and/or
T3). Severe thyrotoxicosis is associated with poor obstetric
outcomes. Women with uncontrolled hyperthyroidism have a
fivefold greater risk of developing severe preeclampsia.27
There is also an increased risk of fetal loss, low birth weight,
prematurity, and placental abruption, which appear to be
directly related to the blood level of maternal T4. Women who
are appropriately treated and euthyroid before pregnancy do
not have increased risk for these poor outcomes.28
2. Physiology of hyperthyroidism
In the first trimester, 10% to 20% of normal women will
have a suppressed TSH due to the effect of hCG. Some
women, especially those with high hCG levels associated with
hyperemesis, molar pregnancy, and twins will have
gestational thyrotoxicosis, also referred to as first-trimester
thyrotoxicosis and hCG-related thyrotoxicosis. This temporary
condition improves as hCG levels decrease and does not
usually require medical intervention. The most common cause
of preexisting hyperthyroidism is Graves disease, an
autoimmune hyperthyroidism secondary to stimulation of the
thyroid gland by TSHrAb. Other causes include (a) toxic
solitary nodule, (b) multiple nodular goiter, (c) ingestions
(e.g., exogenous thyroid hormone, amiodarone, excess
iodine), and (d) subacute thyroiditis (see Table 23.3).
6. Antepartum considerations
a. Medications. The treatment goal is a free T4 level in
the high normal range using the lowest dose of
thioamide possible. For symptom relief, β-blockers can
be used until the free T4 levels come into the normal
range. Table 23.433 summarizes the common drugs and
their action of mechanism for treatment of
hyperthyroidism. Excessive use of antithyroid medication
can result in fetal hypothyroidism and fetal goiter and
must be avoided. Gestational thyrotoxicosis is self-
limiting and does not require thioamide treatment. For
individuals who develop adverse reactions to
thioamides, especially significant leukopenia, or liver
abnormalities, a thyroidectomy in pregnancy may be
required.
5. Intrapartum considerations
Most patients with pituitary disease require no special
anesthetic considerations during labor and delivery.
However, Valsalva maneuvers associated with labor and
delivery may increase intracranial pressure and potentially
increase the risk of pituitary apoplexy in patients with
pituitary macroadenomas. An assisted vaginal delivery
should also be considered. Women should therefore be
monitored for signs and symptoms such as severe headache
and hemodynamic compromise. Stress-dose corticosteroids
may be required in patients with macroadenomas during labor
and delivery. This dose should be discontinued over 2 to 3
days following delivery.
C. Pituitary insufficiency during pregnancy
1. Diagnosis
Pituitary insufficiency is difficult to diagnose during
pregnancy because the symptoms may be attributed to
pregnancy alone. Also, dynamic testing is difficult. Pituitary
insufficiency may be caused by a number of disorders (see
Table 23.5) and can present before pregnancy, or become
clinically significant during pregnancy (i.e., lymphocytic
hypophysitis, diabetes insipidus), or postpartum (i.e.,
Sheehan syndrome).
2. Pathophysiology
In cases of hypopituitarism present before conception, the
destruction of the pituitary results in impairment of its ability
to secrete some or all of the releasing hormones. Deficiencies
of cortisol, thyroid hormone, follicle-stimulating hormone
(FSH), luteinizing hormone (LH), and vasopressin can
result. These women usually present before pregnancy with
infertility. Management of patients with preexisting pituitary
insufficiency should include a review of medications and a
detailed surveillance plan outlined for pregnancy. If the
hormonal deficiency is partial, screening for deficiencies of
other pituitary hormones should be completed. Hormonal
replacement should be optimized. Increased thyroid hormone
may be needed during gestation to meet the increased
requirements for thyroxine during pregnancy. In this case,
measurement of TSH is unhelpful because it will always be
low in patients with hypopituitarism. The free T4 level should
be monitored and kept within the normal range. Cortisol
replacement should be maintained at prepregnancy
replacement doses, unless there is an additional illness or
stress such as hyperemesis, fever, or labor. Periodic
measurements of electrolytes and blood pressure (BP) may
be helpful in determining the adequacy of cortisol
replacement.
a. Lymphocytic hypophysitis
Lymphocytic hypophysitis is an autoimmune disease
causing infiltration of lymphocytes and plasma cells
leading to destruction of the pituitary. Women may
present in pregnancy with signs of an expanding
intrasellar mass (e.g., visual field changes, cranial nerve
palsies, or headache) or hypopituitarism. Immediate
surgical intervention is unnecessary (unless there is
evidence of visual field defects, intractable headaches, or
radiologic evidence of an expanding mass) because
women may undergo a spontaneous regression of the
mass and restoration of pituitary function.47 A short
course of high-dose corticosteroids may produce dramatic
resolution of visual field defects in some women.48
b. Diabetes insipidus
Diabetes insipidus (DI) occurs rarely during pregnancy
with an incidence of 0.004% Polyuria, polydipsia,
extreme thirst, and dehydration are symptoms. The
diagnosis of DI is often made clinically, with laboratory
confirmation. A high urine output, low urine osmolality
<275 mOsm per kg, and high serum osmolality (>290
mOsm per kg) are the hallmarks of DI. Gestational DI is
thought to be caused by increased vasopressinase (a
placental enzyme that metabolizes endogenous arginine-
vasopressin (AVP) but not synthetic DDAVP) activity
with lower renal receptiveness to AVP. Gestational DI has
been associated with preeclampsia, the hemolysis,
elevated liver enzymes, low platelets (HELLP) syndrome,
and fatty liver of pregnancy, likely because the liver is
partially responsible for the metabolism of
vasopressinase. Electrolytes and volume status in
pregnant women with DI should be carefully monitored.49
c. Sheehan syndrome
The pathogenesis of Sheehan syndrome is unclear, but is
felt to be secondary to ischemia of the pituitary gland. The
acute diagnosis of Sheehan syndrome should be
considered in women with postpartum hemorrhage who
are unresponsive to fluid resuscitation. Prolactin levels
that are normally high during pregnancy may be low. An
adrenocorticotropic hormone (ACTH) stimulation test
may be normal as the adrenals have not yet had the time
to atrophy as in other patients with pituitary insufficiency.
Acute pituitary insufficiency, as in the case of acute
Sheehan syndrome, is a medical emergency with
potentially fatal outcomes for both mother and fetus.
Baseline laboratory studies should be obtained, but
therapy should be initiated while awaiting results. Fluid
resuscitation in combination with stress-dose
corticosteroids (i.e., hydrocortisone 100 mg IV every 8
hours) is required. IV glucose may be necessary for
treatment of hypoglycemia. It is important to observe for
polyuria following administration of corticosteroids
because their administration may unmask diabetes
insipidus. If this occurs, an evaluation for DI should be
performed. Thyroid hormone replacement may be
instituted if levels are found to be low.
3. Antepartum risks
Untreated and partially treated hypopituitarism increases the
risk of maternal hypotension and hypoglycemia. Rates of
operative delivery may be increased. Small for gestational
age infants, preterm birth, and intrauterine fetal death have
been reported in patients with preexisting hypopituitarism.
Poorer outcomes may be related to inadequate hormonal
replacement in the mother. Congenital malformations have
not been reported.
4. Intrapartum considerations
Stress corticosteroids will be required for labor and delivery.
Hydrocortisone 100 mg IV q8h should be administered and
decreased to replacement doses over 2 to 3 days following
delivery or postpartum.
C. Pheochromocytoma
Pheochromocytoma is a rare cause of hypertension with an
incidence of approximately 0.1% in the general population.56
Reported prevalence in pregnancy is 0.02%.57 Despite its rarity in
pregnancy, pheochromocytoma is potentially lethal for both
mother and fetus with mortality rates of up to 17% and 40%,
respectively, if untreated; however, prenatal diagnosis with
appropriate therapy dramatically reduces the maternal and fetal
mortality rate to <1% and <15%, respectively.58
Pheochromocytoma should therefore be considered as part of the
differential diagnosis of hypertension during pregnancy.
1. Maternal and fetal risks
a. Maternal risks include hypertensive crisis, which can
lead to (i) myocardial infarction (MI), (ii) stroke, (iii)
dysrhythmias, (iv) CHF, and (v) death. Hypertensive
crisis may be precipitated by general anesthesia, vaginal
delivery, and/or mechanical compression of the tumor
caused by an enlarging uterus.
b. Fetal risks are increased because of severe maternal
hypertension that may lead to IUGR, uteroplacental
insufficiency, fetal hypoxia, and death.
2. When to suspect a pheochromocytoma
Signs that differentiate pheochromocytoma from pregnancy-
related hypertension include (i) paroxysmal nature of the
hypertension; (ii) lack of proteinuria, ankle edema, or elevated
plasma uric acid level; (iii) presence of hypertensive
complications such as heart failure and pulmonary edema; and
(iv) presence of unexplained orthostatic hypotension.58 BP
might paradoxically worsen if a predominately β-blocking
drug such as labetalol were given due to unopposed alpha
adrenergic effect. Women with pheochromocytoma generally
present with labile and difficult to control hypertension. A
20% incidence of proteinuria is seen with
pheochromocytoma, making it difficult to distinguish from
preeclampsia. Pheochromocytoma should be suspected in
women with paroxysmal symptoms such as headache,
tremors, or sweating. Severe and intermittent hypertension,
lack of edema, development of hypertension before 20 weeks,
and the coexistence of GDM may be other clues to the
diagnosis. Most pheochromocytomas are sporadic. However,
there are several familial neuroendocrine disorders of
which pheochromocytoma is a part. These include multiple
endocrine neoplasia (MEN) type 2A, MEN type 2B, von
Hippel-Lindau disease, and neurofibromatosis. Family history
should be obtained and the patient examined for clinical
features of these syndromes.
3. Diagnosis of pheochromocytoma
a. The most sensitive diagnostic test for pheochromocytoma
is differential measurement of metanephrines in urine
or in plasma because these measurements are unaffected
by pregnancy. A baseline 24-hour urine collection should
be obtained. If this sample is normal, a 24-hour collection
starting at the onset of a clinical episode should be
obtained. In general, a value greater than two times
normal indicates pheochromocytoma.
b. Several pharmacologic agents interfere with
measurement of catecholamine levels. These include (i)
tricyclic antidepressants, (ii) labetalol, (iii) methyldopa,
(iv) ethanol, and (v) benzodiazepines, all of which
increase catecholamine values. Use of agents that
interfere with catecholamine measurement should be
discontinued 7 to 14 days before sample collection.59
c. Anatomic localization of the tumor should be performed
by MRI because this imaging modality is not associated
with harm to the fetus. MRI has a sensitivity of close to
100% for lesions >1.5 cm and can be used to document
extraadrenal lesions. Other imaging modalities have been
used, but CT scanning requires ionizing radiation and
contrast dye. Ultrasonography lacks sensitivity.
Metaiodobenzylguanidine (MIBG) scanning is
contraindicated in pregnancy.
c. Neuraxial anesthesia
Epidural anesthesia has been used successfully for
both excisions of pheochromocytoma and CD in
pregnant patients.66 Spinal anesthesia can produce
sudden hemodynamic changes that require vasopressor
therapy and therefore epidural anesthesia is preferred
for most cases. Adequate preoperative α-blockade and
reexpansion of plasma volume are essential before
initiating anesthesia. The use of a combined epidural
technique with general anesthesia must be balanced
against the risk of hypertension during epidural
placement. In addition, sympathetic blockade resulting
from epidural anesthesia does not protect against
catecholamine release during tumor manipulation.67
8. Fetal considerations
Transient neonatal hypotension may be seen in the neonate
following delivery due to transplacental passage of
phenoxybenzamine. Magnesium use can cause neonatal
hypotonia.
V. Conclusion
DM and other endocrine disorders carry significant maternal and fetal
risks during pregnancy. However, the risks can be greatly reduced by
timely diagnosis, optimizing treatment, and involving a
multidisciplinary team in patient care. Communication between
obstetrician, anesthesiologist, and endocrinologist is of utmost
importance. Clear coordinated care plans that are well
documented will aid in producing positive obstetric outcomes for
the affected mothers and their newborns.
REFERENCES
1. American College of Obstetricians and Gynecologists. ACOG Practice Bulletin No. 137: gestational
diabetes mellitus. Obstet Gynecol. 2013;122:406–416.
2. Hedderson MM, Gunderson EP, Ferrara A. Gestational weight gain and risk of gestational diabetes
mellitus. Obstet Gynecol. 2010;115:597–604.
3. Ferrara A. Increasing prevalence of gestational diabetes mellitus: a public health perspective. Diabetes
Care. 2007;30(suppl 2): S141–S146.
4. Kimmerle R, Heinemann L, Delecki A, et al. Severe hypoglycemia incidence and predisposing factors
in 85 pregnancies of type 1 diabetic women. Diabetes Care. 1992;15:1034–1037.
5. Best RM, Chakravarthy U. Diabetic retinopathy in pregnancy. Br J Ophthalmol. 1997;81:249–251.
6. Miodovnik M, Rosenn BM, Khoury JC, et al. Does pregnancy increase the risk for development and
progression of diabetic nephropathy? Am J Obstet Gynecol. 1996;174:1180–1191.
7. Purdy LP, Hantsch CE, Molitch ME, et al. Effect of pregnancy on renal function in patients with
moderate-to-severe diabetic renal insufficiency. Diabetes Care. 1996;19:1067–1074.
8. White P. Classification of obstetric diabetes. Am J Obstet Gynecol. 1978;130:228–230.
9. Casey BM, Lucas MJ, Mcintire DD, et al. Pregnancy outcomes in women with gestational diabetes
compared with the general obstetric population. Obstet Gynecol. 1997;90:869–873.
10. Macintosh MC, Fleming KM, Bailey JA, et al. Perinatal mortality and congenital anomalies in babies
of women with type 1 or type 2 diabetes in England, Wales, and Northern Ireland: population based
study. BMJ. 2006;333:177.
11. Boulet SL, Alexander GR, Salihu HM, et al. Macrosomic births in the United States: determinants,
outcomes, and proposed grades of risk. Am J Obstet Gynecol. 2003;188:1372–1378.
12. Langer O, Levy J, Brutsman L, et al. Glycemic control in gestational diabetes mellitus—how tight is
tight enough: small for gestational age versus large for gestational age? Am J Obstet Gynecol.
1989;161:646–653.
13. Falavigna M, Schmidt MI, Trujillo J, et al. Effectiveness of gestational diabetes treatment: a
systematic review with quality of evidence assessment. Diabetes Res Clin Pract. 2012;98:396–405.
14. Dabelea D. The predisposition to obesity and diabetes in offspring of diabetic mothers. Diabetes Care.
2007;30(suppl 2): S169–S174.
15. American College of Obstetricians and Gynecologists Committee on Practice Bulletins—Obstetrics.
ACOG Practice Bulletin: clinical management guidelines for obstetricians-gynecologists: number 60,
March 2005: pregestational diabetes mellitus. Obstet Gynecol. 2005;105:675–685.
16. Mathiesen ER, Kinsley B, Amiel SA, et al. Maternal glycemic control and hypoglycemia in type 1
diabetic pregnancy: a randomized trial of insulin aspart versus human insulin in 322 pregnant women.
Diabetes Care. 2007;30:771–776.
17. Hirsch IB. Insulin analogues. N Engl J Med. 2005;352:174–183.
18. Lv S, Wang J, Xu Y. Safety of insulin analogs during pregnancy: a meta-analysis. Arch Gynecol
Obstet. 2015. http://www.ncbi.nlm.nih.gov/pubmed/25855052. Accessed May 5, 2015.
19. Ekpebegh CO, Coetzee EJ, van der Merwe L, et al. A 10-year retrospective analysis of pregnancy
outcome in pregestational type 2 diabetes: comparison of insulin and oral glucose-lowering agents.
Diabetic Med. 2007;24:253–258.
20. Su DF, Wang XY. Metformin vs insulin in the management of gestational diabetes: a systematic
review and meta-analysis. Diabetes Res Clin Pract. 2014;104:353–357.
21. Langer O, Yogev Y, Xenakis EM, et al. Insulin and glyburide therapy: dosage, severity level of
gestational diabetes, and pregnancy outcome. Am J Obstet Gynecol. 2005;192:134–139.
22. Carroll MA, Yeomans ER. Diabetic ketoacidosis in pregnancy. Crit Care Med. 2005;33(suppl
10):S347–S353.
23. Nayak S, Lippes HA, Lee RV. Hyperglycemic hyperosmolar syndrome (HHS) during pregnancy. J
Obstet Gynaecol. 2005;25:599–601.
24. Gordon M, Landon MB, Samuels P, et al. Perinatal outcome and long-term follow-up associated with
modern management of diabetic nephropathy. Obstet Gynecol. 1996;87:401–409.
25. Boulvain M, Stan C, Irion O. Elective delivery in diabetic pregnant women. Cochrane Database Syst
Rev. 2001;(2):CD001997.
26. Mendiola J, Grylack LJ, Scanlon JW. Effects of intrapartum maternal glucose infusion on the normal
fetus and newborn. Anesth Analg. 1982;61:32–35.
27. Millar LK, Wing DA, Leung AS, et al. Low birth weight and preeclampsia in pregnancies complicated
by hyperthyroidism. Obstet Gynecol. 1994;84:946–949.
28. Gyamfi C, Wapner RJ, D’Alton ME. Thyroid dysfunction in pregnancy: the basic science and clinical
evidence surrounding the controversy in management. Obstet Gynecol. 2009;113:702–707.
29. Mestman JH. Hyperthyroidism in pregnancy. Curr Opin Endocrinol Diabetes Obes. 2012;19:394–401.
30. Laurberg P, Nygaard B, Glinoer D, et al. Guidelines for TSH-receptor antibody measurements in
pregnancy: results of an evidence-based symposium organized by the European Thyroid Association.
Eur J Endocrinol. 1998;139:584–586.
31. Mandel SJ, Cooper DS. The use of antithyroid drugs in pregnancy and lactation. J Clin Endocrinol
Metab. 2001;86:2354–2359.
32. Mestman JH. Hyperthyroidism in pregnancy. Best Pract Res Clin Endocrinol Metab. 2004;18:267–
288.
33. Pan PH, Tonidandel AM. Anesthesia for pregnant patients with endocrine disorders. In: Suresh MS,
Segal BS, Preston R, et al, eds. Shnider and Levinson’s Anesthesia for Obstetrics. 5th ed. Philadelphia,
PA: Lippincott Williams & Wilkins; 2013: 462–484.
34. Luton D, Le Gac I, Vuillard E, et al. Management of Graves’ disease during pregnancy: the key role of
fetal thyroid gland monitoring. J Clin Endocrinol Metab. 2005;90:6093–6098.
35. Pugh S, Lalwani K, Awal A. Thyroid storm as a cause of loss of consciousness following anaesthesia
for emergency caesarean section. Anaesthesia. 1994;49:35–37.
36. Nayak B, Burman K. Thyrotoxicosis and thyroid storm. Endocrinol Metab Clin North Am.
2006;35:663–686.
37. Sheffield JS, Cunningham FG. Thyrotoxicosis and heart failure that complicate pregnancy. Am J
Obstet Gynecol. 2004;190:211–217.
38. Duggal J, Singh S, Kuchinic P, et al. Utility of esmolol in thyroid crisis. Can J Clin Pharmacol.
2006;13:e292–e295.
39. Bowman ML, Bergmann M, Smith JF. Intrapartum labetalol for the treatment of maternal and fetal
thyrotoxicosis. Thyroid. 1998;8:795–796.
40. Zadra N, Giusti F, Midrio P. Ex utero intrapartum surgery (EXIT): indications and anaesthetic
management. Best Pract Res Clin Anaesthesiol. 2004;18:259–271.
41. Casey BM, Leveno KJ. Thyroid disease in pregnancy. Obstet Gynecol. 2006;108:1283–1292.
42. Leung AS, Millar LK, Koonings PP, et al. Perinatal outcome in hypothyroid pregnancies. Obstet
Gynecol. 1993;81:349–353.
43. Wartofsky L. Myxedema coma. Endocrinol Metab Clin North Am. 2006;35:687–698.
44. Chandraharan E, Arulkumaran S. Pituitary and adrenal disorders complicating pregnancy. Curr Opin
Obstet Gynecol. 2003;15:101–106.
45. Molitch ME. Medical management of prolactin-secreting pituitary adenomas. Pituitary. 2002;5:55–65.
46. Kupersmith MJ, Rosenberg C, Kleinberg D. Visual loss in pregnant women with pituitary adenomas.
Ann Intern Med. 1994;121:473–477.
47. McGrail KM, Beyerl BD, Balck PM, et al. Lymphocytic adenohypophysitis of pregnancy with
complete recovery. Neurosurgery. 1987;20:791–793.
48. Reusch JE, Kleinschmidt-DeMasters BK, Lillehei KO, et al. Preoperative diagnosis of lymphocytic
hypophysitis (adenohypophysitis) unresponsive to short course of dexamethasone: case report.
Neurosurgery. 1992;30:268–272.
49. Kalelioglu I, Kubat Uzum A, Yildirim A, et al. Transient gestational diabetes insipidus diagnosed in
successive pregnancies: review of pathophysiology, diagnosis, treatment, and management of delivery.
Pituitary. 2007;10:87–93.
50. Czeizel AE, Rockenbauer M. Population-based case-control study of teratogenic potential of
corticosteroids. Teratology. 1997;56:335–340.
51. Delibasi T, Ustun I, Aydin Y, et al. Early severe pre-eclamptic findings in a patient with Cushing’s
syndrome. Gynecol Endocrinol. 2006;22:710–712.
52. Orth DN. Cushing’s syndrome. N Engl J Med. 1995;332:791–803.
53. Aron DC, Schnall AM, Sheeler LR. Cushing’s syndrome and pregnancy. Am J Obstet Gynecol.
1990;162:244–252.
54. Buescher MA, McClamrock HD, Adashi EY. Cushing syndrome in pregnancy. Obstet Gynecol.
1992;79:130–137.
55. Glassford J, Eagle C, McMorland GH. Caesarean section in a patient with Cushing’s syndrome. Can
Anaesth Soc J. 1984;31:447–450.
56. Mannelli M, Bemporad D. Diagnosis and management of pheochromocytoma during pregnancy. J
Endocrinol Invest. 2002;25:567–571.
57. Harper MA, Murnaghan GA, Kennedy L, et al. Phaeochromocytoma in pregnancy: five cases and a
review of the literature. Br J Obstet Gynaecol. 1989;96:594–606.
58. Kamoun M, Mnif MF, Charfi N, et al. Adrenal diseases during pregnancy: pathophysiology, diagnosis,
and management strategies. Am J Med Sci. 2014;347:64–73.
59. Young WF Jr. Pheochromocytoma and primary aldosteronism: diagnostic approaches. Endocrinol
Metab Clin North Am. 1997;26:801–827.
60. Botchan A, Hauser R, Kupfermine M, et al. Pheochromocytoma in pregnancy: case report and review
of the literature. Obstet Gynecol Surv. 1995;50:321–327.
61. Fleisher LA, Mythen M. Anesthetic implications of concurrent diseases. In: Miller RD, ed. Miller’s
Anesthesia. 8th ed. Philadelphia, PA: Elsevier; 2015:1156–1225.
62. Dugas G, Fuller J, Singh S, et al. Pheochromocytoma and pregnancy: a case report and review of
anesthetic management. Can J Anaesth. 2004;51:134–138.
63. James MF, Huddle KR, Owen AD, et al. Use of magnesium sulphate in the anaesthetic management of
phaeochromocytoma in pregnancy. Can J Anaesth. 1988;35:178–182.
64. Hudsmith JG, Thomas CE, Browne DA. Undiagnosed phaeochromocytoma mimicking severe
preeclampsia in a pregnant woman at term. Int J Obstet Anesth. 2006;15:240–245.
65. Pullerits J, Ein S, Balfe JW. Anaesthesia for phaeochromocytoma. Can J Anaesth. 1988;35:526–534.
66. Stonham J, Wakefield C. Phaeochromocytoma in pregnancy: caesarean section under epidural
analgesia. Anaesthesia. 1983;38:654–658.
67. Kinney MA, Narr BJ, Warner MA. Perioperative management of pheochromocytoma. J Cardiothorac
Vasc Anesth. 2002;16:359–369.
Thrombophilias/Coagulopathies
James P.R. Brown and M. Joanne Douglas
I. Overview of normal hemostatic coagulation
II. Physiologic changes of coagulation in pregnancy
A. Platelets
B. Coagulation factors
C. Thrombotic control
D. Fibrinolysis
III. Measurements of coagulation in pregnancy
A. Routine tests
B. Bleeding time
C. Near patient testing
IV. Thrombophilia
A. Risk factors for thromboembolic disease in pregnancy
B. Inherited thrombophilias
C. Management of parturients with suspected thrombophilia
D. Acquired thrombophilias
E. Anesthetic implications: neuraxial anesthesia and
anticoagulants
V. Disorders of coagulation
A. Inherited disorders of coagulation
B. Acquired disorders of coagulation
C. Disseminated intravascular coagulation
Summary
KEYPOINTS
1. Hypercoagulability. Normal parturients are hypercoagulable at term.
Parturients are at risk for venous thromboembolic events (VTEs) during
pregnancy and up to 6 weeks’ postpartum.
2. Predicting bleeding risk. No single blood test can predict the risk of
clinically significant bleeding, for example, neuraxial hematoma from
epidural anesthesia or postpartum hemorrhage.
3. Thromboembolic risk reduction. Management of anticoagulation
during pregnancy should be tailored to individual risk. Considerations
include inherited thrombophilias, family, and personal history of previous
VTE.
4. Anesthetic management. When considering neuraxial anesthesia, there
should be an individual assessment of risk versus benefit. Peripartum
management requires multidisciplinary input (obstetrician,
anesthesiologist, hematologist).
B. Inherited thrombophilias
Inherited thrombophilias are responsible for 30% of maternal
VTE; half of these are a result of either Factor V (FV) Leiden or
prothrombin gene abnormalities.15,16 Genes associated with
increased VTE risk are carried by 15% of the Caucasian
population.17
1. Factor V Leiden
a. Most prevalent inherited thrombophilia13
b. Prevalence highest in European Caucasian populations
c. Mutation in FV gene, rendering FV resistant to
inactivation by activated PC (Fig. 24.4). This leads to
slower breakdown of FV, increased thrombin production,
and a prothrombotic state.
d. Heterozygous state common: rarely leads to VTE unless
combined with another risk factor, such as pregnancy
e. Homozygous state increases VTE risk by up to 80
times.17
2. Prothrombin gene mutation
a. Point mutation (G20210A)
b. Increases the circulation of functionally normal
prothrombin
c. Most VTEs occur in those patients with additional risk
factors
d. Commonly inherited with FV Leiden. With both
mutations: 100-fold increase in VTE.16
3. Antithrombin deficiency
a. Previously known as antithrombin (AT) III deficiency
b. Most severe of the inherited thrombophilias
c. AT is a natural anticoagulant that binds and deactivates
thrombin: AT decreases the production and half-life of
thrombin. It also binds and deactivates Factors IXa, Xa,
XIa, and XIIa (Fig. 24.4).
d. Parturients with normal AT activity, but decreased
quantity, have a 400 times increased risk of VTE.15
e. There are greater than 250 mutations that either reduce
quantity or quality of AT production
f. Homozygous individuals are rare: most die in utero.
g. AT deficiency can be acquired, for example, liver disease,
malnutrition, or consumptive coagulopathy.
4. Protein C deficiency
a. Natural anticoagulant, degrades FVa and FVIIIa
b. Strong inhibitor of coagulation, activated by the
thrombin/thrombomodulin complex in the presence of
protein S (a cofactor)18 (Fig. 24.4)
c. PC deficiency results from more than 160 distinct
mutations, with variable quality and quantity of PC
produced.13
5. Protein S deficiency
a. Is a cofactor for activated PC
b. Also directly inactivates FVa and FXa
c. Normal pregnancy increases protein binding of PS,
reducing the availability of unbound active PS.
(1) Individuals homozygous for either PC or PS
deficiency present with neonatal purpura fulminans
and require lifelong anticoagulation.13
C. Management of parturients with suspected thrombophilia
1. Screening for thrombophilia
a. Screening results guide individual risk assessment and
decisions about anticoagulation. Testing for all genetic
mutations listed in Table 24.2 should take place prior to
pregnancy in parturients with:
V. Disorders of coagulation
A. Inherited disorders of coagulation
1. Common factor deficiencies. The anesthetic management of
parturients with common inherited disorders of coagulation is
summarized in Table 24.6 and Table 24.7.
a. von Willebrand disease
(1) Autosomal dominant condition
(2) Most common inherited bleeding disorder
(prevalence 1%)
(3) vWF required for adhesion of platelets to injured
endothelium and also prevents breakdown of
circulating FVIII.
(4) Classified into 3 types25
(a) Quantitative (type I: 75% of cases)
(b) Qualitative (type II)
(c) Complete absence (type III)
(5) vWF levels rise during pregnancy
(a) Patients with type I usually achieve normal
levels by the third trimester.3
(b) vWF levels and activity rarely improve in
patients with types II or III.
(6) A rapid decline in factor levels occurs postpartum
which increases the risk of PPH. Patients with von
Willebrand disease (vWD) are 15 to 20 times more
likely to suffer from delayed (>24 hours) PPH than
normal parturients.26
b. Hemophilia A (FVIII deficiency) and B (FIX
deficiency)
(1) Sex-linked autosomal recessive
(2) Occurs in 1 in 5,000 to 30,000 live male births
(3) Hemophilia A is characterized by deficient or
defective FVIII (activity <35%)3
(4) Hemophilia B (Christmas disease) is characterized by
a deficiency of FIX activity
(5) Female carriers of both conditions have 50% of the
respective factor levels; they are usually
asymptomatic.
(6) FVIII levels significantly increase in pregnancy; FIX
levels do not.
(7) Parturient carriers of hemophilia A or B are at risk for
primary and secondary PPH.
2. Rare factor deficiencies. A review of the management of
inherited bleeding disorders during pregnancy is provided by
the United Kingdom Haemophilia Doctors’ Organization
(UKHCDO).27,28 The suitability of neuraxial anesthesia for
such parturients should be considered on an individual basis,
in discussion with a hematologist. Case reports of successful
neuraxial procedures are described for most deficiencies with
appropriate factor replacement, but individual absolute risk is
difficult to quantify.3
REFERENCES
1. Holmes VA, Wallace JM. Haemostasis in normal pregnancy: a balancing act? Biochem Soc Trans.
2005;33:428–432.
2. Clark P. Changes of hemostasis variables during pregnancy. Semin Vasc Med. 2003;3:13–24.
3. Thornton P, Douglas J. Coagulation in pregnancy. Best Pract Res Clin Obstet Gynaecol. 2010;24:339–
352.
4. Cortet M, Deneux-Tharaux C, Dupont C, et al. Association between fibrinogen level and severity of
postpartum haemorrhage: secondary analysis of a prospective trial. Br J Anaesth. 2012;108:984–989.
5. Samama C, Simon L. Detecting coagulation disorders of pregnancy: bleeding time or platelet count?
Can J Anaesth. 2001;48:515–518.
6. Beilin Y, Arnold I, Hossain S. Evaluation of the platelet function analyzer (PFA-100) vs. the
thromboelastogram (TEG) in the parturient. Int J Obstet Anesth. 2006;15:7–12.
7. Davies JR, Fernando R, Hallworth SP. Hemostatic function in healthy pregnant and preeclamptic
women: an assessment using the platelet function analyzer (PFA-100) and thromboelastograph. Anesth
Analg. 2007;104:416–420.
8. Sharma S, Philip J, Wiley J. Thromboelastographic changes in healthy parturients and postpartum
women. Anesth Analg. 1997;85:94–98.
9. Armstrong S, Fernando R, Ashpole K, et al. Assessment of coagulation in the obstetric population
using ROTEM thromboelastometry. Int J Obstet Anesth. 2011;20:293–298.
10. Huissoud C, Carrabin N, Benchaib M, et al. Coagulation assessment by rotation thrombelastometry in
normal pregnancy. Thromb Haemost. 2009;101:755–761.
11. Oudghiri M, Keita H, Kouamou E, et al. Reference values for rotation thromboelastometry (ROTEM)
parameters following non-haemorrhagic deliveries. Correlations with standard haemostasis
parameters. Thromb Haemost. 2011;106:176–178.
12. Vincelot A, Nathan N, Collet D, et al. Platelet function during pregnancy: an evaluation using the
PFA-100 analyser. Br J Anaesth. 2001;87:890–893.
13. American College of Obstetricians and Gynecologists Women’s Health Care Physicians. ACOG
Practice Bulletin No. 138: inherited thrombophilias in pregnancy. Obstet Gynecol. 2013;122:706–717.
14. Kent N, Leduc L, Crane J, et al. Prevention and treatment of venous thromboembolism (VTE) in
obstetrics. J SOGC. 2000;22:736–749.
15. Kobayashi T. Antithrombin abnormalities and perinatal management. Curr Drug Targets. 2005;6:559–
566.
16. Franchini M, Veneri D, Salvagno GL, et al. Inherited thrombophilia. Crit Rev Clin Lab Sci.
2006;43:249–290.
17. Calderwood CJ, Greer IA. The role of factor V Leiden in maternal health and the outcome of
pregnancy. Curr Drug Targets. 2005;6:567–576.
18. Sugiura M. Pregnancy and delivery in protein C-deficiency. Curr Drug Targets. 2005;6:577–583.
19. Horlocker T, Wedel D, Rowlingson J, et al. Regional anesthesia in the patient receiving antithrombotic
or thrombolytic therapy: American Society of Regional Anesthesia and Pain Medicine Evidence-
Based Guidelines (Third Edition).Reg Anesth Pain Med. 2010;35:64–101.
20. Derksen R, de Groot P. The obstetric antiphospholipid syndrome. J Reprod Immunol. 2008;77:41–50.
21. American College of Obstetricians and Gynecologists. Practice Bulletin No. 132: antiphospholipid
syndrome. Obstet Gynecol. 2012;120:1514–1521.
22. Franchini M. Haemostasis and pregnancy. Thromb Haemost. 2006;95:401–413.
23. Ruppen W, Derry S, McQuay H, et al. Incidence of epidural hematoma, infection, and neurologic
injury in obstetric patients with epidural analgesia/anesthesia. Anesthesiology. 2006;105:394–399.
24. U.S. Food and Drug Administration. FDA Drug Safety Communication: updated recommendations to
decrease risk of spinal column bleeding and paralysis in patients on low molecular weight heparins.
http://www.fda.gov/Drugs/DrugSafety/ucm373595.htm. Accessed August 2014.
25. Kujovich JL. von Willebrand disease and pregnancy. J Thromb Haemost. 2005;3:246–253.
26. James AH. Von Willebrand disease. Obstet Gynecol Surv. 2006;61:136–145.
27. Bolton-Maggs P, Perry D, Chalmers EA, et al. The rare coagulation disorders—review with guidelines
from the United Kingdom Haemophilia Centre Doctors’ Organisation. Haemophilia. 2004;10:593–
628.
28. Lee CA, Chi C, Pavord SR, et al. The obstetric and gynaecological management of women with
inherited bleeding disorders—review with guidelines produced by a taskforce of UK Haemophilia
Centre Doctors’ Organisation. Haemophilia. 2006;12:301–336.
29. McCrae KR. Thrombocytopenia in pregnancy: differential diagnosis, pathogenesis, and management.
Blood Rev. 2003;17:7–14.
30. Boehlen F, Hohlfeld P, Extermann P, et al. Platelet count at term pregnancy: a reappraisal of the
threshold. Obstet Gynecol. 2000;95:29–33.
31. Felbinger TW, Posner M, Eltzschig HK, et al. Laparoscopic splenectomy in a pregnant patient with
immune thrombocytopenic purpura. Int J Obstet Anesth. 2007;16:281–283.
32. Kam PC, Thompson SA, Liew AC. Thrombocytopenia in the parturient. Anaesthesia. 2004;59:255–
264.
33. George JN. The association of pregnancy with thrombotic thrombocytopenic purpura-hemolytic
uremic syndrome. Curr Opin Hematol. 2003;10:339–344. 34. Douglas MJ. Platelets, the parturient
and regional anesthesia. Int J Obstet Anesth. 2001;10:113–120.
35. Montagnana M, Franchi M, Danese E, et al. Disseminated intravascular coagulation in obstetric and
gynecologic disorders. Semin Thromb Hemost. 2010;36:404–418.
36. Stainsby D, MacLennan S, Thomas D, et al. Guidelines on the management of massive blood loss. Br
J Haematol. 2006;135:634–641.
37. Fuller AJ, Bucklin BA. Blood product replacement for postpartum hemorrhage. Clin Obstet Gynecol.
2010;53:196–208.
38. Phillips LE, McLintock C, Pollock W, et al. Recombinant activated factor VII in obstetric hemorrhage:
experiences from the Australian and New Zealand Haemostasis Registry. Anesth Analg.
2009;109:1908–1915.
39. Franchini M, Franchi F, Bergamini V, et al. The use of recombinant activated FVII in postpartum
hemorrhage. Clin Obstet Gynecol. 2010;53:219–227.
40. Levi M. Pathogenesis and management of peripartum coagulopathic calamities (disseminated
intravascular coagulation and amniotic fluid embolism). Thromb Res. 2013;131(suppl 1):S32–S34.
Cardiac Disease in the Obstetric Patient
Nathaen S. Weitzel and Bryan S. Ahlgren
I. Overview
A. Physiology
B. General anesthetic considerations
C. Neonatal risk
D. Obstetric delivery considerations
II. Valvular heart disease: congenital and acquired
A. Mitral stenosis
B. Aortic stenosis
C. Mitral regurgitation
D. Mitral valve prolapse
E. Aortic insufficiency
III. Congenital heart disease in the adult parturient
A. General peripartum considerations
B. Cyanotic lesions
C. Left-to-right shunting
D. Aortic defects
IV. Primary pulmonary hypertension
A. Peripartum considerations
B. Anesthetic management
C. Invasive monitoring
V. Cardiomyopathy
A. Peripartum cardiomyopathy
B. Idiopathic dilated cardiomyopathy
C. Hypertrophic obstructive cardiomyopathy
D. Obstetric care after cardiac transplantation
VI. Ischemic heart disease in pregnancy
A. Peripartum considerations
B. Anesthetic management
VII. Cardiac dysrhythmias and pregnancy
A. Treatment limited to rhythms with hemodynamic
consequences
B. Therapy for dysrhythmias
VIII. Cardiopulmonary resuscitation in the obstetric patient
A. Fetal viability influences therapy
B. Treatment of cardiac arrest from suspected LAST
KEYPOINTS
1. Cardiac disease is present in up to 4% of parturients with 80% of these
cases being congenital in origin.
2. Regardless of origin of cardiac disease, presence of reduced left
ventricular ejection fraction, left-sided obstructive lesions, New York
Heart Association functional class (NYHC) >II, or history of prior
cardiac event are all risk factors for cardiac events in pregnancy.
3. Parturients with a history of congenital heart disease and have received
definitive surgical repair as a child and have good functional results after
surgery will often tolerate pregnancy well. These patients can often be
managed like a normal parturient. Patients presenting with partial
correction, palliative procedures, uncorrected lesions, and residual
cardiac defects need to be managed according to their current
cardiovascular physiology.
4. Peripartum cardiomyopathy is a rare disorder affecting between 1 in
3,000 and 1 in 4,000 live births and has a mortality of up to 20%. Once
identified, anesthetic management should reflect principles for any
patient presenting with advanced heart failure.
I. Overview. During the past century, the care of the parturient with
cardiac disease has changed dramatically. Cardiac disease is present
in up to 4% of total pregnancies with overall cardiac-related mortality
rates estimated to be between 0.5% and 2.7%.1–6 Congenital heart
disease (CHD) is the leading etiology of cardiac disease in pregnancy
in the Western world, owing to advances in early medical and surgical
management and improved survival in women of childbearing age.
CHD accounts for an estimated 80% of parturients with cardiac
disease, yet this number is much lower outside Europe and North
America (<20%).6 In certain subsets of cardiac patients, such as
severe pulmonary hypertension, mortality rates continue to approach
50%.7 Despite advances in care, cardiac disease remains the most
common nonobstetric cause of death among parturients, and it
presents one of the most challenging clinical scenarios for
anesthesiologists to manage.8
A. Physiology. Previous chapters discuss the physiologic changes of
pregnancy, but critical elements for patients with cardiac disease
include1:
1. Increased blood volume of 50%, elevated cardiac output by
40%, and increased heart rate (HR) by 25% to approximately
80 to 100 beats per minute (bpm) (measurements peak by the
end of the second trimester)
2. Reduced systemic vascular resistance (SVR) and pulmonary
vascular resistance (PVR)
3. Labor and delivery itself imposes nearly a 50% increase in
myocardial oxygen demand and cardiac output.
B. General anesthetic considerations. Numerous reports describe
risk factors for cardiac events in pregnancy. Siu et al.3 reported a
prospective trial of 562 patients (CAREPREG Trial) investigating
the relationship between cardiac events during pregnancy and
concomitant cardiac disease. These authors identified four main
predictors of cardiac events3:
1. Prior cardiac event (e.g., heart failure, transient ischemic
attack, stroke, or dysrhythmias)
2. Baseline New York Heart Association (NYHA) functional
class >II or the presence of cyanosis (see Table 25.1)
C. Neonatal risk
Considerations must be made for both the risk of neonatal disease
and for failure of the pregnancy. Figure 25.1 indicates the
distribution of completed pregnancies, elective abortions, and
miscarriages by specific maternal congenital cardiac defect.
Incidence of preterm delivery of neonates born to mothers with
CHD is elevated, with an incidence of 16% to 19%. Rates of
neonatal intensive care unit admission, intraventricular
hemorrhage, small for gestational age, and neonatal mortality are
also all elevated.10,11 Neonates born from mothers with CHD
have a tenfold elevated risk of suffering CHD with an incidence
of 3% to 10%.12 Genetic testing may be useful in select patients
with increased risk profile.
1. Pathophysiology
a. Typical symptoms or signs of severe MS include chest
pain, dyspnea, palpitations, pulmonary edema,
hemoptysis, and thromboembolism.
b. The stenotic valve prevents normal left ventricle filling
and leads to enlargement of the left atria, elevated
pulmonary venous and arterial pressures, and eventually
to right-sided heart failure often with associated tricuspid
valvular insufficiency.
c. Left ventricular stroke volume decreases along with the
cardiac output.
d. Severity is judged by mitral valve area obtained by
echocardiography.16 Normal mitral valve area is 4 to 6
cm2 with mild stenosis typically defined as a valve area of
1.5 to 3 cm2, moderate stenosis as 1.1 to 1.5 cm2, and
severe stenosis as <1.0 cm2. Severe lesions typically
require surgery, which can usually be done at a relatively
low risk to both mother and fetus during the first trimester
of pregnancy.
2. Peripartum considerations. The “hyperdynamic” cardiac
state of pregnancy is poorly tolerated by women with severe
MS.16
a. Increased plasma volume can cause pulmonary edema
and worsen left atrial enlargement.
b. Tachycardia decreases left ventricular diastolic filling
time through the stenotic valve.
c. Atrial fibrillation (AF) is common in MS, and this can be
highly detrimental because the atrial “kick” may account
for as much as 30% of the left ventricular stroke volume
in patients with MS.16 Medical management of AF
typically includes ventricular rate control by using β-
adrenergic blocking agents, unless there is a
contraindication; yet, a resting HR of approximately 90 to
100 bpm is considered normal during pregnancy.
d. Patients with severe disease have improved outcomes
with intervention prior to pregnancy. Typical interventions
include surgical mitral valve commissurotomy, valve
replacement, or percutaneous mitral
commissurotomy.16–19
3. Anesthetic management
Management should be based on the severity of the lesion,
which depends on the mitral valve area and the
hemodynamic stability of the patient.19 Most obstetric
anesthesiologists advocate early pain control using either IV
opioids or epidural analgesia for labor and vaginal
delivery.20–22 Second stage of labor increases cardiac preload
and cardiac output as a result of autotransfusion from the
uterus and relief of aortocaval compression. These alterations
in hemodynamics and cardiac preload can cause acute
pulmonary edema.
Combined spinal-epidural (CSE) technique, where
intrathecal opiates are given early in the first stage of labor
followed by gradual titration of dilute epidural local
anesthetic as labor progresses to the second stage (to cover
T10–S4), provides an excellent hemodynamic profile for most
patients.19
a. Epidural anesthesia is ideal for prolonged second stage
vaginal deliveries where instrumentation is required.
Often the epidural infusion during labor will provide
adequate anesthesia for forceps placement. If the epidural
block is inadequate, administration of epidural fentanyl
along with small doses of 2% lidocaine or 3% 2-
chloroprocaine can provide surgical blockade in a timely
and safe manner.19
b. Reductions in SVR from sympathetic blockade should be
treated preferentially with cautiously titrated
phenylephrine and judicious volume expansion, unless the
patient is relatively bradycardic (HR <70 bpm). In this
situation, ephedrine may be preferable.
4. For CD, epidural anesthesia is the preferred approach if time
allows.1,19
a. “Single-shot” spinal techniques should be avoided in both
moderate and severe MS because abrupt changes in SVR
are poorly tolerated.
b. Slow titration of 2% lidocaine combined with fentanyl
through the epidural catheter will produce surgical
blockade (T4–T6 level). Titration of small doses of
phenylephrine (50 to 100 μg) as needed will counteract
the expected hypotension from sympathectomy. Systolic
BP should ideally exceed 100 mm Hg, although the
potential exists to exacerbate pulmonary hypertension. If
echocardiography in the second or third trimester suggests
pulmonary hypertension or signs of right heart “overload”
(i.e., dilation of right ventricle and/or atrium, often with
tricuspid regurgitation), then placement of a pulmonary
artery catheter (PAC) for labor and delivery should be
considered.
c. The blood volume reduction that accompanies vaginal
delivery and CD are beneficial to the patient with MS, as
long as adequate cardiac preload is maintained.
5. General anesthesia. Consideration of the above
hemodynamic goals should be used in choosing induction and
maintenance anesthetics.1,19
a. Stress response suppression during induction and through
the predelivery period, using opiates such as remifentanil
(0.5 to 1 μg/kg) and a short-acting β-adrenergic blocker
such as esmolol (30 to 50 μg/kg) at induction, should be
considered. Etomidate 0.2 to 0.3 mg/kg is a good choice
for induction, typically accompanied by succinylcholine 1
to 1.5 mg/kg in a rapid-sequence technique accompanied
by cricoid pressure.
b. Drugs that may produce tachycardia should probably be
avoided (e.g., glycopyrrolate, atropine, meperidine,
pancuronium, ketamine).
c. Full stomach and aspiration prophylaxis are the same as
for CD under general anesthesia without mitral stenosis.
d. Methergine, prostaglandins, and oxytocin should be used
cautiously because these drugs may alter systemic or
pulmonary pressures leading to hemodynamic
compromise.22
6. Invasive monitoring including arterial and central venous or
pulmonary arterial pressure monitoring should be considered
for high-risk patients. The risks of placing a PAC, especially
dysrhythmias, are significant, but the information gained in
patients with severe MS may be beneficial to management
during labor.19,20,23 If general anesthesia is selected,
consideration should be given to the use of transesophageal
echocardiography to facilitate decision making about fluid
replacement and/or inotropes and vasopressors.
B. Aortic stenosis
Severe aortic stenosis (AS) is rare among parturients. Rheumatic
heart disease is the most common acquired cause of AS, whereas
a bicuspid aortic valve is the most common congenital cause (see
Fig. 25.3). Left ventricular hypertrophy compensates for AS by
offsetting the increase in left ventricular wall tension imposed by
increased left ventricular pressures. Unlike MS, symptoms of AS
appear late in the course of disease and lead to high mortality.14,24
Patients with severe AS should have surgical correction prior to
pregnancy.1,21
1. Pathophysiology
a. Symptoms of severe AS include syncope, dyspnea, angina
pectoris, and fatigue.1
b. Patients are usually asymptomatic until the valve area is
reduced to 1.0 cm2, which is roughly one third the normal
valve area of 3 to 4 cm2.16,25
c. Severe AS is often defined as a valve area less than 0.7
cm2, which typically is associated with a transvalvular
mean pressure gradient greater than 50 mm Hg derived
from echocardiographic measurements.16,25
2. Peripartum considerations
a. Pregnancy is usually tolerated by women with mild to
moderate AS.8,14
b. Patients with severe AS are unable to accommodate
increased blood volume and tachycardia, and often
experience worsening of symptoms including angina
pectoris and dyspnea. These women have little cardiac
reserve during pregnancy and have a high mortality rate
of 15% to 17%.1,14,24
3. Anesthetic management1,22,24
Vaginal delivery is preferred for mild and moderate AS,
whereas CD is often chosen for patients with severe AS.
Keeping the HR below 90 bpm improves ventricular
emptying against a fixed obstruction, although lower HRs
may be required to avoid subendocardial ischemia in patients
with severe AS and marked LVH. Tachycardia can induce left
ventricular subendocardial ischemia, reduce left ventricular
ejection fraction, and increase myocardial oxygen
consumption. These women also have a limited capacity to
increase left ventricular stroke volume, so the usual LV
preload augmentation that accompanies slower HRs is
impaired. Consequently, an HR below 70 bpm may place
these patients at risk for critically low cardiac output. Left
ventricular hypertrophy places the myocardium at risk for
ischemia under situations of high myocardial oxygen demand
(typically manifested in AS as excessively high left
ventricular systolic pressures or tachycardia) or when SVR is
reduced and coronary perfusion pressure (i.e., diastolic blood
pressure minus left ventricular diastolic pressure) is
compromised.
a. Single-shot spinal anesthesia is contraindicated for
women with severe AS and relatively contraindicated for
those with moderate AS.22
b. Epidural or continuous spinal approaches for labor have
been successfully carried out with slow titration of both
an opioid and a local anesthetic agent.26 Epinephrine
should be avoided because of the risk of causing
tachycardia with intravascular injection.
c. As in MS, early analgesia is beneficial. CSE with an
intrathecal opioid followed by epidural titration of a dilute
local anesthetic solution as labor progresses is ideal.
Management of prolonged second stage instrumented
deliveries is the same as for MS.
4. For general anesthesia, slow titration of opioids and use of
etomidate constitute an excellent approach. As with MS,
avoiding tachycardia is important, so glycopyrrolate, atropine,
pancuronium, and ketamine should be avoided. Propofol
should be used cautiously, if at all, due to reductions in SVR,
cardiac preload, and myocardial depression.14
5. CD considerations are similar to MS except that AS patients
are more afterload dependent and may tolerate the
sympathectomy induced by spinal or epidural anesthesia even
more poorly than MS patients. Both MS and AS patients also
tolerate preload reduction poorly, but AS patients are
probably more likely to experience it, and this may become
clinically significant even with slow onset of an epidural
anesthetic.
6. Invasive monitors
a. Consider arterial catheter placement to closely monitor
changes in blood pressure.
b. Central venous pressure or pulmonary artery catheter.
Use of PAC monitoring is controversial, and placement
decisions should be made on a case-by-case
basis.1,7,14,15,22 Tight volume control is recommended in
these patients, but introduction of the PAC creates some
risk of dysrhythmias, which can be life threatening.
Hypovolemia should probably induce greater concern
with moderate or severe AS than pulmonary edema
because preload reduction can severely reduce cardiac
output. Central venous pressure (CVP) monitoring may be
an adequate substitute for the PAC to help monitor the
volume status in the setting of moderate to severe
disease.1,8 Attempts should be made to keep CVP in the
high normal to slightly elevated range (8 to 12 mm Hg).
With severe AS, neither CVP nor pulmonary artery
occlusion pressures reliably reflect left ventricular
preload, and the use of transesophageal echocardiography
(TEE) should be considered for this purpose if general
anesthesia is utilized.
C. Mitral regurgitation
Consideration of mitral regurgitation (MR) requires division into
acute MR and chronic MR. The most common causes of acute
MR during pregnancy are bacterial endocarditis, trauma, papillary
muscle rupture (see Fig. 25.4), and prosthetic valve dysfunction.
Chronic MR is more common and usually results from
myxomatous degeneration or rheumatic disease.
Echocardiographic imaging is the standard diagnostic modality,
and a case of moderate to severe MR is shown in Figure 25.5. MS
and MR may coexist in pregnant patients. Women with severe
symptomatic MR should consider mitral valve repair prior to
conception.21
1. Pathophysiology
a. Acute MR results in an acute volume load into the left
atrium across the incompetent mitral valve. This results in
elevated pulmonary pressures and reduction in forward
flow. If the patient survives the acute insult, pulmonary
edema and right heart failure may occur and emergency
valve surgery is often required.22
b. With chronic MR, the lesion develops over time and the
left atrium and pulmonary venous system are able to
accommodate the increased blood volume by enlarging.
This predisposes the patient to atrial fibrillation. Elevated
pulmonary pressures are much less common in chronic
MR compared with acute MR, but certainly can occur
with severe chronic MR. Pulmonary congestion is
common.22
c. AF is better tolerated in MR as compared to either MS or
AS because left ventricular preload is better
maintained.8,15
2. Peripartum considerations. MR is well tolerated in
pregnancy, unless the patient has severe MR or is
symptomatic before pregnancy.3
a. Afterload reduction is the treatment for MR, so the
decreased SVR that accompanies pregnancy is helpful.
Moderate tachycardia is also helpful because it limits the
time for regurgitation, so the increase in HR seen with
pregnancy is similarly helpful.
b. Hypertension during pregnancy should be aggressively
treated because increased afterload will increase the
regurgitant fraction.24
c. The risk of thrombus formation is increased as a result of
the enlarged left atrium and the hypercoagulability of
pregnancy. Thrombus is found in up to 20% of patients.1
D. Mitral valve prolapse
Mitral valve prolapse (MVP) is a subset of mitral valve disease
that most often involves early myxomatous degeneration with
minimal MR in women of childbearing age, but advanced disease
with severe MR is possible. This lesion is quite common and is
found in 12% to 17% of women.24 MVP may predispose the
patient to dysrhythmias and may be associated with other
systemic diseases such as Marfan syndrome. In the absence of
concomitant disease, there are no specific alterations in the
anesthetic management of these patients because there are rarely
any cardiac complications that arise from prolapse alone.24 In
contrast to patients with most forms of advanced MR, patients
with early MVP typically have less MR in the presence of high
preload, high afterload, and low HR.
E. Aortic insufficiency
Aortic insufficiency (AI) occurs more commonly than AS, and
rheumatic disease is the causative agent in the majority of
patients. Patients with rheumatic AI often have concomitant
mitral valve disease. AI can also be present in patients with
congenital bicuspid valve disease or with collagen vascular
disorders.24 Acute AI can result from endocarditis, or rarely from
trauma. These patients are critically ill and typically require
urgent surgical correction. There have been reported cases of
medical management of acute AI until CD could be performed,
but it is rare for those patients to survive.27
1. Pathophysiology
a. AI increases left ventricular diastolic volume. Over time,
this causes significant ventricular dilation and eccentric
hypertrophy. This typically precedes the onset of
symptoms, which include dyspnea, diminished exercise
ability, pounding in the chest, and orthopnea.1
b. Progression of disease causes left ventricular failure,
reduced ejection fraction, and further increases in end-
diastolic ventricular volume.
c. In the absence of pregnancy, symptoms typically occur 10
to 20 years after the onset of rheumatic disease.24
2. Peripartum considerations. Mild to moderate AI is well
tolerated during pregnancy.
a. The tachycardia associated with pregnancy reduces the
regurgitant fraction and left ventricular diastolic volume.
The reduction of SVR associated with pregnancy also
promotes forward flow out of the left ventricle.
b. Patients with severe AI who have symptoms of heart
failure prior to pregnancy have diminished cardiac
reserve. Onset of pregnancy-related hypertension or signs
of congenital heart failure (CHF) should prompt treatment
with afterload reduction, diuretics, and/or inotropic
agents.21,24
5. Eisenmenger syndrome
Eisenmenger syndrome (ES) represents the most severe form
of pulmonary artery hypertension seen in adults with CHD.
Chronic high-volume systemic to pulmonary shunting of any
congenital defect results in right ventricular hypertrophy,
elevated PVR, and significant ventricular and arterial
remodeling on the right side of the heart. Eventually,
remodeling and elevation of PVR leads to reversal of shunt
flow and hypoxia. 50 ES carries a maternal mortality ranging
from 30% to 50% along with a high incidence of fetal demise,
so patients are usually counseled against pregnancy (see Fig.
25.1).6,33 Sudden death is common and may be due to stroke,
dysrhythmia, brain abscesses, or heart failure. Twenty-five-
year survival after diagnosis of ES is reported to be 42% in
the absence of pregnancy.39
a. Pathophysiology. ES is defined by a PVR greater than
800 dyn·s/cm5 along with right-to-left or bidirectional
shunt flow.33 Early correction of the shunt may resolve
the progression to pulmonary hypertension, but once
pulmonary arteriolar remodeling (i.e., medial
hypertrophy) develops, the elevated PVR is irreversible,
differentiating ES from primary pulmonary hypertension
(PPH).1
(1) Symptoms: fatigue, dyspnea, cyanosis, edema,
clubbing, and polycythemia
(2) Elevated stroke risk: The underlying right-to-left
shunt, hyperviscosity from polycythemia, and the
development of heart failure promote thrombus
formation and embolization.
b. Peripartum considerations. Patients with ES are often
unable to accommodate the increased oxygen demands of
pregnancy.
(1) Reduced SVR from pregnancy coupled with the
irreversible elevated PVR of Eisenmenger increases
right-to-left shunting and cyanosis.
(2) Reduction in functional residual capacity adds to the
hypoxemia, further reducing oxygen delivery to the
fetus and increasing the risk of fetal demise.1
(3) Management of patients who desire to remain
pregnant should be carried out utilizing a
multidisciplinary approach among obstetricians,
cardiologists, and anesthesiologists trained in high-
risk pregnancies.
c. Anesthetic management for labor. Historically,
neuraxial anesthesia was contraindicated, and general
anesthesia was the standard. A review of cases of
noncardiac surgery including labor and CD in
Eisenmenger syndrome indicates that neuraxial anesthesia
is safe for these patients.51 In addition, it has been shown
that general anesthesia confers a near fourfold increased
risk of maternal mortality in patients with severe PH.52
Regardless of the type of anesthetic, this population
requires utmost vigilance to maintain the aforementioned
hemodynamic goals. Vaginal deliveries will often require
an assisted second stage to reduce cardiac stress. 50,53
d. Labor. Early and effective analgesia is critical to maintain
the balance of SVR and PVR by avoiding catecholamine
surges from pain. Intrathecal opioid administration (CSE)
in first stage of labor is ideal; however, if anticoagulation
is a concern, then a low-dose remifentanil infusion or
patient-controlled analgesia are good options.6
(1) Slow titration of local anesthetic with aggressive
treatment for any reduction in SVR (i.e., systemic
hypotension) using phenylephrine will provide good
results for second stage of labor as well as
instrumental delivery.51
(2) Maintenance of intravascular volume status using
carefully titrated fluid boluses along with use of
phenylephrine for decreased SVR should be used to
prevent onset or exacerbation of cyanosis.
(3) Pudendal blockade may also be employed to avoid
the risk of extending the epidural blockade (and
decreasing SVR) to “cover” stage 2 of labor and for
delivery.
(4) Oxytocin, methergine, and prostaglandins should be
used with extreme caution because of systemic and
pulmonary vascular side effects.
e. Anesthesia for cesarean delivery
(1) Slowly titrated epidural anesthetics maybe preferable
to general anesthesia.
(2) Slow titrated doses of local anesthetic to obtain a
surgical block can be safely used.
(3) Single-shot spinal anesthesia is contraindicated.
(4) Tight hemodynamic monitoring and control is of
utmost importance.51 Avoiding elevations in PVR is
critical (see Table 25.3).
f. For general anesthesia, slow titration of induction agents
is preferred because rapid sequence inductions carry a
high risk of SVR alterations and subsequent
hemodynamic collapse. This places the patient at
increased risk for aspiration, so strict NPO guidelines, use
of pharmacologic prophylaxis against aspiration (e.g.,
sodium citrate, H2-blockade), and mask ventilation using
cricoid pressure are recommended. Propofol should be
avoided or used with extreme caution due to marked
reductions in SVR, whereas ketamine and etomidate seem
appropriate. Inhalational agents should be used with
caution because of their propensity to decrease SVR.
Nitrous oxide should be avoided because of its likelihood
to increase PVR. Maintenance of anesthesia may be
accomplished using careful titration of intravenous agents
such as nondepolarizing neuromuscular blockers, opioids,
and sedative-hypnotic agents (e.g., midazolam or
ketamine) as well as “topping off” with potent
inhalational agents used at concentrations of less than 0.5
MAC.
g. Monitoring
(1) Pulse oximetry may be the most important monitor
because changes in saturation directly correlate with
alterations in shunt flow.1
(2) Intra-arterial monitoring is generally employed and
is used to closely monitor blood pressure.
(3) Central venous catheters are controversial. CVP
catheter placement carries a risk of air embolus,
thrombus, and pneumothorax, which can be
devastating in these patients; however, information
regarding cardiac filling pressures can be useful.
(4) PACs are contraindicated in obstetric patients with
ES for a number of reasons. The anatomic
abnormality causing ES typically renders flow-
directed flotation of PA catheters difficult or
impossible. The risk of dysrhythmia, pulmonary
artery rupture, and thromboembolism are also
elevated. Cardiac output measurements will be
inaccurate due to the large shunt.
(5) If general anesthesia is administered, TEE may
provide the best real-time monitor of cardiac preload
and of the status of right-to-left shunting.
h. Advanced pulmonary artery hypertension therapies
(1) Inhaled nitric oxide. Inhaled nitric oxid (iNO) is a
direct-acting pulmonary vasodilator that avoids
systemic vasodilation, thus reducing shunt flow and
hypoxia. Evidence for use of iNO for labor in
Eisenmenger patients is limited, but several case
reports indicate improvement in oxygenation and
reduced pulmonary pressures.46
(2) Prostacyclin analogues. Prostacyclin analogues act
like iNO and are direct pulmonary vasodilators.
However, their use in pregnancy is not well defined.
As with iNO, it is probably best to institute treatment
prior to hemodynamic collapse and right ventricular
failure.52
(3) Endothelin receptor antagonists. Endothelin
receptor antagonists (ERAs), such as bosentan, work
by competitively binding at the endothelin-A and B
receptors in pulmonary vasculature, limiting vascular
constriction. Animal studies have shown that
bosentan may have teratogenic effects; therefore, its
use in pregnancy is limited.54
(4) Phosphodiesterase inhibitors. Phosphodiesterase
inhibitors (PDE5) such as sildenafil act by inhibiting
the degradation of cGMP, causing pulmonary
vasodilation. Sildenafil’s use in pregnancy and ES is
not well defined. Sildenafil has not been shown to be
teratogenic in animal studies, although its safety
profile is not well elucidated in humans. Patients
presenting on sildenafil therapy probably should be
maintained on the drug during pregnancy.52
C. Left-to-right shunting
1. General considerations. There are some general anesthetic
considerations that apply to lesions causing left-to-right
shunts, particularly in patients with residual defects.32,33,39
a. Left-to-right shunts include defects such as ASD, VSD,
or PDA. Large defects require surgical repair, which, if
done early, result in normal cardiac physiology. These
patients can be managed as typical obstetric patients.
Patients with repaired or small defects tend to tolerate the
physiologic changes of pregnancy well.4
b. Effects of chronic shunting. The balance between SVR
and PVR determines the shunt fraction and the direction
of shunting. Causes of chronic left-to-right shunting
include:
(1) Excessive pulmonary blood flow, which can lead to
pulmonary edema or pulmonary hypertension. As a
result of elevated pulmonary blood flow, PVR
increases over time, which reduces left-to-right
shunting. This eventually causes equilibration of left
and right ventricular pressures and conversion of the
left-to-right shunt into a right-to-left shunt to produce
Eisenmenger physiology.
(2) Once Eisenmenger complex develops, cyanosis
ensues along with variable degrees of heart failure,
which places patients in the highest risk category for
pregnancy (see Table 25.2).
(3) Even without Eisenmenger complex, these patients
may experience heart failure as a result of the high
RV and pulmonary blood flow, which may be as
much as four times systemic blood flow.
c. SVR. Acute changes in SVR from anesthetic
administration or pain can result in alteration or reversal
of the shunt, leading to heart failure or cyanosis. These
alterations depend on where the patient is in the evolution
from large left-to-right shunt into the right-to-left shunting
of Eisenmenger physiology. Overall, anesthetic goals
should be to maintain physiologic balance and avoid
abrupt hemodynamic alterations.
d. Supplemental oxygen should be provided to avoid
hypoxemia and elevation of PVR.
e. Air embolus. Extreme care to avoid an air embolus is
required. Epidural catheter placement should use saline
for loss of resistance, not air, because air entrainment into
an epidural vein can cross to the left sided circulation. IV
lines should be aggressively de-aired and monitored.
f. Single-shot spinal anesthesia is contraindicated for
patients who have or are approaching Eisenmenger
physiology. Spinal anesthesia is theoretically beneficial
for patients with large left-to-right shunts and normal, or
only slightly elevated PVR, that remains far below SVR.
g. Inhalational agents. Uptake should not be affected by
left-to-right shunting. Right-to-left shunting prolongs
inhalation inductions, but this is rarely clinically relevant.
h. Invasive monitors. Central venous or arterial catheters
should be considered in patients with symptomatic heart
failure. PA catheters are considered to be contraindicated
by some because of the difficulty encountered in placing
them and the possibility of increased risk of pulmonary
artery rupture, but this area of practice lacks evidence-
based information.
i. Anticoagulation during pregnancy is common due to
elevated thromboembolic risk, so coagulation status
should be tested prior to neuraxial blockade.
2. Atrial septal defect
ASDs account for nearly one-third of congenital heart defects
and are found in women more commonly than men.39 Small
defects (less than 5 mm) are hemodynamically insignificant,
but large defects (greater than 20 mm) can have severe
hemodynamic consequences.33,34 ASDs do not typically close
spontaneously and are commonly associated with additional
cardiac defects. Modern interventional techniques permit
ASD closure through percutaneous methods with a high rate
of success in long-term outcome studies.55–58
a. Pathophysiology. The overall effect is a left-to-right
shunt along the pressure gradient. This leads to right atrial
and ventricular dilation, increased pulmonary blood flow,
and potentially to pulmonary edema (high-output failure)
and pulmonary hypertension (usually <500 dyn·s/cm5).
The likelihood for development of Eisenmenger complex
over time is much lower for a VSD with similar initial
pulmonary blood flows, but this can also occur.
(1) Development of right heart failure can occur without
repair.
(2) Patients are at risk for dysrhythmias and paradoxical
embolus leading to stroke.
(3) Endocarditis risk is increased, so antibiotic
prophylaxis is required.
3. Ventricular septal defect
This is the most common congenital heart lesion in children,
although nearly 90% close spontaneously by age 10 years.
Those patients with large lesions and who are symptomatic at
birth will usually be surgically corrected, whereas
asymptomatic patients will often be closely monitored for
evidence of spontaneous closure.34,39 Surgical closure
typically involves a right ventricular incision, and this carries
a significant risk of interventricular and atrioventricular
conduction abnormalities. Interventional closure techniques
show definite promise, but lack long-term outcome studies to
support their use.38,39,59
4. Patent ductus arteriosus
PDA accounts for roughly 10% of congenital heart defects
and occurs in females in a ratio of 2:1.34,39 In utero, the
ductus arteriosus connects the aorta and pulmonary artery
allowing fetal circulation to bypass the lungs. Failure of the
ductus arteriosus to close shortly after birth results in a left-to-
right shunt that behaves much like a VSD. If present after
infancy, PDA closure rarely occurs spontaneously and closure
can be accomplished surgically without cardiopulmonary
bypass (CPB). Newer interventional approaches permit PDA
closure in the interventional radiology suite, thus avoiding
surgery and demonstrate >95% clinical success.60
a. Pathophysiology (ventricular septal defect and patent
ductus arteriosus). The predominant effect of left-to-
right shunting depends on the size and the resistance
between the systemic and pulmonary systems. In contrast
to ASD, both VSD and PDA will result in left ventricular
volume overload and dilation.
(1) PVR eventually increases in response to chronic
exposure to aortic pressures, and eventually the flow
equalizes and then converts to a right-to-left shunt to
produce Eisenmenger physiology.
(2) Shunt flow depends on the balance between SVR and
PVR, so acute changes in either of these can alter the
shunt, induce cyanosis, and pulmonary circulatory
and left ventricular volume overload.
b. Anesthetic management. Refer to the general guidelines
at the start of this section. Epidural anesthesia is the
preferred approach for patients with significant shunt
because it reduces alterations in SVR when titrated
slowly.
(1) Either vaginal or CD are acceptable; however, in
patients with severe shunting, vaginal delivery may
provide more stable hemodynamics as a result of
reduced blood loss.
(2) Patients with reduced cardiac function may benefit
from assisted second stage of labor (i.e., assistance
with forceps or vacuum extraction) to reduce the
duration of labor and stress on the cardiovascular
system.
c. General anesthesia can safely be administered in these
patients with close attention to the hemodynamic goals
outlined earlier. Titration of induction agents and careful
ventilatory management minimizes changes in SVR and
PVR (see Table 25.3).
D. Aortic defects
1. Coarctation of the aorta
Coarctation of the aorta is a congenital lesion affecting males
more often than females. It is caused by a constricting band
impinging on the aortic lumen and is most often located just
distal to the left subclavian artery in the area of the
ligamentum arteriosum. Extensive collateral circulation
develops through axillary, subclavian, intercostal, and
scapular arteries. Associated defects include VSD, ASD,
PDA, bicuspid aortic valve, circle of Willis aneurysms, and
mitral valve abnormalities.39,61
a. Pathophysiology. Systolic and mean arterial blood
pressure differences are noted between the upper and
lower extremities, although diastolic pressures are usually
similar. Physiologic consequences include hypertension
proximal to the coarctation, left ventricular hypertrophy
and failure, coronary arterial occlusive disease, stroke,
aortic dissection, and an elevated risk of endocarditis.39
Severity is determined by the gradient across the
coarctation. Surgical repair is typically indicated for mean
arterial pressure gradients greater than 30 mm Hg.
b. Peripartum considerations. Pregnancy is usually well
tolerated in patients with both corrected and uncorrected
lesions, as reported by Beauchesne et al.61 in a series of
118 pregnancies in 50 women. There was one death in
this group from acute aortic dissection. A higher
incidence of CD was also noted.
(1) Significant hypertension above the coarctation can
adversely affect the fetus.61,62 Blood pressure control
is used to reduce the risk of aortic dissection, with β-
adrenergic blockers serving as first-line
antihypertensive drugs. Treatment of hypertension
theoretically serves as a two-edged sword because
adequate uterine perfusion pressure may depend on
high driving pressures above the coarctation,
especially if arterial collaterals are poorly developed.
(2) Alterations in cardiac output and blood volume can
precipitate heart failure.
c. Anesthetic management. Early analgesic administration
reduces hemodynamic stress and theoretically helps
prevent aortic dissection. Vaginal delivery is preferred,
often with a forceps-assisted second stage to reduce risk
of prolonged labor.
(1) For corrected or uncorrected but mild coarctation,
epidural anesthesia is an acceptable approach using
careful titration of opioid and local anesthetic to
avoid large reductions in SVR, which could
compromise uterine perfusion pressure.
(2) Single-shot spinal anesthesia is contraindicated.
(3) Uncorrected coarctation represents one subset of
patients where epidural anesthesia is relatively
contraindicated. In the setting of a high arterial
pressure gradient, placental perfusion pressure can be
reduced significantly following epidural anesthesia.
General anesthesia is preferred in severe coarctation,
with emphasis on hemodynamic stability and
avoidance of tachycardia.
(4) In addition to prevention of substantial reductions in
blood pressure, one should also avoid large increases
in blood pressure because the risk of aortic dissection
and cerebral aneurysm rupture appears to be elevated
in this group of patients.
(5) As in all pregnant women, aortocaval compression
should be avoided and fetal monitoring used because
reduced placental perfusion occur with any chosen
anesthetic.
2. Marfan syndrome
Marfan syndrome (MFS) is a hereditary collagen vascular
disorder that presents diversely in the skeletal, ocular, and
cardiovascular systems, among others.6,21 The increased
stress on the cardiovascular system during pregnancy can
cause aortic dilation or dissection in MFS, which has been
associated with increased mortality during pregnancy. Recent
outcomes studies confirm that MFS continues to carry
increased risk for both the developing fetus and the
mother.63–65
a. Pathophysiology. Development of aortic dissection is the
primary reason for high mortality during pregnancy.
Aortic dissection begins as a tear in the intima, allowing a
path for blood to flow between the intima and media or
adventitia. This creates a false lumen that can spread
distally or proximally along the aorta or into its branch
arteries. Dissections fall into three categories:
(1) Type I dissections involve the ascending aorta, aortic
arch, and descending aorta, whereas Type II
dissections are confined to the ascending aorta. Type
III dissections begin in the descending aorta and
spread distally from there.
(2) Aortic rupture can occur in MFS and is nearly always
fatal.
(3) When the root of the ascending aorta is dilated from
MFS, as can occur even without dissection because
of laxity in the aortic wall, aortic insufficiency (AI)
can occur. This AI can range from asymptomatic and
mild to severe enough to induce left ventricular (LV)
failure even without pregnancy. When Type I or Type
II dissections occur, acute AI often results.
b. Peripartum considerations. Pregnancy presents a
challenge for MFS patients who have significant aortic
involvement and frequently causes increased aortic
dilation or dissection.
(1) If the aortic root diameter (at or distal to the
sinotubular junction) is less than 40 mm, pregnancy
is generally well tolerated.6 A prospective cohort
study from 1995 reported a 1% maternal mortality in
this specific group.65,66 The authors recommend
serial echocardiographic monitoring of aortic size as
well as prophylactic β-adrenergic blockade, which is
a mainstay in MFS both with and without dissection.
(2) The maternal mortality risk is 30% to 40% with a
history of dissection, aortic root size >40 mm, or
aortic valve involvement causing AI.1,3 These
patients are often counseled against pregnancy, and if
pregnancy is attempted, CD is recommended.6
(3) In acute aortic dissection, case reports describe aortic
root repair utilizing normothermic CPB with selective
cerebral perfusion resulting in survival of mother and
fetus.67,68
(4) Data for the management of aortic dissections in
pregnancy is sparse, so no consensus
recommendations exist. Type III dissections should
be managed medically (tight BP and HR control), and
type I or II dissections are managed surgically if the
situation allows.6,64,68
(5) Advances in interventional endovascular
management of aortic pathology are occurring
rapidly. Thoracic Endovascular Aortic Repair
(TEVAR) now has a defined role in the treatment of
thoracic aortic pathology and should be considered
for its management.69 Recently, reports of use of
TEVAR for aortic dissection during pregnancy have
indicated success, however large-scale studies are
lacking.70,71
c. Anesthetic considerations. The main goals for anesthetic
management of labor and delivery are reduction in
cardiovascular stress and afterload reduction.
Currently, there is no consensus on an optimal anesthetic
approach because good outcomes have been reported with
either general or epidural anesthesia.65,72
(1) Epidural placement early in labor can provide a
smooth hemodynamic course and accomplish the
mentioned goals. Prolonged labor and maternal
expulsive efforts during stage II may predispose to
aortic dilation or dissection, so either forceps-assisted
delivery using an epidural catheter to produce solid
perineal anesthesia or CD may be needed.
Communication with the obstetrician is important to
help develop a complete management plan. Spinal
anesthesia is an acceptable and perhaps the most
desirable choice for CD.
d. Acute aortic dissection is a high-risk situation. If the
fetus is mature enough for delivery in the presence of a
Type I or Type II dissection, then a combined CD of the
fetus and aortic repair can be employed using general
anesthesia with full hemodynamic monitoring including
TEE. If the dissection is small and stable and there is a
question of fetal maturity, then medical management with
afterload reduction and β-adrenergic blockade may be the
best option until CD can be performed which conveys a
reasonable expectation of maternal and fetal survival. The
anesthetic technique should emphasize reduction in
cardiovascular stress response and afterload
reduction.65,72
IV. Primary pulmonary hypertension
Although not considered a congenital defect, peripartum management
for PPH is similar to ES. PPH is defined as a syndrome with elevated
pulmonary artery pressures (mean PAP >25 mm Hg at rest) in the
absence of primary cardiac pathology.6,46 PPH is associated with
reduced nitric oxide and prostacyclin synthesis, as well as with
increased thromboxane production. Histologic features include medial
thickening and intimal fibrosis. PPH typically involves pulmonary
vasculature that remains responsive to vasodilators, whereas ES is
characterized by an unresponsive pulmonary vasculature.22
A. Peripartum considerations. Mortality and morbidity is high in
PPH and many authors consider pregnancy to be contraindicated.
Risk for maternal mortality is nearly 30% for PPH.
1. Pregnancy can induce right heart dilation or failure and many
cases of maternal death occur in the initial week postpartum.
2. Thromboembolic events are also common and most patients
require anticoagulation.
B. Anesthetic management.22 Refer to the hemodynamic goals for
ES, with one caveat: Reduction in SVR will not alter the shunt
fraction for PPH patients because there is no defect, but will
cause a marked decrease in cardiac output due to relatively
fixed right-sided pressures. Experience in the management of
these patients is increasing, but reported cases are still quite low.
Consensus recommendations are limited.
1. Vaginal delivery is preferred using epidural anesthesia (or
CSE) with an assisted second stage labor (i.e., forceps
delivery) to minimize stress on the right ventricle.
2. Oxytocin has been used successfully both for induction of
labor and to increase uterine tone postpartum. Careful titration
is required because it can decrease SVR and elevate PVR to
decrease both coronary perfusion pressure and forward flow.
It is important to avoid carboprost and methergine because of
bronchoconstrictive and vasoconstrictive side effects,
respectively.
3. CD has been associated with increased mortality; however,
patients with severe disease causing right heart failure may
require operative delivery.73 The anesthetic should be tailored
to each patient’s underlying cardiac function because epidural
and general anesthesia have similar outcomes.
4. iNO has been used successfully during labor (see discussion
under “Eisenmenger syndrome” section).74 Other therapeutic
agents that are useful in the management of ES are potentially
useful in patients with PPH.
C. Invasive monitoring. Arterial and PACs are generally employed.
The PAC can help guide the vasodilator therapy as well as
monitor the volume status of the patient. Although the risks of
placement discussed in previous sections apply to this subset of
patients, the information gained may outweigh the risk of
placement. TEE should be considered in the setting of general
anesthesia (see Fig. 25.10).
V. Cardiomyopathy
Cardiomyopathy constitutes a rare but devastating cardiac
complication for pregnant women. Etiologies include peripartum
cardiomyopathy, idiopathic dilated cardiomyopathy, and hypertrophic
obstructive cardiomyopathy (HOCM).
A. Peripartum cardiomyopathy. Peripartum cardiomyopathy
(PPCM) occurs with a general incidence of 1:3,000 to 1:4,000
live births.6,75 The current accepted definition is: an idiopathic
cardiomyopathy presenting with heart failure secondary to LV
systolic dysfunction towards the end of pregnancy or in the
months following delivery. It is a diagnosis of exclusion when no
other cause of heart failure is found. The LV may not be dilated,
but the EF is nearly always reduced below 45%.6,76 Recent trends
in the United States suggest an actual incidence of 8.5 to 11 per
10,000 live births, with a major adverse event rate of 13.5%.77
Echocardiography confirms the diagnosis by the presence of
reduced LV ejection fraction (<45%), decreased fractional
shortening (<30%), and/or elevated end diastolic LV diameter
(see Figs. 25.11 and 25.12).
1. Pathophysiology. Symptoms of fatigue, shortness of breath,
and peripheral edema are typical in pregnancy; however,
progressive deterioration and increased severity along with
chest pain, palpitations, and pulmonary edema can indicate
developing heart failure.
a. The etiology of PPCM is unknown, but myocarditis,
maladaptive response to the hemodynamic challenges of
pregnancy, abnormal immune response to pregnancy, and
prolonged tocolysis have been implicated.72,75
b. PPCM is associated with multiple gestations,
preeclampsia, obesity, advanced maternal age, and
coronary vascular disease.
c. Standard therapeutic measures for heart failure should be
initiated including salt and water restrictions, diuretics,
afterload reduction, and inotropic support as needed.75,78
2. Peripartum considerations. Once the diagnosis is made,
collaboration among the cardiologist, obstetrician, and
anesthesiologist is critical for timely decisions about care.
Maternal mortality is nearly 20% with some patients
surviving only if a heart transplant is available.75 There may
be a role for immunosuppressive therapy if myocarditis is
confirmed by biopsy and the patient’s condition is
deteriorating.6,75,78
a. In the setting of heart failure with a fetus that is near term,
delivery should occur because this reduces stress on the
heart.
b. Anticoagulation may be needed to decrease
thromboembolic risk.
c. Standard therapy for heart failure includes angiotensin-
converting enzyme (ACE) inhibitors; however, this needs
to be delayed until after birth because of
teratogenicity.75,78 β-adrenergic blockade, hydralazine,
and nitrates may help allay symptoms during pregnancy,
and there is the potential need for inotropic or “inodilator”
support using drugs such as dobutamine or milrinone.
d. Prognosis depends on the degree of recovery of left
ventricular function by 6 to 12 months postpartum.
Presence of heart failure symptoms at this time indicates
irreversible damage.75 Approximately 50% of women will
recover normal or near-normal cardiac function, but 5-
year mortality with persistent dysfunction can reach
85%.75
e. Repeated pregnancy carries a mortality risk of 1% to 15%
in various populations, with up to 50% of patients
demonstrating recurrance of PPCM.6
3. Anesthetic management should reflect principles for any
patient with heart failure. These patients will have extreme
sensitivity to myocardial depressants including those typically
used for general anesthesia. In this context, even volatile
anesthetics that typically induce vasodilation as their
predominant hemodynamic effect may cause myocardial
depression and decompensation; careful titration and low
concentrations (<0.5 MAC) are recommended if those agents
are chosen.
a. Epidural anesthesia using slow titration of a dilute local
anesthetic solution and an opioid will gently reduce SVR,
so this is a good choice. This should be employed early in
first stage of labor, and often a forceps-assisted delivery
will likely reduce hemodynamic stress on the heart by
expediting delivery.6
b. Coagulation studies should be checked (especially before
epidural placement) because anticoagulation therapy is
common.
4. CD can be managed with epidural anesthesia provided there
is time to slowly titrate the local anesthetic. If general
anesthesia is required, myocardial depressants, such as
inhalational agents, and propofol should be avoided or titrated
cautiously in low doses. Generous use of opioids will reduce
the stress response and remifentanil may be the best choice
because of its short duration of action and low likelihood of
inducing prolonged fetal respiratory depression. Cautious use
of β-adrenergic blockade (e.g., esmolol) can prevent dramatic
increases in HR. Careful attention to intravascular volume and
blood loss is required because both hypovolemia and
hypervolemia are poorly tolerated.
5. Invasive monitors. Arterial catheters should be strongly
considered for patients with PPCM. The decision to place
central venous and PACs should be made on a case-by-case
basis, but they are generally reserved for NYHA class III and
IV patients. In the setting of general anesthesia, TEE provides
rapid, real-time evaluation of cardiac function.
B. Idiopathic dilated cardiomyopathy. Dilated cardiomyopathy
(DCM) is extremely rare in pregnancy with an estimated
incidence of 5:100,000 births per year and has a similar
presentation, risk, and treatment as PPCM. Symptoms are the
same as for PPCM, but the onset of idiopathic dilated
cardiomyopathy (IDC) is not limited to the later stages of
pregnancy. Possible etiologies include idiopathic,
familial/genetic, viral, immune, alcoholic, or toxic causes.79
Treatment goals are the same as for PPCM.
C. Hypertrophic obstructive cardiomyopathy. HOCM is a genetic
disorder causing hypertrophy of the left ventricle and varying
degrees of left ventricular outflow tract obstruction (LVOTO).
The hypertrophy causes a noncompliant or “stiff” ventricle with
varying degrees of diastolic dysfunction, reduction in left
ventricular volumes, and outflow obstruction (see Fig. 25.13).
I. Anatomic disease
A. Scoliosis
B. Spinal surgery
C. Spinal cord injury
D. Spina bifida
E. Intracranial neoplasms
F. Idiopathic intracranial hypertension (pseudotumor cerebri)
G. Maternal central nervous system shunts
II. Vascular disease
A. Intracranial hemorrhage
B. Cortical vein thrombosis
III. Immunologic disease
A. Multiple sclerosis
B. Myasthenia gravis
C. Landry-Guillain-Barré syndrome
IV. Epilepsy
A. Obstetric management
B. Anesthetic management
V. Conclusion
KEYPOINTS
1. In patients with a neurologic condition, thorough preanesthetic
neurologic testing and documentation of preexisting deficits are essential.
2. In the patient with elevated intracranial pressure (ICP), neuraxial
procedures must be considered carefully. Although dural puncture could
cause brainstem herniation, infusion of epidural fluid could also acutely
increase ICP.
3. Although there is no evidence of neuraxial analgesia/anesthesia altering
the course of any neurologic disease, including chronic back pain, a
discussion of the natural history of the patient’s condition should be
included in the preanesthetic evaluation.
I. Anatomic disease
A. Scoliosis
Anesthesiologists seldom encounter parturients with severe
curvature of the spine, due in large part to screening programs
and early intervention. It is more common for patients to have a
vague history and mild curvature that complicates epidural
placement. In addition, patients may have had surgical correction
of the scoliosis with instrumentation, further complicating, but
rarely contraindicating, neuraxial analgesia/anesthesia (see
Section B. Spinal Surgery in subsequent text).
1. Clinical issues
a. Scoliotic curves involve both lateral deviation of the
vertebral column as well as axial rotation of the vertebral
bodies (see Fig. 26.1).
(1) The Cobb angle measures the degree of spinal
curvature (see Fig. 26.2). Minor curves (10 to 20
degrees) occur in approximately 2% of adolescents in
the United States. These are benign and require no
therapy. Curves <40 degrees rarely require fusion, but
thoracic curves >65 degrees will require fusion and,
if untreated, can result in a restrictive respiratory
pattern limiting ventilatory reserve.1
(2) Nonstructural curves are the result of leg length
discrepancies or posture. The spine remains mobile
and the curve is not progressive. Structural curves
are more commonly idiopathic and can result from a
variety of conditions (see Table 26.1).
(2) Prevention/treatment
(a) Prevention is paramount. Early administration
of combined spinal-epidural (CSE), or epidural
analgesia with local anesthetics and opioids can
prevent autonomic hyperreflexia caused by
uterine contractions. Neuraxial analgesia
prevents the reflex by interrupting noxious
sensory input from the viscera. A Foley catheter
should be placed to prevent bladder distension.
(b) Treatment of a hypertensive emergency
includes removal of the offending stimulus, if
possible (e.g., consider bladder catheterization),
and administration of IV nitroprusside,
sublingual nitroglycerin, and/or phentolamine.
2. Obstetric management13,14
a. Traumatic SCI occurring during pregnancy may result in
spontaneous abortion or preterm delivery. For the first
several months after injury, the patient is at increased risk
for deep vein thrombosis (DVT) and pulmonary
embolism. Prophylaxis against these complications may
impact plans for neuraxial anesthesia. Guidelines for
administration of neuraxial anesthesia in patients
receiving anticoagulants can be found at
http://www.asra.com.
b. Patients with SCI who have lesions above T10 will have
painless labor and are at risk for preterm labor and
delivery. Before term, they are monitored closely to
prevent home delivery. Because of inadequate expulsive
efforts in the second stage, assisted delivery may be
necessary.
c. Antepartum consultation in a high-risk obstetric
anesthesia clinic, with delivery at a tertiary care center
that includes appropriate anesthesia coverage, is
recommended.
d. For those at risk for autonomic hyperreflexia,
antihypertensive medications should be available at
the bedside and labor analgesia should be instituted
early. Autonomic hyperreflexia can also confound the
diagnosis of preeclampsia.
e. Temperature should be measured at regular intervals
because hyperthermia (in the absence of infection) can
occur as a result of altered thermoregulation.
f. Patient position should be changed regularly to prevent
decubitus ulcer formation.
g. The American College of Obstetricians and Gynecologists
(ACOG) concluded in their 2002 Committee Opinion:
Obstetric management of patients with spinal cord
injuries, that obstetricians should be aware of the specific
problems related to SCIs. Autonomic dysreflexia is the
most significant medical complication seen in women
with SCIs and precautions should be taken to avoid
stimuli that can lead to this potentially fatal syndrome.
Women with SCIs may give birth vaginally, but when CD
is indicated, adequate anesthesia (spinal or epidural, if
possible) is needed.15
3. Anesthetic management
a. Neuraxial analgesia is the most common approach for the
prevention and treatment of autonomic hyperreflexia.
Although either CSE or epidural analgesia is a good
choice, CSE may be preferable for its rapid onset. Labor
analgesia differs in these patients by (i) need for initiation
very early in labor, (ii) a typical test dose that will not
identify unintentional subarachnoid catheter placement,
and (iii) difficult assessment of level of analgesia. If
neuraxial block placement is impossible, other measures
for treating autonomic hyperreflexia should be at the
bedside (e.g., nitroglycerin, nitroprusside).
b. Sensory levels can only be reliably assessed if the level of
block is more cephalad than the spinal cord lesion.
However, a limited assessment can be made by stroking
the abdomen above and below the umbilicus. If there is
contraction of the abdominal muscles, the level of sensory
block is below that level because sensory blockade will
abolish this reflex.
c. Placement of an arterial catheter may be indicated to
allow beat-to-beat monitoring of blood pressure (BP)
because many of these patients have low baseline BP and
are prone to hemodynamic instability. Pulse oximetry may
also be prudent.
d. For CD, spinal anesthesia is preferable because it
provides better protection from autonomic hyperreflexia
compared to epidural anesthesia.13 If neuraxial anesthesia
is technically impossible, or if the level of cord injury
produces respiratory embarrassment when supine, general
anesthesia will be necessary. During the period of
denervation (i.e., 24 hours to 1 year after injury),
succinylcholine should not be administered due to the risk
of hyperkalemic cardiac arrest, due to the potential for
massive potassium release from postsynaptic and
extrajunctional receptors. However, some have suggested
that succinylcholine should always be avoided in these
patients, and that nondepolarizing muscle relaxants
should be used to facilitate laryngoscopy and intubation,
if necessary.16
D. Spina bifida
Spina bifida results from failure of the vertebrae to fuse
completely around the neural tube contents. There are two major
categories:
1. Spina bifida occulta describes failed fusion of the arch
without herniation of the meninges or spinal cord. Failure
at a single level (usually L5 or S1) is common and occurs in
up to one-third of the population. It is considered to be a
normal variant by some.17 There is a wide range of
presentations, from asymptomatic radiographic findings to
chronic back pain with posterior disc herniation and minor
neurologic problems. In patients who have superficial
stigmata (e.g., tuft of hair or lipoma), there is an increased
likelihood of an underlying occult spinal dysraphism (i.e.,
cord abnormality). Most of these are asymptomatic but there
may be a higher risk for disc herniation.
a. Obstetric management is uncomplicated in patients with
spina bifida occulta.
b. Anesthetic management
(1) Most patients carrying this diagnosis present little
concern, but preanesthetic assessment should include
examination of the back with clear documentation
of neurologic status.18 The patient should also be
informed that analgesia could be incomplete during
delivery because the epidural space may not be
continuous across the defect. There is an increased
risk of dural puncture because of abnormal lamina
and ligamentum flavum formation, especially when
placement is attempted at the site of the lesion. This
risk can be reduced by inserting the needle in a
position remote from the level of the lesion. Most
neuraxial anesthetics are uncomplicated because the
level of the lesion is typically below most neuraxial
needle placements (i.e., L5–S1). In addition, most are
midline defects of the lamina that rarely interfere
with needle placement.
(2) Presence of neurologic deficits or cutaneous
manifestations should concern the
anesthesiologist, particularly for the risk of
tethered cord.19 Tethered cord results from
attachment of the spinal cord to an area of the lumbar
spine. Reports suggest that the incidence of tethered
cord in cases of spinal bifida occulta is low, but
nearly all patients with spina bifida cystica and
meningomyelocele have tethering. If there are
neurologic deficits or cutaneous manifestations,
radiographs, preferably existing ones, should be
obtained.
(3) In a patient with tethered cord who declined
neuraxial analgesia, fentanyl IV PCA with an adjunct
infusion of dexmedetomidine (an α2-agonist with
negligible placental transfer) provided adequate
analgesia without respiratory depression.20
2. Spina bifida cystica (or aperta) includes meningocele (i.e.,
failed closure of the neural arch with herniation of the
meninges) and myelomeningocele (i.e., herniation of both
meninges and neural elements through a vertebral defect).
These cases are much less common (<5 per 1,000 births).
Prenatal folate supplementation has further reduced the
incidence by as much as 50%.21 Improved surgical and
medical care has increased the likelihood that affected women
will present for obstetric interventions. Numerous anomalies
may coexist including Chiari II malformation (i.e., downward
displacement of the medulla, fourth ventricle, and cerebellum
into the cervical spinal canal), hydrocephalus with shunt
placement, and tethered cord. Many of these patients develop
progressive spinal deformities, including kyphoscoliosis.
a. Obstetric management
Recurrent urinary tract infections are the most common
complication, resulting in preterm labor. Patients with
severe kyphoscoliosis may experience respiratory
compromise due to the expanding uterus. CD is reserved
for obstetric indications (e.g., inadequate pelvic
dimensions).
b. Anesthetic management
(1) Preanesthesia consultation. Preanesthetic
consultation helps determine anesthetic options. The
anesthesiologist should determine the level of the
lesion and whether the patient has residual neurologic
function. Most patients with these disorders have
undergone imaging studies to determine the spinal
abnormalities. These will facilitate anesthetic
assessment.
(2) Technical concerns. Parturients with lesions above
T10 are unlikely to experience labor pain, but those
with higher lesions are at risk for autonomic
hyperreflexia (see Section C. Spinal Cord Injury).
Because neuraxial anesthesia is not contraindicated in
these patients,17 such techniques should be utilized
whenever possible to prevent autonomic
hyperreflexia and to avoid the risk of general
anesthesia. However, attempts at identifying the
epidural space can result in dural puncture due to
incomplete formation of the ligamentum flavum. If
epidural placement is successful, one must consider
that the epidural anesthetic may be inadequate due to
discontinuity of the epidural space. In patients with
negligible function below the lesion, concerns for
cord damage are reduced and continuous spinal
anesthesia should be considered.
(3) Other considerations. If a patient has a
ventricular shunt, its function should be assessed.
Symptoms of shunt malfunction or infection include
headache, fever, drowsiness, and convulsions.
Patients with severe kyphoscoliosis may also require
pulmonary function testing. Patients with spina bifida
who lack bladder control often self-catheterize and
are at increased risk (approximately 25%) for latex
sensitivity.22
E. Intracranial neoplasms
The incidence of brain tumors in pregnant patients does not differ
from the age-matched nonpregnant population nor does the
distribution of types: (i) glioma, 38%; (ii) meningioma, 28%; (iii)
acoustic neuroma, 14%; and (iv) pituitary adenoma, 7%. These
tumors may be asymptomatic until hormonal and physiologic
changes of pregnancy (e.g., increased plasma volume) worsen
symptoms through peritumoral edema formation.23 In addition,
some meningiomas contain progesterone receptors, and their
growth can be accelerated during pregnancy.
Metastatic disease to the central nervous system (CNS) from
breast, lung, and skin tumors represents another important source
of intracranial tumors. Hematogenous spread leads to
macroscopic tumor formation in brain parenchyma with increased
edema and breakdown of the blood–brain barrier. Prognosis of
such cases is dependent on the tumor origin.
1. Clinical issues23
Signs and symptoms of an intracranial neoplasm (see Table
26.5)
1. Clinical issues
a. Cerebral angiography can be performed with abdominal
shielding.
b. Despite increased blood volume, it is unclear whether the
risk of aneurysmal bleeding is increased during
pregnancy. However, once a bleed has occurred, early
surgical intervention may improve both maternal and
fetal outcome.33 Cases of endovascular embolization
have also been reported during pregnancy. In some
patients, this approach avoids craniotomy.
c. Similarly, intervention for an AVM that has bled may
reduce the risk of serious hemorrhage.34
2. Obstetric management
Surgically corrected lesions present no special
considerations. Patients with untreated aneurysms or AVMs,
however, require careful management of BP because of the
risk of rebleeding. Although CD should be reserved for
obstetric indications, hypertension, and increased ICP
resulting from expulsive efforts during second stage labor
should be avoided. Excellent labor analgesia with neuraxial
techniques and an instrumental vaginal delivery are
indicated in these patients.35 Cesarean delivery does not
have a definite advantage over vaginal delivery.
3. Anesthetic management
In patients who have undergone surgical repair of either a
cerebral aneurysm or AVM, anesthetic management is not
different from other obstetric patients. However, if the
patient has an uncorrected aneurysm or AVM and vaginal
delivery is expected, epidural analgesia can prevent pain-
induced hypertension and facilitate assisted vaginal delivery.
An arterial line can also be helpful for beat-to-beat monitoring
of the BP. Either spinal or epidural anesthesia are reasonable
choices for CD. In women with elevated ICP, the risk of dural
puncture and brainstem herniation must also be considered
(see preceding text). If general anesthesia is necessary,
anesthetic considerations are similar to nonpregnant patients
undergoing intracranial surgery. In women with a viable fetus,
a combined neurovascular procedure and CD may be
required. Anesthetic management should follow
recommendations for pregnant women undergoing
nonobstetric surgery.
B. Cortical vein thrombosis
1. Cortical vein thrombosis (CVT), although rare, is increased
during pregnancy, particularly the puerperium. It is related to
obstruction of blood flow from the cavernous, sagittal,
cortical, or lateral sinuses. Although the etiology is unclear,
the risk of CVT is increased because of (i) intracerebral blood
flow stasis, (ii) endothelial capillary injury during the second
stage of labor, (iii) dehydration, and (iv) hypercoagulability.
Most cases occur acutely during the second or third week
postpartum.
2. Clinical issues36
a. Risk factors include (i) inherited thrombophilias; (ii)
systemic disease (e.g., lupus); (iii) neoplasia; (iv)
infection, especially locally (e.g., otitis); and (v) use of
oral contraceptives prior to pregnancy. Dural puncture and
intracranial hypotension may also predispose to CVT.
b. Headache is a common presenting symptom, often
associated with nausea and vomiting. The headache may
be constant or positional and cannot be easily
distinguished from a PDPH. A changing character of the
headache over time is more characteristic of CVT.
Patients should undergo a brief neurologic examination
prior to epidural blood patch. Epidural blood patch (EBP)
in a patient with CVT may improve symptoms briefly. In
this setting, imaging may be warranted.37
REFERENCES
1. Asher MA, Burton DC. Adolescent idiopathic scoliosis: natural history and long term treatment
effects. Scoliosis. 2006;1:2.
2. Crosby ET. Disorders of the vertebral column. In: Gambling DR, Douglas MJ, McKay RSF, eds.
Obstetric Anesthesia and Uncommon Disorders. 2nd ed. Cambridge, United Kingdom: Cambridge
University Press; 2008:129–144.
3. Kuczkowski KM. Labor analgesia for the parturient with prior spinal surgery: what does an
obstetrician need to know? Arch Gynecol Obstet. 2006;274:373–375.
4. Yeo ST, French R. Combined spinal-epidural in the obstetric patient with Harrington rods assisted by
ultrasonography. Br J Anaesth. 1999;83:670–672.
5. Ko JY, Leffert LR. Clinical implications of neuraxial anesthesia in the parturient with scoliosis. Anesth
Analg. 2009;109: 1930–1934.
6. Norris MC, Leighton BL, DeSimone CA. Needle bevel direction and headache after inadvertent dural
puncture. Anesthesiology. 1989;70:729–731.
7. Okutomi T, Saito M, Koura M, et al. Spinal anesthesia using a continuous spinal catheter for cesarean
section in a parturient with prior surgical correction of scoliosis. J Anesth. 2006;20:223–226.
8. Smith PS, Wilson RC, Robinson AP, et al. Regional blockade for delivery in women with scoliosis or
previous spinal surgery. Int J Obstet Anesth. 2003;12:17–22.
9. Gerancher JC, D’Angelo R, Carpenter R. Caudal epidural blood patch for the treatment of postdural
puncture headache. Anesth Analg. 1998;87:394–395.
10. Russell R, Comara S. Regional blocks for delivery in women with scoliosis or previous spinal surgery.
Int J Obstet Anesth. 2003;12:308–310.
11. Hirtz D, Thurman DJ, Gwinn-Hardy K, et al. How common are the “common” neurologic disorders?
Neurology. 2007;68: 326–337.
12. Sekhon LH, Fehlings MG. Epidemiology, demographics, and pathophysiology of acute spinal cord
injury. Spine. 2001; 26:S2–S12.
13. Pereira L. Obstetric management of the patient with spinal cord injury. Obstet Gynecol Surv.
2003;58:678–687.
14. Kuczkowski KM. Labor analgesia for the parturient with spinal cord injury: what does an obstetrician
need to know? Arch Gynecol Obstet. 2006;274:108–112.
15. American College of Obstetricians and Gynecologists. ACOG Committee Opinion No. 275: obstetric
management of patients with spinal cord injuries. Obstet Gynecol. 2002;100:625–627.
16. Briggs ED, Kirsch JR. Anesthetic implications of neuromuscular disease. J Anesth. 2003;17:177–185.
17. Avrahami E, Frishman E, Fridman Z, et al. Spina bifida occulta of S1 is not an innocent finding. Spine.
1994;19:12–15.
18. Tidmarsh MD, May AE. Epidural anaesthesia and neural tube defects. Int J Obstet Anesth.
1998;7:111–114.
19. Ali L, Stocks GM. Spina bifida, tethered cord and regional anaesthesia. Anaesthesia. 2005;60:1149–
1150.
20. Palanisamy A, Klickovich RJ, Ramsay M, et al. Intravenous dexmedetomidine as an adjunct for labor
analgesia and cesarean delivery anesthesia in a parturient with a tethered spinal cord. Int J Obstet
Anesth. 2009;18:258–261.
21. Pitkin RM. Folate and neural tube defects. Am J Clin Nutr. 2007;85:285S–288S.
22. Bernardini R, Novembre E, Lombardi E, et al. Risk factors for latex allergy in patients with spina
bifida and latex sensitization. Clin Exp Allergy. 1999;29:681–686.
23. Stevenson CB, Thompson RC. The clinical management of intracranial neoplasms in pregnancy. Clin
Obstet Gynecol. 2005;48:24–37.
24. Alvis JS, Hicks RJ. Pregnancy-induced acute neurologic emergencies and neurologic conditions
encountered in pregnancy. Semin Ultrasound CT MR. 2012;33:46–54.
25. Marx GF, Zemaitis MT, Orkin LR. Cerebrospinal fluid pressures during labor and obstetrical
anesthesia. Anesthesiology. 1961;22:348–354.
26. Hilt H, Gramm HJ, Link J. Changes in intracranial pressure associated with extradural anaesthesia. Br
J Anaesth. 1986;58: 676–680.
27. Su TM, Lan CM, Yang LC, et al. Brain tumor presenting with fatal herniation following delivery
under epidural anesthesia. Anesthesiology. 2002;96:508–509.
28. Thurtell MJ, Wall M. Idiopathic intracranial hypertension (pseudotumor cerebri): recognition,
treatment, and ongoing management. Curr Treat Options Neurol. 2013;15:1–12.
29. Aly EE, Lawther BK. Anaesthetic management of uncontrolled idiopathic intracranial hypertension
during labour and delivery using an intrathecal catheter. Anaesthesia. 2007;62:178–181.
30. Karmaniolou I, Petropoulos G, Theodoraki K. Management of idiopathic intracranial hypertension in
parturients: anesthetic considerations. Can J Anaesth. 2011;58:650–657.
31. Bateman BT, Olbrecht VA, Berman MF, et al. Peripartum subarachnoid hemorrhage nationwide data
and institutional experience. Anesthesiology. 2012;116:324–333.
32. Clark SL, Christmas JT, Frye DR, et al. Maternal mortality in the United States: predictability and the
impact of protocols on fatal postcesarean pulmonary embolism and hypertension-related intracranial
hemorrhage. Am J Obstet Gynecol. 2014;211:32.e1–32.e9.
33. Dias MS, Sekhar LN. Intracranial hemorrhage from aneurysms and arteriovenous malformations
during pregnancy and the puerperium. Neurosurgery. 1990;27:855–865.
34. Magann EF, Doherty DA, Chauhan SP, et al. Pregnancy, obesity, gestational weight gain, and parity as
predictors of peripartum complications. Arch Gynecol Obstet. 2011;284:827–836.
35. Kuczkowski KM. Labor analgesia for the parturient with neurological disease: what does an
obstetrician need to know? Arch Gynecol Obstet. 2006;274:41–46.
36. Demir CF, Inci MF, Özkan F, et al. Clinical and radiological management and outcome of pregnancies
complicated by cerebral venous thrombosis: a review of 19 cases. J Stroke Cerebrovasc.
2013;22:1252–1257.
37. Lockhart EM, Baysinger CL. Intracranial venous thrombosis in the parturient. Anesthesiology.
2007;107:652–658.
38. Portaccio E, Ghezzi A, Hakiki B, et al; for the MS Study Group of the Italian Neurological Society.
Postpartum relapses increase the risk of disability progression in multiple sclerosis: the role of disease
modifying drugs. J Neurol Neurosurg Psychiatry. 2014;85:845–850.
39. Carrithers MD. Update on disease-modifying treatments for multiple sclerosis. Clin Ther.
2014;36:1938–1945.
40. Stangel M, Gold R, Gass A, et al. Current issues in immunomodulatory treatment of multiple sclerosis
—a practical approach. J Neurol. 2006;253(suppl 1):I32–I36.
41. Confavreux C, Hutchinson M, Hours MM, et al; for the Pregnancy Multiple Sclerosis Group. Rate of
pregnancy-related relapse in multiple sclerosis. N Eng J Med. 1998;339:285–291.
42. Ciafaloni E, Massey JM. The management of myasthenia gravis in pregnancy. Semin Neurol.
2004;24:95–100.
43. Levitan R. Safety of succinylcholine in myasthenia gravis. Ann Emerg Med. 2005;45:225–226.
44. Jiang GX, de Pedro-Cuesta J, Strigård K, et al. Pregnancy and Guillain-Barré syndrome: a nationwide
register cohort study. Neuroepidemiology. 1996;15:192–200.
45. Chan LY, Tsui MH, Leung TN. Guillain-Barré syndrome in pregnancy. Acta Obstet Gynecol Scand.
2004;83:319–325.
46. Wiertlewski S, Magot A, Drapier S, et al. Worsening of neurologic symptoms after epidural anesthesia
for labor in a Guillain-Barré patient. Anesth Analg. 2004;98:825–827.
47. Pennell PB. Pregnancy in women who have epilepsy. Neurol Clin. 2004;22:799–820.
48. Shehata HA, Okosun H. Neurological disorders in pregnancy. Curr Opin Obstet Gynecol.
2004;16:117–122.
49. Harden CL. Pregnancy and epilepsy. Continuum. 2014;20:60–79.
Renal and Hepatic Disease in the Pregnant Patient
Quisqueya T. Palacios and M. Susan Mandell
I. Introduction
A. Renal and liver diseases in parturients
B. Multidisciplinary team
II. Renal disease during pregnancy
A. Renal anatomy
B. Renal physiology
C. Assessment of renal function during pregnancy
D. Categories of renal dysfunction and influence on pregnancy
E. Systemic effects of renal disease and prognosis
F. Etiology of renal disease
G. General management strategies
H. Anesthetic implications of renal disease
I. Renal failure associated with pregnancy
J. Renal disease, dialysis, and pregnancy outcomes
III. Liver disease
A. Characteristics of hepatic disease during pregnancy
B. Changes in hepatic anatomy and physiology during pregnancy
C. Assessment of hepatic function
D. Diagnosis of liver disease in pregnancy
E. Anesthetics and hepatic disease
F. Hepatic diseases unique to pregnancy
G. Hepatic diseases exacerbated by pregnancy
KEYPOINTS
1. The incidence of renal disease during pregnancy is 0.1%.
2. Renal disease during pregnancy may be secondary to acute kidney injury
during pregnancy or preexisting chronic renal disease.
3. Pregnancy is associated with hormonally induced anatomic and
physiologic changes in the kidney that directly and indirectly affect renal
function.
4. Maternal and fetal outcomes depend on the degree of renal impairment
and the severity of hypertension and proteinuria. Timely diagnosis and
management of parturients with preexisting chronic renal disease is key
to outcome.
5. Causes of chronic renal disease include diabetes, hypertension, and
systemic lupus erythematosus (SLE).
6. Early management and treatment of preeclampsia, hypertension, sepsis,
and hemorrhage are associated with better outcomes.
7. Careful use of neuraxial techniques and knowledge of systemic effects of
chronic renal disease and safety implications of anesthetic drugs with
dose adjustments are imperative in parturients with renal disease.
8. Causes of acute kidney injury may include hyperemesis gravidarum,
severe preeclampsia, and acute fatty liver of pregnancy (AFLP).
9. Liver disease can be a coincidental finding or caused by the pregnancy.
10. Use of gestational age of the pregnancy is a good guide to the differential
diagnosis of liver disease. For example, hyperemesis gravidarum occurs
mainly in the first trimester, whereas intrahepatic cholestasis occurs in the
third trimester.
11. Liver disease is present in approximately 3% of all pregnancies.
12. In general, mild liver disease has little effect on drug action and
metabolism, whereas advanced liver dysfunction has complex effects on
drug handling.
13. Many patients with hepatic disease also have defects in renal function
marked by a reduction in glomerular filtration and therefore clearance of
renally excreted drugs.
14. Neuraxial and general anesthesia and intraabdominal surgery can
decrease hepatic blood flow by 20% to 30%, which can further prolong
drug activity.
15. Cerebral edema is a leading cause of death in patients with fulminant
hepatic failure of any cause.
16. Acute viral hepatitis is the most common cause of jaundice during
pregnancy.
I. Introduction
Nonobstetric medical disorders (e.g., cardiac and hypertensive
disease, pulmonary disease, and hepatorenal disease) can lead to
increased maternal morbidity and peripartum intensive care unit
(ICU) admission as well as maternal and fetal mortality. Early
diagnosis of associated medical disorders, optimization of preexisting
diseases, and timely delivery of the fetus can improve outcome.
Although maternal mortality rates in the United States are very low,
over the last 30 years, other developed nations have continued to
decrease maternal mortality rates. However, the maternal mortality
rate in the United States has increased when compared to other
developed nations. Many of the causes of maternal mortality are
preventable.1 The overall maternal mortality rate was estimated at
11.5 maternal deaths per 100,000 live births during 1991 to 1997.2 In
2008, maternal mortality rate in the United States has increased and
was estimated at 17 maternal deaths per 100,000 births. In addition,
the increased rates of obesity, hypertension, diabetes, renal disease,
and cesarean delivery (CD) have contributed to increased maternal
morbidity.3
B. Multidisciplinary team
Most critically ill parturients with liver or renal disease will
benefit from the care provided by a multidisciplinary team.
These team members often include a maternal–fetal medicine
specialist, nephrologist or hepatologist, critical care specialist, an
obstetric anesthesiologist, and neonatologist. A combination of
thorough antenatal and intrapartum obstetric anesthesia care,
systematic assessment of peripartum anesthetic considerations, as
well as neonatal intensive care preparation will provide
comprehensive care that can reduce maternal and neonatal
morbidity and mortality.6
2. Potential nephrotoxins
a. There are several potential nephrotoxins that should be
used with caution in parturients with renal insufficiency
(e.g., nonsteroidal anti-inflammatory drugs [NSAIDs],
aminoglycosides, anticholinergics, radiocontrast agents).
NSAIDs, in particular, reduce RBF and should be used
with caution in patients with renal dysfunction.
b. Some inhaled anesthetics (e.g., sevoflurane) increase
inorganic fluoride levels. Although sevoflurane is
metabolized to the nephrotoxic compound A, this has not
been found to be clinically significant.
3. Preoperative preparation and laboratory studies
a. Preoperative evaluation of a parturient with preexisting
renal disease should include a thorough history and
physical examination, paying particular attention to
complications associated with renal disease and any signs
or symptoms of inadequate dialysis, such as uremia and
fluid overload. Table. 27.6 reviews the effects of chronic
renal disease on organ systems.
4. Anesthetic management
Anesthetic management aims to optimize the medical
condition of the patient before labor and delivery. This
includes balancing intravascular volume as well as avoiding
hypotension, renal toxic drugs, and anesthetic complications.
The advantages of neuraxial anesthesia have been discussed
previously (see Chapter 12). Although neuraxial anesthesia
offers many advantages for labor and delivery, the decision to
use a particular anesthetic technique should be based on the
relative risks and benefits. Spinal anesthesia has been used
successfully in parturients with chronic renal disease with
normal coagulation profile for elective CD.26 Relative
contraindications to neuraxial anesthesia may include volume
contraction due to dialysis, hypotension, and coagulation
abnormalities. Serum potassium should be determined before
induction of general anesthesia in patients with renal failure.
Succinylcholine administration increases the serum potassium
level by as much as 0.7 mEq per L. If a patient is already
hyperkalemic, further elevations in potassium could cause
asystole or ventricular dysrhythmia.
1. Hyperemesis gravidarum
Most pregnant women experience nausea and vomiting in the
first trimester, usually resolving by 12 to 16 weeks’ gestation.
In rare cases, pregnant women develop a persistent severe
form of nausea and vomiting called hyperemesis gravidarum.
HG occurs in 0.5 to 10 per 1,000 pregnancies and is the only
pregnancy-induced hepatic disease occurring during the first
trimester of pregnancy. Differential diagnoses include bile
duct stones, cholecystitis, and viral hepatitis.
a. Laboratory evaluation
Mild elevation in the serum transaminases develops in
25% to 67% of cases. In rare circumstances, there is
severe dehydration with marked increases in serum
transaminases of up to 300%, indicating ischemic
hepatocellular injury. Serum unconjugated bilirubin and
ALP rise slightly, whereas serum albumin and
international normalized ratio (INR) remain normal.
These biochemical abnormalities normalize with
treatment.
b. Management and anesthetic implications
Hospitalization for IV hydration, antiemetics, and
monitoring may be required. Initial IV therapy is initiated
for 24 to 48 hours to correct dehydration, electrolyte, and
any acid–base imbalance. Thiamine should be added to
glucose-containing IV solutions when HG is severe and
associated with abnormal hepatic function or in cases
lasting more than 3 weeks.
c. Outcome
Outcomes are usually good with no adverse maternal and
fetal effects (e.g., prematurity, birth defects), provided that
symptoms are adequately treated. The serum
transaminases return to normal with proper supportive
care, and there is rarely any permanent hepatic injury.
Most cases resolve by 20 weeks’ gestation, regardless of
therapy.
2. Cholestasis of pregnancy
a. Intrahepatic cholestasis of pregnancy (ICP) is a rare and
serious complication of pregnancy. It is associated with
fetal mortality rates of 11% to 20% if left untreated. The
cause is unknown, but it is thought that increasing levels
of estrogen during pregnancy play a role in genetically
susceptible patients. Estrogen influences transport across
the bile ducts. Patients usually present with moderate to
severe itching in the third trimester of pregnancy.
Approximately 80% have pruritus alone, whereas 20%
will develop jaundice in addition to itching. The
incidence of ICP is 1 to 2 per 1,000 pregnancies in North
America. Risk factors include older, multiparous women
with multiple gestations, a positive family history of
cholestasis of pregnancy, and cholestasis while on oral
contraceptives. ICP must be distinguished from other
common causes of cholestasis. A careful evaluation by a
medical specialist helps to exclude infectious and
autoimmune causes of cholestasis, which are common in
women of childbearing age.48
b. Laboratory evaluation
The key diagnostic laboratory finding is an increase in
serum bile acids from 10 to 100 times the normal value.
Bilirubin levels rarely exceed 6 mg per dL. The
transaminases can be increased in ICP.
c. Management and anesthetic implications
Patients with ICP can have severe excoriations. This can
cause infection and may make IV access and neuraxial
needle placement difficult. Treatment aims to provide
symptomatic relief from itching. Mild pruritus may
respond to emollients, topical antipruritics, and
antihistamines. Ursodeoxycholic acid (UDCA) is an
effective therapy that provides relief in patients with
moderate or severe pruritus.49 There is rapid resolution
following delivery.
d. Outcomes
ICP is associated with an increase in preterm delivery,
perinatal mortality, and meconium staining. Therefore,
obstetric management involves close fetal surveillance.49
All maternal symptoms resolve rapidly following
delivery. Women with cholestasis of pregnancy have a
60% recurrence in subsequent pregnancies.50
a. Diagnosis
The modified Swansea criteria are used for diagnosis.
Liver biopsy studies show that the criteria have a
sensitivity of 100% and specificity of 57%. The positive
and negative predictive values are 85% and 100%,
respectively. The criteria are shown in Table. 27.11.
d. Outcome
(1) In the absence of fulminant liver failure, liver
function improves rapidly after delivery. Since the
1980s, maternal mortality rates have decreased to
approximately 10% to 20% with early diagnosis,
treatment, and prompt delivery. However, fetal
mortality rates remain high at 23%. With aggressive
therapy, one study reported 100% survival in two
U.S. hospitals over a 15-year period.55
(2) Patients who fail to rapidly emerge from general
anesthesia should be carefully evaluated for
progressive encephalopathy or increased intracranial
pressure. These two conditions are associated with a
poor maternal outcome.51
4. Hemolysis, elevated liver enzymes, low platelets syndrome
HELLP syndrome was previously considered to be a severe
form of preeclampsia. Although many think that HELLP is a
distinct disease, the presentation is very similar to
preeclampsia; there is generalized vasoconstriction and
activation of the coagulation cascade resulting in
thrombocytopenia. This causes microangiopathic hemolytic
anemia and hepatic necrosis. The incidence of HELLP is
approximately 1 in 1,000 pregnancies.52
a. Diagnosis
Women complain of pain in the right upper quadrant,
nausea and vomiting, and weight gain. Some patients may
also present with symptoms of preeclampsia (elevated
blood pressure and proteinuria). Laboratory testing shows
(i) hemolysis, (ii) increase in total bilirubin and lactate
dehydrogenase (LDH), (iii) moderately elevated
transaminases, and (iv) thrombocytopenia. Patients can
develop a microangiopathic hemolytic anemia due to
activation of the coagulation cascade. This can cause
disseminated intravascular coagulation with an increased
risk of bleeding and thrombotic complications. These
findings are useful to distinguish HELLP from other
serious medical conditions with a similar presentation
such as AFLP and SLE. Table. 27.12 reviews the signs
and symptoms of HELLP.
REFERENCES
1. Lang CT, King JC. Maternal mortality in the United States. Best Pract Res Clin Obstet Gynaecol.
2008;22:517–531.
2. Centers for Disease Control and Prevention. Maternal mortality—United States, 1982-1996. MMWR
Morb Mortal Wkly Rep. 1998;47:705–707.
3. Hankins GD, Clark SL, Pacheco LD, et al. Maternal mortality, near misses, and severe morbidity:
lowering rates through designated levels of maternity care. Obstet Gynecol. 2012;120:929–934.
4. Chinnappa V, Ankichetty S, Angle P, et al. Chronic kidney disease in pregnancy. Int J Obstet Anesth.
2013;22:223–230.
5. Nevis IF, Reitsma A, Dominic A, et al. Pregnancy outcomes in women with chronic kidney disease: a
systematic review. Clin J Am Soc Nephrol. 2011;6:2587–2598.
6. Dhir S, Fuller J. Case report: pregnancy in hemodialysis-dependent end-stage renal disease: anesthetic
considerations. Can J Anaesth. 2007;54:556–560.
7. Lindheimer MD, Davison JM. Osmoregulation, the secretion of arginine vasopressin and its
metabolism during pregnancy. Eur J Endocrinol. 1995;132:133–143.
8. Higby K, Suiter CR, Phelps JY, et al. Normal values of urinary albumin and total protein excretion
during pregnancy. Am J Obstet Gynecol. 1994;171:984–989.
9. American College of Obstetricians and Gynecologists. Ob-Gyns Issue Task Force Report on
Hypertension in Pregnancy: preeclampsia diagnosis no longer requires presence of proteinuria.
http://www.acog.org/About-ACOG/News-Room/News-Releases/2013/Ob-Gyns-Issue-Task-Force-
Report-on-Hypertension-in-Pregnancy. Accessed June 19, 2015.
10. Cheung AN, Luk SC. The importance of extensive sampling and examination of cervix in suspected
cases of amniotic fluid embolism. Arch Gynecol Obstet. 1994;255:101–105.
11. Smith MC, Moran P, Ward MK, et al. Assessment of glomerular filtration rate during pregnancy using
the MDRD formula. BJOG. 2008;115:109–112.
12. National Kidney Foundation. K/DOQI clinical practice guidelines for chronic kidney disease:
evaluation, classification, and stratification. Am J Kidney Dis. 2002;39:S1–S266.
13. Podymow T, August P, Akbari A. Management of renal disease in pregnancy. Obstet Gynecol Clin
North Am. 2010;37:195–210.
14. Jones DC, Hayslett JP. Outcome of pregnancy in women with moderate or severe renal insufficiency.
N Engl J Med. 1996;335:226–232.
15. Imbasciati E, Gregorini G, Cabiddu G, et al. Pregnancy in CKD stages 3 to 5: fetal and maternal
outcomes. Am J Kidney Dis. 2007;49:753–762.
16. Wu CC, Chen SH, Ho CH, et al. End-stage renal disease after hypertensive disorders in pregnancy. Am
J Obstet Gynecol. 2014;210:147.e1–147.e8.
17. Fischer MJ. Chronic kidney disease and pregnancy: maternal and fetal outcomes. Adv Chronic Kidney
Dis. 2007;14:132–145.
18. Son KH, Lim NK, Lee JW, et al. Comparison of maternal morbidity and medical costs during
pregnancy and delivery between patients with gestational diabetes and patients with pre-existing
diabetes. Diabet Med. 2015;32:477–486.
19. Landon MB. Diabetic nephropathy and pregnancy. Clin Obstet Gynecol. 2007;50:998–1006.
20. Koh JH, Ko HS, Lee J, et al. Pregnancy and patients with preexisting lupus nephritis: 15 years of
experience at a single center in Korea. Lupus. 2015;24:764–772.
21. Delesalle AS, Robin G, Provôt F, et al. Impact of end-stage renal disease and kidney transplantation on
the reproductive system. Gynecol Obstet Fertil. 2015;43:33–40.
22. López LF, Martínez CJ, Castañeda DA, et al. Pregnancy and kidney transplantation, triple hazard?
Current concepts and algorithm for approach of preconception and perinatal care of the patient with
kidney transplantation. Transplant Proc. 2014;46:3027–3031.
23. Friedrich AD. The controversy of “renal-dose dopamine.” Int Anesthesiol Clin. 2001;39:127–139.
24. Ioscovich A, Orbach-Zinger S, Zemzov D, et al. Peripartum anesthetic management of renal transplant
patients—a multicenter cohort study. J Matern Fetal Neonatal Med. 2014;27:484–487.
25. American Society of Anesthesiologists Task Force on Obstetric Anesthesia. Practice guidelines for
obstetric anesthesia: an updated report by the American Society of Anesthesiologists Task Force on
Obstetric Anesthesia. Anesthesiology. 2007;106:843–863.
26. Modi M, Vora K, Parikh G, et al. Anesthetic management in parturients with chronic kidney disease
undergoing elective caesarean delivery: our experience of nine cases. Indian J Nephrol. 2014;24:20–
23.
27. Bagshaw SM, Bellomo R. Early diagnosis of acute kidney injury. Curr Opin Crit Care. 2007;13:638–
644.
28. Gammill HS, Jeyabalan A. Acute renal failure in pregnancy. Crit Care Med. 2005;33:S372–S384.
29. Lameire N, Van Biesen W, Vanholder R. Acute renal failure. Lancet. 2005;365:417–430.
30. Hoste EA, Kellum JA. Acute kidney injury: epidemiology and diagnostic criteria. Curr Opin Crit
Care. 2006;12:531–537.
31. Nwoko R, Plecas D, Garovic VD. Acute kidney injury in the pregnant patient. Clin Nephrol.
2012;78:478–486.
32. American College of Obstetricians and Gynecologists Committee on Obstetric Practice. Committee
Opinion No. 623: emergent therapy for acute-onset, severe hypertension during pregnancy and the
postpartum period. Obstet Gynecol. 2015;125:521–525.
33. Lindheimer MD, Taler SJ, Cunningham FG; for the American Society of Hypertension. ASH position
paper: hypertension in pregnancy. J Clin Hypertens. 2009;11:214–225.
34. Mehrabadi A, Liu S, Bartholomew S, et al. Hypertensive disorders of pregnancy and the recent
increase in obstetric acute renal failure in Canada: population based retrospective cohort study. BMJ.
2014;349:g4731.
35. Vikse BE, Irgens LM, Leivestad T, et al. Preeclampsia and the risk of end-stage renal disease. N Engl J
Med. 2008;359:800–809.
36. Jonard M, Ducloy-Bouthors AS, Boyle E, et al. Postpartum acute renal failure: a multicenter study of
risk factors in patients admitted to ICU. Ann Intensive Care. 2014;4:36.
37. Dambal A, Lakshmi KS, Gorikhan G, et al. Obstetric acute kidney injury; a three year experience at a
medical college hospital in North Karnataka, India. J Clin Diagn Res. 2015;9:OC01–OC04.
38. Zeeman GG. Obstetric critical care: a blueprint for improved outcomes. Crit Care Med.
2006;34:S208–S214.
39. Neligan PJ, Laffey JG. Clinical review: special populations—critical illness and pregnancy. Crit Care.
2011;15:227.
40. Fontaine-Poitrineau C, Brachereau J, Rigaud J, et al. Renal colic in pregnant women: study of a series
of one hundred and three cases [in French]. Prog Urol. 2014;5:294–300.
41. Elliott MA, Nichols WL. Thrombotic thrombocytopenic purpura and hemolytic uremic syndrome.
Mayo Clin Proc. 2001;76:1154–1162.
42. McDonald SD, Malinowski A, Zhou Q, et al. Cardiovascular sequelae of preeclampsia/eclampsia: a
systematic review and meta-analyses. Am Heart J. 2008;156:918–930.
43. Hussein W, Lafayette RA. Renal function in normal and disordered pregnancy. Curr Opin Nephrol
Hypertens. 2014;23:46–53.
44. Panaye M, Jolivot A, Lemoine S, et al. Pregnancies in hemodialysis and in patients with end-stage
chronic kidney disease: epidemiology, management and prognosis. Nephrol Ther. 2014;10:485–491.
45. Piccoli GB, Attini R, Vigotti FN, et al. Is renal hyperfiltration protective in chronic kidney disease-
stage 1 pregnancies? A step forward unravelling the mystery of the effect of stage 1 chronic kidney
disease on pregnancy outcomes. Nephrology. 2015;20:201–208.
46. Almashhrawi AA, Ahmed KT, Rahman RN, et al. Liver diseases in pregnancy: diseases not unique to
pregnancy. World J Gastroenterol. 2013;19:7630–7638.
47. Bose PD, Das BC, Hazam RK, et al. Evidence of extrahepatic replication of hepatitis E virus in human
placenta. J Gen Virol. 2014;95:1266–1271.
48. Arrese M. Cholestasis during pregnancy: rare hepatic diseases unmasked by pregnancy. Ann Hepatol.
2006;5:216–218.
49. Caughey AB. Cholestasis of pregnancy: in need of a more rapid diagnosis. J Perinatol. 2006;26:525–
526.
50. Schutt VA, Minuk GY. Liver diseases unique to pregnancy. Best Pract Res Clin Gastroenterol.
2007;21:771–792.
51. Holzman RS, Riley LE, Aron E, et al. Perioperative care of a patient with acute fatty liver of
pregnancy. Anesth Analg. 2001;92:1268–1270.
52. Sibai BM. Imitators of severe preeclampsia. Obstet Gynecol. 2007;109:956–966.
53. Gregory TL, Hughes S, Coleman MA, et al. Acute fatty liver of pregnancy; three cases and discussion
of analgesia and anaesthesia. Int J Obstet Anesth. 2007;16:175–179.
54. Horlocker TT, Wedel DJ, Benzon H, et al. Regional anesthesia in the anticoagulated patient: defining
the risks (the second ASRA Consensus Conference on Neuraxial Anesthesia and Anticoagulation).
Reg Anesth Pain Med. 2003;28:172–197.
55. Castro MA, Goodwin TM, Shaw KJ, et al. Disseminated intravascular coagulation and antithrombin
III depression in acute fatty liver of pregnancy. Am J Obstet Gynecol. 1996;174:211–216.
56. Riley C. Liver disease in the pregnant patient. Am J Gastroenterol. 1999;94:1728–1732.
57. Guntupalli SR, Steingrub J. Hepatic disease and pregnancy: an overview of diagnosis and
management. Crit Care Med. 2005;33:S332–S339.
58. Soubra SH, Guntupalli KK. Critical illness in pregnancy: an overview. Crit Care Med. 2005;33:S248–
S255.
59. Czaja AJ. Review article: the management of autoimmune hepatitis beyond consensus guidelines.
Aliment Pharmacol Ther. 2013;38:343–364.
60. Coscia LA, Constantinescu S, Moritz MJ, et al. Report from the National Transplantation Pregnancy
Registry (NTPR): outcomes of pregnancy after transplantation. Clin Transpl. 2010:65–85.
61. Blume C, Sensoy A, Gross MM, et al. A comparison of the outcome of pregnancies after liver and
kidney transplantation. Transplantation. 2013;95:222–227.
Obstetric Anesthesia for Parturients with Respiratory
Diseases
Suzanne K.W. Mankowitz and Stephanie R. Goodman
I. Introduction
II. Asthma
A. Introduction
B. Effect of pregnancy on asthma
C. Effect of asthma on pregnancy
D. Asthma classification and control
E. Medical management
F. Labor management
G. Anesthesia for cesarean delivery
III. Pulmonary embolism during pregnancy
A. Introduction
B. Pregnancy effects on venous thromboembolism
C. Symptoms of venous thromboembolism and pulmonary
embolism
D. Risk factors for venous thromboembolism
E. Imaging studies
F. Treatment
G. Anesthetic implications of venous thromboembolism treatment
IV. Amniotic fluid embolism
A. Background
B. Timing
C. Symptoms and presentation
D. Risk factors
E. Pathophysiology
F. Diagnosis
G. Treatment
V. Venous air embolism
A. Description
B. Timing
C. Symptoms
D. Risk factors
E. Monitoring
F. Treatment
VI. Smoking
A. Pregnancy effects
B. Neonatal effects
C. Offspring effects
D. Pulmonary complications
VII. Obstructive sleep apnea
A. Background
B. Effect on pregnancy
C. Diagnosis
D. Treatment
E. Analgesia and anesthesia
VIII. Sarcoidosis
A. Description
B. Pulmonary sarcoidosis
C. Effect of sarcoidosis on pregnancy
D. Effect of pregnancy on sarcoidosis
E. Diagnosis and treatment
IX. Aspiration pneumonitis
A. Definition
B. Aspiration in parturients
C. Pathophysiology
D. Treatment
E. Prophylaxis
F. Anesthetic management
X. Cystic fibrosis
A. Background
B. Effect of pregnancy on cystic fibrosis
C. Effect of cystic fibrosis on pregnancy
D. Medical management
E. Anesthetic management
XI. Lung transplantation
A. Transplantation and pregnancy
B. Medications
C. Preterm delivery
D. Surveillance
E. Anesthetic management
XII. Restrictive lung disease
A. Pregnancy
B. Spinal cord injury and kyphoscoliosis
C. Evaluation
D. Management
XIII. Acute respiratory distress syndrome and respiratory failure
A. Diagnostic criteria
B. Clinical presentation
C. Causes of acute respiratory distress syndrome
D. Treatment
E. Alternative treatments
F. Effect of acute respiratory distress syndrome on pregnancy
G. Obstetric management
Summary
KEYPOINTS
1. Diagnosis and presentation. Many respiratory diseases are difficult to
diagnose during pregnancy because the normal physiologic changes of
pregnancy resemble the pathologic states seen in these diseases. For
example, embolic disease may present with dyspnea and tachycardia.
Overlapping presentations of respiratory diseases further complicate the
diagnosis. Venous thromboembolism (VTE), amniotic fluid embolism
(AFE), venous air embolism (VAE), obstructive sleep apnea (OSA),
aspiration, severe restrictive lung disease (RLD), cystic fibrosis (CF), and
pulmonary sarcoidosis share similar signs and symptoms.
2. Physiologic changes of pregnancy and disease progression. The
physiologic changes of pregnancy may predispose to respiratory disease
development or exacerbate preexisting disease. For example, the
hematologic changes which occur during pregnancy increase the risk of
VTE. The gastrointestinal and airway changes increase the likelihood of
pulmonary aspiration. OSA symptoms often worsen during pregnancy.
Decreased functional residual capacity and increased tidal volume,
respiratory rate, and blood volume exacerbate disease in patients with
severe RLD, CF, and sarcoidosis.
3. Effect of pregnancy on disease states. Pregnancy may be beneficial to
some parturients with respiratory disease, particularly those with
sarcoidosis and asthma. Other parturients are more likely to suffer
complications during pregnancy, such as lung transplant recipients.
Generally, severe disease prior to pregnancy usually worsens during
pregnancy, particularly in patients with severe asthma, advanced
sarcoidosis, CF, and RLD. Patients with severe cardiorespiratory
compromise from pulmonary embolism (PE), AFE, VTE, and acute
respiratory distress syndrome (ARDS) may require intensive care
admission and may not survive the pregnancy.
4. Perinatal complications and medications. Many respiratory diseases
seen during pregnancy result in perinatal complications. This may result
in maternal pregnancy complications particularly in women with asthma,
smokers, and parturients with OSA. Neonatal complications can also
occur in women with asthma, smokers, lung transplant recipients, and in
parturients with CF and OSA. In addition, the medications which are
administered for the treatment of respiratory disease may have
detrimental fetal effects. However, the overall benefit of medication
administration exceeds their risk in women with severe asthma, transplant
recipients, critically ill patients with ARDS, and after an embolic event.
Sometimes, administered medications may complicate regional anesthetic
management.
5. Anesthetic management. General endotracheal anesthesia is best
avoided in patients with respiratory disease. Neuraxial analgesia during
labor should be encouraged, and neuraxial anesthesia should be used for
most surgical procedures.
F. Treatment36,39
1. Reverse the pressure gradient. Treatment involves reversing
the pressure gradient between the right atrium and the uterus
by increasing right atrial pressure. In order to trap the air in
the right atrium, it is recommended to place the patient in a
left lateral tilt with a 5-degree head-down position.36,37
However, this has not been effective in animal models and
may not be effective in humans.
2. Flood the field. It is also important to flood the field with
fluid such as normal saline and, if possible, eliminate the site
of air entry.
3. Discontinue nitrous oxide. Nitrous oxide should not be used,
and 100% oxygen should be delivered.
4. Central venous catheter. A central venous catheter or PAC
can be used to aspirate air from the heart. As much as 50% of
entrained air can be aspirated via a multiorifice catheter
placed in the right atrium.
5. Positive end-expiratory pressure. The use of positive end-
expiratory pressure (PEEP) and the Valsalva maneuver could
result in a paradoxical embolism.
6. Supportive care. Vasopressors, inotropes, and pulmonary
vasodilators may be necessary.
7. Hyperbaric oxygen. Hyperbaric oxygen therapy has been
useful especially after paradoxical air embolism with cerebral
air embolism.
VI. Smoking. Approximately 14% of women in the United States smoke
during pregnancy. Both perinatal and pregnancy-related
complications occur as a result of smoking.40
A. Pregnancy effects. There are number of pregnancy complications
associated with smoking.40 These include infertility, ectopic
pregnancy, spontaneous abortion (20% to 80% higher in women
who smoke), placental abruption, placenta previa, fetal growth
restriction, and PTD.
B. Neonatal effects. Several fetal malformations have been reported.
In a large meta-analysis, moderate increases in the rates of limb,
cardiac, and musculoskeletal defects were reported. Larger effects
were noted for orofacial clefts, clubfoot, limb, eye, and
gastrointestinal defects.41 These results need to be confirmed in
further studies.
C. Offspring effects. Offspring effects include increased rates of
sudden infant death syndrome and evidence of increased rates of
childhood respiratory disease, such as asthma, as well as
behavioral problems, such as attention deficit disorder has been
reported.
SUMMARY
Normal physiologic changes that occur during pregnancy may worsen many
respiratory diseases. In addition, these changes can make the diagnosis of a
respiratory disease challenging. Many pulmonary diseases seen during
pregnancy have adverse effects on maternal and neonatal outcomes, and
pregnancy sometimes alters the course of a given disease. Some of the
medications used to treat parturients with lung disease have not been fully
studied in terms of their negative impact. Early neuraxial analgesia during labor
is recommended for most parturients with respiratory disease. Larger studies are
needed to elucidate the pathophysiology and outcomes for most of the diseases
discussed in this chapter.
REFERENCES
1. Murphy VE, Clifton VL, Gibson PG. Asthma exacerbations during pregnancy: incidence and
association with adverse pregnancy outcomes. Thorax. 2006;61:169–176.
2. Maselli DJ, Adams SG, Peters JI, et al. Management of asthma during pregnancy. Ther Adv Respir
Dis. 2013;7:87–100.
3. Rocklin RE. Asthma, asthma medications and their effects on maternal/fetal outcomes during
pregnancy. Reprod Toxicol. 2011;32:189–197.
4. Murphy VE, Schatz M. Asthma in pregnancy: a hit for two. Eur Respir Rev. 2014;23:64–68.
5. Murphy VE, Powell H, Wark PA, et al. A prospective study of respiratory viral infection in pregnant
women with and without asthma. Chest. 2013;144:420–427.
6. Wang G, Murphy VE, Namazy J, et al. The risk of maternal and placental complications in pregnant
women with asthma: a systematic review and meta-analysis. J Matern Fetal Neonatal Med.
2014;27:934–942.
7. Murphy VE, Namazy JA, Powell H, et al. A meta-analysis of adverse perinatal outcomes in women
with asthma. BJOG. 2011;118:1314–1323.
8. Enriquez R, Griffin MR, Carroll KN, et al. Effect of maternal asthma and asthma control on pregnancy
and perinatal outcomes. J Allergy Clin Immunol. 2007;120:625–630.
9. Tegethoff M, Olsen J, Schaffner E, et al. Asthma during pregnancy and clinical outcomes in offspring:
a national cohort study. Pediatrics. 2013;132:483–491.
10. Schatz M, Dombrowski MP. Clinical practice. Asthma in pregnancy. N Engl J Med. 2009;360:1862–
1869.
11. National Asthma Education and Prevention Program. Expert Panel Report 3 (EPR-3): Guidelines for
the Diagnosis and Management of Asthma-Summary Report 2007. J Allergy Clin Immunol.
2007;120(suppl 5):S94–S138.
12. Tilley SL, Hartney JM, Erikson CJ, et al. Receptors and pathways mediating the effects of
prostaglandin E2 on airway tone. Am J Physiol Lung Cell Mol Physiol. 2003;284:L599–L606.
13. Woods BD, Sladen RN. Perioperative considerations for the patient with asthma and bronchospasm.
Br J Anaesth. 2009;103(suppl 1):i57–i65.
14. Groeben H. Epidural anesthesia and pulmonary function. J Anesth. 2006;20:290–299.
15. Kuczkowski KM. Labor analgesia for the parturient with respiratory disease: what does an obstetrician
need to know? Arch Gynecol Obstet. 2005;272:160–166.
16. Scavone BM, Ratliff J, Wong CA. Physiologic effects of neuraxial anesthesia. In: Wong C, ed. Spinal
and Epidural Anesthesia. New York, NY: McGraw-Hill; 2007:111–126.
17. Berg CJ, Callaghan WM, Syverson C, et al. Pregnancy-related mortality in the United States, 1998 to
2005. Obstet Gynecol. 2010;116:1302–1309.
18. Heit JA, Kobbervig CE, James AH, et al. Trends in the incidence of venous thromboembolism during
pregnancy or postpartum: a 30-year population-based study. Ann Intern Med. 2005;143:697–706.
19. Greer IA. Thrombosis in pregnancy: updates in diagnosis and management. Hematology Am Soc
Hematol Educ Program. 2012:203–207.
20. Donnelly JC, D’Alton ME. Pulmonary embolus in pregnancy. Semin Perinatol. 2013;37:225–233.
21. Cutts BA, Dasgupta D, Hunt BJ. New directions in the diagnosis and treatment of pulmonary
embolism in pregnancy. Am J Obstet Gynecol. 2013;208:102–108.
22. Saeed G, Möller M, Neuzner J, et al. Emergent surgical pulmonary embolectomy in a pregnant
woman: case report and literature review. Tex Heart Inst J. 2014;41:188–194.
23. Chau DF, Fragneto RY. Maternal embolism. Int Anesthesiol Clin. 2014;52:61–84.
24. Brennan MC, Moore LE. Pulmonary embolism and amniotic fluid embolism in pregnancy. Obstet
Gynecol Clin North Am. 2013;40:27–35.
25. Bourjeily G, Paidas M, Khalil H, et al. Pulmonary embolism in pregnancy. Lancet. 2010;375:500–512.
26. Gray G, Nelson-Piercy C. Thromboembolic disorders in obstetrics. Best Pract Res Clin Obstet
Gynaecol. 2012;26:53–64.
27. Costantine MM. Physiologic and pharmacokinetic changes in pregnancy. Front Pharmacol.
2014;5:65.
28. Tawfik MM, Taman ME, Motawea AA, et al. Thrombolysis for the management of massive
pulmonary embolism in pregnancy. Int J Obstet Anesth. 2013;22:149–152.
29. Mos IC, Klok FA, Kroft LJ, et al. Imaging tests in the diagnosis of pulmonary embolism. Semin Respir
Crit Care Med. 2012;33:138–143.
30. Horlocker TT, Wedel DJ, Rowlingson JC, et al. Regional anesthesia in the patient receiving
antithrombotic or thrombolytic therapy: American Society of Regional Anesthesia and Pain Medicine
Evidence-Based Guidelines (Third Edition). Reg Anesth Pain Med. 2010;35:64–101.
31. Clark SL. Amniotic fluid embolism. Obstet Gynecol. 2014;123(2 pt 1):337–348.
32. Fong A, Chau CT, Pan D, et al. Amniotic fluid embolism: antepartum, intrapartum and demographic
factors. J Matern Fetal Neonatal Med. 2015;28:793–798.
33. Rath WH, Hoferr S, Sinicina I. Amniotic fluid embolism: an interdisciplinary challenge:
epidemiology, diagnosis and treatment. Dtsch Arztebl Int. 2014;111:126–132.
34. Kissko JM III, Gaiser R. Amniotic fluid embolism. Anesthesiology Clin. 2013;31:609–621.
35. McDonnell NJ, Percival V, Paech MJ. Amniotic fluid embolism: a leading cause of maternal death yet
still a medical conundrum. Int J Obstet Anesth. 2013;22:329–336.
36. Kim CS, Liu J, Kwon JY, et al. Venous air embolism during surgery, especially cesarean delivery. J
Korean Med Sci. 2008;23:753–761.
37. Philip J, Sharma SK. Respiratory disorders in pregnancy. In: Gambling D, Douglas MJ, McKay RS,
eds. Obstetric Anesthesia and Uncommon Disorders. 2nd ed. Philadelphia, PA: W.B. Saunders; 2008.
38. Mirski MA, Lele AV, Fitzsimmons L, et al. Diagnosis and treatment of vascular air embolism.
Anesthesiology. 2007;106:164–177.
39. Hadid V, Patenaude V, Oddy L, et al. Sarcoidosis and pregnancy: obstetric and neonatal outcomes in a
population-based cohort of 7 million births. J Perinat Med. 2015;43:201–207.
40. Einarson A, Riordan S. Smoking in pregnancy and lactation: a review of risks and cessation strategies.
Eur J Clin Pharmacol. 2009;65:325–330.
41. Hackshaw A, Rodeck C, Boniface S. Maternal smoking in pregnancy and birth defects: a systematic
review based on 173 687 malformed cases and 11.7 million controls. Hum Reprod Update.
2011;17:589–604.
42. Talbot L, Palmer J. Effects of smoking on health and anesthesia. Anaesth Intensive Care Med.
2013;14:107–109.
43. Morong S, Hermsen B, de Vries N. Sleep-disordered breathing in pregnancy: a review of the
physiology and potential role for positional therapy. Sleep Breath. 2014;18:31–37.
44. Ankichetty SP, Angle P, Joselyn AS, et al. Anesthetic considerations of parturients with obesity and
obstructive sleep apnea. J Anaesthesiol Clin Pharmacol. 2012;28:436–443.
45. Pamidi S, Pinto LM, Marc I, et al. Maternal sleep-disordered breathing and adverse pregnancy
outcomes: a systematic review and metaanalysis. Am J Obstet Gynecol. 2014;210:52.e1–52.e14.
46. Fung AM, Wilson DL, Barnes M, et al. Obstructive sleep apnea and pregnancy: the effect on perinatal
outcomes. J Perinatol. 2012;32:399–406.
47. Gerke AK. Morbidity and mortality in sarcoidosis. Curr Opin Pulm Med. 2014;20:472–478.
48. Stone S, Nelson-Piercy C. Respiratory disease in pregnancy. Obstet Gynaecol Reprod Med.
2010;20:14–21.
49. Freymond N, Cottin V, Cordier JF. Infiltrative lung diseases in pregnancy. Clin Chest Med.
2011;32:133–146.
50. Vahid B, Mushlin N, Weibel S. Sarcoidosis in pregnancy and postpartum period. Curr Resp Med Rev.
2007;3:79–83.
51. Paranjothy S, Griffiths JD, Broughton HK, et al. Interventions at caesarean section for reducing the
risk of aspiration pneumonitis. Cochrane Database Syst Rev. 2014;(2):CD004943.
52. Ng A, Smith G. Gastroesophageal reflux and aspiration of gastric contents in anesthetic practice.
Anesth Analg. 2001;93:494–513.
53. Kluger MT, Short TG. Aspiration during anaesthesia: a review of 133 cases from the Australian
Anaesthetic Incident Monitoring Study (AIMS). Anaesthesia. 1999;54:19–26.
54. Wong CA, McCarthy RJ, Fitzgerald PC, et al. Gastric emptying of water in obese pregnant women at
term. Anesth Analg. 2007;105:751–755.
55. Landay MJ, Christensen EE, Bynum LJ. Pulmonary manifestations of acute aspiration of gastric
contents. AJR Am J Roentgenol. 1978;131:587–592.
56. Kjaer K, Comerford M, Kondilis L, et al. Oral sodium citrate increases nausea amongst elective
cesarean delivery patients. Can J Anaesth. 2006;53:776–780.
57. de Souza DG, Doar LH, Mehta SH, et al. Aspiration prophylaxis and rapid sequence induction for
elective cesarean delivery: time to reassess old dogma? Anesth Analg. 2010;110:1503–1505.
58. Han TH, Brimacombe J, Lee EJ. The laryngeal mask airway is effective (and probably safe) in
selected healthy parturients for elective cesarean section: a prospective study of 1067 cases. Can J
Anaesth. 2001;48:1117–1121.
59. Whitty JE. Cystic fibrosis in pregnancy. Clin Obstet Gynecol. 2010;53:369–376.
60. Budev MM, Arroliga AC, Emery S. Exacerbation of underlying pulmonary disease in pregnancy. Crit
Care Med. 2005;33(suppl 10):S313–S318.
61. Deighan M, Ash S, McMorrow R. Anaesthesia for parturients with severe cystic fibrosis: a case series.
Int J Obstet Anesth. 2014;23:75–79.
62. Cohen R, Talwar A, Efferen LS. Exacerbation of underlying pulmonary disease in pregnancy. Crit
Care Clin. 2004;20:713–730.
63. Muammar M, Marshall P, Wyatt H, et al. Caesarean section in a patient with cystic fibrosis. Int J
Obstet Anesth. 2005;14:70–73.
64. Huffmyer JL, Littlewood KE, Nemergut EC. Perioperative management of the adult with cystic
fibrosis. Anesth Analg. 2009;109:1949–1961.
65. Vos R, Ruttens D, Verleden SE, et al. Pregnancy after heart and lung transplantation. Best Pract Res
Clin Obstet Gynaecol. 2014;28:1146–1162.
66. Gyi KM, Hodson ME, Yacoub MY. Pregnancy in cystic fibrosis lung transplant recipients: case series
and review. J Cyst Fibros. 2006;5:171–175.
67. Thakrar MV, Morley K, Lordan JL, et al. Pregnancy after lung and heart-lung transplantation. J Heart
Lung Transplant. 2014;33:593–598.
68. Cardonick E, Moritz M, Armenti V. Pregnancy in patients with organ transplantation: a review. Obstet
Gynecol Surv. 2004;59:214–222.
69. Halpern SH, Srebrnjak M. Anesthesia for the pregnant patient with immunologic disorders. In: Suresh
M, Roanne L, Preston MD, et al, eds. Shnider and Levinson’s Anesthesia for Obstetrics. 5th ed.
Philadelphia, PA: Lippincott Williams & Wilkins; 2013:626–646.
70. Baker ER, Cardenas DD. Pregnancy in spinal cord injured women. Arch Phys Med Rehabil.
1996;77:501–507.
71. Gupta S, Singariya G. Kyphoscoliosis and pregnancy—a case report. Indian J Anaesth. 2004;48:215–
220.
72. Lapinsky SE, Tram C, Mehta S, et al. Restrictive lung disease in pregnancy. Chest. 2014;145:394–398.
73. Baydur A, Adkins RH, Milic-Emili J. Lung mechanics in individuals with spinal cord injury: effects of
injury level and posture. J Appl Physiol. 2001;90:405–411.
74. Duarte AG. ARDS in pregnancy. Clin Obstet Gynecol. 2014;57:862–870.
75. Cole DE, Taylor TL, McCullough DM, et al. Acute respiratory distress syndrome in pregnancy. Crit
Care Med. 2005;33(suppl 10):S269–S278.
76. Bandi VD, Munnur U, Matthay MA. Acute lung injury and acute respiratory distress syndrome in
pregnancy. Crit Care Clin. 2004;20:577–607.
77. Graves CR. Acute pulmonary complications during pregnancy. Clin Obstet Gynecol. 2002;45:369–
376.
78. Kenn S, Weber-Carstens S, Weizsaecker K, et al. Prone position for ARDS following blunt chest
trauma in late pregnancy. Int J Obstet Anesth. 2009;18:268–271.
79. Nair P, Davies AR, Beca J, et al. Extracorporeal membrane oxygenation for severe ARDS in pregnant
and postpartum women during the 2009 H1N1 pandemic. Intensive Care Med. 2011;37:648–654.
Obesity and Pregnancy
Brenda A. Bucklin and David R. Gambling
I. High-risk patient
A. Anesthetic challenges
B. Maternal comorbidities
C. Obstetric risk and outcomes
II. Obesity: definition and demographics
A. Consensus for definition of obesity
B. Body mass index
C. Increasing incidence of obesity
III. Physiologic changes of obesity and pregnancy
A. Pulmonary
B. Cardiovascular
C. Endocrine
D. Hypercoagulability
E. Gastrointestinal
IV. Anesthesia for the obese pregnant woman
A. Obesity and maternal mortality
B. Patient evaluation
C. General considerations
V. Implications for patients undergoing labor for vaginal delivery
A. Labor analgesia
B. Epidural analgesia/anesthesia in obese patients
C. Placement of neuraxial blocks
D. Placement of epidural catheter
E. Cephalad spread of neuraxial block
VI. Implications for patients undergoing cesarean delivery
A. General considerations
B. Cesarean delivery: maternal morbidity and mortality
C. Anesthetic plan
D. Neuraxial anesthesia
E. General anesthesia
F. Considerations for the operating room
G. Surgical considerations
VII. Postoperative care
A. General considerations
B. Pulmonary considerations
C. Postoperative monitoring
D. Thromboprophylaxis
VIII. Newborn
A. Preterm delivery
B. Fetal anomalies
C. Fetal macrosomia
D. Stillbirth and neonatal deaths
E. Neonatal care
IX. Pregnancy after bariatric surgery
X. Cost
Summary
KEYPOINTS
1. Obesity increases risk of adverse outcomes, including morbidity and
mortality.
2. Anesthesiology consultation is recommended for obese women
antepartum or “early in labor” to allow adequate time for development of
an anesthetic plan.
3. Neuraxial analgesia may precede the onset of labor or a patient’s request
for labor analgesia.
4. Preparation should include blood pressure monitoring and placement of
adequate intravenous access early in labor.
5. Blood products should be readily available.
6. Obese parturients are at risk for cesarean delivery (CD), especially
emergent CD.
7. Continuous catheter techniques are a better choice than single-injection
spinal anesthesia for CD.
8. Tracheal intubation, emergence, extubation, and recovery are critical
periods of anesthetic care in the obese parturient.
9. Multimodal analgesic techniques (e.g., nonsteroidal anti-inflammatory
agents) should be used to decrease total opioid requirements.
10. Communication with the obstetric team is essential for good care.
C. Endocrine
1. Type 2 DM is the paradigm of obesity-related disease. It is
often caused by obesity and will often resolve with weight
loss. Using a baseline BMI of 21, the Nurses’ Cohort Study
found that the risk of developing type 2 DM was increased
fivefold for BMI of 25, 35-fold for BMI of 30, and 93-fold for
BMI >35.2 Other groups have found similar trends of
increasing incidence and insulin use with increasing BMI.3
2. In pregnancy, the placenta secretes contrainsulin hormones
(e.g., human placental lactogen [hPL], human chronic
gonadotropin [hCG], steroids), and insulin resistance becomes
an increasing problem as pregnancy progresses. The
likelihood of developing gestational diabetes mellitus (GDM)
correlates well with prepregnancy BMI. Weiss et al.7 found
that 2.3% of normal-weight parturients developed GDM,
whereas 6.3% of obese and 9.5% of morbidly obese patients
developed GDM. The combination of preexisting insulin
resistance of obesity and the insulin resistance of pregnancy
likely leads to the large amounts of insulin required to achieve
glycemic control. The large insulin requirement, in turn, may
lead to excessive gestational weight gain. Therefore, a vicious
cycle of increasing insulin and weight gain ensues.
3. DM has significant adverse effects on the fetus (see
Chapter 23). Perinatal mortality in both type 1 and type 2 DM
is increased fourfold. The incidence of congenital anomalies
in offspring of mothers with DM is twice that seen in
nondiabetic pregnancies. There is a fourfold increase in neural
tube defects and threefold increase in congenital heart disease.
It is well accepted that maternal obesity is a risk factor for
fetal macrosomia but the cause is unclear. Some believe that
maternal DM and hyperinsulinemia leads to fetal macrosomia,
whereas others have found that maternal obesity without DM
may cause fetal macrosomia.29 Good glycemic control
reduces the risk of adverse perinatal outcomes. HbA1c levels
should be <7%.
D. Hypercoagulability
1. Venous thromboembolism (VTE) is a leading cause of
maternal mortality. The risk of VTE is five times higher in a
pregnant woman than in a nonpregnant woman of similar
age.30 The incidence of VTE in obese parturients is more
than twice that of nonobese controls. Obesity is an
independent risk factor for deep vein thrombosis (DVT).9 In
women younger than 40 years, the relative risk for DVT is
increased sixfold in obese versus nonobese patients. Several
reasons may account for the increased incidence of venous
thrombus formation:
a. Immobility
b. Increased lower extremity venous stasis—secondary to
increased inferior vena cava pressure as a result of
increased intraabdominal pressure
c. Vascular damage occurs with both vaginal deliveries and
CD.
d. Pregnancy-induced hypercoagulable state—increase in
fibrin and factors II, VII, and X and decrease in protein S
and fibrinolytic system
e. Decreased fibrinolytic activity (decreased in individuals
with a combination of hyperlipidemia and insulin
resistance) and increased fibrinogen levels in obesity are
also contributory. Fibrinogen levels increase in proportion
to increasing BMI.
f. Increased thromboxane production, secondary to
hyperlipidemia
g. Polycythemia leading to increased blood viscosity
2. Increased recognition and screening tools (i.e., Doppler
ultrasound) and increased use of thromboprophylaxis have
decreased the incidence of lower extremity venous thrombi.
Both unfractionated and low molecular weight heparin
(LMWH) are mainstays of thromboprophylaxis. An
anticoagulated patient provides a challenge for the
anesthesiologist. The major concern involves the increased
risk of epidural hematoma following placement of a neuraxial
blockade. Variations in the type of anticoagulant and dosing
regimen have led to different recommendations regarding the
appropriate timing for safely administering neuraxial
blockade.31
E. Gastrointestinal
1. The combination of obesity and pregnancy greatly increases
the risk for gastric content regurgitation and possibly
pulmonary aspiration. The frequency of GERD is strongly
correlated with increasing BMI in women.23 Physiologic
changes in normal pregnancy include a decrease in lower
esophageal sphincter tone. In laboring women, gastric
motility decreases and emptying may cease completely.
Parenteral and neuraxial opioid administration contribute to
decreased gastric motility. Obese patients may have larger
gastric volumes and lower gastric pH.32 Obese parturients are
less mobile, which also increases the risk of aspiration. The
increased risk of pulmonary aspiration necessitates strict
nothing by mouth guidelines and the timely administration
of nonparticulate antacids, H2-receptor antagonists, and/or
metoclopramide before induction of anesthesia.33 Because
obese parturients have lower gastroesophageal junction tone
and an increased risk of airway difficulty, their risk for
aspiration is increased.
2. Fatty liver is commonly observed in obese individuals with an
estimated incidence of 60% to 90% among morbidly obese
patients.34 It is the most common cause for elevated
transaminases in the United States. The risk of steatosis
increases with type 2 DM, hyperlipidemia, and/or
hypertension. Unlike the clinically significant microvesicular
steatosis found in fatty liver of pregnancy, the
macrovesicular fatty liver associated with obesity is
usually a benign process. In rare cases, fatty liver associated
with obesity may progress to fibrosis, cirrhosis, and liver
failure. If liver function is compromised, evaluation for
possible coagulopathy must be considered. Decreased liver
function also results in altered drug metabolism and
clearance.
IV. Anesthesia for the obese pregnant woman
A. Obesity and maternal mortality. In the 2006 to 2008
Confidential Enquiries into Maternal Deaths and Child Health in
the United Kingdom, 49% (n = 227) of the women who died from
either direct (e.g., thromboembolic disease, preeclampsia,
hemorrhage) or indirect (e.g., cardiac disease) causes and for
whom the BMI was known were either overweight or obese.13
B. Patient evaluation. Both the physician and the obese parturient
benefit from antenatal anesthetic evaluation. The American
College of Obstetricians and Gynecologists (ACOG) recommends
anesthesiology consultation for obese women antepartum or
“early in labor” to allow adequate time for development of an
anesthetic plan.5 Unfortunately, the availability of antenatal
anesthesia clinics is scarce—approximately 30% in a UK
survey.35 Manpower and financial constraints are factors that
limit availability of antenatal clinics. However, even without a
defined clinic, a sensitive discussion with the patient and her
support group as soon as she arrives on the labor and delivery unit
should include a dialogue about the potential difficulty of
initiating labor analgesia and risk of failure as well as the goals
for patient safety. Although surveys suggest that these discussions
are rare because of concerns of offending the patient,36,37 failure
to address these concerns can contribute to unrealistic patient
expectations or incomplete informed consent. Because obesity is
a social disability and is stigmatized by society, all members of
the health care team should be sensitive in their interactions with
obese parturients.
C. General considerations. Besides the technical challenges in
airway management and placement of neuraxial blocks, there
are associated medical problems and technical considerations.
Obese women should be screened for glucose intolerance early in
pregnancy. BP should be properly measured with correct cuff
size. If this is not possible, then an arterial line should be placed.
Accurate assessment of gestational age should be determined by
serial ultrasonographic examinations. Patients should be made
aware of the increased difficulty in establishing analgesia and
anesthesia, and early neuraxial analgesia should be encouraged.
Venous access may be difficult to obtain and extravasation of an
intravenous (IV) cannula can be easily overlooked. Ultrasound-
guided IV cannulation may facilitate venous access. Obese
parturients have a greater potential for blood loss during CD and
the postpartum period, so well-functioning, large-bore IV access
is essential. Patients need to be informed of the potential need for
central venous access, both for hemodynamic monitoring and
reliable IV access. Blood products also need to be readily
available because of the increased risk of uterine atony and
postpartum hemorrhage.
CLINICAL PEARL Preparation should include BP monitoring
and placement of adequate IV access early in labor. Blood products
also need to be readily available.
D. Thromboprophylaxis
1. Thromboprophylaxis is essential to prevent venous
thrombosis and pulmonary embolism.69 The obese patient is
at especially high risk for perioperative thrombotic events.3,52
They are less mobile due to a combination of obesity and
surgery, and fibrinolytic activity is markedly decreased in
individuals because of a combination of hyperlipidemia and
insulin resistance. Hyperlipidemia may also increase
thromboxane production leading to a greater coagulable state.
Thromboembolic deterrent stockings (TEDSs), LMWH,
adequate hydration, early ambulation, and physical
therapy have all been shown to decrease the incidence of
thromboembolic events. It is essential to communicate with
the surgeon regarding use of LMWH to prevent iatrogenic
mishaps. According to the Third Consensus Conference on
Neuraxial Anesthesia and Anticoagulation,31
a. Patients with postoperative initiation of LMWH
thromboprophylaxis may safely undergo single-injection
and continuous catheter techniques. Management is based
on total daily dose, timing of the first postoperative dose,
and dosing schedule.
(1) Twice-daily dosing. This dosage regimen may be
associated with an increased risk of spinal hematoma.
The first dose of LMWH should be administered
no earlier than 24 hours postoperatively,
regardless of anesthetic technique, and only in the
presence of adequate (surgical) hemostasis.
Indwelling catheters should be removed before
initiation of LMWH thromboprophylaxis. If a
continuous technique is selected, the epidural catheter
may be left in overnight and removed the following
day, with the first dose of LMWH administered at
least 2 hours after catheter removal.
(2) Once-daily dosing. This dosing regimen
approximates the European application. The first
postoperative LMWH dose should be administered 6
to 8 hours postoperatively. The second postoperative
dose should occur no sooner than 24 hours after the
first dose. Indwelling neuraxial catheters may be
safely maintained. However, the catheter should be
removed a minimum of 10 to 12 hours after the last
dose of LMWH. Subsequent LMWH dosing should
occur a minimum of 2 hours after catheter removal.
VIII. Newborn
A. Preterm delivery. Some studies have indicated that premature
infants (deliveries <38 weeks’ gestation) are less likely in obese
parturients.4,12 However, a large population-based cohort study
determined that the risk of preterm, especially extreme preterm
delivery, was increased in overweight and obese women.73
B. Fetal anomalies. Maternal obesity increases the likelihood of
macrosomia and other fetal and perinatal issues.1,51 Macrosomia
causes an increased risk of birth trauma. Most studies also
indicate an increase in incidence of congenital malformations,
including omphalocele, cardiac anomalies, and spina bifida or
other neural tube defects (independent of maternal age, education,
smoking status, alcohol use, and other known risk factors for
neural tube defect). Because the images on fetal ultrasonography
are often suboptimal in obese parturients, there is a higher risk of
failure to diagnose fetal anomalies.13
C. Fetal macrosomia. Macrosomia is defined as a term infant
weighing >4,000 g. An obese parturient has twice the likelihood
of a newborn being >90th percentile in weight versus nonobese
parturients.3,4 The Pedersen hypothesis, maternal hyperglycemia
causing hyperinsulinemia in the fetus resulting in enhanced fetal
growth, has been cited as the reason for fetal macrosomia.
However, studies have found that obese, mothers without diabetes
are more likely to have macrosomic babies compared to
nonobese, mothers with diabetes.29 Therefore, it appears that the
offspring of obese mothers develop fetal macrosomia through an
unknown mechanism. Macrosomic newborns are at increased
risk for birth trauma (e.g., shoulder dystocia and Erb palsy),
asphyxia, meconium aspiration, antepartum stillbirth,
hypoglycemia, hypocalcemia, and erythrocytosis. A term infant
weighing >4,500 g has a more than threefold increase in mortality
compared to an infant of average birth weight.
D. Stillbirth and neonatal deaths. Stillbirth is defined as delivery
of a dead fetus at 28 weeks’ gestation or later. Neonatal death is
defined as death of a live newborn on days 1 to 28 following
delivery.74 A recent meta-analysis and systematic review
evaluating maternal BMI and risk of perinatal mortality
determined that “modest” increases in maternal BMI increased
the risk of fetal death, stillbirth, neonatal, perinatal, and infant
deaths.75 Maternal prepregnancy obesity more than doubles the
risk of stillbirth and neonatal deaths.8,12,25,52 A study that
separated nulliparous women from parous women found that in
parous obese parturients, there was no increase in early neonatal
deaths.25 Cnattingius et al.25 reported an increased risk of late
fetal death among obese women that correlated with
prepregnancy BMI and was independent of the presence or
absence of hypertensive disorders and diabetes. This finding is
contrary to others who found that uncomplicated obese
parturients have no additional risk of poor fetal outcome.12
However, these authors found that obese mothers with
preeclampsia, DM, hypertension, or advanced age had a higher
incidence of perinatal death.76
E. Neonatal care. A recent cohort study of women with BMI >40
determined that the risk of newborn injury to the peripheral
nervous system and skeleton was increased but there was also an
increased risk of respiratory distress syndrome, bacterial sepsis,
convulsions, hypoglycemia in newborns born to mothers with a
BMI >40. In neonates born to obese mothers, admission to a
neonatal intensive care unit was 4 to 10 times greater compared to
neonates born to mothers of normal weight.51 Most of the
admissions were for temperature regulation, glucose monitoring,
and assistance in feeding. Others have noted that neonates born to
obese diabetic parturients have the greatest risk of poor
outcomes.77 Difficulties in monitoring FHR during labor may
also lead to a delayed diagnosis of nonreassuring FHRs.
Furthermore, infants born to obese mothers are less likely to be
breastfed at hospital discharge. This may be related to the
decreased prolactin response to suckling leading to poor
lactogenesis and initiation of breastfeeding or social issues.4
IX. Pregnancy after bariatric surgery
A systematic review on bariatric surgery among women of
reproductive age determined that although rates of adverse maternal
and neonatal outcomes could be lower in women who become
pregnant after bariatric surgery compared with rates in obese pregnant
women, more data are needed.78 In general, a history of bariatric
surgery is not associated with adverse perinatal outcome. However,
these patients should be assessed to rule out cardiomyopathy and
sleep apnea. Maternal anemia may be a result of nutritional
deficiencies following gastric bypass. Iron-deficiency anemia
(microcytic) may result from poor iron absorption. Folate and B12
deficiency may result from poor absorption resulting in macrocytic
anemia. Maternal nutritional deficiencies have resulted in intrauterine
growth restriction12 and neural tube defects. Gastrointestinal bleeding
during pregnancy has been reported following gastric band erosion
(vertical-banded gastroplasty).67
X. Cost
The cost of care for an obese parturient is significantly greater than
that of a normal-weight parturient69,79 and increases in direct
proportion with the BMI.1,12 Obesity predisposes parturients to
hypertension, preeclampsia, DM, insulin use, postpartum
complications, and increased hospitalization. In patients with BMI
>35, there is a 26-fold increase in outpatient visits and two-to fourfold
increase in hospitalizations.1 Other studies have found an increased
use of antepartum services and hospitalizations too. Pathi et al.12
determined that fetal monitoring, ultrasonography, and medical
management of diabetes and hypertension were all increased in this
population. The risk of complications during labor is higher and
includes an increased CD rate. The morbidly obese parturient has an
increased incidence of postoperative complications and antepartum
medical disease, and these factors contribute to longer
hospitalization.12 In one study, the cost of care for a morbidly obese
parturient was estimated at three times that of a normal-weight
parturient. Others have estimated a five-fold to ten-fold increase
depending on the degree of obesity.1 Although no studies evaluated
neonatal costs, the decrease in preterm delivery in the obese
population may offset some of the maternal health costs; however, the
increase in perinatal morbidity, mostly due to maternal DM will
increase medical costs.12
SUMMARY
The anesthesiologist faces significant challenges when caring for the morbidly
obese parturient. Obesity is a significant risk factor for both maternal and
perinatal morbidity and mortality. Overall, the literature suggests that obese
parturients have a 14% to 25% incidence of preeclampsia, a 6% to 14%
incidence of GDM, and a 30% to 47% chance of requiring a CD.7,9 These
numbers are only estimates, and many studies correlate increasing complications
with increasing maternal BMI. The obese parturient may have concurrent
medical problems that need to be addressed. The anesthesiologist needs to
anticipate difficulties during labor and CD and must plan for them accordingly.
Communication with the obstetric team is essential for good care. Every effort
should be made to provide the best possible outcome for the obese parturient and
her fetus, but the life of the mother should never be endangered to save a
compromised fetus.
REFERENCES
1. Galtier-Dereure F, Montpeyroux F, Boulot P, et al. Weight excess before pregnancy: complications and
cost. Int J Obes Relat Metab Disord. 1995;19:443–448.
2. Aubert B, Boutigny D, De Bonis I, et al. Measurement of CP-violating asymmetries in B0 decays to
CP eigenstates. Phys Rev Lett. 2001;86:2515–2522.
3. Johnson SR, Kolberg BH, Varner MW, et al. Maternal obesity and pregnancy. Surg Gynecol Obstet.
1987;164:431–437.
4. Sebire NJ, Jolly M, Harris JP, et al. Maternal obesity and pregnancy outcome: a study of 287,213
pregnancies in London. Int J Obes Relat Metab Disord. 2001;25:1175–1182.
5. American College of Obstetricians and Gynecologists. ACOG Committee Opinion No. 549: obesity in
pregnancy. Obstet Gynecol. 2013;121:213–217.
6. Mhyre JM, Bateman BT, Leffert LR. Influence of patient comorbidities on the risk of near-miss
maternal morbidity or mortality. Anesthesiology. 2011;115:963–972.
7. Weiss JL, Malone FD, Emig D, et al. Obesity, obstetric complications and cesarean delivery rate—a
population-based screening study. Am J Obstet Gynecol. 2004;190:1091–1097.
8. Cedergren MI. Maternal morbid obesity and the risk of adverse pregnancy outcome. Obstet Gynecol.
2004;103:219–224.
9. Saravanakumar K, Rao SG, Cooper GM. Obesity and obstetric anaesthesia. Anaesthesia. 2006;61:36–
48.
10. Ranta P, Jouppila P, Spalding M, et al. The effect of maternal obesity on labour and labour pain.
Anaesthesia. 1995;50: 322–326.
11. Durnwald CP, Ehrenberg HM, Mercer BM. The impact of maternal obesity and weight gain on vaginal
birth after cesarean section success. Am J Obstet Gynecol. 2004;191:954–957.
12. Pathi A, Esen U, Hildreth A. A comparison of complications of pregnancy and delivery in morbidly
obese and non-obese women. J Obstet Gynaecol. 2006;26:527–530.
13. Cantwell R, Clutton-Brock T, Cooper G, et al. Saving mothers’ lives: reviewing maternal deaths to
make motherhood safer: 2006–2008. The Eighth Report of the Confidential Enquiries into Maternal
Deaths in the United Kingdom. BJOG. 2011;118(suppl 1):1–203.
14. Rassmusen KM, Yaktine AL, eds; for the Institute of Medicine. Weight Gain During Pregnancy:
Reexamining the Guidelines. Washington, DC: National Academies Press; 2009.
15. Flegal KM, Carroll MD, Kit BK, et al. Prevalence of obesity and trends in the distribution of body
mass index among US adults, 1999–2010. JAMA. 2012;307:491–497.
16. Unterborn J. Pulmonary function testing in obesity, pregnancy, and extremes of body habitus. Clin
Chest Med. 2001;22:759–767.
17. von Ungern-Sternberg BS, Regli A, Bucher E, et al. Impact of spinal anaesthesia and obesity on
maternal respiratory function during elective caesarean section. Anaesthesia. 2004;59:743–749.
18. Pataka A, Daskalopoulou E, Kalamaras G, et al. Evaluation of five different questionnaires for
assessing sleep apnea syndrome in a sleep clinic. Sleep Med. 2014;15:776–781.
19. Ankichetty SP, Angle P, Joselyn AS, et al. Anesthetic considerations of parturients with obesity and
obstructive sleep apnea. J Anaesthesiol Clin Pharmacol. 2012;28:436–443.
20. Louis JM, Mogos MF, Salemi JL, et al. Obstructive sleep apnea and severe maternal-infant
morbidity/mortality in the United States, 1998–2009. Sleep. 2014;37:843–849.
21. Macavei VM, Spurling KJ, Loft J, et al. Diagnostic predictors of obesity-hypoventilation syndrome in
patients suspected of having sleep disordered breathing. J Clin Sleep Med. 2013;9:879–884.
22. Tomoda S, Tamura T, Sudo Y, et al. Effects of obesity on pregnant women: maternal hemodynamic
change. Am J Perinatol. 1996;13:73–78.
23. Jacobson BC, Somers SC, Fuchs CS, et al. Body-mass index and symptoms of gastroesophageal reflux
in women. N Engl J Med. 2006;354:2340–2348.
24. Bateman BT, Bansil P, Hernandez-Diaz S, et al. Prevalence, trends, and outcomes of chronic
hypertension: a nationwide sample of delivery admissions. Am J Obstet Gynecol. 2012;206:134.e1–
134.e8.
25. Cnattingius S, Bergström R, Lipworth L, et al. Prepregnancy weight and the risk of adverse pregnancy
outcomes. N Engl J Med. 1998;338:147–152.
26. Kenchaiah S, Evans JC, Levy D, et al. Obesity and the risk of heart failure. N Engl J Med.
2002;347:305–313.
27. Alpert MA. Obesity cardiomyopathy: pathophysiology and evolution of the clinical syndrome. Amer J
Med Sci. 2001;321:225–236.
28. Tsueda K, Debrand M, Zeok SS, et al. Obesity supine death syndrome: reports of two morbidly obese
patients. Anesth Analg. 1979;58:345–347.
29. Green JR, Schumacher LB, Pawson IG, et al. Influence of maternal body habitus and glucose
tolerance on birth weight. Obstet Gynecol. 1991;78:235–240.
30. Toglia MR, Weg JG. Venous thromboembolism during pregnancy. N Engl J Med. 1996;335:108–114.
31. Horlocker TT, Wedel DJ, Rowlingson JC, et al. Executive summary: regional anesthesia in the patient
receiving antithrombotic or thrombolytic therapy: American Society of Regional Anesthesia and Pain
Medicine Evidence-Based Guidelines (Third Edition). Reg Anesth Pain Med. 2010;35:102–105.
32. Wong CA, McCarthy RJ, Fitzgerald PC, et al. Gastric emptying of water in obese pregnant women at
term. Anesth Analg. 2007;105:751–755.
33. American Society of Anesthesiologists Task Force on Obstetric Anesthesia. Practice guidelines for
obstetric anesthesia: an updated report by the American Society of Anesthesiologists Task Force on
Obstetric Anesthesia. Anesthesiology. 2007;106:843–863.
34. Fong DG, Nehra V, Lindor KD, et al. Metabolic and nutritional considerations in nonalcoholic fatty
liver. Hepatology. 2000;32:3–10.
35. Rai MR, Lua SH, Popat M, et al. Antenatal anaesthetic assessment of high-risk pregnancy: a survey of
UK practice. Int J Obstet Anesth. 2005;14:219–222.
36. Mhyre JM, Greenfield ML, Polley LS. Survey of obstetric providers’ views on the anesthetic risks of
maternal obesity. Int J Obstet Anesth. 2007;16:316–322.
37. Wen LM, Baur LA, Simpson JM, et al. Mothers’ awareness of their weight status and concern about
their children being overweight: findings from first-time mothers in south-west Sydney. Aust N Z J
Public Health. 2010;34:293–297.
38. Tonidandel A, Booth J, D’Angelo R, et al. Anesthetic and obstetric outcomes in morbidly obese
parturients: a 20-year follow-up retrospective cohort study. Int J Obstet Anesth. 2014;23:357–364.
39. Dresner M, Brocklesby J, Bamber J. Audit of the influence of body mass index on the performance of
epidural analgesia in labour and the subsequent mode of delivery. BJOG. 2006;113:1178–1181.
40. Vricella LK, Louis JM, Mercer BM, et al. Impact of morbid obesity on epidural anesthesia
complications in labor. Am J Obstet Gynecol. 2011;205:370.e1–370.e6.
41. Bloom SL, Spong CY, Weiner SJ, et al. Complications of anesthesia for cesarean delivery. Obstet
Gynecol. 2005;106:281–287.
42. D’Angelo R, Smiley RM, Riley ET, et al. Serious complications related to obstetric anesthesia: the
serious complication repository project of the Society for Obstetric Anesthesia and Perinatology.
Anesthesiology. 2014;120:1505–1512
43. Bauer ME, Kountanis JA, Tsen LC, et al. Risk factors for failed conversion of labor epidural analgesia
to cesarean delivery anesthesia: a systematic review and meta-analysis of observational trials. Int J
Obstet Anesth. 2012;21:294–309.
44. Clinkscales CP, Greenfield ML, Vanarase M, et al. An observational study of the relationship between
lumbar epidural space depth and body mass index in Michigan parturients. Int J Obstet Anesth.
2007;16:323–327.
45. Marroquin BM, Fecho K, Salo-Coombs V, et al. Can parturients identify the midline during neuraxial
block placement? J Clin Anesth. 2011;23:3–6.
46. Arzola C, Davies S, Rofaeel A, et al. Ultrasound using the transverse approach to the lumbar spine
provides reliable landmarks for labor epidurals. Anesth Analg. 2007;104:1188–1192.
47. Bahar M, Chanimov M, Cohen ML, et al. The lateral recumbent head-down position decreases the
incidence of epidural venous puncture during catheter insertion in obese parturients. Can J Anaesth.
2004;51:577–580.
48. Hamilton CL, Riley ET, Cohen SE. Changes in the position of epidural catheters associated with
patient movement. Anesthesiology. 1997;86:778–784.
49. Hodgkinson R, Husain FJ. Obesity and the cephalad spread of analgesia following epidural
administration of bupivacaine for cesarean section. Anesth Analg. 1980;59:89–92.
50. Taivainen T, Tuominen M, Rosenberg PH. Influence of obesity on the spread of spinal analgesia after
injection of plain 0.5% bupivacaine at the L3-4 or L4-5 interspace. Br J Anaesth. 1990;64:542–546.
51. Barau G, Robillard PY, Hulsey TC, et al. Linear association between maternal pre-pregnancy body
mass index and risk of caesarean section in term deliveries. BJOG. 2006;113:1173–1177.
52. Yu CK, Teoh TG, Robinson S. Obesity in pregnancy. BJOG. 2006;113:1117–1125.
53. Mourad M, Silverstein M, Bender S, et al. The effect of maternal obesity on outcomes in patients
undergoing tertiary or higher cesarean delivery. J Matern Fetal Neonatal Med. 2015;28:989–993.
54. Hawkins JL, Chang J, Palmer SK, et al. Anesthesia-related maternal mortality in the United States:
1979–2002. Obstet Gynecol. 2011;117:69–74.
55. Lee Y, Balki M, Parkes R, et al. Dose requirement of intrathecal bupivacaine for cesarean delivery is
similar in obese and normal weight women. Rev Bras Anestesiol. 2009;59:674–683.
56. Carvalho B, Collins J, Drover DR, et al. ED(50) and ED(95) of intrathecal bupivacaine in morbidly
obese patients undergoing cesarean delivery. Anesthesiology. 2011;114:529–535.
57. Pan PH, Bogard TD, Owen MD. Incidence and characteristics of failures in obstetric neuraxial
analgesia and anesthesia: a retrospective analysis of 19,259 deliveries. Int J Obstet Anesth.
2004;13:227–233.
58. Gambling D, Berkowitz J, Farrell TR, et al. A randomized controlled comparison of epidural analgesia
and combined spinal-epidural analgesia in a private practice setting: pain scores during first and
second stages of labor and at delivery. Anesth Analg. 2013;116:636–643.
59. Vricella LK, Louis JM, Mercer BM, et al. Anesthesia complications during scheduled cesarean
delivery for morbidly obese women. Am J Obstet Gynecol. 2010;203:276.e1–e5.
60. Kodali BS, Chandrasekhar S, Bulich LN, et al. Airway changes during labor and delivery.
Anesthesiology. 2008;108:357–362.
61. Collins JS, Lemmens HJ, Brodsky JB, et al. Laryngoscopy and morbid obesity: a comparison of the
“sniff” and “ramped” positions. Obes Surg. 2004;14:1171–1175.
62. Quinn AC, Milne D, Columb M, et al. Failed tracheal intubation in obstetric anaesthesia: 2 yr national
case-control study in the UK. Br J Anaesth. 2013;110:74–80.
63. Dhonneur G, Ndoko S, Amathieu R, et al. Tracheal intubation using the Airtraq in morbid obese
patients undergoing emergency cesarean delivery. Anesthesiology. 2007;106:629–630.
64. Han TH, Brimacombe J, Lee EJ, et al. The laryngeal mask airway is effective (and probably safe) in
selected healthy parturients for elective cesarean section: a prospective study of 1067 cases. Can J
Anaesth. 2001;48:1117–1121.
65. Apfelbaum JL, Hagberg CA, Caplan RA, et al. Practice guidelines for management of the difficult
airway: an updated report by the American Society of Anesthesiologists Task Force on Management
of the Difficult Airway. Anesthesiology. 2013;118:251–270.
66. Mushambi MC, Kinsella SM, Popat M, et al. Obstetric Anaesthetists’ Association and Difficult
Airway Society guidelines for the management of difficult and failed tracheal intubation in obstetrics.
Anaesthesia. 2015;70:1286–1306.
67. Mhyre JM, Riesner MN, Polley LS, et al. A series of anesthesia-related maternal deaths in Michigan,
1985–2003. Anesthesiology. 2007;106:1096–1104.
68. Thornton YS. Caesarean delivery and celiotomy using panniculus retraction in the morbidly obese
patient. J Am Coll Surg. 2001;193:458–461.
69. Alexander CI, Liston WA. Operating on the obese woman—a review. BJOG. 2006;113:1167–1172.
70. American Society of Anesthesiologists Task Force on Neuraxial Opioids, Horlocker TT, Burton AW,
et al. Practice guidelines for the prevention, detection, and management of respiratory depression
associated with neuraxial opioid administration. Anesthesiology. 2009;110:218–230.
71. American Society of Anesthesiologists Task Force on Perioperative Management of Patients with
Obstructive Sleep Apnea. Practice guidelines for the perioperative management of patients with
obstructive sleep apnea: an updated report by the American Society of Anesthesiologists Task Force
on Perioperative Management of Patients with Obstructive Sleep Apnea. Anesthesiology.
2014;120:268–286.
72. Loubert C, Fernando R. Cesarean delivery in the obese parturient: anesthetic considerations. Women’s
Health. 2011;7:163–179.
73. Cnattingius S, Villamor E, Johansson S, et al. Maternal obesity and risk of preterm delivery. JAMA.
2013;309:2362–2370.
74. Kristensen J, Vestergaard M, Wisborg K, et al. Pre-pregnancy weight and the risk of stillbirth and
neonatal death. BJOG. 2005;112:403–408.
75. Aune D, Saugstad OD, Henriksen T, et al. Maternal body mass index and the risk of fetal death,
stillbirth, and infant death: a systematic review and meta-analysis. JAMA. 2014;311:1536–1546.
76. Rahaman J, Narayansingh GV, Roopnarinesingh S. Fetal outcome among obese parturients. Int J
Gynaecol Obstet. 1990;31:227–230.
77. Baron CM, Girling LG, Mathieson AL, et al. Obstetrical and neonatal outcomes in obese parturients. J
Matern Fetal Neonatal Med. 2010;23:906–913.
78. Maggard MA, Yermilov I, Li Z, et al. Pregnancy and fertility following bariatric surgery: a systematic
review. JAMA. 2008;300:2286–2296.
79. Chu SY, Bachman DJ, Callaghan WM, et al. Association between obesity during pregnancy and
increased use of health care. N Engl J Med. 2008;358:1444–1453.
Trauma in the Obstetric Patient
Hen Y. Sela, Lior Drukker, and Sharon Einav
I. Introduction
II. Epidemiology of trauma
A. Epidemiology of obstetric complications during trauma
III. General treatment guidelines
A. Always prefer the mother
B. Adhere to standard care protocols
C. Perform trauma surveys, a pregnancy workup, and assess
fetal viability
IV. Limitations in assessing severity of maternal injury
A. Severity of injury
B. Advanced trauma life support training and use of vital signs
C. Implications of the physiologic changes of pregnancy
D. Clinical assessment of patient severity
V. Principles of radiologic assessment
A. Ultrasound imaging
B. Standard radiology and computed tomography scanning
C. Magnetic resonance imaging
VI. Clinical and test findings versuspregnancy outcome
A. Prenatal maternal injury
B. Minor injuries
C. Kleihauer-Betke testing
D. Flow cytometry
VII. Anesthetic considerations
A. Airway management
B. Anesthetic induction and maintenance
VIII. Specific mechanisms of injury
A. Motor vehicle accidents
B. Falls
C. Domestic violence
D. Burns
E. Penetrating injury
IX. Salvage therapies
A. Extracorporeal membrane oxygenation
B. Perimortem cesarean delivery
Summary
KEYPOINTS
1. Prenatal maternal injury is associated with an increased risk of adverse
pregnancy outcome. Lack of maternal belting is consistently cited as a
risk for poor pregnancy outcome in motor vehicle accidents.
2. Because information regarding management of specific types of trauma
during pregnancy is limited, providers must extrapolate from other
clinical situations (e.g., peripartum hemorrhage) and remain open to the
likelihood that physiologic instability may result not only from
overlooked and/or untreated injuries, but also from baseline differences in
physiology stemming from the presence of pregnancy or from obstetric
complications.
3. Management of maternal trauma is driven primarily by maternal
condition.
4. Because fetal heart rate monitoring may provide early indication of
maternal distress, the fetus should be monitored both prior to and
immediately after surgery whenever possible. Routinely (twice daily if
possible) monitoring is recommended for the duration of maternal
admission as well.
5. The principles guiding care of the pregnant patient with trauma are the
same as in the parturient without trauma: respect toward parental
opinions, which may differ from those of the treating staff, placement of
greater emphasis on fetal/neonatal condition than on gestational age after
24 weeks gestation.
6. Delivery should be considered in an initially unstable patient only if
maternal instability results from a complication of the pregnancy itself
(e.g., hemorrhage from placental abruption).
7. Standard advanced trauma life support (ATLS) shock categories may be
inadequate for classifying the severity of hemorrhage in the pregnant
population.
8. Ultrasound is the imaging mode of choice for all pregnant women.
9. The risk of fetal teratogenesis following exposure to radiation is greatest
during the first trimester of pregnancy.
10. Anesthetic management of the injured pregnant woman should be
directed toward optimization of maternal oxygenation and perfusion.
11. The respiratory reserves of pregnant women are limited. Decisions
regarding the timing of tracheal intubation must balance the increased
likelihood of airway management complications and the increased risk of
maternal/fetal compromise.
12. Regardless of the hospital location of the pregnant patient with trauma, a
birthing kit and heated incubator should always be on location.
13. Perimortem cesarean delivery (PMCD) may be instituted for maternal
salvage, provided aortocaval compression (secondary to the presence of
the gravid uterus) is considered a major contributor to maternal
cardiovascular collapse.
14. The American Heart Association recommends initiation of PMCD within
4 minutes of maternal nontraumatic resuscitation without return of
spontaneous circulation.
I. Introduction
Approximately 1 in 15 pregnant women are involved in some type of
trauma, most commonly a motor vehicle accident (MVA) or a fall.
Health care providers must be able to perform a complete and
thorough trauma workup while evaluating mother and child for life-
threatening obstetric complications. Differences between maternal
physiology and the nonpregnant female physiology challenge
provider ability to distinguish between normal and abnormal clinical
and laboratory findings. Of particular concern is the complexity of
balancing the management of two endangered lives simultaneously.
The presence of the pregnancy itself diverts attention away from the
main trauma victim—the mother. When the fetus is considered viable,
additional challenges present in the form of questions regarding
treatment and salvage priorities.
Appropriate trauma care of the injured pregnant woman entails a
coordinated multidisciplinary effort, which should ideally include
providers with expertise in emergency medicine, trauma surgery,
maternal–fetal medicine, and/or obstetrics, and anesthesiology and
critical care. All providers should act in accordance with accepted
trauma guidelines, which prioritize maternal airway management and
support of breathing and circulation. All should also follow standard
protocols for assessment and management of trauma during
pregnancy.
The current chapter reviews the epidemiology of maternal trauma,
the unique challenges and ethical issues likely to occur during trauma
workup and management in this clinical situation, as well as
recommendations for treatment of the injured pregnant woman. It also
discusses the implications of specific trauma mechanisms and their
associated injuries during pregnancy.
II. Epidemiology of trauma
Trauma complicates approximately 3% to 12% of all pregnancies.1,2
It constitutes a major cause of both maternal2,3 and fetal4–6 mortality.
The incidence of maternal death from trauma is currently estimated to
be 1.4 per 100,000 pregnancies4 and fetal death after maternal trauma
is more than double (3.7 per 100,000 live births).4,5 A particularly
high incidence of fetal death (9.3 per 100,000 live births) has been
observed in trauma cases involving teen mothers (15 to 19 years old)5
possibly due to more severe injury.7 Maternal demise, placental
abruption, maternal shock, and uterine rupture are the most common
risk factors for fetal death.5
Pregnant women are more likely to seek help following trauma
than nonpregnant women,8 and pregnancy at a gestational age greater
than 20 weeks is an indication for transporting the mother to a trauma
center.9 Notwithstanding, the exact prevalence of trauma-related
morbidity and mortality remains unknown; most data are
retrospective, coding systems differ, trauma registries do not include
obstetric information, and some types of injuries are
underreported.2,5,10
Minor trauma is generally much more common than major trauma,
and trauma during pregnancy is no exception. Only 6% of injured
pregnant women are hospitalized, which suggests that most pregnant
women involved in trauma do not sustain severe injuries6; three-
quarters of those hospitalized (77.9%) remain in hospital for no more
than 1 day.8 Major maternal injury occurs in 23 per 100,000
pregnancies. Centers treating hospitalized pregnant women after
trauma report that 12.5% to 19% are severely injured.11–13 Among
these cases, maternal mortality approximates 5.6%.4 MVA, domestic
violence, and assaults are the leading causes of major maternal
trauma,2 whereas falls, burns, homicide, suicide, thermal injuries,
toxic exposure, and drowning account for the remainder.7 Thus,
unintentional injury accounts for most major trauma occurring during
pregnancy. A recent systemic review calculated the approximate
incidence of injuries during pregnancies resulting in live births
according to mechanism; other than domestic violence (incidence
8,307 per 100,000), most types of trauma seem to occur less
frequently among pregnant women than among nonpregnant women.
The incidence of MVAs was estimated at 207 per 100,000, that of
falls 49 per 100,000, toxic exposure 25.8 per 100,000, homicide 2.9
per 100,000, and suicide 2 per 100,000 live births.2
CLINICAL PEARL MVA, domestic violence, and assaults are
the leading causes of major maternal trauma, whereas falls, burns,
homicide, suicide, thermal injuries, toxic exposure, and drowning
account for the remainder.
SUMMARY
Although 1:15 pregnant women are involved in trauma, data on maternal trauma
are lacking. Injury is usually unintentional (e.g., MVA or fall), but health care
providers must also remain alert to the option of intentional injury (i.e., domestic
violence). Appropriate trauma care of the injured pregnant woman requires a
coordinated multidisciplinary effort of professionals acting within accepted
trauma guidelines. Maternal care should be prioritized over treatment of the
fetus. A stable intrauterine environment remains optimal for all but the term
fetus. Thus, maternal and fetal interests generally remain aligned.
Current treatment guidelines for injured pregnant women are generally in
keeping with standard trauma guidelines. The presence of a pregnancy can be
distracting to the trauma team. Therefore, attention should be directed to
completion of maternal primary and secondary surveys before initiating
pregnancy workup and assessing fetal viability. Standard ATLS shock categories
may be inadequate for classifying the severity of hemorrhage in the pregnant
population. Radiologic assessment should follow maternal indications despite
the risk of fetal teratogenesis.
Pregnancy workup includes maternal pelvic examination, and ultrasound and
vaginal examinations (except in mild trauma or placenta previa). Placental
abruption is the most common obstetric complication. Absence of typical
symptoms and signs of abruption does not necessarily rule out occult
hemorrhage from the placenta, particularly in an obtunded patient. CT imaging
and MRI can assist in diagnosing placental abruption. Premature rupture of
membranes, uterine rupture, and PTD are also possible complications. Thus,
regardless of maternal location, a birthing kit should always be kept at hand and
the relevant disciplines notified of patient location.
Fetal monitoring is indicated after maternal workup and stabilization if fetal
gestational age exceeds the limits of viability. Nonreassuring findings may
indicate either maternal instability or fetal distress. Flow cytometry may be used
to seek transplacental fetomaternal transfusion.
Delivery should be considered for maternal instability resulting from a
complication of the pregnancy itself (e.g., hemorrhage from placental abruption)
or for fetal signs of distress despite maternal stability. Antenatal steroids may be
administered if early delivery is being discussed. Anesthesia guidelines are
similar for all pregnant patients; maintenance of maternal oxygenation and
perfusion remain the primary anesthetic goals. Airway management should be
performed by an experienced operator; hypoventilation and/or airway
obstruction may occur during maternal induction, emergence, extubation, or
recovery. Surgical attention may be diverted by the presence of specific injuries.
It is therefore essential that the anesthesia provider consider the overall patient
physiology. In cases of maternal collapse, extracorporeal membrane oxygenation
using heparin-coated tubing and a high-flow technique is a viable salvage option.
PMCD should also be considered if aortocaval pressure secondary to the
presence of the gravid uterus is a major contributor to maternal hemodynamic
compromise. PMCD may also be performed for fetal salvage in cases of failure
to resuscitate the mother. Regardless of the indication, PMCD should ideally be
initiated within 4 minutes of arrest onset.
REFERENCES
1. Hyde LK, Cook LJ, Olson LM, et al. Effect of motor vehicle crashes on adverse fetal outcomes.
Obstet Gynecol. 2003;102: 279–286.
2. Mendez-Figueroa H, Dahlke JD, Vrees RA, et al. Trauma in pregnancy: an updated systematic review.
Am J Obstet Gynecol. 2013;209:1–10.
3. Fildes J, Reed L, Jones N, et al. Trauma: the leading cause of maternal death. J Trauma. 1992;32:643–
645.
4. Kvarnstrand L, Milsom I, Lekander T, et al. Maternal fatalities, fetal and neonatal deaths related to
motor vehicle crashes during pregnancy: a national population-based study. Acta Obstet Gynecol
Scand. 2008;87:946–952.
5. Weiss HB, Songer TJ, Fabio A. Fetal deaths related to maternal injury. JAMA. 2001;286:1863–1868.
6. Fischer PE, Zarzaur BL, Fabian TC, et al. Minor trauma is an unrecognized contributor to poor fetal
outcomes: a population-based study of 78,552 pregnancies. J Trauma. 2011;71:90–93.
7. Cheng HT, Wang YC, Lo HC, et al. Trauma during pregnancy: a population-based analysis of
maternal outcome. World J Surg. 2012;36:2767–2775.
8. Vivian-Taylor J, Roberts CL, Chen JS, et al. Motor vehicle accidents during pregnancy: a population-
based study. BJOG. 2012;119:499–503.
9. National Center for Injury Prevention and Control. 2011 guidelines for field triage of injured patients.
http://www.cdc.gov/fieldtriage/pdf/decisionscheme_poster_a.pdf. Accessed March 24, 2015.
10. Shah PS, Shah J. Maternal exposure to domestic violence and pregnancy and birth outcomes: a
systematic review and meta-analyses. J Womens Health (Larchmt). 2010;19:2017–2031.
11. Curet MJ, Schermer CR, Demarest GB, et al. Predictors of outcome in trauma during pregnancy:
identification of patients who can be monitored for less than 6 hours. J Trauma. 2000;49:18–24.
12. Aboutanos MB, Aboutanos SZ, Dompkowski D, et al. Significance of motor vehicle crashes and
pelvic injury on fetal mortality: a five-year institutional review. J Trauma. 2008;65:616–620.
13. Schiff MA, Holt VL. Pregnancy outcomes following hospitalization for motor vehicle crashes in
Washington State from 1989 to 2001. Am J Epidemiol. 2005;161:503–510.
14. El-Kady D, Gilbert WM, Anderson J, et al. Trauma during pregnancy: an analysis of maternal and
fetal outcomes in a large population. Am J Obstet Gynecol. 2004;190:1661–1668.
15. Weiss HB, Sauber-Schatz EK, Cook LJ. The epidemiology of pregnancy-associated emergency
department injury visits and their impact on birth outcomes. Accid Anal Prev. 2008;40:1088–1095.
16. Williams JK, McClain L, Rosemurgy AS, et al. Evaluation of blunt abdominal trauma in the third
trimester of pregnancy: maternal and fetal considerations. Obstet Gynecol. 1990;75:33–37.
17. Petrone P, Talving P, Browder T, et al. Abdominal injuries in pregnancy: a 155-month study at two
level 1 trauma centers. Injury. 2011;42:47–49.
18. Kuo C, Jamieson DJ, McPheeters ML, et al. Injury hospitalizations of pregnant women in the United
States, 2002. Am J Obstet Gynecol. 2007;196:161.e1–161.e6.
19. Farragher RA, Laffey JG. Maternal brain death and somatic support. Neurocrit Care. 2005;3:99–106.
20. Mallampalli A, Guy E. Cardiac arrest in pregnancy and somatic support after brain death. Crit Care
Med. 2005;33:S325–S331.
21. Esmaeilzadeh M, Dictus C, Kayvanpour E, et al. One life ends, another begins: management of a
brain-dead pregnant mother—a systematic review. BMC Med. 2010;8:74.
22. Mayo TW. Brain-dead and pregnant in Texas. Am J Bioeth. 2014;14:15–18.
23. Nelson LJ, Milliken N. Compelled medical treatment of pregnant women. Life, liberty, and law in
conflict. JAMA. 1988;259:1060–1066.
24. Uppal T, Pickering A, Erasmus K, et al. The legal status of the fetus in New South Wales. J Law Med.
2012;20:178–183.
25. Pinkerton JV, Finnerty JJ. Resolving the clinical and ethical dilemma involved in fetal-maternal
conflicts. Am J Obstet Gynecol. 1996;175:289–295.
26. Thampapillai D. Court-ordered obstetrical intervention and the rights of a pregnant woman. J Law
Med. 2005;12:455–461.
27. National Institutes of Health. NIH policy and guidelines on the inclusion of women and minorities as
subjects in clinical research—amended, October, 2001.
http://grants.nih.gov/grants/funding/women_min/guidelines_amended_10_2001.htm. Accessed
February 23, 2015.
28. U.S. Food and Drug Administration. Women in clinical trials.
http://www.fda.gov/ScienceResearch/SpecialTopics/WomensHealthResearch/ucm131731.htm.
Accessed February 23, 2015.
29. Bowman M, Giles W, Deane S. Trauma during pregnancy—a review of management. Aust N Z J
Obstet Gynaecol. 1989; 29:389–393.
30. Sela HY, Weiniger CF, Hersch M, et al. The pregnant motor vehicle accident casualty: adherence to
basic workup and admission guidelines. Ann Surg. 2011;254:346–352.
31. Brunkhorst J, Weiner J, Lantos J. Infants of borderline viability: the ethics of delivery room care.
Semin Fetal Neonatal Med. 2014;19:290–295.
32. Fanaroff JM, Hascoët JM, Hansen TW, et al. The ethics and practice of neonatal resuscitation at the
limits of viability: an international perspective. Acta Paediatr. 2014;103:701–708.
33. Brownfoot FC, Gagliardi DI, Bain E, et al. Different corticosteroids and regimens for accelerating
fetal lung maturation for women at risk of preterm birth. Cochrane Database Syst Rev. 2013;
(8):CD006764.
34. Lykkedegn S, Sorensen GL, Beck-Nielsen SS, et al. The impact of vitamin D on fetal and neonatal
lung maturation. A systematic review. Am J Physiol Lung Cell Mol Physiol. 2015;308:L587–L602.
35. Gonzalez Garay AG, Reveiz L, Velasco Hidalgo L, et al. Ambroxol for women at risk of preterm birth
for preventing neonatal respiratory distress syndrome. Cochrane Database Syst Rev. 2014;
(10):CD009708.
36. Baker SP, O’Neill B, Haddon W Jr, et al. The injury severity score: a method for describing patients
with multiple injuries and evaluating emergency care. J Trauma. 1974;14:187–196.
37. Osler T, Baker SP, Long W. A modification of the injury severity score that both improves accuracy
and simplifies scoring. J Trauma. 1997;43:922–925.
38. Champion HR, Sacco WJ, Hunt TK. Trauma severity scoring to predict mortality. World J Surg.
1983;7:4–11.
39. Tohira H, Jacobs I, Mountain D, et al. Systematic review of predictive performance of injury severity
scoring tools. Scand J Trauma Resusc Emerg Med. 2012;20:63.
40. Rogers FB, Rozycki GS, Osler TM, et al. A multi-institutional study of factors associated with fetal
death in injured pregnant patients. Arch Surg. 1999;134:1274–1277.
41. Guly HR, Bouamra O, Little R, et al. Testing the validity of the ATLS classification of hypovolaemic
shock. Resuscitation. 2010;81:1142–1147.
42. Mutschler M, Nienaber U, Brockamp T, et al. A critical reappraisal of the ATLS classification of
hypovolaemic shock: does it really reflect clinical reality? Resuscitation. 2013;84:309–313.
43. Mahendru AA, Everett TR, Wilkinson IB, et al. A longitudinal study of maternal cardiovascular
function from preconception to the postpartum period. J Hypertens. 2014;32:849–856.
44. Jensen D, Duffin J, Lam YM, et al. Physiological mechanisms of hyperventilation during human
pregnancy. Respir Physiol Neurobiol. 2008;161:76–86.
45. Schoorl M, Schoorl M, van der Gaag D, et al. Effects of iron supplementation on red blood cell
hemoglobin content in pregnancy. Hematol Rep. 2012;4:e24.
46. DeMaeyer E, Adiels-Tegman M. The prevalence of anaemia in the world. World Health Stat Q.
1985;38:302–316.
47. McLean E, Cogswell M, Egli I, et al. Worldwide prevalence of anaemia, WHO vitamin and mineral
nutrition information system, 1993-2005. Public Health Nutr. 2009;12:444–454.
48. Peña-Rosas JP, De-Regil LM, Dowswell T, et al. Daily oral iron supplementation during pregnancy.
Cochrane Database Syst Rev. 2012;(12):CD004736.
49. Sharma SK, Philip J, Wiley J. Thromboelastographic changes in healthy parturients and postpartum
women. Anesth Analg. 1997;85:94–98.
50. Erez O, Novack L, Beer-Weisel R, et al. DIC score in pregnant women—a population based
modification of the International Society on Thrombosis and Hemostasis score. PLoS One.
2014;9:e93240.
51. Confidential Enquiry into Maternal and Child Health. Why Mothers Die 2000–2002. The Sixth Report
of the Confidential Enquiries into Maternal Deaths in the United Kingdom. London, United Kingdom:
RCOG Press; 2004.
52. Confidential Enquiry into Maternal and Child Health. Saving Mothers’ Lives: Reviewing Maternal
Deaths to Make Motherhood Safer 2003–2005. The Seventh Confidential Enquiry into Maternal
Deaths in the United Kingdom. London, United Kingdom: RCOG Press; 2007.
53. Swanton RD, Al-Rawi S, Wee MY. A national survey of obstetric early warning systems in the United
Kingdom. Int J Obstet Anesth. 2009;18:253–257.
54. Carle C, Alexander P, Columb M, et al. Design and internal validation of an obstetric early warning
score: secondary analysis of the Intensive Care National Audit and Research Centre Case Mix
Programme database. Anaesthesia. 2013;68:354–367.
55. Isaacs RA, Wee MY, Bick DE, et al. A national survey of obstetric early warning systems in the
United Kingdom: five years on. Anaesthesia. 2014;69:687–692.
56. Shah KH, Simons RK, Holbrook T, et al. Trauma in pregnancy: maternal and fetal outcomes. J
Trauma. 1998;45:83–86.
57. Abramowicz JS, Kossoff G, Maršál K, et al. Safety statement, 2000 (reconfirmed 2003): International
Society of Ultrasound in Obstetrics and Gynecology (ISUOG). Ultrasound Obstet Gynecol.
2003;21:100.
58. U.S. Food and Drug Administration. Ultrasound imaging: benefits/risks. http://www.fda.gov/radiation-
emittingproducts/radiationemittingproductsandprocedures/medicalimaging/ucm115357#benefitsrisks.
Accessed March 6, 2015.
59. Salvesen K, Lees C, Abramowicz J, et al. ISUOG statement on the safe use of Doppler in the 11 to 13
+ 6-week fetal ultrasound examination. Ultrasound Obstet Gynecol. 2011;37:628.
60. Svinos H. Best BETs from the Manchester Royal Infirmary. BET 1 using ultrasound to detect
peritoneal fluid in a pregnant patient with abdominal trauma. Emerg Med J. 2009;26:201–202.
61. Goodwin H, Holmes JF, Wisner DH. Abdominal ultrasound examination in pregnant blunt trauma
patients. J Trauma. 2001;50:689–693.
62. Brown MA, Sirlin CB, Farahmand N, et al. Screening sonography in pregnant patients with blunt
abdominal trauma. J Ultrasound Med. 2005;24:175–181.
63. Branney SW, Wolfe RE, Moore EE, et al. Quantitative sensitivity of ultrasound in detecting free
intraperitoneal fluid. J Trauma. 1995;39:375–380.
64. Sirlin CB, Casola G, Brown MA, et al. Use of blunt abdominal trauma: importance of free pelvic fluid
in women of reproductive age. Radiology. 2001;219:229–235.
65. Hussain ZJ, Figueroa R, Budorick NE. How much free fluid can a pregnant patient have? Assessment
of pelvic free fluid in pregnant patients without antecedent trauma. J Trauma. 2011;70:1420–1423.
66. Fleming S, Bird R, Ratnasingham K, et al. Accuracy of FAST scan in blunt abdominal trauma in a
major London trauma centre. Int J Surg. 2012;10:470–474.
67. Miller MT, Pasquale MD, Bromberg WJ, et al. Not so FAST. J Trauma. 2003;54:52–60.
68. Matsushita H, Harada A, Sato T, et al. Fetal intracranial injuries following motor vehicle accidents
with airbag deployment. J Obstet Gynaecol Res. 2014;40:599–602.
69. Aromatario M, Bottoni E, Cappelletti S, et al. Intrauterine fetal decapitation after a high-speed car
crash. Am J Forensic Med Pathol. 2015;36:6–9.
70. Glantz C, Purnell L. Clinical utility of sonography in the diagnosis and treatment of placental
abruption. J Ultrasound Med. 2002;21:837–840.
71. Elsasser DA, Ananth CV, Prasad V, et al. Diagnosis of placental abruption: relationship between
clinical and histopathological findings. Eur J Obstet Gynecol Reprod Biol. 2010;148:125–130.
72. Harrison SD, Nghiem HV, Shy K. Uterine rupture with fetal death following blunt trauma. AJR Am J
Roentgenol. 1995;165:1452.
73. Tremblay E, Thérasse E, Thomassin-Naggara I, et al. Quality initiatives: guidelines for use of medical
imaging during pregnancy and lactation. Radiographics. 2012;32:897–911.
74. Brent RL. What are the reproductive and developmental risks of ionizing radiation? In: Hales B,
Scialli A, Tassinari M, eds. Teratology Primer. 2nd ed. Philadelphia, PA: Thomas Jefferson
University; 2010:56–58.
75. Wakeford R, Little MP. Risk coefficients for childhood cancer after intrauterine irradiation: a review.
Int J Radiat Biol. 2003;79:293–309.
76. Atwell TD, Lteif AN, Brown DL, et al. Neonatal thyroid function after administration of IV iodinated
contrast agent to 21 pregnant patients. AJR Am J Roentgenol. 2008;191:268–271.
77. Kochi MH, Kaloudis EV, Ahmed W, et al. Effect of in utero exposure of iodinated intravenous contrast
on neonatal thyroid function. J Comput Assist Tomogr. 2012;36:165–169.
78. Bourjeily G, Chalhoub M, Phornphutkul C, et al. Neonatal thyroid function: effect of a single
exposure to iodinated contrast medium in utero. Radiology. 2010;256:744–750.
79. Rajaram S, Exley CE, Fairlie F, et al. Effect of antenatal iodinated contrast agent on neonatal thyroid
function. Br J Radiol. 2012;85:e238–e242.
80. Lowdermilk C, Gavant ML, Qaisi W, et al. Screening helical CT for evaluation of blunt traumatic
injury in the pregnant patient. Radiographics. 1999;19:S243–S255.
81. Manriquez M, Srinivas G, Bollepalli S, et al. Is computed tomography a reliable diagnostic modality
in detecting placental injuries in the setting of acute trauma? Am J Obstet Gynecol. 2010;202:611.e1–
611.e5.
82. Wei SH, Helmy M, Cohen AJ. CT evaluation of placental abruption in pregnant trauma patients.
Emerg Radiol. 2009;16: 365–373.
83. Kopelman TR, Berardoni NE, Manriquez M, et al. The ability of computed tomography to diagnose
placental abruption in the trauma patient. J Trauma Acute Care Surg. 2013;74:236–241.
84. Saphier NB, Kopelman TR. Traumatic Abruptio Placenta Scale (TAPS): a proposed grading system of
computed tomography evaluation of placental abruption in the trauma patient. Emerg Radiol.
2014;21:17–22.
85. Raptis CA, Mellnick VM, Raptis DA, et al. Imaging of trauma in the pregnant patient. Radiographics.
2014;34:748–763.
86. Fusco A, Kelly K, Winslow J. Uterine rupture in a motor vehicle crash with airbag deployment. J
Trauma. 2001;51:1192–1194.
87. Dash N, Lupetin AR. Uterine rupture secondary to trauma: CT findings. J Comput Assist Tomogr.
1991;15:329–331.
88. Patenaude Y, Pugash D, Lim K, et al. The use of magnetic resonance imaging in the obstetric patient. J
Obstet Gynaecol Can. 2014;36:349–363.
89. Bergese SD, Puente EG. Anesthesia in the intraoperative MRI environment. Neurosurg Clin N Am.
2009;20:155–162.
90. Masselli G, Gualdi G. MR imaging of the placenta: what a radiologist should know. Abdom Imaging.
2013;38:573–587.
91. Masselli G, Brunelli R, Di Tola M, et al. MR imaging in the evaluation of placental abruption:
correlation with sonographic findings. Radiology. 2011;259:222–230.
92. Banović V, Škrablin S, Banović M, et al. Fetal brain magnetic resonance imaging and long-term
neurodevelopmental impairment. Int J Gynaecol Obstet. 2014;125:237–240.
93. Luley T, Fitzpatrick CB, Grotegut CA, et al. Perinatal implications of motor vehicle accident trauma
during pregnancy: identifying populations at risk. Am J Obstet Gynecol. 2013;208:466.e1–466.e5.
94. Klinich KD, Flannagan CA, Rupp JD, et al. Fetal outcome in motor-vehicle crashes: effects of crash
characteristics and maternal restraint. Am J Obstet Gynecol. 2008;198:450.e1–450.e9.
95. Baerga-Varela Y, Zietlow SP, Bannon MP, et al. Trauma in pregnancy. Mayo Clin Proc.
2000;75:1243–1248.
96. Kissinger DP, Rozycki GS, Morris JA Jr, et al. Trauma in pregnancy. Predicting pregnancy outcome.
Arch Surg. 1991;126: 1079–1086.
97. Poole GV, Martin JN Jr, Perry KG Jr, et al. Trauma in pregnancy: the role of interpersonal violence.
Am J Obstet Gynecol. 1996;174:1873–1877.
98. Garmi G, Marjieh M, Salim R. Does minor trauma in pregnancy affect perinatal outcome? Arch
Gynecol Obstet. 2014;290: 635–641.
99. Muench MV, Baschat AA, Reddy UM, et al. Kleihauer-betke testing is important in all cases of
maternal trauma. J Trauma. 2004;57:1094–1098.
100. Dahmus MA, Sibai BM. Blunt abdominal trauma: are there any predictive factors for abruptio
placentae or maternal-fetal distress? Am J Obstet Gynecol. 1993;169:1054–1059.
101. Tanizaki S, Maeda S, Matano H, et al. Elevated maternal serum α-fetoprotein after minor trauma
during pregnancy may predict adverse fetal outcomes. J Trauma Acute Care Surg. 2014;77:510–513.
102. Dhanraj D, Lambers D. The incidences of positive Kleihauer-Betke test in low-risk pregnancies and
maternal trauma patients. Am J Obstet Gynecol. 2004;190:1461–1463.
103. Wylie BJ, D’Alton ME. Fetomaternal hemorrhage. Obstet Gynecol. 2010;115:1039–1051.
104. Suresh M, Wali A. Failed intubation in obstetrics—airway management strategies. Anesthesiol Clin
North Am. 1998;16:477–498.
105. McKeen DM, George RB, O’Connell CM, et al. Difficult and failed intubation: incident rates and
maternal, obstetrical, and anesthetic predictors. Can J Anaesth. 2011;58:514–524.
106. McDonnell NJ, Paech MJ, Clavisi OM, et al. Difficult and failed intubation in obstetric anaesthesia: an
observational study of airway management and complications associated with general anaesthesia for
caesarean section. Int J Obstet Anesth. 2008;17:292–297.
107. Paranjothy S, Griffiths JD, Broughton HK, et al. Interventions at caesarean section for reducing the
risk of aspiration pneumonitis. Cochrane Database Syst Rev. 2014;(2):CD004943.
108. Peterson GN, Domino KB, Caplan RA, et al. Management of the difficult airway: a closed claims
analysis. Anesthesiology. 2005;103:33–39.
109. McAuliffe F, Kametas N, Costello J, et al. Respiratory function in singleton and twin pregnancy.
BJOG. 2002;109:765–769.
110. Pühringer FK, Sparr HJ, Mitterschiffthaler G, et al. Extended duration of action of rocuronium in
postpartum patients. Anesth Analg. 1997;84:352–354.
111. Gin T, Chan MT, Chan KL, et al. Prolonged neuromuscular block after rocuronium in postpartum
patients. Anesth Analg. 2002;94:686–689.
112. Usui R, Matsubara S, Ohkuchi A, et al. Fetal heart rate pattern reflecting the severity of placental
abruption. Arch Gynecol Obstet. 2008;277:249–253.
113. Vladutiu CJ, Marshall SW, Poole C, et al. Adverse pregnancy outcomes following motor vehicle
crashes. Am J Prev Med. 2013;45:629–636.
114. Bond AL, Edersheim TG, Curry L, et al. Expectant management of abruptio placentae before 35
weeks gestation. Am J Perinatol. 1989;6:121–123.
115. Combs CA, Nyberg DA, Mack LA, et al. Expectant management after sonographic diagnosis of
placental abruption. Am J Perinatol. 1992;9:170–174.
116. World Health Organization. Preterm birth. http://www.who.int/mediacentre/factsheets/fs363/en/.
Accessed March 10, 2015.
117. Thaler I, Manor D, Itskovitz J, et al. Changes in uterine blood flow during human pregnancy. Am J
Obstet Gynecol. 1990;162:121–125.
118. Enakpene CA, Ayinde OA, Omigbodun AO. Incomplete uterine rupture, following blunt trauma to the
abdomen: a case report. Niger J Clin Pract. 2005;8:60–62.
119. van Enk A, van Zwam W. Uterine rupture. A seat belt hazard. Acta Obstet Gynecol Scand.
1994;73:432–433.
120. Dunning K, Lemasters G, Bhattacharya A. A major public health issue: the high incidence of falls
during pregnancy. Matern Child Health J. 2010;14:720–725.
121. McCrory JL, Chambers AJ, Daftary A, et al. Dynamic postural stability in pregnant fallers and non-
fallers. BJOG. 2010; 117:954–962.
122. Inanir A, Cakmak B, Hisim Y, et al. Evaluation of postural equilibrium and fall risk during pregnancy.
Gait Posture. 2014; 39:1122–1125.
123. Ersal T, McCrory JL, Sienko KH. Theoretical and experimental indicators of falls during pregnancy as
assessed by postural perturbations. Gait Posture. 2014;39:218–223.
124. Charlton WP, Coslett-Charlton LM, Ciccotti MG. Correlation of estradiol in pregnancy and anterior
cruciate ligament laxity. Clin Orthop Relat Res. 2001;387:165–170.
125. Schiff MA, Holt VL, Daling JR. Maternal and infant outcomes after injury during pregnancy in
Washington State from 1989 to 1997. J Trauma. 2002;53:939–945.
126. Schiff M. Pregnancy outcomes following hospitalization for a fall in Washington State from 1987 to
2004. BJOG. 2008;115:1648–1654.
127. Bowen E, Heron J, Waylen A, et al. Domestic violence risk during and after pregnancy: findings from
a British longitudinal study. BJOG. 2005;112:1083–1089.
128. Stewart DE, Cecutti A. Physical abuse in pregnancy. CMAJ. 1993;149:1257–1263.
129. Devries KM, Kishor S, Johnson H, et al. Intimate partner violence during pregnancy: analysis of
prevalence data from 19 countries. Reprod Health Matters. 2010;18:158–170.
130. Johnson JK, Haider F, Ellis K, et al. The prevalence of domestic violence in pregnant women. BJOG.
2003;110:272–275.
131. Palladino CL, Singh V, Campbell J, et al. Homicide and suicide during the perinatal period: findings
from the National Violent Death Reporting System. Obstet Gynecol. 2011;118:1056–1063.
132. Chang J, Berg CJ, Saltzman LE, et al. Homicide: a leading cause of injury deaths among pregnant and
postpartum women in the United States, 1991–1999. Am J Public Health. 2005;95:471–477.
133. Krulewitch CJ, Pierre-Louis ML, de Leon-Gomez R, et al. Hidden from view: violent deaths among
pregnant women in the District of Columbia, 1988–1996. J Midwifery Womens Health. 2001;46:4–10.
134. Abdollahi F, Abhari FR, Delavar MA, et al. Physical violence against pregnant women by an intimate
partner, and adverse pregnancy outcomes in Mazandaran Province, Iran. J Family Community Med.
2015;22:13–18.
135. Wiencrot A, Nannini A, Manning SE, et al. Neonatal outcomes and mental illness, substance abuse,
and intentional injury during pregnancy. Matern Child Health J. 2012;16:979–988.
136. Jagoe J, Magann EF, Chauhan SP, et al. The effects of physical abuse on pregnancy outcomes in a
low-risk obstetric population. Am J Obstet Gynecol. 2000;182:1067–1069.
137. Masoodi Z, Ahmad I, Khurram F, et al. Pregnancy in burns: maternal and fetal outcome. Indian J
Burns. 2012;20:36–41.
138. Schubert W, Ahrenholz DH, Solem LD. Burns from hot oil and grease: a public health hazard. J Burn
Care Rehabil. 1990;11:558–562.
139. Lama BB, Duke JM, Sharma NP, et al. Intentional burns in Nepal: a comparative study. Burns.
2015;41:1306–1314. doi: 10.1016/j.burns.2015.01.006.
140. Peck MD. Epidemiology of burns throughout the World. Part II: intentional burns in adults. Burns.
2012;38:630–637.
141. Masoodi Z, Ahmad I, Yousuf S. Routine use of urinary hCG test in adult burn females to detect
“hidden” pregnancies: a review. Burns. 2013;39:803–807.
142. Maghsoudi H, Pourzand A, Azarmir G. Etiology and outcome of burns in Tabriz, Iran. An analysis of
2963 cases. Scand J Surg. 2005;94:77–81.
143. Pacheco LD, Gei AF, VanHook JW, et al. Burns in pregnancy. Obstet Gynecol. 2005;106:1210–1212.
144. Karimi H, Momeni M, Rahbar, H. Burn injuries during pregnancy in Iran. Int J Gynaecol Obstet.
2009;104:132–134.
145. Rezavand N, Seyedzadeh A, Soleymani A. Evaluation of maternal and foetal outcomes in pregnant
women hospitalized in Kermanshah Hospitals, Iran, owing to burn injury, 2003-2008. Ann Burns Fire
Disasters. 2012;25:196–199.
146. Maghsoudi H, Samnia R, Garadaghi A, et al. Burns in pregnancy. Burns. 2006;32:246–250.
147. Akhtar MA, Mulawkar PM, Kulkarni HR. Burns in pregnancy: effect on maternal and fetal outcomes.
Burns. 1994;20:351–355.
148. Jain ML, Garg AK. Burns with pregnancy—a review of 25 cases. Burns. 1993;19:166–167.
149. Gun F, Erginel B, Günendi T, et al. Gunshot wound of the fetus. Pediatr Surg Int. 2011;27:1367–1369.
150. Pasley JD, Demetriades D. Penetrating fetal trauma with late complications: a case report. J Pediatr
Surg. 2012;47:E9–E11.
151. Carugno JA, Rodriguez A, Brito J, et al. Gunshot wound to the gravid uterus with non-lethal fetal
injury. J Emerg Med. 2008;35:43–45.
152. Sakala EP, Kort DD. Management of stab wounds to the pregnant uterus: a case report and a review of
the literature. Obstet Gynecol Surv. 1988;43:319–324.
153. Shehu BB, Ismail NJ, Hassan I, et al. Fetal head injury from intentional penetrating abdominal trauma
in pregnancy. Ann Trop Paediatr. 2010;30:69–72.
154. Gallo P, Mazza C, Sala F. Intrauterine head stab wound injury resulting in a growing skull fracture: a
case report and literature review. Childs Nerv Syst. 2010;26:377–384.
155. Muzumdar D, Higgins MJ, Ventureyra EC. Intrauterine penetrating direct fetal head trauma following
gunshot injury: a case report and review of the literature. Childs Nerv Syst. 2006;22:398–402.
156. Awwad JT, Azar GB, Seoud MA, et al. High-velocity penetrating wounds of the gravid uterus: review
of 16 years of civil war. Obstet Gynecol. 1994;83:259–264.
157. Extracorporeal Life Support Organization. Guidelines for adult respiratory failure.
https://www.elso.org/Portals/0/IGD/Archive/FileManager/989d4d4d14cusersshyerdocumentselsoguidelinesforadultresp
Accessed March 15, 2015.
158. Extracorporeal Life Support Organization. Guidelines for adult cardiac failure.
https://www.elso.org/Portals/0/IGD/Archive/FileManager/e76ef78eabcusersshyerdocumentselsoguidelinesforadultcardi
Accessed March 15, 2015.
159. Extracorporeal Life Support Organization. Guidelines for ECPR cases.
https://www.elso.org/Portals/0/IGD/Archive/FileManager/6713186745cusersshyerdocumentselsoguidelinesforecprcases
Accessed March 15, 2015.
160. Nair P, Davies AR, Beca J, et al. Extracorporeal membrane oxygenation for severe ARDS in pregnant
and postpartum women during the 2009 H1N1 pandemic. Intensive Care Med. 2011;37:648–654.
161. Robertson LC, Allen SH, Konamme SP, et al. The successful use of extra-corporeal membrane
oxygenation in the management of a pregnant woman with severe H1N1 2009 influenza complicated
by pneumonitis and adult respiratory distress syndrome. Int J Obstet Anesth. 2010;19:443–447.
162. Smith IJ, Gillham MJ. Fulminant peripartum cardiomyopathy rescue with extracorporeal membranous
oxygenation. Int J Obstet Anesth. 2009;18:186–188.
163. Weinberg L, Kay C, Liskaser F, et al. Successful treatment of peripartum massive pulmonary
embolism with extracorporeal membrane oxygenation and catheter-directed pulmonary thrombolytic
therapy. Anaesth Intensive Care. 2011;39:486–491.
164. Leeper WR, Valdis M, Arntfield R, et al. Extracorporeal membrane oxygenation in the acute treatment
of cardiovascular collapse immediately post-partum. Interact Cardiovasc Thorac Surg. 2013;17:898–
899.
165. Arlt M, Philipp A, Iesalnieks I, et al. Successful use of a new hand-held ECMO system in
cardiopulmonary failure and bleeding shock after thrombolysis in massive post-partal pulmonary
embolism. Perfusion. 2009;24:49–50.
166. Biderman P, Einav S, Fainblut M, et al. Extracorporeal life support in patients with multiple injuries
and severe respiratory failure: a single-center experience? J Trauma Acute Care Surg. 2013;75:907–
912.
167. Wen PH, Chan WH, Chen YC, et al. Non-heparinized ECMO serves a rescue method in a multitrauma
patient combining pulmonary contusion and nonoperative internal bleeding: a case report and
literature review. World J Emerg Surg. 2015;10:15.
168. Zhou R, Liu B, Lin K, et al. ECMO support for right main bronchial disruption in multiple trauma
patient with brain injury—a case report and literature review. Perfusion. 2015;30:403–406.
169. Muellenbach RM, Redel A, Küstermann J, et al. Extracorporeal membrane oxygenation and severe
traumatic brain injury. Is the ECMO-therapy in traumatic lung failure and severe traumatic brain
injury really contraindicated? Anaesthesist. 2011;60:647–652.
170. Plotkin JS, Shah JB, Lofland GK, et al. Extracorporeal membrane oxygenation in the successful
treatment of traumatic adult respiratory distress syndrome: case report and review. J Trauma.
1994;37:127–130.
171. Vanden Hoek TL, Morrison LJ, Shuster M, et al. Part 12: cardiac arrest in special situations: 2010
American Heart Association Guidelines for Cardiopulmonary Resuscitation and Emergency
Cardiovascular Care. Circulation. 2010;122:S829–S861.
172. Einav S, Kaufman N, Sela HY. Maternal cardiac arrest and perimortem caesarean delivery: evidence
or expert-based? Resuscitation. 2012;83:1191–1200.
173. Chibber R, Al-Harmi J, Fouda M, et al. Motor-vehicle injury in pregnancy and subsequent feto-
maternal outcomes: of grave concern. J Matern Fetal Neonatal Med. 2015;28:399–402.
Management of the Opioid Dependent Parturient
Jessica L. Young, Ellen M. Lockhart, and Curtis L. Baysinger
I. Introduction
II. Obstetric management
A. Risks of opioid dependence in pregnancy
B. Identification
C. Treatment
D. Chronic pain in pregnancy
E. Peripartum obstetric management
III. Neonatal abstinence syndrome
A. Definition and incidence
B. Symptoms
C. Diagnosis
D. Risk factors
E. Treatment of neonatal abstinence syndrome
IV. Pain management during the peripartum period
A. Opioid tolerance
B. Baseline long-acting opioid maintenance
C. Pain management for labor and delivery
D. Anesthetic management of cesarean delivery
E. Postcesarean delivery analgesia
Summary
KEYPOINTS
1. Opioid dependent (OD) parturients should have long-acting opioid
therapy either instituted or maintained during pregnancy because it
improves obstetric outcomes. Acute detoxification is not recommended.
2. Postdelivery breastfeeding is safe and should be encouraged.
3. Pain management is complicated by opioid tolerance, hyperalgesia to
sensory stimuli, psychosocial issues, and possible use of other illicit
substances.
4. Neuraxial analgesia for labor and vaginal delivery is safe and effective
when similar doses of local anesthetics are used in OD women compared
to non-OD women.
5. The therapeutic window between analgesia and respiratory depression of
acutely administered opioids may be narrowed in OD women. Tolerance
to other side effects of opioid therapy does not imply a decrease in risk of
respiratory depression.
6. Treatment of pain after cesarean delivery should employ a multimodal
approach to pain control including division of daily maintenance opioid
into several doses and non-opioid analgesics.
I. Introduction
Opioid use has increased dramatically among women of childbearing
age over the past 15 years, with a concomitant rise in OD parturients
who present for labor and delivery. Nonmedical use of prescription
opioids and opioid-related deaths tripled between 1999 and 2010 (see
Fig. 31.1). Recent reports cite that more than 12% of parturients with
commercial insurance were prescribed opioids during their pregnancy
in 2011 (see Fig. 31.2) with an increase of 5% between 2000 and
2007; among parturients on Medicaid, this increase was nearly 20%.
Neonatal abstinence syndrome (NAS) has increased proportionately
as well. Parturients who take opioids chronically pose obstetric and
social challenges to all obstetric care providers.
II. Obstetric management (see Fig. 31.3)
A. Risks of opioid dependence in pregnancy
1. Opioid dependence in pregnancy is associated with poor
maternal and fetal outcomes including miscarriage, preterm
labor, preterm delivery, premature rupture of membranes,
intrauterine growth restriction, and NAS.
2. Withdrawal symptoms during pregnancy can be dangerous for
the fetus because it causes maternal tachycardia,
hypertension, decreased placental perfusion, and uterine
contractions.1
3. Women in the cycle of intoxication and withdrawal are at
higher risk for intrauterine growth restriction, insufficient
nutrition, or exposure to illicit opioids such as heroin.
4. Rates of infectious disease, such as hepatitis C, are higher in
this population.2,3
5. Due to a combination of psychosocial risk factors, this patient
population is at risk for delayed or inadequate prenatal care.
6. Poor fetal outcomes and increased risk of neonatal abstinence
can be attributed to the concurrent use of other substances
including tobacco, alcohol, benzodiazepines, and other illicit
drugs.4–6
7. Concurrent psychiatric disorders are also common in this
population, with one 2010 study reporting up to 65% of these
patients have symptoms of mental illness.7
8. Opioids are not thought to be teratogenic, particularly with
short-term use.
a. Most large retrospective studies have shown no
association between opioid exposure and congenital
anomalies8; however, one retrospective study suggests an
association between codeine exposure in the first trimester
and cleft palate, cardiac defects, and pyloric stenosis.9
b. More recently, the 2011 National Birth Defects
Prevention Study showed an association between cardiac,
spinal, and abdominal wall defects with opioid exposure
in the first trimester, but these results have not been
replicated.10
D. Risk factors
1. The dose of methadone does not influence the rate of NAS
diagnosis but does affect its severity and the need for
treatment.30–32
2. Preterm birth does not influence the odds of the infant
receiving treatment.
3. Timing and mode of delivery as well as timing of the last
maternal methadone dose are significant risk factors for the
development of NAS requiring treatment.24
4. The need for pharmacologic therapy is affected by genetics,
other drug exposures, gestational age, breastfeeding, and
maternal rooming-in.28
5. Maternal buprenorphine use may reduce the incidence and
severity of NAS when compared to other opioids.17
E. Treatment of neonatal abstinence syndrome
1. Pharmacologic intervention is required for 50% to 70% of
infants with NAS who demonstrate escalating symptoms.
2. Once symptoms resolve, treatment can be weaned over a
period of days to weeks.33
3. Methadone and morphine are most commonly used to treat
NAS.
4. The efficacy of clonidine has not been proven, but can be
used as an adjuvant to other therapies.27
5. Barbiturates and benzodiazepines have been used but are not
currently recommended as primary agents.27
6. The use of naloxone is contraindicated because it may induce
neonatal seizures.27
IV. Pain management during the peripartum period (see Fig. 31.3)
A. Opioid tolerance. Opioid tolerance, opioid hyperalgesia,
physical dependence with risk of withdrawal, and concomitant
use of illegal drugs create challenges to effective pain
management.
1. Women with opioid dependence exhibit hypersensitivity to
pain stimuli such as cold pressor tests and electrical
stimulation.34
2. Opioid tolerance is thought to be due to opioid receptor
downregulation and decreases in central glutamate receptor
activity (i.e., decreased antinociception). Opioid-induced
hypersensitization is due to increased N-methyl-D-aspartate
receptor activity. In addition, there are increases in spinal cord
dynorphin concentrations (i.e., increased pronocioception).
Both mechanisms result in an increase in the amount of opioid
required to achieve a given level of pain control.34
3. Studies of peripartum pain management are limited. Hence,
any recommendations for pain management in OD pregnant
women are based on reports from nonpregnant women with
opioid dependence who have undergone anesthesia and
surgery. Although these reports commonly measure changes
in opioid use, the results may not be applicable in the OD
patient (see Table 31.1).
CLINICAL PEARL Clinical studies to guide pain control during
the peripartum period are virtually nonexistent in OD patients.
Guidelines for management can be extrapolated from studies in
nonpregnant surgical patients and non-OD parturients.
B. Baseline long-acting opioid maintenance. The baseline opioid
consumed by the OD parturient should be continued through the
peripartum period. Use of mixed agonist/antagonist opioids
may precipitate withdrawal and are to be avoided.35
1. Once daily dosing of long-acting opioids prevents withdrawal,
but dividing daily doses of methadone and buprenorphine into
6 to 8 hour intervals will improve the quality of analgesia.36
2. Small doses of methadone and buprenorphine are often
effective in opioid-naive surgical patients, but even the large
doses commonly seen in OD patients may be inadequate after
cesarean delivery in a woman with opioid dependence.
C. Pain management for labor and delivery37,38
1. Studies of pain management and neuraxial analgesia in
laboring women with opioid dependence are lacking.
2. Neuraxial techniques commonly used in women without
opioid dependence are likely to be effective in most women
with opioid dependence because local anesthetic efficacy is
not affected.34
3. Opioid requirements for analgesia after vaginal delivery have
not been shown to be significantly increased in OD
women.37,38
4. Use of non-opioid analgesics following vaginal delivery
should be similar to that used in parturients without opioid
dependence.37,38
SUMMARY
The OD parturient requires a multidisciplinary approach to
management. Screening for opioid dependence early in pregnancy
should occur in women not already identified as OD, and appropriate
long-term therapy should be prescribed. Patient fears of opioid
withdrawal and pain during the peripartum period require reassurance
and counseling. Neuraxial techniques for pain relief during vaginal
delivery are similar to those used in non-OD women. Postcesarean
delivery analgesia requires a multimodal approach utilizing
appropriate neuraxial analgesic techniques, additional opioid
administration, and non-opioid supplements. Prospective trials
evaluating optimal pain management in these patients do not exist; all
recommendations are based on retrospective studies with small
numbers of patients and reports from nonpregnant surgical patients.
REFERENCES
1. Bolnick JM, Rayburn WF. Substance use disorders in women: special considerations during
pregnancy. Obstet Gynecol Clin North Am. 2003;30:545–558, vii.
2. Wu L-T, Ling W, Burchett B, et al. Gender and racial/ethnic differences in addiction severity, HIV
risk, and quality of life among adults in opioid detoxification: results from the National Drug Abuse
Treatment Clinical Trials Network. Subst Abuse Rehabil. 2010;2010:13–22.
3. Rondinelli AJ, Ouellet LJ, Strathdee SA, et al. Young adult injection drug users in the United States
continue to practice HIV risk behaviors. Drug Alcohol Depend. 2009;104:167–174.
4. Jones HE, Heil SH, O’Grady KE, et al. Smoking in pregnant women screened for an opioid agonist
medication study compared to related pregnant and non-pregnant patient samples. Am J Drug Alcohol
Abuse. 2009;35:375–380.
5. Green TC, Grimes Serrano JM, Licari A, et al. Women who abuse prescription opioids: findings from
the Addiction Severity Index-Multimedia Version Connect prescription opioid database. Drug Alcohol
Depend. 2009;103:65–73.
6. Stine SM, Heil SH, Kaltenbach K, et al. Characteristics of opioid-using pregnant women who accept
or refuse participation in a clinical trial: screening results from the MOTHER study. Am J Drug
Alcohol Abuse. 2009;35:429–433.
7. Benningfield MM, Arria AM, Kaltenbach K, et al. Co-occurring psychiatric symptoms are associated
with increased psychological, social, and medical impairment in opioid dependent pregnant women.
Am J Addict. 2010;19:416–421.
8. Chen CH, Lin HC. Prenatal care and adverse pregnancy outcomes among women with depression: a
nationwide population-based study. Can J Psychiatry. 2011;56:273–280.
9. Brennan MC, Rayburn WF. Counseling about risks of congenital anomalies from prescription opioids.
Birth Defects Res A Clin Mol Teratol. 2012;94:620–625.
10. Broussard CS, Rasmussen SA, Reefhuis J, et al. Maternal treatment with opioid analgesics and risk for
birth defects. Am J Obstet Gynecol. 2011;204:314.e1–314.e11.
11. American College of Obstetricians and Gynecologists. ACOG Committee Opinion No. 524: opioid
abuse, dependence, and addiction in pregnancy. Obstet Gynecol. 2012;119:1070–1076.
12. Chang G, Orav EJ, Jones JA, et al. Self-reported alcohol and drug use in pregnant young women: a
pilot study of associated factors and identification. J Addict Med. 2011;5:221–226.
13. Sarkar M, Burnett M, Carrière S, et al. Screening and recording of alcohol use among women of child-
bearing age and pregnant women. Can J Clin Pharmacol. 2009;16:e242–e263.
14. Roberts SC, Nuru-Jeter A. Women’s perspectives on screening for alcohol and drug use in prenatal
care. Women’s Health Issues. 2010;20:193–200.
15. Kaltenbach K, Berghella V, Finnegan L. Opioid dependence during pregnancy: effects and
management. Obset Gynecol Clin North Am. 1998;25:139–151.
16. Winklbaur B, Kopf N, Ebner N, et al. Treating pregnant women dependent on opioids is not the same
as treating pregnancy and opioid dependence: a knowledge synthesis for better treatment for women
and neonates. Addiction. 2008; 103:1429–1440.
17. Jones HE, Kaltenbach K, Heil SH, et al. Neonatal abstinence syndrome after methadone or
buprenorphine exposure. N Engl J Med. 2010;363:2320–2331.
18. McNicholas L. Clinical Guidelines for the Use of Buprenorphine in the Treatment of Opioid
Addiction. Rockville, MD: U.S. Department of Health and Human Services; 2004.
19. Cleary BJ, Donnelly JM, Strawbridge JD, et al. Methadone and perinatal outcomes: a retrospective
cohort study. Am J Obstet Gynecol. 2011;204:139.e1–139.e9.
20. Stewart RD, Nelson DB, Adhikari EH, et al. The obstetrical and neonatal impact of maternal opioid
detoxification in pregnancy. Am J Obstet Gynecol. 2013;209:267.e1–267.e5.
21. Sharpe C, Kuschel C. Outcomes of infants born to mothers receiving methadone for pain management
in pregnancy. Arch Dis Child Fetal Neonatal Ed. 2004;89:F33–F36.
22. Young JL, Martin PR. Treatment of opioid dependence in the setting of pregnancy. Psychiatr Clin
North Am. 2012; 34:441–460.
23. Raith W, Kutschera J, Müller W, et al. Active ear acupuncture points in neonates with neonatal
abstinence syndrome (NAS). Am J Chin Med. 2011;39:29–37.
24. Liu AJ, Jones MP, Murray H, et al. Perinatal risk factors for the neonatal abstinence syndrome in
infants born to women on methadone maintenance therapy. Aust N Z J Obstet Gynaecol. 2010;50:253–
258.
25. Jansson LM, Velez M. Neonatal abstinence syndrome. Curr Opin Pediatr. 2012;24:252–258.
26. Patrick SW, Schumacher RE, Benneyworth BD, et al. Neonatal abstinence syndrome and associated
health care expenditures: United States, 2000-2009. JAMA. 2012;307:1934–1940.
27. Hudak ML, Tan RC; and the Committee on Drugs; Committee on Fetus and Newborn. Neonatal drug
withdrawal. Pediatrics. 2012;129:e540–e560.
28. Logan BA, Brown MS, Hayes MJ. Neonatal abstinence syndrome: treatment and pediatric outcomes.
Clin Obstet Gynecol. 2013;56:186–192.
29. Welle-Strand GK, Skurtveit S, Jansson LM, et al. Breastfeeding reduces the need for withdrawal
treatment in opioid-exposed infants. Acta Paediatr. 2013;102:1060–1066.
30. Cleary BJ, Donnelly J, Strawbridge J, et al. Methadone dose and neonatal abstinence syndrome-
systematic review and meta-analysis. Addiction. 2008;105:2071–2084.
31. Seligman NS, Almario CV, Hayes EJ, et al. Relationship between maternal methadone dose at delivery
and neonatal abstinence syndrome. J Pediatr. 2010;157:428–433.
32. Dryden C, Young D, Hepburn M, et al. Maternal methadone use in pregnancy: factors associated with
the development of neonatal abstinence syndrome and implications for healthcare resources. BJOG.
2009;116:665–671.
33. McCarthy JJ. Intrauterine abstinence syndrome (IAS) during buprenorphine inductions and methadone
tapers: can we assure the safety of the fetus? J Matern Fetal Neonatal Med. 2012;25:109–112.
34. Mitra S, Sinatra RS. Perioperative management of acute pain in the opioid-dependent patient.
Anesthesiology. 2004;101:212–227.
35. Stromer W, Michaeli K, Sandner-Kiesling A. Perioperative pain therapy in opioid abuse. Eur J
Anaesthesiol. 2013;30:55–64.
36. Alford DP, Compton P, Samet JH. Acute pain management for patients receiving maintenance
methadone or buprenorphine therapy. Ann Intern Med. 2006;144:127–134.
37. Meyer M, Paranya G, Keefer Norris A, et al. Intrapartum and postpartum analgesia for women
maintained on buprenorphine during pregnancy. Eur J Pain. 2010;14:939–943.
38. Meyer M, Wagner K, Benvenuto A, et al. Intrapartum and postpartum analgesia for women
maintained on methadone during pregnancy. Obstet Gynecol. 2007;110(suppl 1):261–266.
39. Rapp SE, Ready LB, Nessly ML. Acute pain management in patients with prior opioid consumption: a
case-controlled retrospective review. Pain. 1995;61:195–201.
40. Walker JM, Farney RJ, Rhondeau SM, et al. Chronic opioid use is a risk factor for the development of
central sleep apnea and ataxic breathing. J Clin Sleep Med. 2007;3:455–461.
41. Mishriky BM, George RB, Habib AS. Transversus abdominis plane block for analgesia after cesarean
delivery: a systematic review and meta-analysis. Can J Anaesth. 2012;59:766–778.
42. Rackelboom T, Le Strat S, Silvera S, et al. Improving continuous wound infusion effectiveness for
postoperative analgesia after cesarean delivery: a randomized controlled trial. Obstet Gynecol.
2010;116:893–900.
43. Vercauteren M, Vereecken K, La Malfa M, et al. Cost-effectiveness of analgesia after caesarean
section: a comparison of intrathecal morphine and epidural PCA. Acta Anaesthesiol Scand.
2002;46:85–89.
44. de Leon-Casasola OA, Myers DP, Donaparthi S, et al. A comparison of postoperative epidural
analgesia between patients with chronic cancer taking high doses of oral opioids versus opioid-naive
patients. Anesth Analg. 1993;76:302–307.
45. Pan PH. Post cesarean delivery pain management: multimodal approach. Int J Obstet Anesth.
2006;15:185–188.
46. Young JL, Lockhart EM, Baysinger, CL. Anesthetic and obstetric management of the opioid-
dependent parturient. Int Anesthesiol Clin. 2014;52:67–85.
47. American Academy of Pediatrics Committee on Drugs. Neonatal drug withdrawal. Pediatrics.
1998;101:1079–1088.
48. Jahr JS, Lee VK. Intravenous acetaminophen. Anesthesiol Clin. 2010;28:619–645.
49. Moore A, Costello J, Wieczorek P, et al. Gabapentin improves postcesarean delivery pain
management: a randomized, placebo-controlled trial. Anesth Analg. 2011;112:167–173.
50. Yanagidate F, Hamaya Y, Dohi S. Clonidine premedication reduces maternal requirement for
intravenous morphine after cesarean delivery without affecting newborn’s outcome. Reg Anesth Pain
Med. 2001;26:461–467.
51. Bauchat JR, Higgins N, Wojciechowski KG, et al. Low-dose ketamine with multimodal postcesarean
delivery analgesia: a randomized controlled trial. Int J Obstet Anesth. 2011;20:3–9.
Maternal Morbidity and Mortality
Jill M. Mhyre
I. Maternal mortality
A. Definitions
B. Epidemiology
C. Etiologies
D. Risk factors
II. Severe maternal morbidity
A. Definitions
B. Epidemiology
III. Prevention and lessons learned
A. Confidential enquiries into maternal death
B. Preventable factors
C. Morbidity surveillance systems
IV. Anesthesia-related maternal mortality
A. Definitions
B. Epidemiology
C. Etiologies
KEYPOINTS
1. Anesthesia is rarely the direct cause of maternal death; nevertheless,
anesthesia providers have the potential to improve overall patient safety
through multidisciplinary collaboration to deliver high-quality peripartum
care.
2. Cardiovascular disease is the most common cause of maternal death, but
hemorrhage, hypertensive disorders of pregnancy, venous
thromboembolism, and sepsis appear to be the most preventable.
3. Preexisting medical conditions increase maternal risk, but deaths occur in
previously healthy women.
4. Failed airway management is the most common cause of maternal death
with general anesthesia. Airway management drills can help maintain
optimal skills, equipment, and preparation for actual airway management
emergencies.
5. High neuraxial block is the leading cause of maternal death from
neuraxial anesthesia. The potential need for airway rescue and
resuscitation should be anticipated with every neuraxial block placement.
OVER THE PAST 50 YEARS, anesthesia has become remarkably safe; however,
pregnancy continues to lead to unacceptable rates of maternal harm. The primary
reason to analyze maternal mortality and morbidity is to identify opportunities to
improve safety for future patients. Patterns of preventable maternal morbidity
and mortality point to priorities for individual clinical care as well as health
system improvements. Anesthesiologists, with expertise in physiology,
resuscitation, critical care, and high reliability systems, are well-positioned to
lead multidisciplinary teams to implement solutions that will ensure future
maternal and perinatal patient safety.
I. Maternal mortality
A. Definitions
1. A maternal death is defined by the World Health
Organization (WHO) as a death of women while pregnant or
within 42 days of termination of pregnancy, irrespective of the
duration and site of the pregnancy, from any cause related to
or aggravated by the pregnancy or its management, but not
from accidental or incidental causes.1
2. Late maternal death transpires more than 42 days, but less
than 1 year after termination of pregnancy. Late maternal
deaths are excluded from official maternal mortality ratio
(MMR) statistics for the purpose of international comparison.
3. A direct maternal death results from obstetric complications
of the pregnant state or from interventions, omissions,
incorrect treatment, or a chain of events resulting from any of
the above.
4. An indirect maternal death results from previous existing
disease or disease that developed during pregnancy and which
is not due to direct obstetric causes, but was aggravated by the
physiologic effects of pregnancy.
5. A pregnancy-related death, classified by the United States
Centers for Disease Control and Prevention, refers to the
death of a woman while pregnant or within 1 year of the end
of her pregnancy, from any cause related to or aggravated by
her pregnancy, its management, but not from accidental or
incidental causes.
B. Epidemiology
1. The WHO defines the MMR as the number of direct and
indirect maternal deaths per 100,000 live births. The MMR
declined to 210 maternal deaths per 100,000 live births in
2013, from 380 in 1990.1
2. A total of 289,000 women died while pregnant or within 42
days of the end of pregnancy in 2013.1 The majority of global
maternal deaths take place in Sub-Saharan Africa (62%) or
South Asia (24%).1
3. The 2013 MMR was 16 per 100,000 live births in the
developed world and 230 per 100,000 in developing regions,
a 14-fold difference.1
4. The U.S. MMR was 13.8 between 2006 and 2010, whereas
the pregnancy-related mortality ratio was 16.2
5. Contemporaneous MMRs in other developed countries were
5.6 in the United Kingdom (2009 to 2011),3 10.3 in France
(2007 to 2009),4 6.8 in Australia (2006 to 2010), and 6.1 in
Canada (2009 to 2011).
C. Etiologies
Although direct causes of maternal death claim the most lives
globally (e.g., hemorrhage, hypertensive disorders, and infection),
an increasing proportion deaths in the developed world are
attributed to indirect causes (e.g., cardiovascular disease and
other medical conditions exacerbated by pregnancy). Figure 32.1
illustrates cause-specific proportional pregnancy-related mortality
for the United States between 1987 and 2010.
D. Risk factors
The most potent risk factors for maternal mortality and
severe morbidity are clinically significant medical conditions
that predate the pregnancy.3,8,13,14
1. Three-quarters of women who died in the United Kingdom
between 2009 and 2012 had a preexisting medical condition.3
2. Preconception counseling and intensive multidisciplinary
antepartum and intrapartum care may improve outcomes for
women with serious medical or mental health conditions that
may be aggravated by pregnancy.3,15
3. Insufficient prenatal care may result in suboptimal
management of existing medical conditions or delay the
diagnosis of pregnancy complications.2,8
4. Advanced maternal age increases maternal risk,16 with a
linear trend evident for each 5-year increase in maternal age
beyond 34 years.15
5. Racial and ethnic minority groups experience increased
risk.8,15
a. In the United States, non-Hispanic black race confers a
threefold increase in the risk of maternal death compared
with non-Hispanic white women.2,17 The disparity is
exacerbated by increasing maternal age. Non-Hispanic
black women older than 25 years have a fourfold
increased risk of death, as illustrated in Figure 32.2.2
b. Immigrants and nonnative speakers experience high
rates of both maternal death and substandard care.15,18,19
6. Obesity (body mass index [BMI] ≥30 kg per m2) increases
risk of death when severe pregnancy complications develop.18
Conversely, very low body weight (<60 kg) may increase risk
of hemorrhage-related mortality due to extremely small
circulating blood volume.3,8
7. Multifetal pregnancies increase risk of serious complications
of pregnancy and maternal death.15,20
8. Cesarean delivery is associated with maternal death.21,22
a. Most maternal deaths following CD are caused by the
underlying medical or obstetric disease that indicated the
CD; rarely, cesarean surgery causes fatal infection,
hemorrhage, or venous thromboembolism.9
b. Cesarean deliveries increase the risk of placenta accreta in
subsequent pregnancies and thus contribute to the overall
population level risk of maternal death.23
II. Severe maternal morbidity
A. Definitions
1. Severe morbidity refers to serious complications in
pregnancy with the potential to cause end-organ injury or
maternal death. A practical definition of severe morbidity is
either intensive care unit (ICU) admission or the
administration of ≥4 units of erythrocytes or whole blood, or
both.3
2. Near-miss morbidity includes life-threatening complications
that result in severe end-organ dysfunction or failure.14,24
B. Epidemiology
1. Severe morbidity complicates 1.6% of pregnancies in the
United States, corresponding to more than 60,000 pregnancies
per year.25
2. Near-miss morbidity develops in approximately 0.13% to
0.2% of US hospitalizations for delivery.14,26
CLINICAL PEARL For each maternal death, approximately 50
to 100 women experience a severe morbidity.
C. Etiologies
1. Potential etiologies of anesthesia-related maternal death are
listed in Table 32.3.
REFERENCES
1. World Health Organization. Trends in Maternal Mortality: 1990 to 2013: Estimates by WHO,
UNICEF, UNFPA, The World Bank and the United Nations Population Division. Geneva, Switzerland:
Department of Reproductive Health and Research; 2014.
2. Creanga AA, Berg CJ, Syverson C, et al. Pregnancy-related mortality in the United States, 2006–2010.
Obstet Gynecol. 2015;125:5–12.
3. Callaghan WM, Grobman WA, Kilpatrick SJ, et al. Facility-based identification of women with severe
maternal morbidity: it is time to start. Obstet Gynecol. 2014;123:978–981.
4. Saucedo M, Deneux-Tharaux C, Bouvier-Colle MH; for French National Experts Committee on
Maternal Mortality. Ten years of confidential inquiries into maternal deaths in France, 1998-2007.
Obstet Gynecol. 2013;122:752–760.
5. Knight M, Kenyon S, Brocklehurst P, et al, eds. Saving Lives, Improving Mothers’ Care: Lessons
Learned to Inform Future Maternity Care from the UK and Ireland Confidential Enquiries into
Maternal Deaths and Morbidity 2009-2012. Oxford, United Kingdom: National Perinatal
Epidemiology Unit, University of Oxford; 2014.
6. Grotegut CA, Kuklina EV, Anstrom KJ, et al. Factors associated with the change in prevalence of
cardiomyopathy at delivery in the period 2000-2009: a population-based prevalence study. BJOG.
2014;121:1386–1394.
7. Acosta CD, Kurinczuk JJ, Lucas DN, et al. Severe maternal sepsis in the UK, 2011–2012: a national
case-control study. PLoS Med. 2014;11:e1001672.
8. Nair M, Kurinczuk JJ, Brocklehurst P, et al. Factors associated with maternal death from direct
pregnancy complications: a UK national case-control study. BJOG. 2015;122:653–662.
9. Clark SL, Belfort MA, Dildy GA, et al. Maternal death in the 21st century: causes, prevention, and
relationship to cesarean delivery. Am J Obstet Gynecol. 2008;199:36.e1–36.e5.
10. Berg CJ, Harper MA, Atkinson SM, et al. Preventability of pregnancy-related deaths: results of a
state-wide review. Obstet Gynecol. 2005;106:1228–1234.
11. California Department of Public Health. The California Pregnancy-Associated Mortality Review:
report from 2002 and 2003 Maternal Death Reviews. Stanford, CA: California Maternal Quality Care
Collaborative; 2011.
12. Clark SL, Christmas JT, Frye DR, et al. Maternal mortality in the United States: predictability and the
impact of protocols on fatal postcesarean pulmonary embolism and hypertension-related intracranial
hemorrhage. Am J Obstet Gynecol. 2014;211:32.e1–32.e9.
13. Bateman BT, Mhyre JM, Hernandez-Diaz S, et al. Development of a comorbidity index for use in
obstetric patients. Obstet Gynecol. 2013;122:957–965.
14. Mhyre JM, Bateman BT, Leffert LR. Influence of patient comorbidities on the risk of near-miss
maternal morbidity or mortality. Anesthesiology. 2011;115:963–972.
15. Cantwell R, Clutton-Brock T, Cooper G, et al. Saving mothers’ lives: reviewing maternal deaths to
make motherhood safer: 2006-2008: The Eight Report of the Confidential Enquiries into Maternal
Deaths in the United Kingdom. BJOG. 2011;118(suppl 1):1–203.
16. Callaghan WM, Berg CJ. Pregnancy-related mortality among women aged 35 years and older, United
States, 1991-1997. Obstet Gynecol. 2003;102:1015–1021.
17. Creanga AA, Bateman BT, Kuklina EV, et al. Racial and ethnic disparities in severe maternal
morbidity: a multistate analysis, 2008-2010. Am J Obstet Gynecol. 2014;210:435.e1–435.e8.
18. Kayem G, Kurinczuk J, Lewis G, et al. Risk factors for progression from severe maternal morbidity to
death: a national cohort study. PLoS One. 2011;6:e29077.
19. Creanga AA, Berg CJ, Syverson C, et al. Race, ethnicity, and nativity differentials in pregnancy-
related mortality in the United States: 1993-2006. Obstet Gynecol. 2012;120:261–268.
20. Walker MC, Murphy KE, Pan S, et al. Adverse maternal outcomes in multifetal pregnancies. BJOG.
2004;111:1294–1296.
21. Deneux-Tharaux C, Carmona E, Bouvier-Colle MH, et al. Postpartum maternal mortality and cesarean
delivery. Obstet Gynecol. 2006;108:541–548.
22. Liu S, Liston RM, Joseph KS, et al; for the Maternal Health Study Group of the Canadian Perinatal
Surveillance System. Maternal mortality and severe morbidity associated with low-risk planned
cesarean delivery versus planned vaginal delivery at term. CMAJ. 2007;176:455–460.
23. Solheim KN, Esakoff TF, Little SE, et al. The effect of cesarean delivery rates on the future incidence
of placenta previa, placenta accreta, and maternal mortality. J Matern Fetal Neonatal Med.
2011;24:1341–1346.
24. Say L, Pattinson RC, Gülmezoglu AM. WHO systematic review of maternal morbidity and mortality:
the prevalence of severe acute maternal morbidity (near miss). Reprod Health. 2004;1:3.
25. Callaghan WM, Creanga AA, Kuklina EV. Severe maternal morbidity among delivery and postpartum
hospitalizations in the United States. Obstet Gynecol. 2012;120:1029–1036.
26. Geller SE, Rosenberg D, Cox S, et al. A scoring system identified near-miss maternal morbidity during
pregnancy. J Clin Epidemiol. 2004;57:716–720.
27. Lewis G, ed. Saving Mothers’ Lives: Reviewing Maternal Deaths to Make Motherhood Safer — 2003-
2005. The Seventh Report of the Confidential Enquiries into Maternal Deaths in the United Kingdom.
London, United Kingdom: Confidential Enquiry into Maternal and Child Health; 2007.
28. MacKay AP, Berg CJ, Liu X, et al. Changes in pregnancy mortality ascertainment: United States,
1999-2005. Obstet Gynecol. 2011;118:104–110.
29. Callaghan WM. State-based maternal death reviews: assessing opportunities to alter outcomes. Am J
Obstet Gynecol. 2014;211:581–582.
30. Geller SE, Koch AR, Martin NJ, et al. Assessing preventability of maternal mortality in Illinois: 2002-
2012. Am J Obstet Gynecol. 2014;211:698.e1–698.e11.
31. Farquhar C, Sadler L, Masson V, et al. Beyond the numbers: classifying contributory factors and
potentially avoidable maternal deaths in New Zealand, 2006-2009. Am J Obstet Gynecol.
2011;205:331.e1–331.e8.
32. Mhyre JM. Maternal mortality. Curr Opin Anaesthesiol. 2012;25:277–285.
33. American College of Obstetricians and Gynecologists. Obstetric Care Consensus No. 2: levels of
maternal care. Obstet Gynecol. 2015;125:502–515.
34. Hankins GD, Clark SL, Pacheco LD, et al. Maternal mortality, near misses, and severe morbidity:
lowering rates through designated levels of maternity care. Obstet Gynecol. 2012;120:929–934.
35. D’Alton ME, Main EK, Menard MK, et al. The national partnership for maternal safety. Obstet
Gynecol. 2014;123:973–977.
36. Main EK, Menard MK. Maternal mortality: time for national action. Obstet Gynecol. 2013;122:735–
736.
37. Mhyre JM, D’Oria R, Hameed AB, et al. The maternal early warning criteria: a proposal from the
national partnership for maternal safety. Obstet Gynecol. 2014;124:782–786.
38. Kilpatrick SJ, Berg C, Bernstein P, et al. Standardized severe maternal morbidity review: rationale and
process. J Obstet Gynecol Neonatal Nurs. 2014;43:403–408.
39. Hawkins JL, Chang J, Palmer SK, et al. Anesthesia-related maternal mortality in the United States:
1979-2002. Obstet Gynecol. 2011;117:69–74.
40. Davies JM, Posner KL, Lee LA, et al. Liability associated with obstetric anesthesia: a closed claims
analysis. Anesthesiology. 2009;110:131–139.
41. D’Angelo R, Smiley RM, Riley ET, et al. Serious complications related to obstetric anesthesia: the
serious complication repository project of the Society for Obstetric Anesthesia and Perinatology.
Anesthesiology. 2014;120:1505-1512.
42. Mhyre JM, Riesner MN, Polley LS, et al. A series of anesthesia-related maternal deaths in Michigan,
1985-2003. Anesthesiology. 2007;106:1096–1104.
43. Lofsky A. Doctors company reviews maternal arrest cases. Anesth Patient Saf Found Newsl.
2007;22:28–30.
44. Mhyre JM. Why do pharmacologic test doses fail to identify the unintended intrathecal catheter in
obstetrics? Anesth Analg. 2013;116:4–5.
Guidelines
from National
Organizations
Guidelines from National Organizations
Kathryn J. Zuspan
I. Terms used to label guidance documents
II. American Society of Anesthesiologists as a leader in developing
guidance documents
III. “Practice Parameter” is the American Society of Anesthesiologists
term for guidance documents
A. Evidence-based practice parameters
B. Consensus-based American Society of Anesthesiologist
Practice Parameters
C. Other—statements, positions, and protocols
IV. How should Practice Parameters be used?
V. Limitations of guidance documents
VI. How to judge/compare documents
VII. National organizations with guidance relevant to obstetric
anesthesia
VIII. American Society of Anesthesiologists website access and relevant
documents
A. American Society of Anesthesiologist documents specific to
obstetric anesthesia practice
IX. American College of Obstetricians and Gynecologists
X. American Academy of Pediatrics and American College of
Obstetricians and Gynecologists Collaboration: Guidelines for
Perinatal Care
XI. Relevant documents from the American Society of Regional
Anesthesia and Pain Medicine
XII. Relevant document from the American Heart Association
XIII. Relevant document from the Society for Obstetric Anesthesia and
Perinatology
KEYPOINTS
1. Guidelines from organizations provide valuable information but cannot
guarantee a specific outcome. Interpretation and application of guidelines
takes place at the local level. A departure from recommendations may be
appropriate if the facts and circumstances show a physician’s care met his
or her duty to the patient.
2. All guidelines are not created equal. They can be based on evidence,
consensus, expert opinion, or any combination of these. They can provide
minimum requirements, recommendations for a range of strategies, and
statements to help in decision-making.
3. In evidence-based documents, every researched topic is discussed
followed by a grade referring to the amount of science supporting the
relationship (linkage) between the clinical intervention and clinical
outcome. Level 1 (supportive) has sufficient scientific evidence; level 2
(suggestive) has some evidence; level 3 (equivocal) has little or unclear
evidence; inconclusive, insufficient, or silent grades mean a linkage lacks
supporting evidence.
4. Knowledge of relevant guidelines is an important part of medical
practice.
5. Not all documents agree. The reader is encouraged to compare
documents to discover the reasons for discrepancies; to examine the dates
of acceptance, revision, and its references; and to check the source of
information.
REFERENCES
1. Williams MS, Davies JM. Medicolegal issues in obstetric anesthesia. In: Chestnut DH, ed. Chestnut’s
Obstetric Anesthesia: Principles and Practice. 5th ed. Philadelphia, PA: Elsevier Saunders; 2014:780–
781.
2. American Society of Anesthesiologists. Standards, guidelines, and statements: policy statement on
practice parameters. https://www.asahq.org/For-Members/Standards-Guidelines-and-Statements.aspx.
Accessed November 13, 2014.
3. American Society of Anesthesiologists. Potential conflict of interest policy.
https://www.asahq.org/For-Members/Publications-and-Research/Newsletter-
Articles/2010/May2010/competing-allegiances-equals-a-conflict-of-interest.aspxm. Accessed
November 18, 2014.
4. Weiskopf RB. Conflicts of interest in expert-authored practice parameters, standards, guidelines,
recommendations. Anesthesiology. 2010;113:751–752.
5. Djulbegovic B, Guyatt GH. Evidence-based practice is not synonymous with delivery of uniform
health care. JAMA. 2014;312:1293–1294.
6. American Society of Regional Anesthesia and Pain Medicine. Advisories and guidelines.
http://www.asra.com/advisory-guidelines. Accessed November 20, 2014.
7. Horlocker TT, Wedel DJ, Rowlingson JC, et al. Regional anesthesia in the patient receiving anti-
thrombotic or thrombolytic therapy: American Society of Regional Anesthesia and Pain Medicine
Evidence-Based Guidelines (Third Edition). Reg Anesth Pain Med. 2010;35:64–101.
8. Geerts WH, Berggvist D, Pineo GF, et al. Prevention of venous thromboembolism: American College
of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest. 2008;133(suppl
6):381S–453S.
9. American College of Obstetricians and Gynecologists. 2014 Compendium of selected publications.
http://www.sales.acog.org/2014-compendium-of-selected-publications-cd-rom-P498.aspx. Accessed
November 20, 2014.
10. Freeman RK, Cohen AW, Depp R III, et al. American College of Obstetrics and Gynecology Task
Force on Cesarean Delivery: evaluation of cesarean delivery.
http://archive.poughkeepsiejournal.com/assets/pdf/BK15725757.PDF. Accessed November 23, 2014.
11. American Society of Anesthesiologists. Standards, guidelines, and statements. Guidelines for
Neuraxial Anesthesia in Obstetrics. www.asahq.org/quality-and-practice-management/standards-and-
guidelines.aspx. Accessed November 9, 2015.
12. American Society of Anesthesiologists. Practice parameters (practice guidelines).
http://www.asahq.org/for-members/practice-management/practice-parameters.aspx. Accessed
November 20, 2014.
13. American Academy of Pediatrics, American College of Obstetricians and Gynecologists. Guidelines
for Perinatal Care. 7th ed. Washington, DC: American Academy of Pediatrics; 2012.
http://shop.aap.org/Guidelines-for-Perinatal-Care-7th-Edition-eBook. Accessed October 30, 2015.
14. Neal JM, Bernards CM, Butterworth JF IV, et al. ASRA practice advisory on local anesthetic systemic
toxicity. Reg Anesth Pain Med. 2010;35:152–161.
15. Neal JM, Bernards CM, Hadzic A, et al. ASRA practice advisory on neurologic complications in
regional anesthesia and pain medicine. Reg Anesth Pain Med. 2008;33:404–415.
16. Vanden Hoek TL, Morrison LJ, Shuster M, et al. Part 12: cardiac arrest in special situations: 2010
American Heart Association Guidelines for Cardiopulmonary Resuscitation and Emergency
Cardiovascular Care. Circulation. 2010;122(18 suppl 3):S829–S861.
17. Lipman S, Cohen S, Einav S, et al. The Society of Obstetric Anesthesia and Perinatology consensus
statement on the management of cardiac arrest in pregnancy. Anesth Analg. 2014;118:1003–1016.
Index
Page numbers followed by f and t indicate figures and tables, respectively.
Dalteparin, 487t
DCM. See Dilated cardiomyopathy
D-dimer, 592
Decreased placental perfusion, 438–439
Deep venous thrombosis, 216, 697
Desflurane, 475, 573
Dexamethasone, 462t
Diabetes insipidus (DI), 469
Diabetes mellitus (DM)
effect of, on pregnancy, 453–454
hyperglycemia, 453–454
effect of pregnancy on, 452–453
complications of, 452–453
diabetic nephropathy, 453
glycemic control, 452
GDM, 452
introduction, 451–452
management, during labor and delivery, 456–458
anesthetic management, 457–458
cesarean delivery, 457–458
glucose management, 457
labor analgesia, 457–458
timing of delivery, 456–457
management, during pregnancy, 454–456
diabetic ketoacidosis, 455–456
insulin, 454, 455t
with oral hypoglycemic agents, 454–455
renal disease and, 456
retinopathy and, 456
postpartum care, 458
insulin, 458
neonatal hypoglycemia, 458
renal disease and, 564
Type 1, 452
Type 2, 452
Diabetic ketoacidosis (DKA), 455–456
Diamorphine (heroin), 147–148, 366
DIC. See Disseminated intravascular coagulation
Diethylstilbestrol, 85t
Difficult airway management
airway assessment, 266–271
atlanto-occipital joint extension, 268, 269f
history, 266
jaw protrusion or mandibular protrusion test, 268, 268f
Mallampati classification, 266–267, 267f
mentohyoid distance, 268, 269f, 270f
mouth opening, 268
physical examination, 266
specific individual tests, 266
thyromental distance, 268
anatomic and physiologic changes contributing to, 265–266
airway changes, 265
cardiovascular changes, 266
gastrointestinal changes, 266
recommendations for airway management and, 266
respiratory changes, 266
anesthesia-related morbidity and mortality, 263–264
experience in United Kingdom, 264
experience in United States, 263–264
anesthetic management in obstetric patients with predicted difficult airway,
273–277
ASA practice guidelines, 273
with emphasis on awake intubation, 275–277
undergoing labor or operative delivery, 273–275
aspiration of gastric contents, 272
conclusion, 283
definitions, 261
difficult and failed intubation, 262
difficult laryngoscopy, 261
extubation and postanesthesia care unit airway issues, 283
failed intubation, 261
goals for airway management during pregnancy, 261–262
introduction, 260
management of pregnant patient with unanticipated difficult airway, 277–
283
failed intubation, 279–281
GlideScope, C-MAC VL and Airtraq VL, 279
maintenance of oxygenation/ventilation, 279–281
management of “cannot intubate, cannot ventilate” situation, 281–282
management with patient with critical airway and hypoxemia, 282–
283
tracheal intubation, step one, 277–279, 278f
tracheal intubation, step two, 277–279, 278f
maternal deaths and airway-related issues, 265
United Kingdom data, 265
United States data, 265
morbid obesity in pregnancy and airway, 271–272
supraglottic airway, 261
Digoxin, 31t
Dilated cardiomyopathy (DCM), 526
Dilutional anemia, 9
Dilutional coagulopathy, 319–320
Dinoprostol, 67
Dinoprostone (Cervidil), 68
Diphenhydramine, 370
Direct maternal death, 667
Disseminated intravascular coagulation (DIC), 320, 321t, 494–495
DKA. See Diabetic ketoacidosis
DM. See Diabetes mellitus
Domestic violence, 645
Doppler ultrasound, 312
Doppler velocimetry, 101–102
Doulas, 138
Droperidol, 475
Dührssen incisions, 299
Dyspnea, 197
Dysrhythmias. See Cardiac dysrhythmias
Halothane, 475
HELLP syndrome. See Hemolysis, elevated liver enzymes, low platelets
syndrome
Hemabate. See Carboprost
Hematologic disorders, 124–125
Hematologic system
in multiple gestation pregnancy, 291
in pregnancy, 9–10
Hemolysis, elevated liver enzymes, low platelets syndrome (HELLP syndrome),
577–578, 578t
Hemolytic uremic syndrome (HUS), 571
Hemophilia A, 490–491, 491t
Hemophilia B, 490–491, 491t
Hemorrhage. See Maternal hemorrhage; specific types of hemorrhage
Heparin, 31t, 411t, 488, 489t, 590, 592–593
Hepatic disease
anesthetics and, 573
characteristics, 572, 572t
diagnosis of liver disease, in pregnancy, 573
diseases exacerbated by pregnancy, 578–579
autoimmune hepatitis, 578
organ transplantation and, 578–579
viral hepatitis, 578
diseases unique to pregnancy, 574–578
acute fatty liver of pregnancy (AFLP), 571, 575–577, 576t
cholestasis of pregnancy, 574–575
hemolysis, elevated liver enzymes, low platelets syndrome (HELLP
syndrome), 577–578, 578t
hyperemesis gravidarum, 574
hepatic anatomy and physiology, in pregnancy, 573
hepatic function assessment, 573
introduction, 558–559
multidisciplinary team, 559
Hepatic function, 11, 12t, 573
Hepatitis
acute hepatitis, 122
anesthetic considerations, 123–124
autoimmune and viral, 578
chronic hepatitis, 123
neuraxial anesthesia for, 123
Hepatobiliary system, 442
Heroin. See Diamorphine
Herpes simplex virus (HSV-1 and HSV-2), 126
Herpes zoster (shingles), 128
HIV. See Human immunodeficiency virus
HOCM. See Hypertrophic obstructive cardiomyopathy
HSV-1. See Herpes simplex virus
HSV-2. See Herpes simplex virus
Human immunodeficiency virus (HIV)
anesthetic considerations, 125–126
cardiovascular disease, 124–125
endocrine disorders, 124–125
gastrointestinal involvement, 124–125
general anesthesia, 126
general considerations, 124
hematologic disorders, 124–125
neuraxial anesthesia, 125
neurologic disease, 124–125
pulmonary complications, 124
renal disease, 124–125
systemic manifestations, 124–125
transmission, from mother to fetus, 124
HUS. See Hemolytic uremic syndrome
Hydralazine, 31t, 444, 444t, 472
Hydromorphone, 366, 367–368, 367t
recommended single doses, for postcesarean analgesia, 368t, 369t
Hydrotherapy, 141–142
Hypercoagulability, 616–617
Hyperemesis gravidarum, 574
Hyperfibrinogenemia, 10
Hyperglycemia, 453–454. See also Diabetes mellitus
Hypertensive disorders
anesthetic considerations, 445–446
assessment of volume status, 445–446
blood pressure control, 446
coagulation in, 441, 446
delivery mode and anesthetic technique, 446–447
cesarean delivery, 447
vaginal delivery, 446–447
differential diagnosis and definitions, 436–437
chronic hypertension, 436
chronic hypertension with superimposed preeclampsia, 437
gestational hypertension, 436
preeclampsia, 436–437
epidemiology, 438
incidence, 438
maternal mortality, 438
neonatal mortality, 438
etiology, 438–439
antioxidants, 439
decreased placental perfusion, 438–439
genetic influences, 439
inflammatory mediators, 439
lifestyle modifications, 439
oxidative stress, 439
placental ischemia outcome, 439
prostaglandin imbalance, 439
vasoactive substances, 439
maternal death from, 669
obesity and, 615
obstetric management, 442–445
antihypertensive agents, 444t
blood pressure control, 443, 444t
prediction and prevention of preeclampsia, 442
seizure prophylaxis, 443
timing and route of delivery, 442–443
pathophysiology, 439–442
placenta and, 442
postpartum care, 448
analgesia, 448
blood pressure control, 448
fluid balance, 448
long-term sequelae, 448
seizure prophylaxis, 448
pregnancy-induced hypertension, 492–493
risk factors, 438, 438t
Hyperthermia, 186
Hyperthyroidism
antepartum considerations, 461–462
fetal goiter and Graves disease, 461, 464, 465
medications, 461
thyroid storm, 461–463
causes of, 460, 460t
clinical presentation, 460
definition and pathophysiology, 459–460
drugs, 462t
fetal outcome, 461
intrapartum and anesthetic considerations
cesarean delivery, 463–464
labor analgesia, 463
major concerns, 463
management, 461
physiology of, 460
Hypertrophic obstructive cardiomyopathy (HOCM), 526–527
Hypnosis, 140
Hypoglycemia, neonatal, 458
Hypopituitarism, 468, 468t
Hypoplastic left heart syndrome, 514–517
anesthetic management, 517
invasive monitoring, 517
pathophysiology, 515
peripartum considerations, 517
surgical correction, 515, 517
Hypotension, 184
focused cardiac ultrasound, 196–197
obesity and, 615–616
spontaneous intracranial, 382, 408
unexplained, 471
Hypothyroidism
antenatal care, 465
clinical presentation, 464
intrapartum and anesthetic considerations, 465–466
obstetric risk and complications, 465
pathophysiology, 464
treatment, 464
Hypoxemia, 7–8, 14
fetal compensatory measures to, 25
management of patients with critical airway, 282–283
transient, 344–346
Hysterectomy, 329
Ibuprofen, 429
Idiopathic dilated cardiomyopathy (IDC), 526
Idiopathic intracranial hypertension (pseudotumor cerebri), 546–547
Iliohypogastric and ilioinguinal peripheral nerve blocks, 371, 663
Immune function, 10
Implied consent, 75
Incomplete breech, 296–297, 296f
Indirect maternal death, 667
Indomethacin, 61
Influenza, 120
Informed consent
background on, 74–75
delegation of, 77
exceptions to, 78–79
failure to obtain, 79
implied consent, 75
laboring women and, 75
law and, 79
from minors, 77
presentation of information and risk, 76
purpose of informed consent discussion, 76
refusal to be informed, 76–77
refusal/withdrawal of consent, 77
withholding information, 76
written consent, 78
Inhalation agents, 428
Inhalation techniques, 143–145
Inherited thrombophilias, 485–486, 487t
Instrumental vaginal delivery, 182–183
neuraxial analgesia and, 227–229
Insulin
diabetes mellitus (DM) management with, 454, 455t
glucose management, in labor and delivery, 457
postpartum care, 458
Intermittent auscultation, 110
Intermittent bolus injection, 180
Intra-arterial monitoring, 519
Intracerebral hemorrhage, 381
Intracranial hemorrhage, 380–381
anesthetic management, 548
clinical issues, 547–548
obstetric management, 548
overview, 547
Intracranial neoplasms
anesthetic management, 545–546
clinical issues, 544–545
obstetric management, 545
signs and symptoms, 544–545, 545t
Intracranial pressure measurement, 209–211
Intraoperative cell salvage, 317–318
Intrapartum emergencies
amniotic fluid embolism (AFE), 336–338, 337t
preterm labor and delivery, 333–334
shoulder dystocia, 334–336
umbilical cord compression/prolapse, 336
Intrapartum evaluation, 347–348
Intrathecal catheter, 383
Intrathecal opioids, 368–369, 369t
Intrathecal saline, 383
Ischemic heart disease, in pregnancy, 529
Isoflurane, 573
Joint mobility, 13
Nalbuphine, 158
Neonatal abstinence syndrome (NAS), 659–660
Neonatal hypoglycemia, 458
Neonatal resuscitation. See Newborn resuscitation
Neonatal shock, 356
Neonate evaluation
Apgar score, 348–349, 348t
umbilical cord blood gas measurements, 349
Neoplasm, 381
intracranial, 544–546
Neostigmine, 112, 180, 369
Nerve conduction studies, 401–402
Neuraxial analgesia
ambulation during labor, 183
advantages, 183
disadvantages, 183
suggested guidelines for, 183t
analgesia for vaginal delivery, 182–183
instrumental vaginal delivery, 182–183
spontaneous vaginal delivery, 182
in cesarean delivery, with obesity, 621
contraindications, 165, 165t
description of technique, 169–175
combined spinal-epidural technique, 173–174, 501–502
continuous spinal technique, 175
epidural technique, 169–173
fetal malposition, labor, and, 229
impact on obstetric outcomes, 221–234
ambulation, 232
benefits, 222t
breastfeeding success rates, 233
cesarean delivery, 230–231
conclusion, 234
effects of neuraxial analgesia on progress of labor, 222–223
first stage of labor, 223–224
instrumental vaginal delivery, 227–229
introduction, 221–222
maternal fever rates, 232–233
oxytocin augmentation, 231–232
second stage of labor, 225–226
indications, 164–165
local anesthetics, choice of, 177–180
concentration and dose considerations, 177–179
use of adjuvants in labor analgesia, 179–180
maintenance of analgesia, 180–182
continuous epidural infusion, 180–181
continuous spinal analgesia, 182
intermittent bolus injection, 180
patient-controlled epidural analgesia, 181, 181t
maternal perineal injury and, 229
maternal request, 164–165
preparation, 167–169
aspiration prophylaxis, 167t
evaluation and consent, 167–168
intrapartum platelet counts and blood type and screen, 168
intravenous access, 168
monitoring, 168–169
procedure checklist, 167t
rationale for choice of technique, 166–167
anesthetic considerations, 167
maternal coexisting disease, 166
obstetric considerations, 166
side effects and complications, 184–187
accidental IV local anesthetic, 186–187
back pain, 186
epidural hematoma and abscess, 187
failed analgesia, 184–185
fetal heart rate abnormalities, 186
hypotension, 184
maternal hyperthermia, 186
meningitis, 187
motor block, subdural, and high/total spinal block, 186
neurologic deficits, 187
treatment for pruritus, 184
unintended dural puncture, 185
urinary retention, 186
Neuraxial anatomy
difficult identification of ligamentum flavum, 164
engorgement of epidural veins, 164
higher level of apex of thoracic kyphosis, 164
reduction in intervertebral gap, 162–163
widening and rotation of pelvis, 163
Neuraxial anesthesia, 13
anesthetic management in obstetric patients with predicted difficult airway,
274
ASA on, 693–694
for chorioamnionitis, 119
for febrile parturient, 131
for hepatitis, 123
HIV, 125
obesity and cesarean delivery, 621
pain pathways, during labor, 162
thoracic, 587–588, 588f
thrombophilia and anticoagulated patient, 488, 489t
tubal sterilizations and, 423
ultrasound-guided neuraxial anesthesia, 202–208
Neuraxial blocks
guidelines for administering, 411–412, 411t
infectious complications of, 408–410
in vaginal delivery, with obesity, 619, 620
Neurologic and neuromuscular diseases
anatomic disease
idiopathic intracranial hypertension (pseudotumor cerebri), 546–547
intracranial neoplasms, 544–546
maternal central nervous system shunts, 547
scoliosis, 535–540
spina bifida, 543–544
spinal cord injury, 540–543
spinal surgery, 540
conclusion, 554
epilepsy
anesthetic management, 554
classification of seizure disorders, 553t
obstetric management, 553–554
immunologic disease
Landry-Guillain-Barré, 552–553
multiple sclerosis (MS), 549–550
myasthenia gravis (MG), 550–552
vascular disease
cortical vein thrombosis, 548
intracranial hemorrhage, 547–548
Neurologic deficits, following labor and delivery
basic anatomy, 393–394
common obstetric neuropathies, 394–397
femoral nerve, 395
lateral femoral cutaneous nerve of thigh, 394–395
obstetric nerve injury and implications, 395t
obturator nerve, 395
peripheral nerve injuries, 394t
sciatic nerve, 395–397
sensory innervation of lower extremities, 396f
diagnosis and treatment of neuropathies, 400–402
electromyography, 400–401, 401f, 401t
electrophysiologic studies, 402
nerve conduction studies, 401–402
epidural hematoma, 410–411
history and initial evaluation, 392–393
causes of postpartum neurologic injury, 393
extent of injury, 393
relevant questions during evaluation, 392–393
infectious complications of neuraxial blocks, 408–410
epidural abscess, 409–410
postdural puncture meningitis, 408–409
ischemic injury to spinal cord, 398–399
artery of Adamkiewicz, 398–399
blood supply to spinal cord, 398, 398t
diagnosis of spinal cord ischemia, 399
lumbar arteries, 399
lesion types, 399–400
chemical injury, 399
direct nerve trauma, 399–400
neurologic injury, 390–392
frequency of transient and permanent neurologic deficits, 391t
incidence, 390–392
peripheral nerve injuries, 402–403
differential diagnosis, 403t
neurologist consultation and imaging studies, 402–403
preexisting causes, 402
recommendations, 412
spinal fluid leakage, 404–408
autologous epidural blood patch, 405
complications related to, 404–408
imaging studies, 405
intracranial hematomas, 407–408
postdural puncture headache, 404, 405f, 406–408
prophylactic epidural blood patch, 407
seizures, 408
spontaneous intracranial hypotension, 408
Neurosurgical procedures, 88–89
Newborn resuscitation
anticipating depressed newborn, 346–348
antepartum evaluation, 346–347
antepartum factors associated with resuscitation, 347t
intrapartum evaluation, 347–348
intrapartum factors associated with resuscitation, 347t
discontinuation of resuscitative efforts, 357
evaluating neonate, 348–349
medications, 355–357, 355t
epinephrine, 355–356
sodium bicarbonate, 356–357
volume expanders, 356
neonatal adaptations to extrauterine life, 344–346
fetal cardiovascular and pulmonary physiology, 344, 345f
normal peripartum transition to extrauterine life, 344
prolonged hypoxemia/acidosis and failure to transition, 344–346
neonatal resuscitation
assisted ventilation, 350–353
in breech presentation, 303
chest compression, 354, 355f
considerations for establishing ventilation, 353
endotracheal intubation, 351–353, 352f, 353t
initial resuscitation, 350
maintainance of normothermia, 350
oxygen administration, 353–354
preparation, 349, 350t
protocol, 351f
resuscitation algorithm, 350
special resuscitation circumstances, 357–360
magnesium toxicity, 360
meconium-stained amniotic fluid, 357–358
opioid-induced respiratory depression, 359
premature infants, 358–359
umbilical vein catheterization, 354–355
New York Heart Association (NYHA), 498, 498t
Nifedipine, 59, 444t, 445
Nitric oxide, 519
Nitrofurantoin, 31t
Nitroglycerin, 444t, 445, 475
anesthetic considerations, 63
in breech presentation, 298
mechanism of action, 62
route of administration/dose, 62
toxicity/side effects, 63
uses, 62
Nitroprusside, 474
Nitrous oxide, 85, 519, 695
Non-neuraxial analgesic techniques, 137
non-neuraxial pharmacologic methods of pain relief, 143–158
inhalation techniques, 143–145
non-opioid analgesia and sedatives, 145–147
opioid analgesia, 147–158, 179
nonpharmacologic methods of pain relief, 138–143
antenatal health education, 138
complementary therapies, 139–141
hydrotherapy, 141–142
relaxation techniques, 138
sterile water injection, 142–143, 143f
support, during labor, 138
transcutaneous electrical nerve stimulation (TENS), 142
Non-neuraxial pharmacologic methods of pain relief, 143–158
Nonobstetric surgery, during pregnancy
alterations in maternal physiology, 84
incidence and anesthetic concerns, 83–84
intraoperative anesthetic management, 87–88
maintenance of fetal oxygenation, 84
postoperative care, 88
preoperative plan and counseling, 86–87
prevention and treatment of preterm labor, 84
principles for anesthetic management, 86t
special situations, 88–91
cardiac surgery requiring cardiopulmonary bypass, 89
fetal interventions, 89–91, 90t
laparoscopic surgeries, 89
neurosurgical procedures, 88–89
trauma, 88
teratogenicity of anesthetic agents, 84–85, 86t
Non-opioid analgesia and sedatives, 145–147
Nonpharmacologic methods of pain relief, 138–143
Nonreassuring fetal status
diagnosis, 310–313
Doppler ultrasound, 312
fetal heart rate interpretation, 310
fetal heart rate monitoring, 310–311
fetal heart rate pattern, 310
presence of meconium, 311
umbilical cord gases, 311
fetal distress, 307–308
pathophysiology, 308–310
basic causes of fetal compromise, 309t
fetal acid–base balance and asphyxia, 309–310
fetal asphyxia causes, 309
fetal response to asphyxia, 310
treatment, 312–315
anesthetic management, 313–315
maternal safety, 314–315
obstetric management, 312–313
Nonsteroidal anti-inflammatory drugs (NSAIDs), 372, 490t, 664
Nonstress test (NST), 99
NSAIDs. See Nonsteroidal anti-inflammatory drugs
NST. See Nonstress test
NYHA. See New York Heart Association
Obesity, in pregnancy
anesthesia for obese pregnant women, 617–618
general considerations, 618
obesity and maternal mortality, 617
patient evaluation, 617
asthma and, 614
cardiomyopathy and, 615
cesarean delivery and, 620–624
anesthetic plan, 620–621
general anesthesia, 621–623
general considerations, 620
maternal morbidity and mortality, 620
neuraxial anesthesia, 621
operating room considerations, 623–624
surgical considerations, 624
cost and, 627
definition and demographics, 611–612
high-risk patient, 610–612
anesthetic challenges, 610
maternal comorbidities, 610, 611t
obstetric risk and outcomes, 610–611, 612t
hypertensive disorders and, 615
hypotension and, 615–616
morbid obesity and airway, 271–272
newborns and, 626–627
fetal anomalies, 626
fetal macrosomia, 626
neonatal care, 627
preterm delivery, 626
stillbirth and neonatal deaths, 626–627
physiologic changes, 612–617
cardiovascular system, 615–616
endocrine system, 616
gastrointestinal system, 617
hypercoagulability, 616–617
pulmonary system, 612–614
postoperative care, 625–626
general considerations, 625
postoperative monitoring, 625
pulmonary considerations, 625
thromboprophylaxis, 625–626
postoperative wound infection, 624
pregnancy, after bariatric surgery, 627
projected prevalence of, by 2025, 271f
summary of, 627–628
vaginal labor and delivery, 618–620
epidural analgesia, 618–619
epidural catheter placement, 619–620
labor analgesia, 618
neuraxial blocks, cephalad spread of, 620
neuraxial blocks, placement, 619
venous thromboembolism and, 616–617
Obesity hypoventilation syndrome (OHS), 614
Obstetric emergencies
categories of, 307
intrapartum emergencies, 333–338
nonreassuring fetal status, 307–315
peripartum bleeding, 315–333
Obstetric medications. See Tocolytic medications; Uterotonic medications;
specific medications
Obstetric nerve injury and implications, 395t
Obstructive sleep apnea (OSA), 598–599, 614f
Obturator nerve, 395
OCT. See Oxytocin challenge test
OHS. See Obesity hypoventilation syndrome
Ondansetron, 31t
ONSD. See Optic nerve sheath diameter
Opioid analgesia, 147–158, 179
dose ranges, 179t
Opioid dependent parturient, management of
introduction, 655
neonatal abstinence syndrome (NAS), 659–660
obstetric management, 655–659
algorithm for, 657f
chronic pain, in pregnancy, 659
identification, 658
peripartum, 659
rates of dependency, 656f
risk of dependency, in pregnancy, 655, 658
treatment, 658–659
pain management during peripartum period, 660–664
anesthetic management of cesarean delivery, 661
baseline long-acting opioid management, 661
opioid tolerance, 660–661
pain management for labor and delivery, 661
postcesarean delivery analgesia, 661, 664
summary of studies reporting peripartum pain management, 662t
summary, 664
Opioid-induced respiratory depression, 359
Opioid partial agonists, 157–158
Opioids, 39, 110
adjuncts, 368, 369
epidural, 367–368, 368t
for epidural anesthesia, in cesarean delivery, 250
intrathecal, 368–369, 369t
in postcesarean analgesia, 366–370
side effects, 369–370
for spinal anesthesia, in cesarean delivery, 253
systemic, 366–367
Optic nerve sheath diameter (ONSD). See Intracranial pressure measurement
Oral fiberoptic intubation technique, 276–277
Oral hypoglycemic agents, 454–455
OSA. See Obstructive sleep apnea
Ovarian arteries, 21
Oxycodone, 157
Oxymorphone, 366, 367t
Oxytocin, 13
anesthetic considerations, 65
mechanism of action, 64
neuraxial analgesia and augmentation of, 231–232
route of administration/dose, 64
toxicity/side effects, 65
uses, 64
for uterine atony, 328
Oxytocin antagonist (atosiban), 63
Oxytocin challenge test (OCT), 99
Pain, 137
ASA Statement on Pain Relief during Labor, 693
back pain, 49, 186
gate control theory, 139f
non-neuraxial pharmacologic methods of pain relief, 143–158
nonpharmacologic methods of pain relief, 138–143
opioid dependency and management of, 659–664
pathways, during labor, 162
uncontrolled maternal pain, 8
Pancreatic function, 12–13
Pancuronium, 475
Panniculus, 624
Paracervical block, 112
Paresthesia, 399–400
Parvovirus B19, 127
Patent ductus arteriosus (PDA), 521
Patient-controlled epidural analgesia, 181, 181t
PDA. See Patent ductus arteriosus
Pelvic/abdominal ultrasound, 213–215
Penetrating injury, 646–647
Penicillin, 31t
Pentazocine, 475
Perimortem cesarean delivery, 647
Peripartum bleeding
antepartum hemorrhage, 321–327
management of obstetric hemorrhage, 315–321
acute normovolemic hemodilution, 318
antepartum donation and autologous transfusion, 317
clinical signs of hemorrhagic shock, 315t
complications, 318
fundamentals of obstetric hemorrhage, 315–316
intraoperative cell salvage, 317–318
key management considerations, 316
massive blood loss and transfusion, 318–321, 318t
transfusion, 316–317
postpartum hemorrhage, 327–333
Peripartum cardiomyopathy (PPCM), 524–526
Peripheral nerve fiber classification, 38
Peripheral nerve injuries, 394t, 402–403, 403t
Peroneal nerve, 397f
Pharmacokinetics, 40–41
Phenobarbital, 462t
Phenothiazines, 31t, 110
Phenoxybenzamine, 474
Phentolamine, 475
Phenylephrine, 39
Phenytoin, 85t
Pheochromocytoma, 178, 473–475
Phosphodiesterase inhibitors, 519
PI. See Pulsatility index
Pickwickian syndrome. See Obesity hypoventilation syndrome
Pituitary disorders
introduction, 466
pituitary adenomas, 466–468
acromegaly, 466–467
antepartum maternal risk, 467
classification, 466
clinical considerations, 466–467
intrapartum considerations, 467–468
macroadenoma expansion, 466
management, 467
prolactinomas, 466
pituitary insufficiency, during pregnancy, 467, 468–469
antepartum risks, 469
diabetes insipidus, 469
hypopituitarism, 468, 468t
intrapartum considerations, 469
lymphocytic hypophysitis, 469
Sheehan syndrome, 469
Pituitary function, 13
Placenta, 19–20. See also Uteroplacental anatomy
abnormal placentation types, 331f
abruptio placentae, 324–326
hypertensive disorders and, 442
local anesthetics and, 41
macroscopically, 21
microscopically, 21
retained, 329–330
Placenta accreta
anesthetic management, 331–332
definition, 330
diagnosis, 330–331
epidemiology, 330
interventional radiology techniques, 332
obstetric management, 331
Placental abruption, 643
Placental ischemia, 439
Placenta previa
anesthetic management, 323
cesarean delivery and, 322–323, 322t
definition, 321
diagnosis, 322
epidemiology, 322
obstetric management, 322–323
Plasma progesterone concentrations, 418
Platelet count and function, 9, 168, 481, 493
Pneumonia, 119–120
Postcesarean analgesia
epidural anesthesia for, 367–368, 370–371
introduction, 365–366
medications, 366–373
acetaminophen, 372
in breast milk, 373
gabapentin, 373
ketamine, 369, 372
local anesthetics, 370–371
magnesium, 369, 372
nonsteroidal anti-inflammatory drugs, 372
opioids, 366–370
multimodal therapy, 366
summary, 373–374
Postdural puncture headache
characteristics of, 404
diagnosis, 379–380
clinical characteristics, 379–380
occurrence of, 379
differential diagnosis, 380–382, 380t
benign intracranial hypertension (pseudotumor cerebri), 382
cerebral venous and sinus thrombosis, 381
intracranial hemorrhage, 380–381
lactation headache, 382
medications/substance withdrawal, 381
meningitis, 382
migraine, 380
neoplasm, 381
posterior reversible leukoencephalopathy syndrome, 382
preeclampsia, 381
spontaneous intracranial hypotension, 382
tension headache, 380
incidence of, 404f
intracranial hematomas and, 407–408
location of, 405f
managing, 406–407
pathophysiology, 378
prevention and treatment, 382–384
recommendations, 384–385
ambulation, 384
analgesics, 384
epidural blood patch, 385
hydration, 384
pharmacologic therapy, 384–385
prevention, after known accidental dural puncture, 385
risk factors, 378–379
scope of problem, 378
spinal fluid leakage and, 404, 405f, 406–408
Postdural puncture meningitis, 408–409
Posterior reversible leukoencephalopathy syndrome, 382
Postpartum hemorrhage
definition, 327
placenta accreta, 330–332
retained placenta, 329–330
uterine atony, 327–329
uterine inversion, 332–333
Postpartum infection (endometritis), 121–122
Postpartum tubal ligation (PPTL), 417
anesthetic considerations, 422–429
anesthesia provider and operating room times, 426f
anesthetic risk, 422–423
aspiration risk and prophylaxis, 424
breastfeeding and anesthesia, 425
general anesthetics, 428
local anesthetics, 428–429
neuraxial anesthesia and tubal sterilizations, 423
preoperative assessment, 424
regional anesthetics, 425–428
ASA Practice Guidelines for Postpartum Sterilization, 418
Pomeroy method, 422f
postpartum anatomic and physiologic changes, 418–419
cardiovascular changes, 418
gastrointestinal changes, 418–419
summary of, 429
surgical clip method, 423f
surgical considerations relevant to anesthesiologist, 421–422
overweight or obese patients, 421–422
tubal sterilization, 421
timing of tubal sterilization, 419–421
factors affecting, 419t
for high risk patients, 421
interval versus delayed postpartum sterilization, 419–420
from obstetrician’s perspective, 420–421
Potassium iodide, 462t
PPCM. See Peripartum cardiomyopathy
PPTL. See Postpartum tubal ligation
Preeclampsia, 381
chronic hypertension with superimposed preeclampsia, 437
differential diagnosis and definition, 436
epidemiology, 438
etiology, 438–439
FoCUS and, 198
in multiple gestation pregnancy, 291
obstetric management, 442–445
optic nerve sheath diameter in preeclamptic patients, 210–211, 211f
pathophysiology, 439–442
prediction and prevention, 442
renal failure and, 569–570
risk factors, 438, 438t
signs and symptoms, 437t, 440t
Pregabalin, 383
Pregnancy. See also specific topics
after bariatric surgery, 627
fever in, 116–129
requirements for labeling of prescription drugs and biological products, 30t
Pregnancy, physiological changes of
cardiovascular system in, 4–6
aortocaval compression, 6
blood volume, 4, 4t
cardiac output, 4
ECG changes, 5
myocardial contractility, 5
SVR, 5
central nervous system in, 13–14
biochemical changes, 12
inhalation anesthetics and minimum alveolar concentration, 13
neuraxial anesthesia/local anesthetics, 13
endocrine system in, 12–13
pancreatic function and glucose metabolism, 12–13
pituitary function, 13
thyroid function, 12
gastrointestinal system in, 10–11
gastric emptying, 11
gastric position and pressure, 10
gastric secretion, 10–11
lower esophageal sphincter tone, 10
hematologic system in, 9–10
coagulation factors, 9–10
dilutional anemia, 9
leukocytes and immune function, 10
platelet count and function, 9
hepatic function in, 11, 12t
hepatic blood flow, 11
liver function tests, 12t
serum albumin concentration, 11
serum estrogen in, 11
serum progesterone in, 11
splanchnic, portal, and esophageal venous pressure, 11
maternal physiologic changes, anesthetic implications, 14–15
aortocaval compression, 14
blood loss replacement, 15
endotracheal intubation, 14
epidural doses, 15
hypoxemia, 14
morbidity, 15
mortality, 15
muscle relaxants, 15
subarachnoid doses, 15
upper airway changes, 14
ventilation, 14
musculoskeletal changes, 13
joint mobility, 13
lumbar lordosis, 13
renal system in, 11–12
anatomic changes, in renal blood flow, 11, 12
GFR, 11–12
respiratory system in, 6–9
arterial blood gases, 6, 7t
FRC, 8
lung volumes, 7
mechanisms of hypoxemia, 7–8
oxygen delivery, 8–9
respiratory consequences of uncontrolled maternal pain, 8
Pregnancy-induced hypertension, 492–493
Pregnancy-related death, 668
Premature infants, resuscitation for, 358–359
Preterm birth
anesthetic management, 333–334
in breech presentation, 299
in multiple gestation pregnancy, 293–294
obesity and, 626
prevention and treatment, of preterm labor, 84
risk factors, 333
trauma and, 644
Prilocaine, 48
Primary apnea, 346
Primary pulmonary hypertension, 523–524
Procaine, 37t, 44t, 48
Professional negligence, 79
Progesterone, 11, 14
Prolactin, 13
Prolactinomas, 466
Prone position, 382
Prophylactic antibiotics, 245
Prophylactic epidural blood patch, 384, 407
Propofol, 504, 519, 642
Propranolol, 475
Propylthiouracil (PTU), 462t, 463
Prostacyclin analogues, 519
Prostaglandin imbalance, 439
Prostaglandins
anesthetic considerations, 68
drugs, 67
mechanism of action, 67
route of administration/dose, 67–68
toxicity/side effects, 68
uses, 67
Prostaglandin synthase. See Cyclooxygenase inhibitors
Protein C deficiency, 485
Protein S deficiency, 485–486
Prothrombin gene mutation, 485
Pruritus, 184, 370
Pseudoephedrine, 31t
Pseudotumor cerebri. See Benign intracranial hypertension; Idiopathic
intracranial hypertension
PTU. See Propylthiouracil
Pulmonary artery catheter, 504
Pulmonary embolism, during pregnancy
guidelines on, 697
imaging studies, 590–592, 591f
introduction, 589
treatment, 592
venous thromboembolism
maternal death from, 669
obesity and, 616–617
pregnancy effects on, 589
risk factors for, 590
treatment and anesthetic implications, 592–593
Pulmonary function tests, 583f, 584–585
Pulmonary hypertension, 508, 523–524
Pulmonary physiology, fetal, 344, 345f
Pulmonary system, 612–614
Pulmonary ultrasound
applications, 200–201
introduction, 199
sensitivity and specificity of auscultation, chest radiography, and lung
ultrasonography, 201t
technique, 199–200
Pulmonary vascular resistance (PVR), 507
Pulsatility index (PI), 101
Pulse oximetry, 519
PVR. See Pulmonary vascular resistance
Pyelonephritis, 570–571
Pyridostigmine, 550
Radiation, 85t
Radioactive iodine 131, 461, 462t
Rapid sequence intubation, 622–623
Recombinant factor VII, 321, 494
Reflexology, 139
Relaxation techniques, 138
Remifentanil, 148, 149–150, 150f–152f, 153, 502
maternal and neonatal effects, 155t
for pheochromocytoma, 178
studies for labor analgesia, 154t
Renal disease
anesthetic implications
altered responses to drugs, 566
potential nephrotoxins, 566
preoperative preparation and laboratory studies, 566–567
anesthetic management, 467
categories of renal dysfunction and influence on pregnancy, 561–562, 561t
DM and, 456
etiology
collagen vascular diseases, 564
diabetes mellitus and hypertension, 564
primary renal diseases, 563–564
renal transplant patients, 564–565
general management strategies, 565
HIV and, 124–125
introduction, 558–559
loop diuretics and, 565
monitoring, during pregnancy, 563t
multidisciplinary team, 559
renal anatomy, 559
renal changes, in pregnancy, 560t
renal disease, dialysis, and pregnancy outcomes, 571–572
renal failure associated with pregnancy
acute fatty liver of pregnancy (AFLP), 571, 575–577, 576t
acute kidney injury, 568–569, 568t, 569t
hemolytic uremic syndrome (HUS), 571
preeclampsia and, 569–570
pyelonephritis, 570–571
renal colic, 571
renal function assessment, in pregnancy, 560–561
renal physiology, 559–560
signs and symptoms of acute renal decompensation, 565t
systemic effects, 562, 563t
of chronic renal disease, 567t
Renal system, 11–12, 441–442
Resistance index (RI), 101
Respiratory diseases, obstetric anesthesia for parturients with
acute respiratory distress syndrome (ARDS) and respiratory failure, 604–
605, 604t
amniotic fluid embolism (AFE), 593–595
aspiration pneumonitis, 599–600
asthma, 583–589, 614
cystic fibrosis, 601–602
introduction, 582
lung transplantation, 602–603
obstructive sleep apnea (OSA), 598–599, 614f
pulmonary embolism, during pregnancy, 589–593
restrictive lung disease, 603–604
sarcoidosis, 599
smoking, 597
summary, 605
venous air embolism (VAE), 595–597
Respiratory gas exchange, 24–26
Respiratory system
in hypertensive disorders, 441
in multiple gestation pregnancy, 291
in pregnancy, 6–9
Respiratory tract infection, 119–120
Restrictive lung disease, 603–604
Resuscitation, 169, 530–531. See also Newborn resuscitation
Retained placenta, 329–330
Retinopathy, 456
RI. See Resistance index
Risk disclosure, 80
Ritodrine, 56, 57
Rocuronium, 428
Ropivacaine, 37t, 41, 43, 44t, 179
for spinal anesthesia, in cesarean delivery, 253