You are on page 1of 160

Advanced Life Support in Obstetrics

PROVIDER MANUAL

EIGHTH EDITION
Provider Manual
June 2017

ALSO© Editorial Advisory Board


Larry Leeman, MD, MPH
Professor, University of New Mexico School of Medicine
Departments of Family and Community Medicine and Obstetrics and Gynecology
Albuquerque, NM

Jeffrey D. Quinlan, MD, CAPT, MC, USN


Associate Professor and Chair Department of Family Medicine
Uniformed Services University
Bethesda, MD

Lee T. Dresang, MD
Professor, University of Wisconsin School of Medicine and Public Health
Department of Family Medicine
Madison, WI

David S. Gregory, MD
Program Director, Centra-Lynchburg Family Medicine Residency
Associate Clinical Professor, University of Virginia
Charlottesville, VA

Larry Howell, Medical Illustrator


Howell Graphics and Illustration
Blue Springs, MO
ALSO Status Life Cycle

ALSO Provider ALSO Instructor ALSO Approved ALSO Advisory


2-year status Candidate Instructor Faculty
Must attend an ALSO Attend an ALSO 5-year status Must maintain
Provider Course and Instructor Course, Must instruct in three Approved Instructor
successfully complete then instruct and courses (ALSO and/or status to maintain
the testing elements be evaluated at one BLSO) every 5 years Advisory Faculty
course (ALSO or to maintain status status
BLSO) within 1 year
and receive Approved
Instructor status

ALSO Providers and Instructors are responsible for their own status maintenance and expiration
dates. Due to the ALSO database being programmed with the above requirements, no exceptions
can be made. Please call or email ALSO Staff if you would like to verify your status at any time.

ALSO Provider - Participation in an ALSO Provider Course provider completes the ALSO Instructor course. Provider
and completion of course testing requirements designates status is automatically extended to meet the approved
the learner as an ALSO Provider. To maintain Provider instructor expiration date, and individual will be exempt from
status, the individual must attend another Provider Course taking the Provider Course as long as they maintain ALSO
before their 2-year expiration date. An ALSO Provider is Approved Instructor status by teaching in three courses
eligible to be course director for an ALSO Provider Course. every 5 years. If the Instructor teaching requirements are not
The ALSO Advisory Board recommends that the course met, the ALSO status will expire and the individual will need
director be an ALSO Approved Instructor or Instructor to start the ALSO status life cycle again.
Candidate, although it is not mandatory. ALSO Advisory Faculty - To be considered for Advisory
Instructor Candidate - An ALSO Provider is eligible to attend Faculty status, the individual must be recommended by an
an ALSO Instructor Course. Participation in an ALSO Advisory Faculty, complete the required Advisory Faculty
Instructor Course designates the provider as an ALSO Application, and submit it with other required documents to
Instructor Candidate. The Instructor Candidate must teach ALSO Staff. Maintaining ALSO Approved Instructor status is
in an ALSO or BLSO Provider course within 1 year of taking required for applying for an ALSO Advisory Faculty status.
the Instructor Course and be evaluated and recommended This faculty position requires that the person be considered
by an ALSO Advisory Faculty for Approved Instructor status. an “expert” on all aspects of the ALSO Provider Course.
The Instructor Candidate Evaluation form must be submitted It is mandatory that there be an ALSO Advisory Faculty
by the Instructor Candidate to the ALSO Staff before the present at every ALSO Provider Course to maintain quality,
1-year expiration date of the candidate. consistency, and ensure that the course adheres to all the
ALSO Approved Instructor - An Instructor Candidate whose requirements set forth by the ALSO Advisory Board. An
teaching skills have been evaluated by an Advisory Faculty individual serving as Advisory Faculty may not also serve
can then be recommended and approved to be an ALSO as the course director at a course. In addition, the Advisory
Approved Instructor. To maintain Approved Instructor status, Faculty will not get credit for the course unless they are also
the individual must teach in three courses (ALSO and/or teaching and are listed on the faculty page of the Participant
BLSO) every 5 years. The 5-year cycle begins the day the and Faculty Form submitted to the ALSO Staff.

To find out more about the ALSO program and


upcoming courses, please visit www.aafp.org/also.

Copyright 2017© American Academy of Family Physicians. All rights reserved. — Status Life Cycle  iii
ALSO Table of Contents

Overview of the Program . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1 – 4


A First-Trimester Pregnancy Complications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1 – 16
B Medical Complications of Pregnancy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1 – 29
C Vaginal Bleeding in Late Pregnancy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1 – 14
D Preterm Labor and Premature Rupture of Membranes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1 – 21
E Intrapartum Fetal Surveillance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1 – 21
F . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1 – 14
G Malpresentations, Malpositions, and Multiple Gestation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1 – 26
H Assisted Vaginal Delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1 – 13
I Shoulder Dystocia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1 – 20
J Postpartum Hemorrhage . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1 – 16
K Maternal Resuscitation and Trauma . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1 – 20
L Safety in Maternity Care . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1 – 11
M OB Cases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . (Handouts)
N Third and Fourth Degree Perineal Lacerations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1 – 12
O Diagnostic Ultrasound in Labor and Delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1 – 15
P Neonatal Resuscitation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1 – 15
Q Cesarean Delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1 – 34
R 
Birth Crisis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1 – 11

— Table of Contents  v
ALSO Program

ALSO Advisory Board, 2017-2018


Jennifer Frank, MD, Chair
Ann Evensen, MD Sajjini Thomas, MD
Gretchen Heinrichs, MD Janice Taleff, CNM
Sarah Jones, MD Barbara True, RNC-OB
Tony Ogburn, MD Johanna Warren, MD

ALSO Staff
Robyn Brumble, BSN, RNC-OB Ruth Fleming
Manager of Maternity Services ALSO Program Specialist

Brian Doty Jennifer Head


Associate Editor ALSO Program Specialist

Carla Cherry
ALSO Program Specialist

The American Academy of Family Physicians (AAFP) references used in the new curriculum. The curriculum
wishes to acknowledge the initial development of demonstrates the evidence, and quality of that evidence,
the ALSO Program by the University of Wisconsin on which any recommendations of care are based.
Department of Family Medicine and the original The current ALSO Provider Manual continues to
national ALSO Development Group of family physi- be an ongoing process. Each chapter is reviewed on
cians, obstetricians, and nurses, which formed in 1991. a 3-year cycle. Revisions will be published annually
The ALSO Program, originally conceptualized by and 1-page addendums will only be published if an
James R. Damos, MD, was developed under the leader- important/key evidence-based practice recommenda-
ship of Dr Damos and John W. Beasley, MD. The tion becomes available that would significantly alter a
AAFP acquired the ALSO Program in 1993. change in practice.
The 2000 edition of the ALSO Course curriculum
added levels of evidence for recommendations and

© 2017 American Academy of Family Physicians


11400 Tomahawk Creek Parkway, Leawood, Kansas 66211-2672
1-800-274-2237 • 913-906-6000

— Overview  1
Managing Editor and Author Brendon Cullinan, MD Susanna R. Magee, MD, MPH
Larry Leeman, MD, MPH Senior Medical Director for Primary Care Associate Professor of Family Medicine
Hennipen County Medical Center, Albert Medical School of Brown University
Professor
Minneapolis, Minnesota Assistant Professor,
University of New Mexico School of Medicine
Albuquerque, New Mexico
University of Minnesota Kristi K. Miller, MS, RN
Department of Family Medicine Quality Consultant
Medical Teamwork Consultants LLC
Associate Editors and Authors Stan E. Davis, MD, FACOG
Lee T. Dresang, MD Healthcare Consultant Neil J. Murphy, MD, FACOG
Professor TeamSTEPPS and In Situ Simulation Southcentral Foundation
University of Wisconsin, Department of Alaska Native Medical Center
Family Medicine & Community Health Mark Deutchman, MD
Professor Stephen Ratcliffe, MD, MSPH
Jeffrey D. Quinlan, MD, CAPT, MC, University of Colorado School of Medicine Program Director
USN Lancaster General Family Medicine
Associate Professor and Chair Department
Ann Evensen, MD Residency Lancaster, Pennsylvania
of Family Medicine, Uniformed Services Assistant Professor, CHS
University, Bethesda, Maryland University of Wisconsin School of Medicine William G. Sayres, Jr, MD
and Public Health Smith Family Endowed Chair in Medicine
Verona, WI Assistant Dean for Foundations
Medical Illustrator University of Washington School of Medicine
Larry Howell Patricia Fontaine, MD, MS
Howell Graphics and Illustration Blue Senior Clinical Investigator Barbara A. True, MN, CNS,
Springs, Missouri HealthPartners Institute for Education and RNC-OB, C-EFM
Research Clinical Nurse Specialist
Current Authors Texas Health Arlington Memorial
Jennifer Frank, MD
Karen Ailsworth MD, MS, FAAP Senior Medical Director Sara G. Shields, MD, MS, FAAFP
Professor Baraboo Family Practice ThedaCare Physicians Clinical Professor of Family Medicine and
Residency Rural Training Program Community Health
St. Mary’s/Dean Venture, Baraboo, Robert W. Gobbo, MD University of Massachusetts
Wisconsin Affiliate University of Wisconsin Program Director Family Health Center of Worcester
Department of Family Medicine-Madison Providence Oregon Family Medicine,
Hood River Rural Residency Program. Mary Beth Sutter, MD
Janice M. Anderson, MD Family Medicine Faculty
Associate Program Director Kim Hinshaw, MB, BS, FRCOG University of New Mexico School of Medicine
Forbes Family Medicine Residency Program Consultant Obstetrician & Gynaecologist,
Director of Research, Sunderland Royal J. Ely Walker, MD, MPH
Lesley A. Atwood, MD, FAAP Hospital and Visiting Professor, Dept of Rural Obstetrics Fellow
Clinical Associate Professor Health Sciences, University of Sunderland, University of Colorado School of Medicine
Family Medicine UK
University of Minnesota Johanna B. Warren, MD
Allina Health, Hastings Caroline Homer RM, Faculty Physician
MMedSci(ClinEpi), PhD Providence Oregon Family Medicine
Zachary Baeseman, MD Professor of Midwifery Residency Program
ThedaCare Physicians University of Technology Sydney, Australia
Waupaca, WI Helen Welch MSN, CNM
Sarah K. Jorgensen, DO Kaiser Permanente Northwest
R. Eugene Bailey, MD Associate Program Director
Associate Professor, Department of Family National Capitol Consortium Family Karen Wildman, MD, FAAFP
Medicine Medicine Residency Program Clinical Associate Professor Family
Assistant Professor, Department of Fort Belvoir Community Hospital Medicine, UWSOM
Obstetrics and Gynecology Fort Belvoir, VA Family Medicine Residency Spokane
Timothy P. Canavan, MD, MSc Paul Lewis, OBE Colleen Zimmermann, MD
Attending Perinatologist Greenwich Hospital, Chair of ALSO UK Executive Board of Faculty, Clinical Instructor
Yale New Haven Health System Trustees Family Medicine Residency Spokane
Emeritus Professor of Midwifery Practice and
Professional Development Bournemouth
University
Bournemouth, England

Provider Manual Disclosures — It is the policy of the AAFP that all individuals in a position to control content disclose any relationships with
commercial interests upon nomination/invitation of participation. Disclosure documents are reviewed for potential conflicts of interest and,
if identified, conflicts are resolved prior to confirmation of participation. Only those participants who had no conflict of interest or who
agreed to an identified resolution process prior to their participation were involved in this CME activity. All individuals in a position to control
content for this Provider Manual have indicated that they have no relevant financial relationships to disclose.

2 Overview—
Preface tially infectious. Universal precautions should always be
The ALSO Provider Course is an educational program followed in pregnancy and birth care.
designed to assist health professionals in developing
and maintaining the knowledge and skills needed Copyright
to effectively manage the emergencies which arise in The American Academy of Family Physicians (AAFP)
maternity care. The course includes required reading, owns the ALSO copyright and trademark on all of the
lectures, and hands-on workstations. Evaluation is by a course materials, including the Provider Manual, slide
written exam and skills assessment stations. There are set, and written exam. Use of portions of the materials
many appropriate ways of managing emergencies. The outside of an authorized ALSO course is strictly pro-
treatment guidelines presented in ALSO do not neces- hibited without prior written approval from the AAFP.
sarily represent the only way to manage problems and
emergencies. Instead, these guidelines are presented as Course Disclaimer
reasonable methods of management in obstetrical emer- The material presented at this course is being made
gencies. Each maternity care clinician must ultimately available by the AAFP for educational purposes only.
exercise his or her own professional judgement in This material is not intended to represent the only, nor
deciding on appropriate action in emergency situations. necessarily best, methods or procedures appropriate for
Completion of the ALSO Provider Course does not the medical situations discussed, but rather is intended
imply competency to perform the procedures discussed to present an approach, view, statement, or opinion
in the course materials. of the faculty which may be helpful to others who
face similar situations. The AAFP disclaims any and
Overall Course Objectives all liability for injury, or other damages, resulting to
1. Discuss
  methods of managing pregnancy and any individual attending this course and for all claims
birth urgencies and emergencies, which may help which may arise out of the use of the techniques dem-
standardize the skills of practicing maternity care onstrated therein by such individuals, whether these
clinicians. claims shall be asserted by physician, or any other per-
2.  Demonstrate content and skill acquisition as demon- son. Every effort has been made to ensure the accuracy
strated by successful completion of the course testing of the data presented at this course. Physicians may care
requirements. to check specific details, such as drug doses and con-
traindications, etc, in standard sources prior to clinical
CDC Recommendation application. This Course has been determined to be a
The Centers for Disease Control and Prevention Level 2 classification under the AMA/PRA Credit and
recommend universal precautions be used in all situa- Classification Guidelines. The AAFP does not certify
tions where a risk of exposure to blood or bodily fluids competence upon completion of the ALSO Provider
is present, and the potential infectious status of the Course, nor does it intend this course to serve as a basis
patient is unknown. All bodily fluids (blood, urine for requesting new or expanded privileges.
stool, saliva, vomitus, etc) should be treated as poten-

—Overview  3
2006–Current Edition
Evidence-Based Curriculum
Strength of Recommendation Taxonomy (SORT)

The current edition of the ALSO Provider Manual and one-page evidence-based addendums use the
Strength of Recommendation Taxonomy (SORT) to determine evidence based recommendtions:
A • Recommendation based on consistent and good-quality patient-oriented evidence.
B • Recommendation based on inconsistent or limited-quality patient-oriented evidence.
C • Recommendation based on consensus, usual practice, opinion, disease-oriented evidence, or case series for studies
of diagnosis, treatment, prevention, or screening.
Patient-oriented evidence measures outcomes that matter to patients: morbidity, mortality, symptom improvement, cost
reduction, and quality of life. Disease-oriented evidence measures intermediate, physiologic, or surrogate end points that
might not reflect improvement in patient outcomes (eg, blood pressure, blood chemistry, physiologic function, pathologic
findings).
A summary of recommendations will follow each chapter and will form the basis for the one-page addendums. In deciding
which points to choose for summary recommendations, authors and editors gave priority to recommendations that were
high quality (SORT A).
SORT B recommendations were also included when they had important clinical implicaitons or were related to common
clinical practices. SORT C recommendations only warrant mention in the summary, if the author/editor felt there was
exceptional relevance for clinical practice.

From Ebell MH, Siwek J, Weiss BD, et al. Strength of recommendation taxonomy (SORT): a patient-centered approach to grading evi-
dence in the medical literature. Am Fam Physician. 2004;69(3):548-556.

4 Overview—
Chapter A
First-Trimester Pregnancy Complications

Mark Deutchman, MD, J. Ely Walker, MD


Revised January 2016

Learning Objectives
At the end of this activity, learners will be able to:
1. Describe the process, diagnosis, and management 3. Describe the spectrum of psychological reactions
of miscarriage, ectopic pregnancy, and gestational to early pregnancy loss.
trophoblastic disease. 4. Describe the techniques of uterine aspiration for
2. Discuss the value of human chorionic gonadotropin the treatment of incomplete miscarriage (Optional).
(hCG) levels and sonographic discriminatory
criteria in diagnosing first-trimester pregnancy
complications.

Introduction as low as 5 mIU/mL. The rate of rise in quantitative


Complications during the first trimester of pregnancy serum hCG levels may be used to monitor patients
are common. Approximately 15% of clinically recog- with pain or bleeding whose initial ultrasound exami-
nized pregnancies result in spontaneous miscarriage, nation did not yield a definitive diagnosis. A large study
and estimates of miscarriage before clinical recognition of such patients before 10 weeks’ gestation showed
are as high as 50%.1 In addition to miscarriage, vaginal that a viable intrauterine pregnancy with hCG levels
bleeding can be associated with ectopic pregnancy, greater than 5,000 mIU/mL had an hCG rise of 53%
trophoblastic disease, or cervical bleeding from causes in 48 hours. However, women with a miscarriage or an
unrelated to pregnancy, or bleeding may occur in preg- ectopic pregnancy also can have a rise that is within this
nancies that proceed without further complications. range. Therefore, an adequately rising hCG does not
rule out nonviable pregnancy.3,5,6
Normal First-Trimester Pregnancy Progress The gestational sac first becomes visible on transvagi-
Pregnancy is clinically dated from the first day of the nal ultrasound during the 5th menstrual week as a 2- to
last normal menstrual period, which is an observable 5-mm sonolucent area surrounded by an echogenic
event, instead of the conception date. Conception ring of chorionic villi. This early gestational sac is only
occurs approximately 2 weeks later. All gestational visible using a high-frequency transducer (5 MHz or
landmarks in this chapter are based on menstrual dat- greater) and the transvaginal scan route. A small sono-
ing; embryology textbooks commonly use conception lucent fluid collection, or pseudosac, can also be pres-
dating, which is 2 weeks less. ent in cases of ectopic pregnancy, so additional features
The placenta produces human chorionic gonadotro- of a normal gestational sac can be sought, particularly
pin (hCG) after implantation. Implantation occurs at the eccentric location of the gestational sac indicating
approximately 23 menstrual days, which is approxi- that it is implanted within the endometrium rather
mately 8 days after conception. Commonly available than being located in the endometrial stripe. The yolk
over-the-counter urine pregnancy tests are approxi- sac appears during transvaginal scanning during the 6th
mately 100% sensitive and specific at detecting the menstrual week and provides clear evidence of an intra-
beta subunit of hCG at levels of 25 mIU/mL, which uterine pregnancy. By the end of the 6th menstrual
may allow detection of pregnancy around the time of week, the fetal pole becomes visible during transvaginal
the first missed period.2,3 Serum tests can detect hCG scanning as a 2 to 8 mm pole with embryonic cardiac

Copyright 2017© American Academy of Family Physicians. All rights reserved. — Chapter A  1
Chapter A

activity. These sonographic landmarks are visible


Table 1. Early Pregnancy Landmarks with transabdominal scanning approximately 1
week later than with transvaginal scanning.7
Menstrual
Age (weeks) Embryologic Event/Sonographic/hCG Correlation
Embryologic, clinical, hCG, and sonographic
findings are closely correlated and are displayed
3 to 4 Implantation site – Decidual thickening in Table 1.8,9 Ultrasonography is so valuable that,
4 Trophoblast – Peritrophoblastic flow on color flow
when it is readily available, many clinicians use
Doppler it as a primary tool in evaluating first-trimester
complications, leaving serum hCG testing as a
4 to 5 Gestational sac typically visible when hCG reaches
1,500 to 2,000 mlU/mL secondary tool used only if sonographic findings
are equivocal. A routine first-trimester ultrasound
5 to 6 Yolk sac
in early pregnancy appears to enable better assess-
5 to 6 Embryo and cardiac activity ment of gestational age and earlier detection of
multifetal pregnancies. However, the benefits for
other substantive outcomes are less clear.10
The use of specific sonographic findings as
Table 2. Crown-Rump Length and Menstrual Age discriminatory criteria to diagnose early pregnancy
loss is presented in detail in the pathophysiology
Crown-Rump Menstrual Crown-Rump Menstrual
Length (mm) Age (days) Length (mm) Age (days) section of this chapter.
After the embryo is sonographically visible,
2 42 16 57 first-trimester menstrual age is calculated from
4 44 20 60 crown-rump length using parameters such as those
shown in Table 2, which can be commonly found
6 47 25 65 in textbooks and are normally included in the
8 49 30 69 calculation software of ultrasound equipment.
10 51 35 72
13 54 40 76

2  Chapter A —
First-Trimester Pregnancy Complications

Early Pregnancy Loss: Pathophysiology, Discriminatory Criteria, Clinical Course


and Prognosis

Table 3. Terms Applied to Early Pregnancy Loss11,13

Embryonic demise — an embryo with a crown-rump length >7 mm without cardiac activity.
Anembryonic pregnancy — presence of a gestational sac >25 mm without evidence of embryonic tissues
(ie, yolk sac, embryo). This term is preferred to the older and less accurate phrase blighted ovum.
Spontaneous abortion — spontaneous loss of a pregnancy before 20 weeks’ gestation. Can be further
described as:
• Incomplete — occurs when some but not all of the products of conception have passed.
• Complete — all products of conception have passed through the external cervical os.
• Septic — incomplete abortion associated with ascending infection of the endometrium, parametrium, adnexa,
or peritoneum.
• Inevitable — bleeding in the presence of a dilated cervix, indicating that passage of the conceptus is
unavoidable.
• Missed — the fetus or embryo is dead but no tissue has been passed. The cervix is closed. These patients
often present with no growth in uterine size or no audible fetal heart tones.
Threatened abortion — bleeding before 20 weeks’ gestation in the presence of an embryo with cardiac activity
or a gestational/yolk sac and a closed cervix.
Subchorionic hemorrhage — ultrasonographic finding of blood between the chorion and uterine wall, typically
seen in the setting of vaginal bleeding.
Recurrent pregnancy loss — more than two consecutive pregnancy losses. The phrase habitual aborter has
been used but is no longer appropriate.
Ectopic pregnancy — pregnancy outside the uterine cavity, most commonly in the fallopian tube but can occur
in the broad ligament, ovary, cervix, or elsewhere in the abdomen.
Heterotopic pregnancy — simultaneous intrauterine and ectopic pregnancy. Incidence is rare, thought to occur
in 1/30,000 spontaneous pregnancies, but occur in 1.5/1,000 pregnancies involving assisted reproductive
techniques.12,13
Gestational trophoblastic disease, or hydatidiform mole — complete mole: placental proliferation in the
absence of a fetus. Most have a 46,XX chromosomal composition, all derived from paternal source. Partial
mole: molar placenta occurring together with a fetus. Most are genetically triploid (69,XXX).
Vanishing twin — A multi-fetal pregnancy is identified and one or more fetuses later disappear. More commonly
seen now that early ultrasound scanning is common. If this occurs early in pregnancy, the embryo is often
reabsorbed. Later occurrence results in a compressed or mummified fetus or amorphous material.

setting of anembryonic pregnancy or embryonic


Spontaneous Abortion (Miscarriage) demise. Spontaneous abortion may also be dis-
The cause of spontaneous abortion is rarely covered incidentally when asymptomatic patients
determined in clinical practice, but it is known
that approximately half are because of major
genetic abnormalities, typically trisomy, triploidy, Table 4. Environmental Factors Linked to Spontaneous
or monosomy.12 Environmental factors linked to Abortion
spontaneous abortion are shown in Table 4.
Spontaneous abortion can manifest clinically in Uterine anomalies Irradiation
Leiomyomata Maternal diethylstilbestrol
several different ways. Most commonly, vaginal exposure
Incompetent cervix
bleeding and cramping are present. But occasion- Advanced maternal age
Tobacco, alcohol, or cocaine use
ally, regression of pregnancy symptoms or lack Progesterone deficiency because Infections
of Doppler-detected fetal heart tones by 10 to 12 of luteal phase defect Occupational chemical exposure
weeks’ gestation are the first clinical signs in the

— Chapter A  3
Chapter A

undergo early ultrasound examinations for other performed. To examine for chorionic villi, rinse
reasons such as pregnancy dating or genetic and float the tissue in saline. Low magnification,
screening. backlighting, and teasing the tissue can help. Passed
Clinical examination should include palpation tissue should be submitted for pathologic examina-
of the abdomen and pelvis. The size and position tion, which is definitive in questionable cases.
of the uterus, the location of any tenderness, the If products of conception are seen at the cervi-
presence of rebound tenderness, and the presence cal os, ring forceps can be used to gently remove
of masses should be noted. Adnexal tenderness the tissue. More aggressive attempts to remove
and any masses should raise suspicion for ectopic partially expelled tissue should be preceded by
pregnancy, although a normal corpus luteum cyst discussion with the patient, informed consent, and
can also be the cause of either. If the patient’s last administration of analgesia or sedation.
menses was at least 9 to 10 weeks prior, and ultra- When the diagnosis is not clear based on clini-
sound is not readily available, consider listening cal findings, transvaginal scanning is essential for
for the fetal heartbeat during the bimanual pelvic accurate diagnosis. Specific sonographic character-
examination while elevating the uterus with the istics of the gestational sac, yolk sac, and embryo
intravaginal hand. can be reliably used to make an accurate and
A speculum examination will reveal non-uterine timely diagnosis. Table 5 presents guidelines for
causes of bleeding, the degree of cervical dilation use of sonographic findings when discriminating
and, if present, tissue being passed. The quantity of between viable and failed early pregnancy. Preg-
blood in the vault and the source of bleeding (from nancy failure can be reliably diagnosed if the gesta-
the os versus other sites) should be noted. If an tional sac is 25 mm or greater without an embryo,
intact gestational sac, an embryo, or the character- if an embryo fails to appear by 11 days after a yolk
istic fronds of chorionic villi are seen, miscarriage sac appears, or if an embryo of crown-rump length
is proven and ectopic pregnancy is virtually ruled greater than 7 mm does not show a heartbeat.13
out, except in the rare case of heterotopic preg- When ultrasound reveals a fetal heartbeat in a
nancy. If there is doubt about the origin of expelled patient with bleeding, the probability of miscar-
tissue, an examination for chorionic villi can be riage is only 2.1% in women younger than 35

Table 5. Guidelines for Transvaginal Ultrasonographic Diagnosis of Pregnancy Failure in a Woman


with an Intrauterine Pregnancy of Uncertain Viability

Findings Diagnostic of Pregnancy Failure Findings Suspicious for, but Not Diagnostic of, Pregnancy Failure*

Crown–rump length of >7 mm and no heartbeat Crown–rump length of <7 mm and no heartbeat
Mean sac diameter of >25 mm and no embryo Mean sac diameter of 16–24 mm and no embryo
Absence of embryo with heartbeat >2 wk after a scan Absence of embryo with heartbeat 7–13 days after a scan that showed
that showed a gestational sac without a yolk sac a gestational sac without a yolk sac
Absence of embryo with heartbeat >11 days after a Absence of embryo with heartbeat 7–10 days after a scan that showed
scan that showed a gestational sac with a yolk sac a gestational sac with a yolk sac
Absence of embryo >6 wk after last menstrual period
Empty amnion (amnion seen adjacent to yolk sac, with no visible embryo)
Enlarged yolk sac (>7 mm)
Small gestational sac in relation to the size of the embryo (<5 mm
difference between mean sac diameter and crown-rump length)

Criteria are from the Society of Radiologists in Ultrasound Multispecialty Consensus Conference on Early First Trimester Diagnosis of Miscar-
riage and Exclusion of a Viable Intrauterine Pregnancy, October 2012.
*When there are findings suspicious for pregnancy failure, follow-up ultrasonography at 7 to 10 days to assess the pregnancy for viability is gen-
erally appropriate.
Reprinted from Doubilet PM, Benson CB, Bourne T, et al; Society of Radiologists in Ultrasound Multispecialty Panel on Early First Trimester
Diagnosis of Miscarriage and Exclusion of a Viable Intrauterine Pregnancy. Diagnostic criteria for nonviable pregnancy early in the first trimes-
ter. N Engl J Med. 2013;369(15):1443-1451.

4  Chapter A —
First-Trimester Pregnancy Complications

years, but increases to 16.1% in women older between expectant, medical, or surgical manage-
than 35 years.14 ment. The majority of first-trimester miscarriages
Complete spontaneous miscarriage might result in occur completely and spontaneously without
an empty uterus with a bright endometrial stripe as intervention. Although surgical intervention in the
a result of the uterine walls having collapsed against form of uterine aspiration has traditionally been
each other. Echogenic material within the endo- used liberally, expectant management and medical
metrial cavity commonly creates an endometrial treatment are valid options. Women with excessive
stripe greater than 15 mm after treatment of early bleeding, pain, or infection benefit from medi-
pregnancy failure with misoprostol.15 Therefore, cal or surgical intervention.11 In an observational
endometrial thickness alone is not an indication study of 451 cases, 91% of those with incomplete
of the need for surgical intervention after medical miscarriage, 76% of those with missed miscarriage,
management of miscarriage with misoprostol. and 66% of those with anembryonic pregnancy
Septic abortion should be assumed when the completed their miscarriage without surgical
patient is febrile or has excessive uterine or adnexal intervention. Overall, 70% of women completed
tenderness, or signs of peritonitis. The clinician their miscarriage within 14 days of classification.20
should ask about a history of attempted therapeu- Women who have not completed the miscarriage
tic abortion or illegal abortion that might have for an emotionally uncomfortable period of time
left tissue behind or perforated the uterus. Septic may prefer medical or surgical intervention. The
abortion is a potentially life-threatening condition woman’s emotional state and personal preferences
requiring prompt resuscitation, uterine evacuation, are important in choosing the course of action.
and broad-spectrum antibiotic treatment.16,17 Ultimately the decision should be primarily driven
Sonography may reveal a hematoma between by the patient’s desires after being well informed
the chorion and uterine wall in subchorionic hem- of her options.21
orrhage and a gestational sac and embryo will be Clinical trials comparing expectant management
present. When subchorionic hemorrhage is visible with misoprostol and misoprostol with surgical
on ultrasound, the likelihood of miscarriage aver- treatment have shown:22-24
ages approximately 10% even when a heartbeat • In incomplete miscarriage, expectant and medi-
is detected, but varies by maternal age, size of the cal management with misoprostol are highly
hematoma, and gestational age.18 Therefore, the successful.
patient should be advised to expect bleeding. • In missed abortion, medical management with
The quantity of bleeding predicts pregnancy misoprostol and surgical treatment are more
loss only when it is heavy. A prospective analysis effective than expectant treatment.
of 4,510 women who were followed in the early • Typical misoprostol doses are 600 µg orally or
first trimester showed that 1,204 (27%) had some 600 to 800 µg vaginally.
bleeding or spotting. There was no increase in • Women treated with misoprostol have more
miscarriage risk in early pregnancy when there was bleeding but less pain than those treated
spotting or light bleeding. However, miscarriage surgically.
risk increased significantly in the 8% of women • Women treated expectantly have more outpa-
who reported heavy bleeding. This was the only tient visits than those treated with misoprostol.
group to have an increased risk of miscarriage • Surgery is associated with more trauma and infec-
(adjusted odds ratio = 2.84, 95% confidence inter- tion complications than misoprostol treatment.
val = 1.82 to 4.43).19 • Misoprostol has fewer gastrointestinal adverse
effects when administered vaginally than when
Management of Spontaneous Abortion administered orally.
If an ultrasound reveals an intrauterine pregnancy As a result of trials conducted on the medi-
with cardiac motion, the patient should be fol- cal management of miscarriage, a change to less
lowed with cautious optimism and an explanation surgical management has been suggested, even
that there are no known interventions to prevent though this is an off-label use of misoprostol.25 It
miscarriage. is also reasonable for a woman to change treat-
When the clinician’s examination reveals ment course and mix the three treatment options.
incomplete miscarriage, the patient must choose Women commonly choose a period of expectant

— Chapter A  5
Chapter A

management followed by medical therapy with


misoprostol if they no longer desire to wait. This Table 6. Ectopic Pregnancy Risk Factors
could also be followed by uterine aspiration if
medical therapy is not successful. History of tubal surgery, including tubal ligation and/
or reanastomosis of the tubes after tubal ligation
There is no evidence supporting the use of
prophylactic antibiotics in early pregnancy fail- History of tubal infection including pelvic
inflammatory disease
ure,26 although there is such evidence for uterine
aspiration for induced abortion. When misopro- Contraception with progestin-only pills
stol is used for medical abortion, the incidence Contraception with intrauterine devices
of infection complications may be reduced by
History of in utero exposure to diethylstilbestrol
administering the drug by the buccal route and by
administering doxycycline 100 mg twice daily for History of ectopic pregnancy
7 days. However, this study is low-quality evidence
because it compares two different time periods.27 It
is unclear whether it is the use of antibiotics or the Signs, Symptoms, and Diagnosis
change in the route of misoprostol administration Pain and vaginal bleeding are the hallmark
that resulted in the lowered incidence of infection. symptoms of ectopic pregnancy. Pain is almost
It is also unclear whether this is also true when universal; it is typically unilateral and in the lower
misoprostol is used to manage early pregnancy loss. abdomen. Bleeding is also common after a short
After a miscarriage, it is customary to recom- period of amenorrhea. Physical examination may
mend a brief period of contraception before reveal a tender adnexal mass, often mentioned
attempting another pregnancy. However, this in texts, but only noted clinically in a minority
practice is not supported in the literature. A of cases. Furthermore, it may easily be confused
prospective study showed no statistically signifi- with a tender corpus luteum of a normal intra-
cant difference of recurrent miscarriage in women uterine pregnancy. Finally, signs and symptoms of
who had interpregnancy intervals of less than 6 hemoperitoneum and shock can occur, including a
months versus those who had longer intervals.28 distended silent, doughy abdomen, shoulder pain,
For patients desiring long-term contraception, bulging cul-de-sac into the posterior fornix of the
an intrauterine device placed immediately after vagina, and hypotension.
spontaneous or induced first-trimester abortion is Initially, the serum hCG level in ectopic preg-
safe and effective.29 nancy rises but then typically plateaus or falls.
Transvaginal ultrasound scanning is a key diagnos-
Ectopic Pregnancy tic tool and can rapidly make these diagnoses:
Pathophysiology and Risk Factors
Ectopic pregnancy is typically located in the fal- 1. Ectopic pregnancy is ruled out by the presence
lopian tube, but rarely can occur in the broad of an intrauterine pregnancy with the exception
ligament, ovary, cervix at the site of a prior hys- of rare heterotopic pregnancy.
terotomy, or elsewhere in the abdomen. Ectopic
pregnancy can result in impairment or loss of fer- 2. Ectopic pregnancy is proven when a gestational
tility and, because of internal hemorrhage, remains sac and an embryo with a heartbeat are seen
the second most common cause of maternal outside of the uterus.
mortality. Early diagnosis is the key to preventing
morbidity and mortality and preserving fertility. 3. Ectopic pregnancy is highly likely if any adnexal
All health care professionals who care for women mass distinct from the corpus luteum or a sig-
of childbearing age should have an active work- nificant amount of free pelvic fluid is seen.30
ing knowledge of ectopic pregnancy and should When ultrasound findings are not definitive,
have a high index of suspicion in any woman who the location of the pregnancy is unknown. Table 7
presents with bleeding and/or pain during early provides guidelines for using hCG measurements
pregnancy. Risk factors are shown in Table 6, but in combination with transvaginal ultrasound find-
many ectopic pregnancies occur in women with- ings to establish a diagnosis and avoid interrupting
out risk factors. a potentially viable pregnancy. When transvaginal

6  Chapter A —
First-Trimester Pregnancy Complications

ultrasound shows no intrauterine fluid (gesta- a ruptured ovarian cyst, which can be managed
tional sac) and the hCG is above a discriminatory expectantly from the frank hemorrhage because of
zone threshold, viable intrauterine pregnancy is ruptured ectopic pregnancy. However, improved
unlikely, so the clinician should have a high index use of ultrasound and highly sensitive hCG
of suspicion for ectopic pregnancy. This threshold have decreased the need for this procedure. The
for hCG is in question; some sources use 3,000 presence of any cul-de-sac fluid indicates ectopic
mIU/mL,15 some use 3,510 mIU/mL31 and the pregnancy until proven otherwise.
threshold is dependent on the quality of the ultra- When hCG levels are not rising normally and
sound equipment and the sonographer performing ultrasound cannot confirm pregnancy location,
the ultrasound. In stable patients, repeat hCG test- a uterine aspiration may yield chorionic villi or a
ing and transvaginal ultrasound are prudent before gestational sac. When this happens, a failed intra-
diagnosing and treating for ectopic pregnancy.13,31 uterine pregnancy is diagnosed and treatment for
In some cases of ectopic pregnancy, a small fluid an ectopic pregnancy avoided. When suspicion for
collection within the uterus can be mistaken for ectopic pregnancy is high but cannot be confirmed
a true gestational sac. However, this pseudoges- with noninvasive testing, laparoscopy can confirm
tational sac lacks a surrounding echogenic ring of the diagnosis and accomplish surgical treatment or
chorionic villi, a yolk sac, or fetal pole. An unrup- methotrexate can be administered if a viable intra-
tured corpus luteum cyst can be mistaken for an uterine pregnancy has been definitively excluded.
ectopic gestational sac, and a ruptured corpus
luteum cyst can produce free pelvic fluid suggest- Management
ing ruptured ectopic pregnancy. Culdocentesis can With early diagnosis, the management of ectopic
be helpful in differentiating the thin pink fluid of pregnancy occurs most frequently in the outpa-

Table 7. Diagnostic and Management Guidelines Related to the Possibility of a Viable


Intrauterine Pregnancy in a Woman with a Pregnancy of Unknown Location*

Finding Key Points

No intrauterine fluid collection • A single measurement of hCG, regardless of its value, does not reliably
and normal (or near-normal) distinguish between ectopic and intrauterine pregnancy (viable or nonviable).
adnexa on ultrasonography† • If a single hCG measurement is <3,000 mIU/mL, presumptive treatment
for ectopic pregnancy with the use of methotrexate or other pharmacologic
or surgical means should not be undertaken, in order to avoid the risk of
interrupting a viable intrauterine pregnancy.
• If a single hCG measurement is >3,000 mIU/mL, a viable intrauterine
pregnancy is possible but unlikely. However, the most likely diagnosis is a
nonviable intrauterine pregnancy, so it is generally appropriate to obtain at
least one follow-up hCG measurement and follow-up ultrasonogram before
undertaking treatment for ectopic pregnancy.
Ultrasonography not yet • The hCG levels in women with ectopic pregnancies are highly variable,
performed often <1,000 mIU/mL, and the hCG level does not predict the likelihood of
ectopic pregnancy rupture. Thus, when the clinical findings are suspicious
for ectopic pregnancy, transvaginal ultrasonography is indicated even when
the hCG level is low.

*Criteria are from the Society of Radiologists in Ultrasound Multispecialty Consensus Conference on Early First Trimester
Diagnosis of Miscarriage and Exclusion of a Viable Intrauterine Pregnancy, October 2012.
† Near-normal (ie, inconsequential) adnexal findings include corpus luteum, a small amount of free pelvic fluid, and para-
tubal cyst.
hCG = human chorionic gonadotropin.
Reprinted from Doubilet PM, Benson CB, Bourne T, et al; Society of Radiologists in Ultrasound Multispecialty Panel on
Early First Trimester Diagnosis of Miscarriage and Exclusion of a Viable Intrauterine Pregnancy. Diagnostic criteria for
nonviable pregnancy early in the first trimester. N Engl J Med. 2013;369(15):1443-1451.

— Chapter A  7
Chapter A

tient setting by a clinician with experience and


confidence in ectopic management. Current Table 8. Indications for Surgical
treatment options favor medical and laparoscopic Management of Ectopic Pregnancy
surgical management with expectant management
reserved for cases with a declining quantitative Unstable vital signs or signs of hemoperitoneum
hCG level less than 1,000 mIU/mL.32,33 Uncertain diagnosis
Open surgical management is limited to tubal Advanced ectopic pregnancy (elevated hCG levels,
rupture and hemoperitoneum if a surgeon with large mass, cardiac activity)
laparoscopic training is available. Surgical man-
Unreliable follow-up
agement via laparoscopy or open laparotomy
can involve complete removal of the fallopian Contraindication to observation or methotrexate
tube (salpingectomy) or removal of the ectopic
pregnancy and conservation of the tube (salpin- hCG = human chorionic gonadotropin.
gostomy). Ectopic pregnancies located in the
tubal cornua, interstitial area, or uterine cervix are
dangerous and difficult to manage. Table 8 shows Table 9. Criteria for Expectant
indications for surgical management. Management of Ectopic Pregnancy33-36
Expectant or medical management are options
for hemodynamically stable women who are care- Minimal pain or bleeding
fully selected and informed according to the cri-
Patient reliable for follow-up
teria in Tables 9 and 10.33-36 hCG level is the best
predictor of successful management with metho- No evidence for tubal rupture
trexate. A systematic review of several studies Starting hCG level <1,000 mlU/mL and decreasing
showed that failures with single-dose methotrexate
Ectopic or adnexal mass <3 cm, or not detected
occurred 3.7% of the time when hCG levels were
below 5,000 mIU/mL versus 14.3% when above No embryonic heartbeat
this cut-off level. Thus, methotrexate is used only
in special circumstances when hCG levels exceed hCG = human chorionic gonadotropin.

5,000 mIU/mL.37 Appendix A is a protocol for use


of methotrexate. Single and multiple dose proto-
cols are available; multiple dose protocols are more Table 10. Criteria for Medical
commonly used in cases of high hCG levels.38 Management of Ectopic Pregnancy
Expectant management is used most often when with Methotrexate33-36
the location of the pregnancy cannot be deter-
mined. Medical management with methotrexate, Stable vital signs and low level of symptomatology
a folic acid antagonist, is appropriate for properly No medical contraindication for methotrexate
selected patients and has been shown in random- therapy (ie, normal liver enzymes, complete blood
ized trials to be safe and effective; it also can be count, platelet count)
less costly and result in equal or better subsequent Unruptured ectopic pregnancy
fertility than conservative surgical treatment.38-40
Absence of embryonic cardiac activity
Gestational Trophoblastic Disease Ectopic mass <4 cm
Pathophysiology and Risk Factors41 Starting hCG levels <5,000 to 10,000 mlU/mL
Gestational trophoblastic disease, or molar preg-
nancy, is an occasional cause of first-trimester hCG = human chorionic gonadotropin.
bleeding in the United States (1:1,000 to 1:1,500),
but is more common in Southeast Asia.
A complete mole consists of placental prolifera- bunches of grapes. Most complete moles have
tion in the absence of a fetus. Risk factors include a 46,XX chromosomal composition, all derived
extremes of age and previous molar pregnancy. from paternal sources. Mole recurrence may prog-
The placental villi are swollen and often resemble ress to metastatic choriocarcinoma. A partial mole

8  Chapter A —
First-Trimester Pregnancy Complications

is a molar placenta occurring together with a fetus,


which is typically nonviable. Genetic testing typi- Table 11. Trophoblastic Disease Signs
cally reveals triploidy (69,XXY). Partial mole is less and Symptoms
common than a complete mole, and has a lower
risk of recurrence. Uterus larger than expected for gestational age
Absent fetal heartbeat
Signs, Symptoms, and Diagnosis
Higher than expected hCG levels (except in cases
The signs and symptoms of gestational trophoblas- of partial mole)
tic disease are shown in Table 11.
Hyperemesis, pregnancy-induced hypertension at
an early gestational age, and/or thyrotoxicosis
Treatment
Ovarian enlargement, caused by theca-lutein cysts
Prompt evacuation of the uterus is the primary resulting from ovarian hyperstimulation because of
treatment. After evacuation of a complete mole, high hCG levels
all patients should have serial monitoring of hCG
Vaginal bleeding in the first or early second trimester,
levels for 6 months to 1 year with use of a highly which is often dark and may cause anemia
effective method of contraception.42 If the hCG
Grape-like vesicles are passed in cases that
level plateaus or increases, then recurrence must be progress into the second trimester
assumed, investigated, and treated with chemother-
apy (methotrexate). Because of the relative rarity of hCG = human chorionic gonadotropin.
this disease and the many possible complications,
consultation is recommended when the HCG level
is not falling appropriately. Theca-lutein ovarian sional to include them in the care plan. Available
cysts (functional ovarian cysts which are typically evidence suggests that although the vast majority
bilateral and caused by elevated hCG levels) do not of women desire support from their health care
require management and will resolve after evacu- professional, many do not receive the quantity
ation of the molar tissue. Approximately 20% or quality of support they desire. The types of
of women with a complete mole will experience interventions that are most effective in managing
recurrence in the form of a mole that invades the psychological symptoms are uncertain, but the fol-
myometrium or becomes aggressively metastatic. lowing approaches may be practical ways to miti-
gate the normal grief after early pregnancy loss.
Grief and Psychological Management Acknowledge and attempt to dispel guilt. Many
of Early Pregnancy Loss43,44 women think that some action on their part
Miscarriage represents a major loss to the preg- caused or contributed to the miscarriage. This
nant woman and her family. The grief reaction guilt can revolve around sexual activity, food,
that often follows is similar in intensity to that minor trauma, physical activity, or emotional
experienced after other major losses, though stress. Women whose losses can be ascribed to
women experience and describe it in varied ways. a definite cause have lower levels of anxiety and
Although healing will occur, the time to recovery grief. Therefore, evaluation of available tissue for
also varies. The feelings of loss tend to be strongest chromosomal anomalies is recommended when
in the first 6 months after the miscarriage, but possible. Even when a definitive cause cannot
can be persistent and pervasive enough to cause be ascertained, reassurance that the patient did
long-term symptoms or even affect the patient’s nothing to cause the loss is appropriate. This
next pregnancy. Women at risk of a stronger grief reassurance may need to be repeated several
reaction include those who experience a missed times. Women should be counseled that genetic
abortion, with loss at a later gestational age, with a or developmental errors likely occurred early in
longer time to conception of their next pregnancy, the pregnancy, and there was no possibility of the
and with a critical self-perception. pregnancy progressing to produce a live infant.
Partners also experience grief with pregnancy The post-loss follow-up visit is not the time to
loss. Because partners are often a primary social focus on modifiable risk factors that might have
support for patients and attend many post-loss contributed to the loss (ie, alcohol or tobacco use).
visits, it is important for the health care profes- Addressing these issues before future pregnancies is

— Chapter A  9
Chapter A

indicated, but is better done after the acute trauma Warn patients of the anniversary phenomenon. A
of loss fades. The patient’s religious belief system recurrence of grief feelings on their due date or the
can be called on during this counseling. anniversary of the miscarriage can occur. This can
Acknowledge and legitimize grief. Allowing also occur at the birth of a friend’s baby or during
patients to discuss their emotions surrounding the patient’s subsequent pregnancy. Posttraumatic
their loss might be the most important aspect stress disorder should be considered in women
of psychological care. One study suggests that experiencing prolonged grieving, anxiety, or other
women who had a medical follow-up visit at symptoms that affect their general or reproductive
which they were not provided an opportunity to functioning.
discuss their feelings had more anxiety and depres- Include the partner in your psychological care.
sion than those who had no follow-up. Patients Partners often feel the pain of loss and should be
and their partners should be allowed to cry or feel included in counseling and decisions. Men’s reac-
sad. Minimizing their feelings can isolate them tions to pregnancy loss are more strongly influ-
and decrease the medical professional’s credibility. enced by the status of the marital relationship than
Legitimize their feelings by confirming that mis- are women’s. Offering couples counseling and
carriage is the death of a baby. Comments such as including men in the healing process can speed
“you can try again” or “at least it happened early” resolution for both partners.
are inappropriate. Simple measures that validate Assess level of grief and adjust counseling accord-
grief should not be underestimated. Listening to ingly. Many women are ambivalent or distressed
the patient, holding her hand, or telling her how by the pregnancy and may experience mixed
sad you feel for her can help her through this trau- feelings or profound relief at the loss. A history of
matic period. The patient should be seen within 1 abortion, failed birth control, or rape may further
to 2 weeks in the office, or called on the phone a complicate the emotional response. Allowing the
few days after the miscarriage. patient to express her emotions in a supportive
Reassure about the future. Grief will fade with and nonjudgmental atmosphere is always an
time. Most patients have an excellent likelihood appropriate intervention.
of a subsequent normal pregnancy. With fewer
than three miscarriages, the risk of miscarriage in Rh Prophylaxis and Future Conception
future pregnancies is no greater than usual. It is after Pregnancy Loss
important to explain that the next pregnancy will Several follow-up issues must be addressed after
not need to be managed differently because of the any type of pregnancy loss. Rh-negative women
miscarriage. This is an excellent time to encourage who miscarry during the first trimester should
the initiation of a prenatal vitamin. receive 50 µg of anti-D immune globulin.45 Con-
Counsel the patient how to tell family and friends traception should be discussed and started imme-
about the miscarriage. If family members and diately if conception is not desired; all methods
friends knew about the pregnancy, a designated are equally safe immediately after spontaneous
individual can inform them of the loss. This abortion or ectopic pregnancy. There is no good
allows them to express their sympathy and provide evidence suggesting an ideal interpregnancy inter-
emotional support, and may avoid embarrassing val.46 Folic acid supplementation before future
encounters in which others assume the pregnancy conception attempts substantially reduces the risk
is progressing. If the pregnancy was unknown to of neural tube defects.47
family and friends, they may recognize and be
concerned about external signs of grief or distress. Summary
A decision must be made whether to tell them. First-trimester pregnancy complications are
Informing other children in the family can also common and the differential diagnosis includes
be difficult. However, families often find comfort life-threatening conditions such as ectopic preg-
in allowing children to share in the grieving and nancy. Knowledge and application of discrimina-
remembering process. Parents should be encour- tory criteria can significantly aid in distinguishing
aged to discuss the loss in honest and develop- among normal early pregnancy, miscarriage, and
mentally appropriate ways, just as they would the ectopic pregnancy. Medical treatment of ectopic
death of another family member. pregnancy is possible in properly selected cases. In

10  Chapter A —
First-Trimester Pregnancy Complications

incomplete miscarriage, nonsurgical management Contraindications to Uterine Aspiration


has a high likelihood of success depending on the 1. Medical contraindications are rare but include
diagnosis. In embryonic demise or anembryonic active pelvic infection.
pregnancy, misoprostol or surgical management
are significantly more effective than expectant 2. Pregnancy loss is not proven to the patient’s
management. satisfaction, the physician’s satisfaction, or both.
Because there is a lack of clear superiority of
expectant versus surgical management of mis- 3. The patient prefers to await spontaneous
carriage, the woman’s preference should play a completion for any reason (eg, religious beliefs,
dominant role in decision-making.48 When the cost, desire to avoid surgical procedures, etc).
choice is made to manage early pregnancy failure Uterine aspiration is not appropriate if the spon-
by other than expectant means, vaginal misopro- taneous abortion appears to be complete based on
stol is highly effective, safe, and well-accepted by the following criteria:
women, with fewer gastrointestinal adverse effects • The uterus is small and firm.
than the oral route. Evidence does not support the • Scant or no bleeding is occurring.
use of antibiotics in all women with incomplete • Tissue has been passed and is available for
abortion. After any type of first-trimester preg- inspection, and appears complete.
nancy loss, Rh-negative women should receive • The patient is reliable for follow-up.
50 µg of anti-D imm une globulin. Acknowledge- • Ultrasound examination (preferably transvaginal)
ment of grief and demonstrations of empathy and shows an empty uterus.
reassurance are useful techniques in counseling
women after miscarriage. Uterine Aspiration Performed Under Local
Anesthesia
Optional Section 1. P
 lace an intravenous (IV) line if patient is
Surgical Management of Miscarriage: bleeding heavily or if IV drugs will be used.
Dilation and Curettage and Manual
Vacuum Aspiration 2. A
 n Rh test should be obtained in women
The majority of first-trimester miscarriages occur whose status is unknown. A hematocrit or
completely and spontaneously without interven- hemoglobin should be obtained if there is
tion. And when intervention is selected, medical suspicion of anemia or excessive blood loss.
management is highly effective. Uterine aspiration A white blood cell count, prothrombin time,
by electric suction or manual vacuum aspiration partial thromboplastin time, fibrin split
may be indicated when: products, and blood type and screen may be
obtained depending on clinical circumstances
1. Heavy bleeding is present (greater than one pad (eg, heavy bleeding).
per hour).
3. S edation and analgesia should be adminis-
2. Patient is clinically stable (no bleeding or tered. Two to 5 mg of IV midazolam and
cramping), but pregnancy loss is shown con- 50 to 100 µg of IV fentanyl are commonly
clusively and the patient prefers intervention to used. Alternatively, other opioids or benzodi-
expectant management. azepines may be used. In many situations, the
patient’s partner or another support individual
3. Ectopic pregnancy needs to be ruled out. In may be present during the procedure.
certain situations, a clinical distinction cannot
be made between an ectopic and an intrauterine 4. The size and position of the uterus should
pregnancy. If tissue from a dilation and curet- be identified by bimanual examination.
tage (D&C) procedure contains chorionic villi, If the fetal measurements and uterine size
the pregnancy was intrauterine. Rarely, an intra- are greater than those for 14 weeks’ gesta-
uterine and an ectopic pregnancy (heterotopic tion, a dilation-and-extraction procedure is
pregnancy) may coexist, creating a confusing indicated, which requires advanced train-
and dangerous clinical situation. ing beyond that required for first-trimester

— Chapter A  11
Chapter A

aspiration. If a procedure is required beyond axis of the uterus, until slight resistance is felt,
14 weeks’ gestation, consider adding 20 units while exerting slight traction on the tenaculum
of oxytocin to IV fluids. to stabilize the cervix and straighten out the
cervico-vaginal angle. The curette should never
5. The cervix should be exposed with a specu- be forced after passing the internal os because
lum. A medium Graves speculum typically perforation is the most serious potential com-
suffices. The cervix and posterior fornix are plication of the procedure.
cleansed with an antiseptic solution. The
anterior lip of the cervix is then grasped with a 10. After the curette is in place, the suction hosing
single-toothed tenaculum. is attached and the suction machine is turned
on. Then the suction valve on the handle of
6. A paracervical block can be achieved with the hose. Sixty cm of mercury (Hg) or greater
20 cc of 1% lidocaine or another local anes- must be achieved for adequate suction.
thetic agent via a 20-gauge spinal needle.
One-fourth of the amount of the block is 11. With the suction on, the curette is rotated sev-
administered at 3:00, 5:00, 7:00, and 9:00; eral times in one direction, then several times
or one-half the amount of the block at 4:00 in the other direction, with a slight in-and-
and 8:00 where the cervix meets the vagina. out motion. Most of the pressure should be
A superficial wheal is raised and the syringe is maintained lateral, and forceful jabbing at the
aspirated before injecting to avoid intravascu- uterine fundus should be avoided because per-
lar injection. Several variants of the paracervi- foration is a risk. The amount and nature of
cal block exist, and all are equally satisfactory. tissue that appears in the plastic curette should
be observed carefully. Products of conception
7. If the cervix is closed, or insufficiently dilated often appear tan or grey, admixed with blood
to easily admit the required suction curette, and clots. Yellowish fluid can be noted. The
it can be progressively dilated using cervical curette is withdrawn slowly, avoiding the vagi-
dilators. Dilation should occur to the num- nal side wall while the suction is operating.
ber of mm that is equivalent to the estimated
gestational age in weeks or 1 mm less (eg, dilate 12. The suction and rotation sequence can be
to 9 or 10 mm to aspirate a 10-week missed repeated after inserting the curette into the
abortion). Control is indicated for this por- uterus again.
tion of the procedure, as dilators and uterine
sounds cause the largest number of uterine 13. M
 anual vacuum aspiration is accomplished
perforations. If the patient is clinically stable, with a simple handheld plastic syringe that
overnight dilation with laminaria is an option. generates its own suction mechanically. This
Another means of dilation that has been suc- device is inexpensive, easy to use, and does not
cessful, but is not approved by the US Food require electricity. It is particularly appropriate
and Drug Administration, is the buccal, sublin- for completions of early gestations (eg, less than
gual, or vaginal administration of misoprostol 8 to 10 weeks’ menstrual age). It can be used
400 µg 2 to 3 hours before the procedure. in the office setting where a suction machine is
not accessible. It is also appropriate in develop-
8. If the os is open, ring forceps can be used to ing countries where electricity is not available.
remove any loose tissue that is encountered.
14. A light, sharp curettage of the uterus can
9. The suction curette should be equivalent to the be performed to determine that it is empty,
size of the uterus in weeks (eg, a no. 10 curette followed by one more pass of the suction
for a 10 weeks’ sized uterus) is appropriate. A curette. This is no longer routinely indicated
curved curette is used if the uterus is anteflexed because of the increased pain and increasing
or retroflexed. A straight curette can be used if use of postprocedure vaginal ultrasound to
the uterus is midposition. The suction curette confirm completion in association with tissue
is inserted along the previously determined examination.

12  Chapter A —
First-Trimester Pregnancy Complications

15. After examination of the tissue, it should trum antibiotics, such as a cephalosporin, should
undergo examination for confirmation of be considered for any perforation.
diagnosis. To confirm an intrauterine preg-
nancy, chorionic villi must be identified. The 2. I ncomplete Evacuation — Incomplete evacu-
tissue must be sent to a laboratory for pathol- ation is identified by continued bleeding and
ogy unless the presence of villi or an embryo cramping after the procedure, or ultrasound
is conclusively confirmed by the physician evidence of retained tissue or endometritis.
performing the uterine aspiration. Incomplete evacuation can be managed by
repeating the procedure. Ultrasonic guidance
16. After the uterine aspiration is completed, the or general anesthesia is often helpful. Antibiot-
patient should be monitored for excessive ics are recommended if the second procedure
bleeding. Misoprostol can be administered by occurs more than a few hours after the first.
the rectal, buccal, or sublingual route in a dose
of 400 to 800 µg. Methergine® 0.2 mg may be 3. Bleeding — The differential diagnosis of bleed-
administered intramuscularly or orally. Trans- ing includes perforation, incomplete evacuation
fusions are required rarely. with retained tissue, cervical or uterine injury, or a
bleeding disorder. Methylergonovine 0.2 mg four
17. If the patient is Rh negative, 50 µg (mini dose) of times a day for 2 days is commonly administered
Rh immune globulin should be administered.45 to patients with more than average bleeding dur-
ing and after a procedure. An alternative is miso-
18. Doxycycline 100 mg is administered orally prostol 200 µg four times a day for 2 days. This is
pre-procedure to decrease the likelihood of an off-label use of misoprostol, but is effective in
endometritis. practice because of its powerful uterotonic effect.24

Complications of Uterine Aspiration 4. Infection — Infection may be referred to as sep-


Complications of uterine aspiration (suction D&C) tic abortion, endometritis, paraendometritis or
can occur. Careful performance of the procedure, pelvic peritonitis. It is diagnosed by the presence
consultation with more experienced physicians when of fever, uterine and parauterine tenderness,
needed, and a high index of suspicion for identifying peritonitis, and an elevated leukocyte count.
complications can prevent complications. Management is with antibiotics. For ill patients
who require hospitalization, an IV cephalospo-
1. Perforation — Perforation occurs when an rin or triple antibiotics (ampicillin, gentamycin,
instrument passes through the uterine wall. The and clindamycin or metronidazole) may be
diagnosis is typically apparent when a sound or required. Less ill patients can be treated in the
dilator passes through the cervix to a significantly outpatient setting. Clear guidelines for antibiotic
greater depth than expected. Occasionally, a suc- regimens are lacking. When tissue is found to be
tion curette or a sharp curette will draw maternal retained, repeating the uterine evacuation may
abdominal contents such as omentum or bowel be necessary. Oxytocics should be administered
out through the cervix. Heavy bleeding, signs as described previously in 2. Incomplete Evacua-
of peritonitis, or evidence of intra-abdominal tion. Rarely, in very ill patients, hospitalization
bleeding also can help identify perforation. If and hysterectomy may be necessary.
perforation occurs with a blunt instrument, such
as a uterine sound, and if the D&C has been 5. L
 ate Sequelae — Intrauterine synechiae (Asher-
completed, observation alone for a minimum of man syndrome) is often discussed but rarely
2 hours may suffice. If perforation has occurred seen. It is most likely to occur when a suction
with a sharp instrument, such as a curette or D&C is performed in the presence of infection,
with a suction curette, laparoscopy or laparot- a prolonged missed abortion, or postpartum.
omy may be indicated. If the uterine aspiration Incompetent cervix can occur rarely because of
has not been completed at the time the perfora- cervical injury. The most common late sequelae
tion is identified, it can be completed under to suction D&C are depression and related psy-
ultrasonic or laparoscopic guidance. Broad-spec- chological reactions to the loss of the pregnancy.

— Chapter A  13
Chapter A

SORT: Key Recommendations for Practice


Evidence
Clinical Recommendation Rating References

Pregnancy failure can be reliably diagnosed if the gestational sac is 25 mm or C 13


greater without an embryo, if an embryo fails to appear by 11 days after a yolk
sac appears, or if an embryo of crown-rump length greater than 7 mm does
not show a heartbeat.
Success of miscarriage management depends on diagnosis. In an incomplete A 22-24
miscarriage, nonsurgical management has a high likelihood of success. In
an embryonic demise or anembryonic pregnancy, misoprostol or surgical
management is considerably more effective than expectant management.
There is a lack of clear superiority of expectant versus surgical management of A 48
miscarriage. Therefore, the woman’s preference should play a dominant role
in the decision-making process.
When the choice is made to manage early pregnancy failure by other than A 22,23
expectant means, vaginal misoprostol is highly effective and safe, and well-
accepted by women, with fewer gastrointestinal adverse effects than the
buccal or oral routes.
Evidence does not support the use of antibiotics in all women with incomplete A 26
abortion.
Women undergoing induced abortion benefit from a single dose of doxycycline A 50
to prevent infection.
After any type of first-trimester pregnancy loss, Rh-negative women should C 45
receive 50 µg of anti-D immune globulin.
Acknowledgement of grief and demonstration of empathy and reassurance are C 43,44
useful techniques in counseling women after miscarriage.

Strength of Recommendation Taxonomy (SORT)

Strength of Recommendation Definition


A • Recommendation based on consistent and good-quality patient-oriented evidence.
B • Recommendation based on inconsistent or limited-quality patient-oriented evidence.
C • Recommendation based on consensus, usual practice, opinion, disease-oriented evidence, or case series for studies
of diagnosis, treatment, prevention, or screening.
Patient-oriented evidence measures outcomes that matter to patients: morbidity, mortality, symptom improvement, cost
reduction, and quality of life. Disease-oriented evidence measures intermediate, physiologic, or surrogate end points
that might not reflect improvement in patient outcomes (eg, blood pressure, blood chemistry, physiologic function,
pathologic findings).
From Ebell MH, Siwek J, Weiss BD, et al. Strength of recommendation taxonomy (SORT): a patient-centered approach to
grading evidence in the medical literature. Am Fam Physician. 2004;69(3):548-556.

14  Chapter A —
First-Trimester Pregnancy Complications

References
1. Wilcox AJ, Weinberg CR, O’Connor JF, et al. Incidence of 18. Bennett GL, Bromley B, Lieberman E, Benacerraf BR.
early loss of pregnancy. N Engl J Med. 1988;319(4):189-194. Subchorionic hemorrhage in first-trimester pregnancies:
2. Cole LA, Sutton-Riley JM, Khanlian SA, Borkovskaya M, prediction of pregnancy outcome with sonography.
Rayburn BB, Rayburn WF. Sensitivity of over-the-counter Radiology. 1996;200(3):803-806.
pregnancy tests: comparison of utility and marketing mes- 19. Hasan R, Baird DD, Herring AH, Olshan AF, Jonsson
sages. J Am Pharm Assoc (2003). 2005;45(5):608-615. Funk ML, Hartmann KE. Association between first-
3. Ehrenkranz JR. Home and point-of-care pregnancy trimester vaginal bleeding and miscarriage. [Level II].
tests: a review of the technology. Epidemiology. 2002; Obstet Gynecol. 2009;114(4):860-867.
13(Suppl 3):S15-S18. 20. Luise C, Jermy K, May C, Costello G, Collins WP,
4. Barnhart KT, Sammel MD, Rinaudo PF, Zhou L, Hummel Bourne TH. Outcome of expectant management of
AC, Guo W. Symptomatic patients with an early viable spontaneous first trimester miscarriage: observational
intrauterine pregnancy: HCG curves redefined. Obstet study. BMJ. 2002;324(7342):873-875.
Gynecol. 2004;104(1):50-55. 21. Allison JL, Sherwood RS, Schust DJ. Management of
5. Barnhart KT. Clinical practice. Ectopic pregnancy. first trimester pregnancy loss can be safely moved into
N Engl J Med. 2009;361(4):379-387. the office. Rev Obstet Gynecol. 2011;4(1):5-14.

6. Seeber BE, Barnhart KT. Suspected ectopic pregnancy. 22. Zhang J, Gilles JM, Barnhart K, Creinin MD, Westhoff C,
Obstet Gynecol. 2006;107(2 Pt 1):399-413. Erratum in Frederick MM; National Institute of Child Health Human
Obstet Gynecol. 2006;107(4):955. Development (NICHD) Management of Early Pregnancy
Failure Trial. A comparison of medical management
7. Laing FC, Frates MC, Benson CB. Ultrasound Evalua- with misoprostol and surgical management for early
tion During the First Trimester of Pregnancy. In: Callen pregnancy failure. N Engl J Med. 2005;353(8):761-769.
PW, ed. Ultrasonography in Obstetrics and Gynecology.
4th ed. Philadelphia, PA: W.B. Saunders Co; 2000. 23. Bagratee JS, Khullar V, Regan L, Moodley J, Kagoro H.
A randomized controlled trial comparing medical and
8. Paspulati RM, Bhatt S, Nour SG. Sonographic evalu- expectant management of first trimester miscarriage.
ation of first-trimester bleeding. Radiol Clin North Am. Hum Reprod. 2004;19(2):266-271.
2004;42(2):297-314. Erratum in Radiol Clin North Am.
2008;46(2):437. 24. Weeks A, Alia G, Blum J, et al. A randomized trial of
misoprostol compared with manual vacuum aspiration
9. Kadar N, Bohrer M, Kemmann E, Shelden R. The dis- for incomplete abortion. Obstet Gynecol. 2005;106(3):
criminatory human chorionic gonadotropin zone for 540-547.
endovaginal sonography: a prospective, randomized
study. Fertil Steril. 1994;61(6):1016-1020. 25. Winikoff B. Pregnancy failure and misoprostol—time for
a change. N Engl J Med. 2005;353(8):834-836.
10. Whitworth M, Bricker L, Neilson JP, Dowswell T.
Ultrasound for fetal assessment in early pregnancy. 26. May W, Gülmezoglu AM, Ba-Thike K. Antibiotics for
Cochrane Database Syst Rev. 2010;(4):CD007058. incomplete abortion. Cochrane Database Syst Rev.
2007;(4):CD001779.
11. Chen BA, Creinin MD. Contemporary management of
early pregnancy failure. Clin Obstet Gynecol. 2007;50(1): 27. Fjerstad M, Trussell J, Sivin I, Lichtenberg ES, Cullins V.
67-88. Rates of serious infection after changes in regimens for
medical abortion. N Engl J Med. 2009;361(2):145-151.
12. Goddijn M, Leschot NJ. Genetic aspects of miscar-
riage. Baillieres Best Pract Res Clin Obstet Gynaecol. 28. Buss L, Tolstrup J, Munk C, et al. Spontaneous abor-
2000;14(5):855-865. tion: a prospective cohort study of younger women
from the general population in Denmark. Validation,
13. Doubilet PM, Benson CB, Bourne T, et al; Society of occurrence and risk determinants. Acta Obstet Gynecol
Radiologists in Ultrasound Multispecialty Panel on Early Scand. 2006;85(4):467-475.
First Trimester Diagnosis of Miscarriage and Exclusion
of a Viable Intrauterine Pregnancy. Diagnostic criteria 29. Grimes DA, Lopez LM, Schulz KF, Stanwood NL.
for nonviable pregnancy early in the first trimester. Immediate postabortal insertion of intrauterine devices.
N Engl J Med. 2013;369(15):1443-1451. Cochrane Database Syst Rev. 2010;(6):CD001777.

14. Smith KE, Buyalos RP. The profound impact of patient 30. Sawyer E, Jurkovic D. Ultrasonography in the diagnosis
age on pregnancy outcome after early detection of fetal and management of abnormal early pregnancy. Clin
cardiac activity. Fertil Steril. 1996;65(1):35-40. Obstet Gynecol. 2007;50(1):31-54.

15. Creinin MD, Harwood B, Guido RS, Fox MC, Zhang 31. Connolly A, Ryan DH, Stuebe AM, Wolfe HM. Reevalu-
J; NICHD Management of Early Pregnancy Failure ation of discriminatory and threshold levels for serum
Trial. Endometrial thickness after misoprostol use β-hCG in early pregnancy. Obstet Gynecol. 2013;121(1):
for early pregnancy failure. Int J Gynaecol Obstet. 65-70.
2004;86(1):22-26. 32. van Mello NM, Mol F, Ankum WM, Mol BW, van der
16. Eschenbach DA. Treating spontaneous and induced Veen F, Hajenius PJ. Ectopic pregnancy: how the diag-
septic abortions. Obstet Gynecol. 2015;125(5):1042-1048. nostic and therapeutic management has changed. Fertil
Steril. 2012;98(5):1066-1073.
17. Stubblefield PG, Grimes DA. Septic abortion. N Eng J
Med. 1994;331(5):310-314.

— Chapter A  15
Chapter A

33. Cohen MA, Sauer MV. Expectant management of ecto- 42. Berkowitz RS, Goldstein DP. Clinical practice. Molar
pic pregnancy. Clin Obstet Gynecol. 1999;42(1):48-54, pregnancy. N Engl J Med. 2009;360(16):1639-1645.
quiz 55-56. 43. Brier N. Grief following miscarriage: a comprehensive
34. Mashiach S, Carp HJ, Serr DM. Nonoperative manage- review of the literature. J Womens Health (Larchmt).
ment of ectopic pregnancy. A preliminary report. 2008;17(3):451-464.
J Reprod Med. 1982;27(3):127-132. 44. Murphy FA, Lipp A, Powles DL. Follow-up for improving
35. Adoni A, Milwidsky A, Hurwitz A, Palti Z. Declining psychological well being for women after a miscarriage.
beta-HCG levels: an indicator for expectant approach in Cochrane Database Syst Rev. 2012;3:CD008679.
ectopic pregnancy. Int J Fertil. 1986;31(1):40-42. 45. American College of Obstetrics and Gynecology.
36. Garcia AJ, Aubert JM, Sama J, Josimovich JB. Expect- ACOG practice bulletin. Prevention of Rh D alloimmuni-
ant management of presumed ectopic pregnancies. zation. Number 4, May 1999 (replaces educational bul-
Fertil Steril. 1987;48(3):395-400. letin Number 147, October 1990). Clinical management
37. Menon S, Colins J, Barnhart KT. Establishing a human guidelines for obstetrician-gynecologists. Int J Gynae-
chorionic gonadotropin cutoff to guide methotrexate col Obstet. 1999;66(1):63-70.
treatment of ectopic pregnancy: a systematic review. 46. Wong LF, Schliep KC, Silver RM, et al. The effect of
Fertil Steril. 2007;87(3):481-484. a very short interpregnancy interval and pregnancy
38. American College of Obstetricians and Gynecolo- outcomes following a previous pregnancy loss. Am J
gists. ACOG Practice Bulletin No. 94: Medical man- Obstet Gynecol. 2015;212(3):375.e1-375.e11.
agement of ectopic pregnancy. Obstet Gynecol. 47. Cordero AM, Crider KS, Rogers LM, Cannon MJ, Berry
2008;111(6):1479-1485. RJ. Optimal serum and red blood cell folate concentra-
39. Stovall TG, Ling FW. Single-dose methotrexate: an tions in women of reproductive age for prevention of neu-
expanded clinical trial. Am J Obstet Gynecol. 1993;168(6 ral tube defects: World Health Organization guidelines.
Pt 1):1759-1762, discussion 1762-1765. MMWR Morb Mortal Wkly Rep. 2015;64(15):421-423.

40. Lipscomb GH, McCord ML, Stovall TG, Huff G, Portera 48. Nanda K, Lopez LM, Grimes DA, Peloggia A, Nanda
SG, Ling FW. Predictors of success of methotrexate G. Expectant care versus surgical treatment for miscar-
treatment in women with tubal ectopic pregnancies. N riage. Cochrane Database Syst Rev. 2012;3:CD003518.
Engl J Med. 1999;341(26):1974-1978. 49. Barnhart KT; Supplement to. Clinical practice. Ectopic
41. Lurain JR. Gestational trophoblastic disease I: epidemi- pregnancy. N Engl J Med. 2009;361(4):379-387.
ology, pathology, clinical presentation and diagnosis of 50. Achilles SL, Reeves MF; Society of Family Planning.
gestational trophoblastic disease, and management of Prevention of infection after induced abortion: release
hydatidiform mole. Am J Obstet Gynecol. 2010;203(6): date October 2010: SFP guideline 20102. Contracep-
531-539. tion. 2011;83(4):295-309.

16  Chapter A —
Objectives
First‐Trimester Pregnancy 
• Describe the process, diagnosis, and management of miscarriage,
Complications ectopic pregnancy, and gestational trophoblastic disease
• Discuss the value of human chorionic gonadotropin (hCG) levels and 
Revised January 2017 sonographic discriminatory criteria in diagnosing first‐trimester 
pregnancy complications
• Describe the spectrum of psychological reactions to early 
pregnancy loss
• Describe the techniques of uterine aspiration for the treatment of 
incomplete miscarriage (Optional)

Diagnosis of Pregnancy Etiologies of Early Pregnancy Bleeding
• Clinical:  • Miscarriage: occurs in 15% of clinically recognized
– History: missed menses pregnancies
– Physical examination
• Laboratory: • Ectopic pregnancy
– Qualitative hCG • Gestational trophoblastic disease
– Quantitative hCG
• Cervical bleeding from causes unrelated to pregnancy
• Ultrasound:
– Doppler auscultation • Pregnancies that proceed as otherwise uncomplicated
– Imaging

Early Pregnancy Landmarks Patterns of hCG Change


Menstrual Age  Embryologic/Sonographic/hCG Correlation
(weeks)

3 to 4 Implantation site ‐ Decidual thickening

4 Trophoblast ‐ Peritrophoblastic flow on color flow Doppler


Minimum rise in viable 
Gestational sac typically visible when hCG reaches 
4 to 5
1,500 to 2,000 mIU/mL
intrauterine pregnancy is 
53% in 48 hours
5 to 6 Yolk sac

5 to 6 Embryo and cardiac activity Reprinted from Barnhart KT. Clinical practice. Ectopic pregnancy. 


N Engl J Med. 2009;361(4):379‐387. 

Copyright 2017© American Academy of Family Physicians. All rights reserved. 1


Intradecidual Sign Normal Gestational Sac
Uterus
• Round shape
• Location in the
uterine fundus
SAC
• Echogenic ring
surrounding sac

5th menstrual week, transvaginal scan

Yolk Sac 6 to 7 Week Embryo with Heartbeat

GA by Crown‐Rump Length Pregnancy Loss Terminology
• Spontaneous abortion • Recurrent pregnancy loss
 Incomplete  • Ectopic pregnancy
 Complete  • Heterotopic pregnancy
 Septic
• Gestational trophoblastic
 Inevitable 
 Missed  disease
• Threatened abortion • Vanishing twin
Menstrual age* (weeks) = CRL (cm) + 6.5 • Subchorionic hemorrhage
*Accurate between 8 and 13 weeks

2
Pathophysiology of Miscarriage Pathophysiology of Miscarriage
• Major genetic anomaly • External environmental factors
• Internal environmental factors  Substance use (eg, tobacco, alcohol, cocaine)
 Uterine: anomalies, leiomyomata, incompetent cervix  Irradiation
 Maternal diethylstilbestrol exposure  Infection
 Luteal phase defect  Occupational chemical exposure
 Immunologic factors • Advanced maternal age

Clinical Course Physical Examination
• Missed menses, pregnancy symptoms • Abdominal examination
• Positive beta hCG – Pain location, rebound, distension
• Vaginal bleeding – Speculum examination
– Assess dilation
• Beta hCG falls or plateaus
• Bimanual examination
• Lower abdominal cramping, backache
– Uterine size, adnexal tenderness
• Products of conception passed

Pregnancy in Interstitial/Cornual
Doppler Detection of Fetal Heartbeat
Portion of Fallopian Tube
• Listen after 9 to 10 weeks  • Sac does not have 
with handheld Doppler myometrium in all directions 
• Sensitivity enhanced by  • No continuity with 
elevating the uterus  endometrial stripe 
internally during 
examination  • Rupture may not occur until 
13 weeks gestation
• Caution – Hearing a 
heartbeat DOES NOT rule  • Fetal heart tones may be 
Green = gestational sac  /  Yellow = fetal pole 
out ectopic  Purple = echogenic margins of fallopian tube heard with Doppler cornual
E = endometrium / Red = echogenic line abuts  ectopic
gestational sac  

3
Ultrasound Discriminatory Findings 
Float Test for Chorionic Villi
of Early Pregnancy Failure
• Place products of Diagnostic Suspicious
Mean sac diameter >25 mm without  Mean sac diameter 16 to 24 mm without 
conception in normal embryo embryo
saline Embryo ≥7 mm without heartbeat Embryo <7 mm without heartbeat

• Absence of chorionic Absent embryo with heartbeat 11  Absent embryo with heartbeat 7 to 10 days 


days after yolk sac seen after yolk sac seen
villi raises suspicion for
ectopic Absent embryo ≥6 weeks after last normal 
menstrual period 
Yolk sac >7 mm

Confirmed Fetal Loss Completed Miscarriage
Anembryonic Pregnancy Dead Embryo
• U = Empty uterus
• CX = Cervix
• B = Bladder

Subchorionic Hemorrhage Management of Miscarriage


• 50% probability of loss when bleeding is present,
• E= Embryo
though presence of fetal heartbeat is reassuring 
• YS = Yolk sac (2% to 16% probability of loss)
• SCH = subchorionic • Majority do not require intervention, unless at risk
hemorrhage for infection or excessive bleeding
• Routine antibiotics are not indicated
• Address contraceptive needs

4
Miscarriage Treatment Options Medical and Expectant Management
• Surgical • Incomplete Miscarriage ‐ Expectant and medical management 
are both highly successful
– Dilation and Curettage (D&C)
• Missed Abortion ‐ Medical and surgical management are 
– Manual Vacuum Aspiration (MVA) more effective than expectant management
• Medical • Women managed expectantly have more outpatient visits 
than those treated with misoprostol
– Misoprostol (off‐label use) • Women treated with misoprostol have more bleeding but less 
• Expectant – Wait for spontaneous completion pain than those treated surgically
• Surgery is associated with more trauma and infectious 
complications than misoprostol treatment

Misoprostol (Off‐Label Use) Ectopic Pregnancy
• Typical dosages: • Pregnancy outside the uterus
– 600 mcg orally – Usually in fallopian tube
– 600 to 800 mcg vaginal or buccal • Occurs in 1% of pregnancies
• Fewer gastrointestinal adverse effects when given  • Second most common cause of maternal mortality
vaginally or buccal route than when given orally • Early diagnosis is critical!
• Highly efficacious and well‐accepted 
by women

Risk Factors for Ectopic Diagnosis of Ectopic
• History of previous ectopic pregnancy • Failure of beta hCG to rise appropriately (53% in 48 hours)
• Prior tubal surgery • Clinical: Pelvic pain, tubal rupture, intraperitoneal hemorrhage, 
abdominal and shoulder pain, shock, death
• Prior tubal infection(s) • Ultrasound (transvaginal)
– Intrauterine pregnancy rules out ectopic except in rare cases of 
• Progestin‐only contraception heterotopic pregnancy
• Contraceptive intrauterine device – No gestational sac + beta hCG >3,000 to 3,510 mIU/mL highly suggestive 
– Gestational sac/embryo outside of uterus confirms ectopic
• In utero diethylstilbestrol exposure – Pitfalls: pseudogestational sac, ruptured corpus luteum
• Many occur in women with no risk factors! • Laparoscopy – gold standard

5
Pregnancy of Unknown Location Ectopic Pregnancy – Extrauterine Mass
Transvaginal Ultrasound Guidelines
Finding


Empty uterus and no 
adnexal mass or free 
Single hCG measurements do not reliably 
distinguish between ectopic and intrauterine
Uterus
pelvic fluid • hCG >3,000 or 3,510 mIU/mL is highly suspicious 
for ectopic pregnancy, but if patient is stable, 
repeat hCG and ultrasound before treating

Ultrasound not yet  • Even when hCG levels are <1,000 mIU/mL, 


performed proceed with transvaginal ultrasound because  Extrauterine
hCG levels vary widely in ectopic pregnancy 
cases and do not predict risk of rupture  mass

Pseudosac of Ectopic Pregnancy Extrauterine Signs of Ectopic


Finding Risk of Ectopic 
No mass or free fluid 20%
Any free fluid 71%
Echogenic mass 85%
Moderate to large amount 
of fluid  95%
Echogenic mass with fluid 100%
Information from Mahony BS, Filly RA, Nyberg DA, Callen PW. Sonographic evaluation of ectopic pregnancy.
Ultrasound Med. 1985;4(5):221-228.

Free Pelvic Fluid Culdocentesis
• 18 or 20 gauge needle passed 
through the posterior fornix 
and aspirated for fluid
• Bloody fluid with hematocrit 
>15% represents active 
intraperitoneal bleeding  
• Predominantly used in sites 
without onsite access to 
transvaginal ultrasound

6
Ectopic Treatment Options Surgical Management
• Surgical salpingectomy or salpingostomy by: • Mainstay of treatment
– Open laparotomy • Conservative – conservation of tube
– Laparoscopy • Surgical extirpation – removal of tube
• Criteria for selecting surgery
• Medical – Unstable vital signs or hemoperitoneum
– Methotrexate – Uncertain diagnosis
• Expectant – wait for spontaneous resolution – Advanced ectopic pregnancy
– Unreliable follow‐up
– Contraindication to expectant management or methotrexate

Medical Management: Methotrexate Criteria for Expectant Management
• Safe, effective, and less costly than surgery • Minimal pain or bleeding
• Equal or better fertility preservation • Patient reliable for follow‐up
• Criteria for use:
– Stable vital signs, few symptoms • No evidence for tubal rupture
– No contraindication to drug • Starting hCG level <1,000 mIU/mL and falling
– Unruptured ectopic
– Absence of embryonic cardiac activity • Ectopic or adnexal mass <3 cm, or not detected
– Ectopic mass ≤4 cm • No embryonic heartbeat
– Starting  beta hCG levels <5,000 to 10,000 mIU/mL

Gestational Trophoblastic Disease Clinical Manifestations
• Incidence = 1:1,000 to 1,500 pregnancies • Vaginal bleeding first/early second trimester
• Predisposing factors • Higher than expected beta hCG levels
– previous molar disease 
– pregnancy at end of reproductive life • Uterine size > dates without heart tones
• Complete hydatidiform mole • Hyperemesis
– Placental proliferation in absence of a fetus; 46,XX
– Placental villi swollen, grape‐like • Early gestational hypertension
• Partial mole  • Thyrotoxicosis
– Molar placenta + nonviable fetus; 69,XXY
• Ovarian enlargement
• Recurrence  metastatic choriocarcinoma

7
Monitoring and Treatment of 
Molar Pregnancy
Trophoblastic Disease
• Prompt evacuation of the uterus
• Serial beta hCG monitoring
• One year of contraception 
• Recurrence 
– Occurs in 20% with complete mole 
– Invades myometrium or becomes metastatic
– Treated with methotrexate
• Most can conceive, carry normal pregnancy

Grief and Loss after Miscarriage  Psychological Management
• A major loss to the pregnant woman and her family • Acknowledge, dispel guilt
• Grief reaction similar in intensity to other major losses • Legitimize grief
• Strongest feelings occur in first 6 months, but can be  • Provide comfort and ongoing support
persistent enough to cause long‐term symptoms
• Reassure about the future
• Risk factors for a stronger grief reaction:
– Missed abortion • Counsel patient on how to tell family and friends
– Loss at a later gestational age • Warn about anniversary phenomenon
– Longer time to conception of their next pregnancy • Include partner in psychological care
– Critical self‐perception
• Assess level of grief and adjust counseling accordingly
• Partners also experience grief with pregnancy loss

Rh Prophylaxis and 
Summary
Future Conception
• Discriminatory criteria can significantly aid in distinguishing normal 
• Rh‐negative women who miscarry during the first early pregnancy from miscarriage and ectopic pregnancy
trimester should receive 50 mcg of anti‐D immune • For incomplete miscarriage, nonsurgical treatments have a high 
globulin. Full dose acceptable. likelihood of success
• There is no good evidence suggesting an ideal • In embryonic demise or anembryonic pregnancy, misoprostol or 
interpregnancy interval surgical treatment are considerably more effective than expectant
treatment
• Folic acid supplementation before future conception • When choosing expectant versus surgical management of miscarriage,
attempts substantially reduce the risk of neural the woman's preference should play a dominant role in decision 
tube defects making

8
Surgical Management of Miscarriage
Summary
Optional Slides
• When the choice is made to treat early pregnancy failure other than by 
expectant means, vaginal or buccal misoprostol is highly efficacious, safe,  • Dilation and curettage (D&C)
and well‐accepted by women, with fewer gastrointestinal adverse effects 
than the oral route • Manual Vacuum Aspiration (MVA)
• Ectopic pregnancy can be treated medically or expectantly in properly 
selected cases
• Evidence does not support the use of antibiotics in all women with 
incomplete abortion
• After first trimester pregnancy loss, Rh‐negative women should receive 50 
mcg of anti‐D immune globulin
• Acknowledgement of grief, empathy, and reassurance are useful in 
counseling women after miscarriage

Suction D&C Surgery is Not Required
• Indications • Uterus is small and firm
– Heavy bleeding
– Fetal demise conclusive and patient desires not to await spontaneous  • Bleeding is scant or absent
loss
– Need to rule out ectopic pregnancy
• Tissue passed appears complete
• Contraindications • Patient is reliable for follow‐up
– Active pelvic infection
– Coagulopathy • Ultrasound (transvaginal) shows empty uterus
– Fetal demise not proven
– Patient preference to await spontaneous abortion

D&C Procedure, Part 1 Paracervical Block
• Place intravenous line
• Obtain hemoglobin and Rh factor
• Administer sedation/analgesia
• Identify size, position of uterus
• Expose cervix, grasp with tenaculum
• Administer paracervical block

9
Dilating the Cervix D&C Procedure, Part 2
• Necessary when the cervical  • Progressively dilate cervix, if needed
canal will not allow passage 
of appropriate cannula
• Insert suction curette along axis of uterus
• Gently dilate with dilators  • Avoid perforation
and cannula of increasing  • Attach suction; check valve pressure
size
• Or give misoprostol several  • Rotate curette with in‐and‐out motion
hours before the procedure • Observe and submit tissue

Curette in Place Manual Vacuum Aspiration (MVA)
• Simple handheld plastic syringe
• Generates suction mechanically
• Appropriate for very early gestations
• Useful for office procedures or where electricity is
not available

MVA Instruments and Supplies Inserting Cannula

10
Attaching MVA Cannula Creating Vacuum – First Step

Creating Vacuum – Second Step Emptying the Uterus


• Gently rotate the
syringe, taking care 
not to withdraw
cannula aperture(s)
beyond cervical os
• Do not grasp syringe
by the plunger arms

Releasing the Pinch Valve D&C Procedure, Part 3
• When the pinch valve is • Repeat suction curettage sequence
released, the vacuum is 
• Sharp curettage optional
transferred through the 
cannula to the uterus • Withdraw suction without touching vaginal side wall
• Blood, tissue, and  • Monitor for bleeding
bubbles will flow 
through the cannula  • Administer Rh immune globulin if Rh negative
into the syringe

11
Uterine Perforation Complications of D&C
• Perforation
• Incomplete evacuation
• Bleeding
• Infection
• Late sequelae
– Intrauterine synechiae is rare without sharp curettage
– Depression and psychological reactions

12
Chapter B
Medical Complications of Pregnancy

Larry Leeman, MD, MPH, Lee Dresang, MD, Patricia Fontaine, MD


Revised February 2015

Learning Objectives
At the end of this activity, learners will be able to:
1. Describe the following potentially life-threatening c. Peripartum cardiomyopathy.
medical complications of pregnancy: d. Deep venous thrombosis.
a. Hypertensive disorders. e. Pulmonary embolism.
b. Acute fatty liver of pregnancy. 2. Formulate a plan for diagnosis and treatment.

Introduction ing up to 10 percent of gestations.1 As defined by the


The presence of a fetus complicates many medi- National High Blood Pressure Education in Pregnancy
cal problems as a result of the complex interactions (NHBPEP) Working Group, hypertension in preg-
between the mother, the disease, and the treatment. nancy may be chronic (occurring prior to 20 weeks’ ges-
Understanding these interactions is crucial in optimiz- tation or persisting beyond 12 weeks post partum), may
ing outcomes for both mother and baby. The mother arise de novo during the pregnancy (gestational hyper-
is the first priority in any medical emergency since tension or preeclampsia), or may represent a superim-
the fetus is dependent on her for physiologic sup- position of preeclampsia on chronic hypertension.1,2
port. In managing an eclamptic seizure or pulmonary
embolism, for example, every effort is directed toward Chronic Hypertension
supporting maternal vital functions and using neces- Chronic hypertension is defined as an elevated blood
sary critical care interventions. Concern for the fetus pressure greater than 140/90 mm Hg on two occasions
is demonstrated by choosing expectant management at least four hours apart prior to or during the first 20
for preeclampsia without severe features when the fetal weeks of pregnancy. Chronic hypertension is associated
gestational age is < 37 weeks, treating the mother with with adverse perinatal outcomes including preeclamp-
medications that minimize adverse fetal effects, and sia, intrauterine growth restriction and placental abrup-
by choosing tests that limit direct fetal x-ray exposure. tion. The severity of maternal blood pressure at 20
This chapter focuses on four potentially life-threatening weeks is associated with worse outcomes.3 Treatment
medical complications: hypertensive disorders, acute of mild to moderate chronic hypertension in pregnancy
fatty liver of pregnancy (AFLP), peripartum cardio- has no proven fetal benefit, nor has it been shown to
myopathy (PPCM), and thromboembolic disease. The prevent preeclampsia.4,5 Excessively lowering the blood
hypertensive disorders are the most common medical pressure may result in decreased placental perfusion and
complications of pregnancy, while AFLP and PPCM adverse perinatal outcomes.6 However, when the blood
are uncommon disorders unique to pregnancy that pressure is persistently greater than 150 to 160/100
cause significant morbidity and mortality. Thrombo- to 110 mm Hg, pharmacologic treatment is indicated
embolic disease is a leading cause of maternal mortality in order to prevent maternal end organ damage.1,2,7 A
in more developed countries. lower threshold is appropriate for treating women who
already manifest target organ damage such as renal
Hypertensive Disorders of Pregnancy insufficiency and left ventricular hypertrophy.1
Worldwide, hypertensive disorders represent the most Methyldopa, labetalol, and nifedipine are the oral
common medical complication of pregnancy, affect- agents most commonly used for severe, chronic hyper-

Copyright 2017© American Academy of Family Physicians. All rights reserved. — Chapter B  1
Chapter B

tension in pregnancy in the 2013 report of the Gestational Hypertension


American College of Obstetricians and Gynecolo- The NHBPEP Working Group has recommended
gists (ACOG) Hypertension in Pregnancy task that “gestational hypertension” replace the term
force.2 ACE inhibitors and angiotensin II receptor “pregnancy-induced hypertension.”1 Pregnant
antagonists should not be used due to associa- women who develop hypertension after 20 weeks
tion with intrauterine growth restriction (IUGR), and do not have significant, preeclampsia-level pro-
neonatal renal failure, oligohydramnios, effects of teinuria should be diagnosed with gestational hyper-
oligohydramnios (including limb abnormalities, tension. Gestational hypertension is a provisional
cranial ossification defects and pulmonary hyper- diagnosis used for a heterogeneous group of women
plasia) and death.2 The beta-blocker atenolol, has including 1) those that will eventually develop
been associated with IUGR.2 Thiazide diuret- proteinuria during the pregnancy and be diagnosed
ics may be continued if used prior to pregnancy with preeclampsia, 2) those who will have persistent
however they can exacerbate the intravascular hypertension after 12 weeks postpartum and be
fluid depletion of preeclampsia if chronic hyper- diagnosed with chronic hypertension, and 3) those
tension becomes complicated by superimposed who do not develop preeclampsia and whose blood
preeclampsia and in the scenario must be discon- pressures normalize postpartum. Women in the
tinued.8,9 For this reason thiazide diuretics are last group are ultimately diagnosed as having had
not first line antihypertensive agents for chronic “transient hypertension of pregnancy.”
hypertension in pregnancy. Gestational hypertension is not a benign cat-
Women in active labor with uncontrolled egory. Approximately 50 percent of women
severe chronic hypertension require treatment diagnosed with gestational hypertension between
with intravenous labetalol or hydralazine in doses 24 to 35 weeks ultimately develop preeclampsia.11
similar to those used for preeclampsia with severe Expectant management of gestational hyperten-
features as described below.9 Although intrave- sion can reduce the increased cesarean delivery rate
nous medications have traditionally been recom- that occurs with inductions.12 If the blood pressure
mended rather than oral medications, a small progresses to the severe range (systolic greater than
2013 RCT demonstrated a more rapid lowering 160 mm Hg or diastolic greater than 110 mm Hg),
of blood pressure with oral nifedipine compared then management similar to a preeclamptic with
to intravenous labetalol10 and oral nifedipine is severe features is required even if the patient does
considered to be a therapeutic option for severe not have proteinuria, because women with severe
acute hypertension in the 2013 ACOG Task gestational hypertension have worse perinatal
Force recommendations.2 outcomes than women with preeclampsia without
Women with chronic hypertension should severe features.13 ACOG recommends induction at
be monitored carefully for the development of 37 weeks if not delivered sooner.8 A retrospective
super-imposed preeclampsia and IUGR.2 The analysis of women with gestation hypertension,
development of proteinuria or a sudden sustained mild preeclampsia and mild chronic hypertension
increase in proteinuria, a sudden increase in blood demonstrated higher rates of maternal ICU admis-
pressure in a woman whose hypertension has sion, postpartum hemorrhage and blood transfu-
previously been well controlled, or development sion in the gestational hypertension group.14
of the severe features of preeclampsia (RUQ pain,
headache, visual changes, pulmonary edema, rise Preeclampsia
in creatinine or transaminases, or development of Definitions
thrombocytopenia (< 100,000/mcL), are diagnos- Preeclampsia is a multi-organ disease process char-
tic for superimposed preeclampsia.2 Fetal growth acterized by hypertension and either proteinuria or
should be assessed by serial ultrasounds starting severe features of preeclampsia. To meet diagnos-
after 24 weeks of gestation to screen for develop- tic criteria for preeclampsia, systolic blood pressure
ing IUGR.2 Although we lack evidence for an must be 140 mm Hg or greater, or diastolic blood
optimal interval for fetal growth ultrasound assess- pressure 90 mm Hg or greater, on at least two
ments, every four weeks is a reasonable option if occasions no less than fours hours apart.2 Blood
no evidence of intrauterine growth restriction or pressure should be measured at each prenatal visit
superimposed preeclampsia. using an appropriate sized cuff with the patient

2  Chapter B —
Medical Complications of Pregnancy

seated in an upright position. If the initial blood specific abnormal lab findings, or the presence of
pressure is elevated then a repeat measurement is clinical symptoms resulting from involvement of
checked after at least a 5-minute rest. If the blood the kidneys, brain, liver and cardiovascular system.
pressure demonstrates a systolic of ≥ 160 mm Hg Proteinuria is no longer a criteria for preeclampsia
or a diastolic ≥ 110 mm Hg then a blood pressure with severe features as higher levels of protein is
can be confirmed within minutes and the diag- not an indicator of disease severity.2
nosis of preeclampsia may be rapidly made. An The etiology of preeclampsia remains unknown
increase in blood pressure of 30 mm Hg systolic or and no single causal factor links all theories
15 mm Hg diastolic is no longer included in the (Table 1).16 Growing evidence suggests the disease
definition of preeclampsia2 as similar increases are is a multi-organ disease and not just high blood
common in uncomplicated pregnancies. pressure and proteinuria. It is evident that the pla-
The diagnostic threshold for proteinuria is centa plays a central role in pre-eclampsia.2 Despite
300 mg in a 24-hour specimen or a urine protein/ the identification of a number of bio-markers and
creatinine ratio > 0.3.2 A dipstick reading lacks clinical risk factors a recent study of nulliparous
sensitivity or specificity for proteinuria, however women found the predictive benefit of these factors
if a 24 hour analysis or urine protein/creatinine to be modest and none are indicated outside of
ratio is not available then two random urine research studies.17 Risk factors are listed in Table 2.
dipstick measurements greater than or equal to 1+
(30 mg/dl) six hours apart indicates the presence
of proteinuria. However, a 24 hour determina- Table 1. Theories Associated with the
tion is the gold standard because urine dipsticks Pathophysiology of Preeclampsia
can be affected by dehydration and bacteriuria. A
catheterized UA may avoid protein due to con- Genetic predisposition (maternal, paternal,
thrombophilia)18,19
tamination, though a traumatic catheterization
Immunologic phenomena19
can introduce protein from blood. In selected
clinical circumstances a 12 hour urine collection Abnormal placental implantation (defects in
trophoblasts and spiral arterioles)22-23
to quantitate protein is another alternative. A
random urine can rule out significant proteinuria Vascular endothelial damage24 and oxidative stress
if the protein/creatinine ratio is less than 0.19.15 Angiogenic factors (low level of placental growth
Since proteinuria occurs late in the course of pre- factor)25,26
eclampsia it is not useful for screening.
The diagnosis of preeclampsia can be made
without proteinuria if any of the following Table 2. Preeclampsia Risk Factors27-29
severe features of pregnancy are present: platelets
< 100,000/mL, serum creatinine > 1.1 mg/dLor Family history of preeclampsia (1st generation
a doubling of serum creatinine from baseline (if relative) 3X
known) without another etiology, pulmonary Nulliparity 3X
edema, or elevation of transaminase to twice the Maternal age greater than 40 1.6X
normal level. The presence of cerebral or visual
Multiple gestation 3X
symptoms is also sufficient to diagnose pre-
Preeclampsia in a prior pregnancy (particularly if
eclampsia in the setting of elevated blood pressure.2 severe or prior to 32 weeks) 7X
Edema supports the diagnosis of preeclampsia
Chronic hypertension and/or renal disease
when it is pronounced and generalized (affecting
the face or hands), but is no longer a diagnostic Systemic Lupus Erythematosus/Antiphospholipid
syndrome
criteria. One third of preeclamptic women never
have edema, while non-dependent edema is seen Elevated body mass index 2X
in a significant proportion of women without Diabetes mellitus (preexisting) 3X
preeclampsia.1
Note: Previously, young maternal age was considered a
The distinction between preeclampsia and risk factor, but this was not supported by a systematic
preeclampsia with severe features is based upon review.27
the degree of blood pressure elevation, presence of

— Chapter B  3
Chapter B

Prevention task force recommended against supplementation


Randomized controlled trials fail to support a except in populations with low calcium intake.2,36
role for routine prenatal supplementation with An association has been shown between low
calcium, omega three fatty acids, or antioxidant levels of Vitamin D and the subsequent develop-
vitamins E and C to prevent preeclampsia.28,30-35 ment of preeclampsia however it is unknown if
Calcium supplementation may decrease the inci- supplementation will decrease the incidence of
dence of hypertension, preeclampsia and maternal preeclampsia. Antiplatelet agents (e.g. low dose
death among women at high risk of developing aspirin), have small to moderate benefits for pre-
those conditions and women with low calcium vention. A Cochrane analysis of low-dose aspirin
intakes.36 The World Health Organization has for women at increased risk for preeclampsia dem-
recommend routine supplementation with of onstrated a 17% reduction in the risk of develop-
1.5 to 2.0 grams of elemental calcium for women ing preeclampsia with a NNT of 72 to prevent one
with low calcium intake.37 Women in the United case of preeclampsia. In the subgroup of women
States or other high resource countries are unlikely at highest risk due to a history of previous pre-
to have low calcium intake due to the widespread eclampsia with severe features, diabetes, chronic
supplementation of food and the ACOG 2013 hypertension, renal or autoimmune disease, only
19 women needed to be treated with low-dose
aspirin to prevent one case of preeclampsia.38
Figure 1. Recommended Management of Gestational The 2013 ACOG taskforce recommended
Hypertension or Preeclampsia Without Severe Features39 initiation of 60 to 80 mg of aspirin in the late first
trimester in women with history of preeclampsia
Maternal and fetal findings in a prior pregnancy requiring delivery before
34 0/7 weeks or had preeclampsia in more than
one prior pregnancy.2
37 0/7 weeks or more of gestation
or Management of Preeclampsia without
37 0/7 weeks or more of gestation with: Severe Features
Labor or rupture of membranes
Abnormal maternal-fetal test results
Expectant management of women with pre-
Ultrasonographic estimate of fetal eclampsia without severe features may include
weight less than fifth percentile twice weekly blood pressures, weekly lab tests
Suspected abruptio placentae
(CBC, ALT and/or AST, LDH, uric acid, and
creatinine), twice weekly non-stress tests (NSTs)
No Yes and weekly amniotic fluid indices (AFIs) or weekly
biophysical profiles (BPPs), and ultrasounds for
Less than 37 0/7 weeks of gestation
fetal growth every three weeks.1,2,39 Fetal umbili-
Inpatient or outpatient management cal artery Doppler studies are recommended to be
Maternal evaluation: twice weekly included in antenatal surveillance for women with
Fetal evaluation
With preeclampsia: twice weekly
preeclampsia when IUGR has been detected.2
nonstress test The decision to bring about delivery by induc-
With gestational hypertension: tion or cesarean section involves balancing pre-
weekly nonstress test maturity-related risks with the risk of worsening
preeclampsia. Delivery is generally indicated for
women with preeclampsia or gestational hyperten-
37 0/7 weeks or more of gestation Delivery sion at 37 weeks based on the HYPITAT RCT
Yes
Worsening maternal or fetal condition Prostaglandins
Labor or premature rupture of if needed for
of induction vs., expectant management and the
membranes induction recommendations of a 2011 workshop sponsored
by the Society of Maternal Fetal Medicine and the
National Institute of Child Health and Human
Development.40.41 A secondary analysis of HYPI-
Information from Roberto, JM, August, PA, Bakris, G, et al. Task force on Hyper-
tension in Pregnancy: American College of Obstetrics and Gynecology, 2013. TAT showed greater benefit of labor induction
on preventing high-risk maternal situations and

4  Chapter B —
Medical Complications of Pregnancy

reducing the cesarean delivery rate in women with treatment goals are to 1) prevent seizures, 2) lower
an unfavorable cervical exam presumably because blood pressure in order to prevent maternal cere-
these women were more remote from spontane- bral hemorrhage, and 3) expedite delivery based
ous labor.42 An economic analysis of HYPITAT on a decision that takes into account disease sever-
demonstrated cost saving from labor induction as ity and fetal maturity.
compared to expectant monitoring.43
Maternal Evaluation and Stabilization
Severe Features of Preeclampsia Sample admitting orders for preeclampsia with
Diagnostic criteria for preeclampsia with severe severe features are outlined in Table 4. Fluid
features are listed in Table 3. Preeclampsia with management requires special care. Excessive fluid
severe features may result in multi-system dete-
rioration that can be gradual or fulminant. Severe
headache, visual disturbances, and progressive Table 4. Admitting Orders for Severe Preeclampsia with
hyperreflexia may signal impending generalized Severe Features
seizures (eclampsia). Increasing peripheral vascu-
lar resistance stresses the cardiovascular system, Bed rest with seizure precautions
and pulmonary edema may result. A decreased Vital signs (blood pressure, pulse, respiration), deep tendon reflexes,
glomerular filtration rate may progress to oliguria and neurologic checks every 15 minutes until stable
and acute renal failure. Hemodilution usually Accurate intake and output; Foley catheter if needed
lowers pregnancy creatinine levels; levels above Intravenous: Lactated Ringer at 50 to 125 ml per hour to maintain urine
0.9 mg/dl in pregnancy are abnormal. Liver mani- output of 30 to 40 ml per hour. Total intake (intravenous and oral)
festations include elevated transaminases, sub- should not exceed 125 ml per hour or 3000 ml per day
capsular hemorrhage with right upper quadrant External monitor for contractions and fetal heart rate
pain, and capsular rupture with life-threatening Labs:
intraabdominal bleeding. Preeclampsia-related Dipstick urine for protein on admission (not needed if have definitive
coagulopathies include HELLP syndrome and finding of severe features)
disseminated intravascular coagulation (DIC). Begin 24 hour urine for total protein (not needed if have definitive finding
of severe features)
Obstetric complications include IUGR, abrup-
Complete blood count and platelet count
tion, and fetal or maternal demise.
Creatinine
AST or ALT
Management of Preeclampsia with
Uric acid
Severe Features LDH
The progression of preeclampsia is only reversed Peripheral blood smear
by delivery. Patients with preeclampsia with severe Medications:
features should be admitted to the hospital, placed 1. Magnesium sulfate (see Table 5 for dosing)
on bedrest, and carefully monitored.39 The overall 2. For systolic BP ≥ 160 mm Hg or diastolic BP ≥ 110 mm Hg on two
measurements 15 minutes apart, give one of the following:
Hydralazine 5 to 10 mg IV over two minutes, if after 20 minutes BP
Table 3. Diagnostic Criteria for Severe remains elevated, then give additional 10 mg IV. If above threshold
Preeclampsia with Severe Features after 20 minutes then change to IV Labetalol.9
or
Blood pressure equal to or exceeding 160 mm Hg Labetalol 20 mg IV initial dose over 2 minutes. If BP remains elevated
systolic or 110 mm Hg diastolic on at least two after 10 minutes, double the dose to 40 mg and then to 80 mg at 10
occasions four hours apart to 30 minute intervals until target blood pressure is reached. If SBP
is ≥ 160 mm Hg or DPB ≥ 100 mm Hg after the 80 mg dose, then
Any of the following signs and symptoms: change to IV Hydralazine.1,39 The maximum dose of IV labetalol is
Progressive renal insufficiency (Serum creatinine 300 mg in a 24 hour period.9
> 1.1 mg/dL or double baseline) or
Cerebral or visual disturbances Nifedipine 10 to 20 mg orally. May repeat dose every 30 minutes if
Pulmonary edema needed until 40 mg po has been given then administer 10 to 20 mg
Impaired liver function (transaminases 2x normal), po every 4 to 6 hours.
right upper quadrant pain or epigastric pain 3. Calcium gluconate one gram IV: keep at bedside in case of respiratory
Thrombocytopenia (< 100,000/ml) depression due to magnesium sulfate.

— Chapter B  5
Chapter B

administration can result in pulmonary edema, severe features should be monitored closely and
ascites and cardiopulmonary overload, while magnesium started if they develop severe features.2
too little fluid can exacerbate an already con- Magnesium sulfate works by slowing neuromus-
stricted intravascular volume and lead to further cular conduction and depressing central nervous
end-organ ischemia. Urine output should be system irritability. It does not have significant
maintained above 30 ml per hour using intra- effects on lowering blood pressure. A quarter of
venous lactated ringers or normal saline.44 Total women have side effects, most commonly flush-
intravenous fluid intake should be limited to ing.56 Table 5 presents a standard dosing regimen.
100 ml per hour44,45 and total oral and intrave- Magnesium sulfate is excreted by the kidneys.
nous fluid intake should not exceed 125 ml per Women with normal renal function do not require
hour or 3,000 ml per day. A Foley catheter allows routine monitoring of serum magnesium levels,
accurate monitoring of urine output. A Swan- however women with absent reflexes, elevated
Ganz catheter may optimize fluid management serum creatinine or decreased urine output
if pulmonary edema and renal failure are present (< 30 cc/hr), should have magnesium levels drawn
but should not be routinely used.44 every six hours after the loading dose has been given
Plasma volume is reduced among women with and the infusion rate adjusted accordingly.55,59
preeclampsia, suggesting that increasing plasma Magnesium toxicity can lead to respiratory
volume with colloid solution might improve paralysis, central nervous system depression and
uteroplacental circulation, and perinatal outcomes. cardiac arrest. With magnesium overdose, vital
However, risk/benefit data regarding this practice functions are lost in a predictable sequence. If
is lacking.46 DTRs are present, magnesium concentrations are
In addition to the basic laboratory investigation rarely toxic.59 The magnesium sulfate infusion
for preeclampsia, the woman with signs of severe should be discontinued and a magnesium level
disease may be evaluated with, LDH, peripheral checked immediately if absent DTRs are noted,
blood smear and labs for evidence of hemolysis the respiratory rate is less than 12 per minute,
and disseminated intravascular coagulation (DIC) or urine output is less than 30 ml per hour.55,59
depending upon clinical scenario. Maternal deaths have resulted from overdoses due
to administration of improperly prepared solu-
Magnesium Sulfate (MgSO4) tions.60 The antidote for magnesium sulfate over-
Magnesium sulfate helps prevent seizures in dose is one gram of calcium gluconate (10 ml of
women with preeclampsia47-49 and is more effec- a 10 percent solution) infused intravenously over
tive in preventing recurrent seizures in eclamptic two minutes.44 Avoid rapid intravenous admin-
patients than phenytoin, diazepam or a lytic cock- istration or extravasation. Use calcium gluconate
tail (chlorpromazine, promethazine and meperi-
dine).48,50-53 The Magpie trial demonstrated that
63 women with severe preeclampsia need to Table 5. Magnesium Sulfate in
receive magnesium sulfate prophylaxis to prevent Preeclampsia with Severe Features or
one eclamptic seizure.47 Severe Gestational Hypertension57
Should magnesium sulfate be used for women
Loading dose: four to six grams mixed in 100 ml,
with preeclampsia without severe features? given IV over 15 to 20 minutes, followed by a
Assuming 50 percent of seizures are prevent- continuous infusion of two grams per hour
able by MgSO4,47 then 400 women with mild Monitor:
preeclampsia need to be treated to prevent one Reflexes
eclamptic seizure.54 The 2013 ACOG Task Force Mental status
report recommended that preeclamptic women Respiratory status
who are not symptomatic and have blood pressure Urine output
under 160/110 mm Hg should not universally Magnesium levels (therapeutic range = 4 to 8 mg/dl)58
receive magnesium sulfate for seizure prophylaxis. should be checked if renal dysfunction elevated
creatinine 70.9 mg/dL, urine output < 30 cc/hr,
However, blood pressure is only mildly elevated loss of reflexes or other symptoms of magnesium
in 30 to 60 percent of women who develop toxicity
eclampsia;55 Women with preeclampsia without

6  Chapter B —
Medical Complications of Pregnancy

with caution in women with renal failure, severe these three antihypertensive agents is supported by a
hypophosphatemia, or acidosis. 2013 ACOG task force report on HTN in preg-
nancy,2 the United Kingdom’s NICE guidelines7
Antihypertensive Medications and a Cochrane review.64 Oral labetalol at a dose of
The optimal level of blood pressure control in 200 mg is considered an alternative by ACOG for
pregnancies complicated by hypertension is lowering severe blood pressure when intravenous
unknown.61,62 Less tight control may decrease the medications are not an option and is recommended
risk of infants being small for gestational age, but in the NICE guidelines.7,9 If the blood pressure
may potentially increase the risk of respiratory remains ≥ 160/110 mm Hg and intravenous medi­
distress syndrome, severe hypertension, antenatal cations are still not an option then 200 mg oral
hospitalization, and proteinuria at delivery.6,61 labetalol can be repeated.9 We recommend that each
Although traditional recommendations are based maternity care unit choose a single first line medica-
on diastolic blood pressures, a retrospective review tion as well as have alternatives available for women
of 28 women with preeclampsia with severe fea- who are refractory to the selected agent.
tures who experienced a cerebrovascular accident For women with preeclampsia with severe
demonstrated that over 90 percent had systolic features undergoing expectant management at a
BP over 160 mm Hg, but only 12.5 percent had gestational age under 34 weeks, oral labetalol and
diastolic BP over 110 mm Hg.63 nifedipine are acceptable options.39 Delivery is
There are several possible choices for the antihy- recommended for women with preeclampsia with
pertensive agent depending on whether the goal is severe features at 34 0/7 weeks or greater.2
acute or chronic control. For acute management,
intravenous labetalol and hydralazine are com- Fetal Surveillance
monly used.1,64 Doses for intravenous labetalol Assessment for uteroplacental insufficiency may be
and hydralazine and oral nifedipine are given in achieved utilizing non-stress tests (NSTs), amni-
Table 4, which is based on the 2013 ACOG report otic fluid measurements and biophysical profiles.
on Hypertension in Pregnancy.2 A Cochrane Umbilical artery Doppler systolic-to-diastolic
review of medications for treating severe hyperten- ratios may detect early uteroplacental insufficiency
sion in pregnancy showed no evidence that one and this examination is indicated for fetuses with
agent had superior effectiveness.64 The role of intrauterine growth restriction. The presence of
hydralazine as a first line choice has been ques- reversed end diastolic umbilical artery flow is an
tioned by a meta-analysis showing more maternal indication for delivery (after corticosteroids are
hypotension, tachycardia, and headaches com- administered if < 34 weeks EGA).2 Monitoring
pared to other antihypertensives.62 The need for frequency varies depending on the clinical context.
intravenous antihypertensives, either in repeated A common regimen for preeclampsia without
doses or by continuous infusion, indicates an severe features at less than 37 0/7 weeks includes
unstable patient who is likely to need continuous twice weekly NSTs and weekly measurement of
monitoring and careful management. amniotic fluid index with biophysical profile for
Oral nifedipine or labetalol are alternatives to follow-up of non-reactive NSTs.1,39 Ultrasound
intravenous medications when severe range blood for assessment of fetal growth should be repeated
pressures require treatment. Traditionally intra- every three weeks.2 Women with gestational
venous medications have been preferred to allow hypertension at less than 37 0/7 weeks may receive
for rapid lowering of blood pressure and careful antenatal surveillance with weekly NSTs and
titration to avoid maternal and fetal effects of an amniotic fluid volume measurement. Women with
excessive decrease in blood pressure. In two studies preeclampsia with severe features are admitted
nifedipine has been shown to control blood pressure to a hospital and may receive daily monitoring.
more rapidly than intravenous labetelol,10,65 and a Corticosteroids are administered to accelerate lung
third trial demonstrated equivalent time to adequate maturity for fetuses between 24 and 34 weeks
blood pressure control.66 Nifedipine has been shown gestation, either betamethasone (two doses of
to cause a greater increase in both cardiac index66 12 mg given intramuscularly 24 hours apart) or
and urinary output65 than labetalol, as well as a dexamethasone (four doses of 6 mg given intra-
decreasing systemic vascular resistance. The use of muscularly 12 hours apart).39

— Chapter B  7
Chapter B
Figure 2. Management of Preeclampsia and HELLP Syndrome

• Observe in labor and delivery for first 24 to 48 hours


• Corticosteroids, magnesium sulfate prophylaxis, and anti­
hypertensive medications
• Ultrasonography, monitoring of fetal heart rate, symptoms,
and laboratory tests

Contraindications to continued expectant management


• Eclampsia
• Pulmonary edema
• Disseminated intravascular coagulation
Delivery once maternal Yes
• Uncontrollable severe hypertension
condition is stable
• Nonviable fetus
• Abnormal fetal test results
• Abruptio placentae
• Intrapartum fetal demise

No

Are there additional expectant complications?


• Greater than or equal to 33 5/7 weeks of gestation
• Persistent symptoms
Corticosteroids for fetal • HELLP or partial HELLP syndrome
Yes
maturation • Fetal growth restriction (less than fifth percentile)
Delivery after 48 hours • Severe oligohydramnios
• Reversed end-diastolic flow (umbilical artery Doppler studies)
• Labor or premature rupture of membranes
• Significant renal dysfunction

No

Expectant management
• Facilities with adequate maternal and neonatal intensive care
resources
Yes • Fetal viability—33 6/7 weeks of gestation
Delivery
• Inpatient only and stop magnesium sulfate
• Daily maternal—fetal tests
• Vital signs, symptoms, and blood tests
• Oral antihypertensive drugs

No

• Achievement of 34 0/7 weeks of gestation


• New-onset contraindications to expectant management
• Abnormal maternal-fetal test results
• Labor or premature rupture of membranes

HELLP = hemolysis, elevated liver enzymes, and low platelet count.


From Roberto, JM, August, PA, Bakris, G, et al. Task force on Hypertension in Pregnancy: American College of Obstetrics
and Gynecology, 2013.

Delivery Decisions in Preeclampsia with compromise on antenatal assessment. Maternal fac-


Severe Features tors include the degree to which the hypertension
Delivery is the only known cure for preeclamp- is controllable and any clinical or laboratory signs
sia. Decisions regarding the timing and mode of of impending decompensation. For patients with
delivery are based on a combination of maternal resistant severe hypertension, eclampsia pulmo-
and fetal factors. Fetal factors include gestational nary edema, placental abruption or other signs of
age, evidence of lung maturity and signs of fetal maternal or fetal deterioration delivery is indicated

8  Chapter B —
Medical Complications of Pregnancy

after maternal stabilization without waiting the full indications for cesarean delivery may include status
48 hours for antenatal corticosteroids, irrespective epilepticus, severe range blood pressures resistant to
of gestational age. Women at < 34 weeks gestation medication management, or other situations indicat-
should be delivered after 48 hours of antenatal ing worsening of maternal condition remote from
corticosteroids for the indications of thrombocyto- delivery (e.g. pulmonary edema, severe thrombocy-
penia with platelets < 100,000/ml, transaminases topenia). Some experts recommend cesarean deliv-
twice the normal value, IUGR (< 5 percent), severe ery for fetuses under 30 weeks when the cervix is not
oligohydramnios (AFI < 5 cm), umbilical artery ripe, but a trial of induction may be considered.1,39
reversed end-diastolic flow or new or worsening
renal dysfunction. If maternal and fetal conditions Postpartum Management of Preeclampsia
allow, trying to delay labor and give corticosteroids Most patients with preeclampsia respond
is recommended for preeclampsia in the setting promptly to delivery, with decreased blood pres-
of preterm premature rupture of membranes or sure, diuresis, and general clinical improvement.
preterm labor.2 Eclampsia may occur postpartum with the greatest
There is limited data regarding the optimal risk for postpartum eclampsia occurring in the first
management of women with preeclampsia with 48 hours.55 Magnesium sulfate should be contin-
severe features between 24 and 34 weeks with the ued for 12 to 24 hours, or occasionally longer if
2013 Cochrane review based on only four RCTS the clinical situation warrants.39,55,73 Patients on
with a total of 425 women.67-70 The use of expect- magnesium sulfate require ongoing monitoring
ant management with close maternal and fetal of blood pressure and urine output, as they are at
surveillance in a hospital with perinatal and neona- risk for pulmonary edema due to intravenous fluid
tology services appears to decrease neonatal mor- overload, mobilization of third space fluids, and
bidity and length of stay in the newborn intensive decreased renal function.
care unit (NICU), however many women are not Hypertension may worsen in the days follow-
candidates for expectant management or may need ing delivery as fluid in the “third space” returns
urgent delivery due to complications including to the vasculature. For this reason ACOG recom-
eclampsia, HELLP syndrome, pulmonary edema, mends observation in the hospital for 72 hours
renal insufficiency, concerning fetal surveillance, after delivery with gestational hypertension and
or placental abruption.67-70 In one study, bed rest preeclampsia or the equivalent monitoring at
and close monitoring of women between 28 to 32 home. As there are no longer fetal concerns with
weeks with preeclampsia prolonged pregnancy by regard to blood pressure lowering the postpartum
an average of 15 days, resulting in fewer days in threshold to SBP of ≥ 150 or DBP ≥ 100 at least
the NICU and fewer cases of respiratory distress four hours apart. If blood pressure is ≥ 160 or
syndrome and necrotizing enterocolitis, without DBP ≥ 110, recheck within 15 minutes and if BP
increasing maternal morbidity.69 The largest RCT remains elevated initiate antihypertensive treat-
is the Mexpre Latin study which was a multi- ment within 60 minutes of diagnosis. Women
site study of 8 centers in Latin America, which with persistent blood pressure elevation greater
despite a delay in delivery of 10.3 vs. 2.2 days, did than 24 hours after delivery should not be treated
not show neonatal benefit.71 Of note the varied with nonsteroidal anti-inflammatory agents as
criteria for intervention led to many more deliver- these may worsen blood pressure.2 Although we
ies for uncontrolled BP than in Sibai’s smaller US lack high quality studies on postpartum hyperten-
study.69,71 A commentary accompanying the Mex- sive management,74 oral nifedipine or labetalol are
pre study recommended that a woman with pre- commonly used and if needed intravenous labet-
eclampsia with severe features should be delivered alol or hydralazine may be used as described for
after administration of corticosteroids in countries intrapartum management.9,39 Patients should be
with limited resources rather than attempting evaluated in the office 7 to 10 days after hospital
continued expectant management.71,72 discharge or sooner if they are symptomatic.2
Attempted vaginal delivery is recommended for
women with preeclampsia with severe features if Eclampsia
there is no evidence of maternal or fetal compro- The generalized seizures of eclampsia represent a
mise, or other obstetric contraindication.1 Potential life-threatening emergency, requiring immediate

— Chapter B  9
Chapter B

attention while honoring the concept of “primum left side and suctioning her mouth. Summon
non nocere” or “first do no harm.” someone skilled in intubation to be immediately
available.77 The adult CPR recovery position
Pathophysiology involves the patient being in as lateral a position
Preeclampsia is characterized by a loss of regula- as possible. Allow for observation of breathing and
tion of cerebral blood flow and plasma exudation avoiding any pressure on the chest.78 This posi-
into the brain. The precise mechanism leading tion helps a semiconscious or unconscious person
to seizures is unknown, but may include cerebral breathe and permits fluids to drain from the nose
edema, transient vasoconstriction, ischemia, or and throat to avoid aspiration; in addition, it
microinfarcts.55 maximizes venous return.

Clinical Course 3. Prevent maternal injury. Falls from the bed


Eclampsia may be preceded by worsening of the can result in contusions or fractures, and head
signs and symptoms of preeclampsia with severe injury may result from violent seizure activity.
features, or may appear unexpectedly in a patient Close observation, soft padding and use of side rails
whose preeclampsia lacked severe features and had on the bed may help prevent these complications.
minimally elevated or normal blood pressure. In
one large series, 15 percent of the cases had dia- 4. Give magnesium sulfate to control seizures.
stolic blood pressure below 90 mm Hg.75 It is rare If the patient with preeclampsia has already
for eclampsia to occur prior to 20 weeks gestation received a prophylactic loading dose of magne-
in the absence of gestational trophoblastic disease. sium sulfate and is receiving a continuous infusion
Neurological symptoms often precede eclamptic when the seizure occurs, an additional two grams
seizures as demonstrated by a study of 46 eclamp- should be infused intravenously. Otherwise, a six-
tic women at a Tanzanian hospital. Eighty percent gram loading dose of magnesium sulfate should
of these women had a preceding headache and be given intravenously over 15 to 20 minutes,
forty five percent had visual changes.76 followed by a maintenance dose of two grams per
Eclamptic seizures usually last from 60 to hour. A total of more than eight grams magnesium
90 seconds, during which time the patient is with- sulfate should not be exceeded over a short period
out respiratory effort. A postictal phase may follow of time.55,77 A serum magnesium level may be
with confusion, agitation, and combativeness. The obtained four to six hours after the loading dose,
timing of an eclamptic seizure can be antepartum and the maintenance infusion adjusted accord-
(53 percent), intrapartum (19 percent), or post- ingly. After the seizure has ended, administer
partum (28 percent).75 supplemental oxygen. When the patient has stabi-
lized, plan for prompt delivery. Avoid the tempta-
Management tion to perform immediate cesarean delivery for a
An eclamptic seizure is dramatic and disturbing. self-limited seizure episode.
The attending clinician is challenged to remain
calm and avoid unnecessary interventions that can Maternal and Fetal Outcomes in Eclampsia
result in iatrogenic complications.55,77 The perinatal mortality from an eclamptic seizure in
high resource area is less than one percent, however
1. Do not attempt to shorten or abolish the in low resource settings, rates of 6.7 to 7.5 percent
initial seizure by using drugs such as diazepam were demonstrated in a 2008 study in Morrocco79
or phenytoin. These drugs can lead to respiratory and 2011 study in Nigeria, respectively.
depression, aspiration, or frank respiratory arrest, From 2006-2010, 9.4 percent of US preg-
particularly when they are given repetitively or used nancy-related deaths were due to hypertensive
in combination with magnesium sulfate. Further, disorders of pregnancy.81 An early study found
phenytoin is less effective than magnesium sulfate approximately 50 percent of US preeclampsia/
in preventing recurrent eclamptic seizures.50 eclampsia related deaths occurred in women with
eclampsia.82 Abruption (seven to 10 percent),
2. Protect the airway and minimize the risk disseminated intravascular coagulation (seven to
of aspiration by placing the woman on her 11 percent), aspiration pneumonia (two to three

10  Chapter B —
Medical Complications of Pregnancy

percent) and cardiopulmonary arrest (two to five gastrointestinal bleeding. Physical findings include
percent) are serious causes of morbidity and mor- right upper quadrant and epigastric tenderness. As
tality in eclamptic women.79 12 to 18 percent of women with HELLP are nor-
Most fetal eclampsia-related morbidity and motensive and 13 percent do not have proteinuria,
mortality result from prematurity, growth restric- clinicians must consider HELLP in patients who
tion and placental abruption. During an eclamptic lack these classic findings of preeclampsia.85
seizure, the fetus will frequently manifest hypoxia-
related bradycardia. In the absence of other serious Differential Diagnosis of HELLP Syndrome
medical or obstetric complications, the fetus usu- One of the most difficult challenges posed by
ally recovers. HELLP syndrome is its extensive differential diag-
In rural or remote areas, maternity care provid- nosis. The differential of right upper quadrant pain
ers need to balance the risk of transfer versus the includes cholecystitis, hepatitis, acute fatty liver of
benefits of tertiary maternal and neonatal care. pregnancy, gastroesophageal reflux, gastroenteritis
When the patient is adequately treated with mag- and pancreatitis. Urinalysis or kidney function
nesium sulfate and stabilized, a successful transfer abnormalities may suggest pyelonephritis, hemo-
can be made. Close coordination with consultants lytic uremic syndrome, or ureteral calculi. Other
at the receiving institution is mandatory. causes of thrombocytopenia in pregnancy include:
gestational thrombocytopenia, pseudothrombocy-
HELLP Syndrome topenia, HIV, immune thrombocytopenic purpura,
The acronym HELLP describes a variant of severe systemic lupus erythematosus, antiphospholipid
preeclampsia with severe features characterized by syndrome, hypersplenism, DIC, thrombotic throm-
Hemolysis, Elevated Liver enzymes, and Low Plate- bocytopenic purpura, hemolytic uremic syndrome,
lets.83 HELLP syndrome poses significant chal- congenital thrombocytopenias and medications.86
lenges to maternity care providers: first, to maintain A high index of suspicion is the key to diagnosing
a high index of suspicion for the diagnosis, par- HELLP syndrome. Any patient with complaints
ticularly in pregnant patients who are remote from of right upper quadrant or epigastric pain, nausea,
term and may not be hypertensive; and second, to vomiting, or any signs of preeclampsia should be
manage the life-threatening, multi-organ system evaluated with a complete blood count, platelet
complications. Research has yet to elucidate why count, and liver enzyme determinations.87
a small subset of women with preeclampsia with
severe features develop the HELLP syndrome. Laboratory Diagnosis and Classification of
HELLP Syndrome
Risk Factors and Clinical Presentation of Laboratory tests are used both for diagnosis and
HELLP Syndrome as an indicator of severity in HELLP syndrome.
HELLP syndrome occurs in less than one percent A falling platelet count and rising serum LDH
of pregnancies, but up to 20 percent of pregnan- (indicative of both hemolysis and liver dysfunc-
cies are complicated by preeclampsia with severe tion) reflect the severity of the disease. Thrombo-
features.84 The clinical presentation of HELLP cytopenia also forms the basis of a commonly used
syndrome is quite variable. classification system.45 Table 6 lists some com-
At the time of diagnosis, 30 percent of women monly used laboratory criteria for the diagnosis of
were postpartum, 18 percent were term, 42 HELLP syndrome.85
percent are preterm (27 to 37 weeks gestation) In addition, when the platelet count is less than
and 11 percent are extremely preterm (less than 50,000 per mm,61 or concerns develop regarding
27 weeks).84 The most common presenting active bleeding due to a coagulopathy, then fibrin-
complaints are right upper quadrant or epigastric ogen, fibrin degradation products or d-dimer, pro-
pain, nausea, and vomiting. Many patients will thrombin and partial thromboplastin times should
give a history of malaise or non-specific symptoms be assessed to rule out superimposed DIC.
suggesting an acute viral syndrome.85 A subset
presents with headache and visual disturbances Management of HELLP Syndrome
consistent with preeclampsia with severe features. Management of HELLP follows the general guide-
Advanced coagulopathy may cause hematuria or lines for preeclampsia with severe features. All

— Chapter B  11
Chapter B

Regional anesthesia is safe with platelet counts


Table 6. Criteria for Laboratory above 100,000/mcL and should be avoided if
Diagnosis of HELLP Syndrome2,45,87 platelet counts are less than 50,000/mcL. Between
50,000/mcL and 100,000/mcL, regional anesthe-
Hemolysis sia may be safe, but “its use in such patients will
Abnormal peripheral blood smear (evidence of
damaged erythrocytes – schistocytes, burr cells,
require a consensus among the obstetrician, anes-
helmet cells) thesiologist, and patient.”86 Platelet transfusions
Serum bilirubin greater than, or equal to, 1.2 mg/dL usually increase platelet counts by 10,000/mcL per
LDH greater than 600 IU/L unit and are given 6 to 10 units at a time.86
Elevated Liver Enzymes
Transaminases (AST and/or ALT) > twice upper limit 3. Spontaneous rupture of a subcapsular liver
of normal hematoma: This is a life threatening complication
Low Platelet Count that must be suspected in any patient with HELLP
Less than 100,000/mcL who develops shock and massive ascites. Emer-
gent laparotomy may be life saving. A subcapsular
hematoma may be suggested by right upper quad-
women with HELLP should receive magnesium rant, epigastric or shoulder pain. The diagnosis is
sulfate from the time of hospital admission until confirmed by CT or US. If unruptured, the hema-
24 to 48 hours postpartum.85 toma may be monitored with serial US or CT
Management issues specific to HELLP syn- scans in a facility with a readily available vascular
drome include the following: or general surgeon and a blood bank aware of the
potential need for massive transfusions.87
1. Corticosteroids: Although a few small random-
ized controlled trials have demonstrated improve- Delivery and Postpartum Management
ment in laboratory measurements, particularly The decision regarding timing of delivery is
platelet counts, with the use of high dose steroids,85 weighted toward earlier delivery for women
a Cochrane analysis did not demonstrate improved with HELLP than for women with preeclampsia
maternal or fetal outcomes beyond the known with severe features without HELLP. Specifi-
benefits of corticosteroids for fetuses less than 34 cally, infants greater than 28 weeks gestation are
weeks.88 The only randomized, double-blind, pla- routinely delivered 24 to 48 hours after the first
cebo controlled clinical trial failed to demonstrate maternal dose of dexamethasone or betamethasone
any improved maternal outcomes with antepar- is administered.87 Conservative management of
tum or postpartum use of dexamethasone except HELLP remains experimental and in most women
for a shorter time to platelet count recovery in the clinical course is too rapid to wait for the com-
women with platelet counts below 50,000/mcL.89 plete steroid course before initiating delivery.2,85
Increased platelet counts may permit the use of The choice between vaginal and cesarean
regional anesthesia.90 High dose corticosteroids delivery should be based on obstetric factors (e.g.
are not recommended for routine use in women parity and cervical ripeness), fetal maturity, and
beyond 34 weeks gestational age or postpartum. the severity of medical complications.85,87 Cesar-
ean delivery carries special risks, such as bleeding
2. Blood products: Fresh frozen plasma, plate- due to thrombocytopenia and difficulty control-
lets, and packed red blood cells may be needed to ling blood pressure due to depleted intravascular
correct coagulation defects or acute hemorrhage. volume. The surgeon may elect to place a sub-
Women with platelets greater than 50,000/mcL fascial drain or perform secondary skin closure
are unlikely to bleed, but intrapartum plate- due to expected continued oozing. After delivery,
let transfusions are indicated if the count dips some women with HELLP syndrome experience
below 20,000/mcL prior to anticipated vaginal a period of clinical and laboratory deterioration
delivery or in the presence of significant bleeding before recovery. Magnesium sulfate infusion is
(e.g. ecchymosis, bleeding from puncture sites, continued for at least 24 hours. The platelet count
bleeding gums). Platelets may be considered prior typically reaches its nadir and the LDH its peak
to a cesarean delivery if platelets re < 50,000/mcL. 24 to 48 hours after delivery.91 Unfortunately,

12  Chapter B —
Medical Complications of Pregnancy

postpartum deterioration sometimes progresses Differential Diagnosis


to include hepatic rupture, renal failure, pulmo- Most women with AFLP are misdiagnosed on ini-
nary edema, ascites, pleural effusion, postpartum tial hospital admission: preeclampsia and hepatitis
hemorrhage, acute respiratory distress syndrome are the most common initial diagnoses.92
(ARDS) or DIC. These patients may require Many clinical features of AFLP overlap those
prolonged intensive care with continuous cardiac of preeclampsia and HELLP syndrome, and
monitoring, central lines, respirator care, dialysis patients may have both diseases. Approximately
and other major interventions. There is a one per- half of patients with AFLP will have hypertension,
cent risk of maternal mortality in high-resources proteinuria or edema. Acute hepatitis and liver
settings.8 Clinical signs of recovery include a damage secondary to drugs or toxins should also
decreasing blood pressure, mobilization of fluid be considered in the differential diagnosis.
from peripheral edema, ascites, or pleural effu- The diagnosis of AFLP is heavily dependent on
sions, and subsequent diuresis. laboratory findings. Early in the disease course,
bilirubin is elevated and may be detected in the
Acute Fatty Liver of Pregnancy urine, the international normalized ratio (INR)
Acute fatty liver of pregnancy (AFLP) is a rare and activated partial thromboplastin time (aPTT)
condition that occurs in the third trimester and are prolonged, while the platelet count is only
may be initially diagnosed as HELLP syndrome mildly decreased (100,000 to 150,000/mcL). This
due to similarities in clinical and laboratory find- contrasts with HELLP, where significant throm-
ings. The incidence of AFLP is approximately bocytopenia is an early finding and bilirubin is
one in 7,000 to 16,000 pregnancies.92,93 In the usually normal.45,95 In AFLP, the AST and ALT
past and today in low-resource settings, fetal are usually elevated, but not to the extent that
and maternal mortality were each as high as 85 would be expected with acute infectious hepatitis.
percent, but with earlier recognition and prompt Appropriate serologic tests for acute infectious
delivery mortality is now less than 15 percent.92,93 hepatitis can further clarify the diagnosis. In one
The pathophysiology of AFLP involves abnor- case series, all women with AFLP had laboratory
mal hepatic mitochondrial function that leads evidence of DIC, including markedly decreased
to accumulation of fat droplets in hepatocytes, antithrombin III levels.92 Although hypoglyce-
and culminates in fulminant hepatic failure if mia was found in all patients in one study, it was
left untreated. The etiology is unknown. Women only present in 50 percent of patients in another
carrying infants with a mutation affecting fatty study and its absence does not exclude AFLP.
acid oxidation, Long-Chain 3 Hydroxyacyl CoA A case series of 51 cases at Parkland Hospital
Dehydrogenase deficiency (LCHAD), have an demonstrated kidney injury in almost all cases,
increased incidence of AFLP. Infants of moth- with 76 percent having a creatinine over 1.5 mg/
ers with AFLP should be tested for LCHAD, dl.96 Radiologic tests are of limited usefulness in
as 19 percent of AFLP cases are associated with diagnosing AFLP, as ultrasound studies, computed
this mutation. Affected infants have a 75 to 90 tomography (CT) scans and magnetic resonance
percent mortality rate, which can be decreased imaging (MRI) of the liver all have high false
dramatically through dietary treatment.94 negative rates.92 Liver biopsy can confirm the
AFLP presents in the third trimester with vom- diagnosis of AFLP but is invasive and not usually
iting (71 to 75 percent of cases), upper abdominal necessary in order to proceed with treatment.92,95
pain (43 to 50 percent), malaise (31 percent) and
jaundice (28 to 37 percent).92,93 Physical exami- Treatment
nation findings are non-specific, and the liver The most important treatment for AFLP is
size is normal or small. With disease progression, delivery, since the disease never remits and severe
liver failure develops with signs of coagulopathy, complications can develop if delivery is delayed.
asterixis, encephalopathy and coma. There may As is the case with preeclampsia and HELLP
be ascites (due to portal hypertension), pancre- syndrome, the choice between vaginal and cesar-
atitis, and gastrointestinal bleeding secondary ean delivery should be based on obstetric fac-
to severe vomiting, esophagitis, and associated tors, fetal maturity and the severity of medical
coagulation disorders. complications.92 Hepatotoxic general anesthet-

— Chapter B  13
Chapter B

ics should be avoided. Coagulopathy should be pulmonary embolism, amniotic fluid embolism or
corrected although infusion of antithrombin has pneumonia as well as iatrogenic fluid overload.
not been shown to improve clinical outcomes.92 The management of PPCM during pregnancy
Hypoglycemia may be corrected with infusions of differs from standard congestive heart failure
10 percent dextrose, supplemented by boluses of treatment, because ACE inhibitors are contrain-
50 percent dextrose.92 If diagnosis and delivery are dicated in pregnancy and care must be taken to
accomplished early, postpartum improvement is avoid excess diuresis with its accompanying risk of
generally rapid. The Parkland study demonstrated uteroplacental insufficiency. Close collaboration
resolution of ongoing hepatic necrosis within a between maternal fetal medicine and cardiology
few days of delivery and clinical improvement specialists is recommended when the diagnosis
common by 3 to 4 days postpartum, however, is made prior to delivery. Severe cases that do
laboratory evidence of AFLP can persist for 7 to not improve with at least two weeks of standard
10 days or more.96 Rarely, liver transplantation has therapy may be treated with immunosuppressive
been required for multisystem failure that does not therapy if an endomyocardial biopsy demonstrates
improve with delivery.97 When patients continue myocarditis.99,102 The prognosis for women with
to worsen after delivery plasmapheresis has been PPCM depends on the degree of myocardial
used with promising results in a case series of 29 dysfunction.101 Future pregnancies are at risk for
women from China, however the rarity of AFLP recurrent, life-threatening PPCM. Women whose
and usual postpartum clinical improvement make cardiac function does not recover fully should be
clinical trials unlikely.98 discouraged from conceiving again.99,101

Peripartum Cardiomyopathy (PPCM) Venous Thromboembolism (VTE) During


The incidence of PPCM is between one in 3,000 Pregnancy
to 4,000 births.99 Its importance lies in its high Definitions
mortality rate, which has been estimated at five to Venous thromboembolism (VTE) refers to both
20 percent.100,101 Cardiomyopathy accounted for deep venous thrombosis (DVT) –a blood clot in
11.8 percent of US pregnancy-related deaths from the venous system of the lower extremities – and
2006 to 2010.81 By definition, PPCM is heart pulmonary embolism (PE) – a clot that breaks
failure developing in the last month of pregnancy loose and lodges in the pulmonary arteries.103
or within five months of delivery in a woman
without another identifiable cause of the heart Incidence and Clinical Significance
failure.81 Left ventricular systolic dysfunction is VTE complicates 0.5 to 2.0 per 1,000 pregnan-
documented with echocardiography.99 cies and is a leading cause of maternal mortality
The etiology of PPCM remains unknown but in developed countries, accounting for 9 percent
evidence points to myocarditis, perhaps due to of US maternal deaths.103 The incidence of VTE
a weakened immune response to viral infection is higher postpartum than antenatal with the peak
of the myocardium.99 Other etiologies that have incidence in the first week after delivery.104 The
been suggested but not proven include malad- importance of timely diagnosis is underscored
aptation to the normal hemodynamic stress of by the fact that up to 25 percent of patients with
pregnancy, stress-activated cytokines and genetic untreated DVT develop PE, and undiagnosed PE
factors. There are cases of familial PPCM99 and an has a mortality rate of 30 percent.105 Morbidity is
increased rate in African Americans.101 also common: following DVT, 29 to 79 percent
Initial diagnosis may be delayed because the signs of women suffer post-thrombotic syndrome, with
and symptoms of systolic dysfunction, including chronic leg pain and swelling, varicose veins, skin
dyspnea, fatigue, tachypnea, and lower extremity discoloration, and ulceration.106
edema, are common in the last month of preg-
nancy and immediate postpartum period. The Pathophysiology and Risk Factors
differential diagnosis includes cardiomyopathy due VTE develops as a result of multiple interacting
to other etiologies such as valvular disease, ischemia risk factors.107 The classic predisposing factors
or myocardial dysfunction secondary to preeclamp- of hypercoagulation, venous stasis, and vascu-
sia, dyspnea from respiratory disease including lar damage are present in every pregnancy and

14  Chapter B —
Medical Complications of Pregnancy

postpartum.108 Hypercoagulability of pregnancy


results from an increased concentration of pro- Table 7. Risk Factors for VTE103,112,113
coagulant factors VII, VIII, X, Von Willebrand
factor, Plasminogen activator inhibitor-1 and Personal or family history of VTE
Plasminogen activator inhibitor-2, combined with Thrombophilic disorders
decreased anticoagulant free Protein S.103 Stasis Multiparity (more than four deliveries)
results from increased venous distension and from Age greater than 35 years
obstruction of the inferior vena cava by the gravid Obesity
uterus. Reduction in venous flow is evident by 13
Severe varicose veins
weeks gestation, reaches a nadir at 36 weeks, and
returns to non-pregnant levels approximately 6 Hyperemesis
weeks postpartum.109 Vascular damage may occur Hypertensive disorders of pregnancy
during vaginal or cesarean delivery. Table 7 lists Prolonged bed rest or immobility during travel
additional risk factors for VTE in pregnancy. The Infection/sepsis
most important risk factor for VTE is a personal Dehydration
history of VTE: 15 to 25 percent of VTEs in
Major medical problems (mechanical heart valve,
pregnancy are recurrent events.103,110 Overall, inflammatory bowel disease, nephrotic syndrome,
the risk of VTE is five to fifty times higher for a sickle cell disease, myeloproliferative disorders)
pregnant woman than a non-pregnant woman of Cesarean delivery, especially if emergent
the same age.111
Postpartum hemorrhage
Thrombophilic Disorders Smoking
Inherited or acquired thrombophilic disorders are Multiple pregnancy
among the important risk factors for VTE, which
may be inherited or acquired. Approximately
50 percent of women with VTE in pregnancy
Table 8. Inherited Thrombophilias108
have a thrombophilic disorder, compared to
only 10 percent of the general population in the
Factor V Leiden mutation
United States.109
The inherited thrombophilias are listed in Prothrombin G20210A mutation
Table 8. Factor V Leiden and prothrombin Methylene tetrahydrofolate reductase (MTHFR)
mutation
G20210A mutations are the most common.108
Women with protein C and protein S deficiencies Antithrombin deficiency
have an eight-fold increased risk of pregnancy- Protein C deficiency
related VTE.114 Protein S deficiency
Universal screening for thrombophilia is not
recommended; however, testing is recommended
for women with a personal history of VTE that for the MTHFR mutation or homocysteine levels
is not attributable to a non-recurrent risk factor because of a lack of evidence that these factors
(such as trauma, surgery, travel or immobilization) affect maternal or fetal outcomes.108
or who have a first degree family member with a Accurate interpretation of screening tests
high-risk thrombophilia (antithrombin deficiency; requires knowledge of the effects of pregnancy and
double heterozygous for prothrombin G20210A other disorders. Normal pregnancy decreases pro-
mutation and factor V Leiden; factor V Leiden tein S levels.116 Antithrombin and Protein C levels
homozygous or prothrombin G20210A mutation remain normal throughout pregnancy, but protein
homozygous) or a venous thrombosis before age C resistance increases during the second and
50 in the absence of other risk factors.108 Throm- third trimesters.116 Massive thrombus decreases
bophilia screeing is no longer recommended for antithrombin levels; nephrotic syndrome and
women with pregnancy complications such as pre-eclampsia are associated with decreased anti-
intrauterine growth restriction (IUGR) and pre- thrombin levels, and liver disease with decreased
eclampsia.115 ACOG recommends not screening protein C and S levels.117

— Chapter B  15
Chapter B

Antiphospholipid antibodies are the most com- presentation in the first trimester was a significant
mon and clinically important acquired thrombo- predictor of DVT.120
philic defects. Antiphospholipid syndrome (APS) Less than 10 percent of women with signs and
in pregnancy is defined by at least one clinical symptoms of DVT have the diagnosis confirmed
and one laboratory criteria.118 Clinical criteria by objective testing. Definitive diagnosis is essen-
include 1) arterial, venous or small vessel throm- tial due to the need for acute treatment, evaluation
bosis of any tissue or organ, 2) unexplained fetal for underlying thrombophilia, and prophylaxis in
death beyond 10 weeks, 3) birth before 34 weeks future pregnancies.
of a normal appearing fetus due to preeclampsia/
eclampsia or placental insufficiency, or 4) three Diagnostic Testing
or more unexplained, consecutive spontaneous When DVT is strongly clinically suspected,
miscarriages before 10 weeks. Laboratory criteria anticoagulation should be given until results
include 1) lupus anticoagulant, 2) anticardiolipin of confirmatory tests are available.103,115,117 The
antibody, or 3) anti-B2-glycoprotein I on at least first-line diagnostic test for DVT is Doppler
two occasions twelve or more weeks apart.118 ultrasound.103,115,117 A Doppler study indicating
Lupus anticoagulant is more specific but less sensi- deep vein thrombosis in the affected leg is suf-
tive than the other two laboratory criteria.118 ficient to recommend a full course of therapeutic
anticoagulation.103,115,117 Negative Doppler results
Deep Venous Thrombosis with low clinical suspicion do not require anti-
Clinical Signs and Symptoms coagulation. If iliac vein thrombosis is suspected
Unlike DVTs in non-pregnant patients, most and the Doppler is negative, magnetic resonance
DVTs in pregnant women occur in the left leg, imaging (MRI) or empiric anticoagulation is indi-
perhaps because of the gravid uterus compress- cated.103,122 If empiric anticoagulation is chosen,
ing the left iliac vein. A 2010 systematic review venous Doppler should be repeated in a week.117
of 6 studies (not all RCTs) involving 124 women Because of its high false positive rate in preg-
found that 88 percent of DVTs in pregnancy nancy, D-dimer is not recommended in the evalu-
occurred on the left and 71 percent were restricted ation for acute VTE in pregnancy.103,117 A low level
to proximal veins without involving calf veins.119 D-dimer, however, does make VTE unlikely.117,123
Deep venous thrombosis may have a subtle
clinical presentation and may be difficult to dis- Treatment
tinguish from gestational edema. Typical symp- Low-molecular weight heparins (LMWHs) are the
toms are unilateral leg pain and swelling. Two or treatment of choice for VTE in pregnancy.103,115
more centimeters difference in lower leg circum- LMWH is discussed in more detail in the section
ference is associated with a higher risk of DVT Anticoagulation in Pregnancy.
[adjusted odds ratio (OR) 13.62; 95 percent CI
4.56 to 40.67].120 Pulmonary Embolism
The mnemonic “LEFt” can help identify Clinical Signs and Symptoms
pregnant women at higher risk of having a DVT. In contrast to DVT, which is as common during
“L” stands for Left leg symptoms; “E” stands for pregnancy as postpartum, at least two thirds of
Edema (2 or more centimeter leg circumference PEs occur postpartum.104 Dyspnea and tachypnea
discrepancy) and “Ft” stands for First trimester are the most common presenting symptoms of PE.
symptoms. The “LEFt” mnemonic has a better The clinical picture varies from mild dyspnea and
negative than positive predictive value. A study tachypnea accompanied by chest pain to dramatic
of the “LEFt” mnemonic effectiveness found that cardiopulmonary collapse.
DVT in pregnancy was diagnosed in 13 of 111 Clinical pre-test probability assessments, such
(11.7 percent; 95 percent CI 8.3 to 20.9) with at as the Wells score, have not been validated in
least one of the LEFt criteria versus none of 46 (0 pregnancy.
percent; 95 percent CI 0.0 to 7.9 percent) with
none of the LEFt criteria.121 Although only 29.4 Diagnostic Testing
percent of DVTs in the LEFt study were in the An approach to the diagnosis of suspected PE
first trimester, a multivariate analysis found that using non-invasive testing is outlined in Figure 3.

16  Chapter B —
Medical Complications of Pregnancy

Figure 3. Algorithm for Diagnosis of Pulmonary Embolism103,117

Clinical suspicion of PE

Stablilization and consideration of anticoagulation

Venous Doppler lower extremities (optional)

Negative and low suspicion Negative and ongoing Thrombosis


suspicion of PE

Clinical surveillance Therapeutic


Chest X-ray anticoagulation

Normal and ongoing Abnormal


suspicion of PE

CTPA or V/Q scan* CTPA

Abnormal and minimal Normal and ongoing Abnormal


suspicion of PE suspicion of PE

Anticoagulation Consider anticoagulation Therapeutic


not indicated and repeat imaging anticoagulation

*Consideration of factors including: availablity of CT vs. V/Q scan; higher radiation to breast tissue with spiral CT; higher
sensitivity and specificity with spiral CT; low but higher fetal radiation with V/Q scan
CTPA = Computed tomography pulmonary angiogram; PE = pulmonary embolism; V/Q scan = Ventillation-perfusion scan.

When a patient presents with possible PE, puted tomography pulmonary angiogram (CTPA)
stabilization should be the first priority. Please see in order to avoid the radiation of these tests.117 If
Chapter K: Maternal Resuscitation and Trauma a DVT is diagnosed, anticoagulation is recom-
for more details on stabilization. Consideration mended whether or not there is a PE.117
should be given to anticoagulation until a more A chest x-ray may help in deciding whether to
definitive diagnosis is made.117 order a V/Q scan or CTPA. A cohort study found
Some experts recommend ordering a venous a V/Q scan to be preferable to CTPA for diagnosis
Doppler prior to considering a V/Q scan or com- of PE in women who have a negative chest x-ray;

— Chapter B  17
Chapter B

CTPA was more likely to be diagnostic for women Arterial blood gas determination and electrocar-
with an abnormal chest x-ray.124 V/Q scans are less diogram may help determine the clinical likeli-
likely to be non-diagnostic in pregnant women hood of PE or may suggest other conditions.
as they are young with fewer comorbidities.125 When there is high suspicion of PE and V/Q
The choice between V/Q scan and CTPA may scan or CTPA is negative or low-probability/
be affected by availability of each in a particular equivocal, consideration should be given to con-
medical center. tinuing anticoagulation and repeating imaging.117
Fetal radiation exposure with CTPA is less than
10 percent of that with a V/Q scan but the abso- Treatment
lute fetal risk for both is low. The fetal radiation As discussed above, treatment should begin with
dose for CTPA is equivalent to a < 1 in 1,000,000 stabilization, which is covered in detail in Chapter
risk of cancer at age 15 compared with a 1 in K: Maternal Resuscitation and Trauma. Antico-
280,000 risk for a V/Q scan.126 agulation may be started empirically while await-
Maternal breast radiation exposure is a concern ing diagnostic tests. Most women who die from
with CTPA.103 CTPA involves 2.5 to 3.0 rads PE die in the first 30 minutes, so prompt action is
of radiation; 1 rad is estimated to increase the essential.136
lifetime risk of breast cancer by 13.6 percent for LMWH is the treatment of choice for PE as
a woman younger than 35.127 Some feel that this well as DVT.103,115 For more details, please see the
is an overestimation and 1 rad would increase a Anticoagulation in Pregnancy section. If antico-
30-year-old woman’s lifetime risk of breast cancer agulation is contraindicated or repeat PE occurs
by 0.2 percent.128 The patient should be involved despite adequate anticoagulation, it may be neces-
in choosing between a CTPA and V/Q scan sary to insert a filter in the inferior vena cava.117
when possible.129 Anticoagulation is continued after the filter is
The sensitivity of CTPA has increased with placed unless contraindicated.
technological advances. First-generation single- In the case of life-threatening massive PE,
detector-row CT scanners have had a positive thrombolytic therapy, percutaneous catheter
predictive value of only 85 percent130 and are only thrombus fragmentation or surgical embolectomy
30 percent sensitive for subsegmental defects, may be utilized, depending on local expertise.117,137
which account for 20 percent of symptomatic A 2010 Cochrane Review of eight trials involving
PE.130 Newer multidetector-row CT scanners 679 non-pregnant women did not find thrombo-
allow improved visualization of the segmental and lytic therapy better than heparin for PE.138
subsegmental pulmonary arteries131 and have posi-
tive and negative predictive values of 99 percent, Anticoagulation in Pregnancy
comparable to pulmonary angiography which is When clinical findings and the results of diagnos-
now rarely used.132 Multidetector-row CT scan- tic testing show DVT or PE, therapeutic antico-
ners allow quicker scanning of the lung, avoiding agulation is indicated. Anticoagulation options
respiratory movement and artifact: the 16-slice include low molecular weight heparins (LMWHs)
CT can image the entire chest with submillime- such as dalteparin, enoxaparin and tinzaparin;
ter resolution in less than 10 seconds.133 CTPA unfractionated heparin (UFH); and, in the post-
can identify an alternative diagnosis in about two partum period, warfarin (Coumadin®).
thirds of cases in which PE is not present; how- The 2012 American College of Chest Physician
ever, it may detect suspicious-appearing abnormal- (ACCP) Guidelines state, “For pregnant patients,
ities that require further evaluation or even biopsy we recommend LMWH for the prevention and
but actually are benign.134 treatment of VTE, instead of UFH.”115 This is
MRI for diagnosis of pulmonary embolism is supported by a 2010 Cochrane Review and a
an attractive option because it does not expose 2005 systemic review of 64 studies (not all RCTs).
the fetus to ionizing radiation, and it is as sensi- Two other 2010 Cochrane Reviews did not find
tive and specific as CTPA in diagnosing PE.105,135 enough evidence to recommend one form of
Disadvantages of MRI include expense, questions thromboprophylaxis over another.139,140
about accessibility, and the fact that it is relatively Heparin is considered safe for use during preg-
unstudied in pregnancy.105,134 nancy because it does not cross the placenta and is

18  Chapter B —
Medical Complications of Pregnancy

ommend against using direct thrombin and factor


Table 9. Baseline Laboratory Tests for Xa inhibitors in pregnancy.115 Society of Obstetri-
Initiating Anticoagulation129 cians and Gynaecologists (SOGC) guidelines also
recommend against direct thrombin and factor
Thrombophilia profile (See Table 8) — controversial; Xa inhibitors in pregnancy.112 Exceptions include
best when not pregnant
women with a severe heparin allergy or heparin-
Creatinine (LMWHs require dose adjustment with
abnormal renal function)
induced thrombocytopenia.103,115 A prospective
cohort study of 12 pregnancies in 10 women
Liver function tests (warfarin is contraindicated with
significantly abnormal liver function) involving fondaparinux thrombophrophylaxis
because of allergy to LMWH, found no maternal
Complete blood count with platelet count
or fetal complications.
PT/INR Table 9 lists the baseline laboratory evalua-
Activated partial thromboplastin time (aPTT) tion that should be considered prior to initiating
Anti-Xa level (not recommended routinely; indicated anticoagulation.
with women < 50 kg or > 90 kg, recurrent VTE, Therapeutic anticoagulation is recommended
recurrent VTE or renal insufficiency)
for VTE in pregnancy; it should continue for at
least three months from diagnosis.112,115,117 After
three months of therapeutic dosing, anticoagula-
not secreted in breast milk.141 Neither LMWH nor tion can be reduced to intermediate or prophylac-
UFH cause significant bone loss in pregnancy.142,143 tic dosing through at least six weeks postpartum.
LMWHs are at least as effective as UFH and are Acceptable therapeutic doses for LMWH are
less likely to cause allergy or result in infection listed in Table 10. A 2013 Cochrane Review of
from contaminated multidose vials.103 There is no 5 trials involving 1,508 participants found that,
evidence favoring one LMWH over another.144,145 in non-pregnant patients, once per day LMWH
LMWH is compatible with breastfeeding.146-148 is as effective as twice-daily LMWH.112,151 No
Warfarin should be avoided during pregnancy. RCTs have looked at once vs twice daily dosing
It crosses the placenta and increases the risk of in pregnancy. Some lower quality studies sup-
miscarriage and stillbirth, embryopathy — nasal port once-daily dosing in pregnancy, while others
hypoplasia and/or stippled epiphyses — when do not.103 A retrospective study of 126 pregnant
used in the first trimester, CNS abnormalities women with a history of prior VTE, 66 percent
when used in any trimester, and maternal and fetal of whom received once-daily LMWH found no
hemorrhage when used near time of delivery.115 recurrent VTE in either group.152 A retrospec-
However, warfarin is safe for breastfeeding.117,149,150 tive study of once-daily tinzaparin in 37 pregnant
There is limited evidence on direct thrombin women found two thrombotic events. Dosages of
inhibitors [e.g., Argatroban, bivalirudin, dabi- LMWH should be adjusted in the setting of renal
gatran and lepirudin] and factor Xa inhibitors insufficiency, notably with preeclampsia with
[e.g., apixaban, rivaroxaban and fondaparinux] severe features.145
during pregnancy and breastfeeding. Direct Hospitalization may be indicated for initial
thrombin inhibitors and factor Xa inhibitors have anticoagulation for VTE in pregnancy, especially
been assigned to FDA pregnancy categories B or if a woman is unstable, has a large thrombus or
C, but both classes of anticoagulants should be has co-morbidities.103 Initial IV UFH may be
avoided in pregnancy until more evidence on their preferable if a woman may need delivery, surgery
safety is available. The 2012 ACCP guidelines rec- or thrombolysis.103

Table 10. Therapeutic Dosing of Low Molecular Weight Heparin108

Enoxaparin (100 units/mg) Dalteparin Tinzaparin

Therapeutic dose 1 mg/kg subcutaneously 100 units/kg SQ every 12 hrs or 175 units/kg SQ
(SQ) every 12 hrs 200 units/kg SQ every 24 hours every 24 hrs

— Chapter B  19
Chapter B

The optimal protocol for monitoring treat- the initial treatment of DVT or PE in pregnancy.
ment with LMWHs has not been established. It UFH may be chosen over LMWH in some set-
is not necessary to follow the aPTT as with UFH. tings for reasons of cost or availability. Recom-
Whether to follow Anti-Xa levels is controversial, mended dosages and monitoring are described in
and the target range is not well established.153 The Table 11.
use of Anti-Xa levels are generally not followed For postpartum DVT or PE, warfarin may be
unless a woman is < 50 kg or more than 90 kg, started concomitantly with heparin.117 Because
has renal insufficiency or high-risk factors such of an initial inhibition of Protein C, warfarin can
as recurrent VTE.117 With twice daily therapeu- cause a hypercoagulable state for the first 3 to 5
tic LMWH, the target anti-Xa level is 0.6 to 1.0 days of therapy.155 The LMWH or UFH should
units/ml; with once daily dosing the target should be continued until the target INR of 2.0-3.0 is
be slightly higher.108 Platelet counts are monitored achieved for two consecutive days.117 Typically,
initially after injection for women taking UFH; this level of anticoagulation is obtained within
if thrombocytopenia occurs, it is usually between five days.156 LMWH and warfarin therapy can
7 to 14 days from therapy initiation.154 Recom- be started concomitantly in an outpatient setting
mendations vary regarding the need to recheck for selected postpartum patients who are medi-
platelets counts after the initial baseline, with cally stable, with a supportive home environment
SOGC recommending a platelet count one week and access to daily monitoring until the INR is
after initiating LMWH while the Royal College of therapeutic.157 Patients only requiring six weeks of
Obstetricians and Gynecologists (RCOG) states anticoagulation may opt to continue on LMWH
it is not necessary to recheck platelet counts after rather than transitioning to warfarin.103
LMWH initiation.112,117
Intravenous (IV) and/or subcutaneous (SQ) Delivery Management of the
forms of UFH may be used instead of LMWH for Anticoagulated Patient
Delivery issues include how to alter heparin dos-
ing during labor and under what conditions to
Table 11. Therapeutic Dosages and Monitoring of use epidural analgesia or spinal anesthesia. The
Intravenous (IV) and Subcutaneous (SQ) UFH American Society of Regional Anesthesia (ASRA)
recommends delaying regional anesthesia until
IV regimen117 10 to 12 hours after last prophylactic dose of
IV bolus of 80 international units (IU)/kilogram (kg) LMWH or 24 hours after last therapeutic dose of
Followed by a continuous infusion of 18 IU/kg/hour LMWH.158 The 2012 ACCP guidelines suggest
aPTT 4 to 6 hours after the loading dose and 6 hours after any dose discontinuing LMWH 24 hours before an induc-
change
tion or scheduled cesarean.115
Thereafter, check the aPTT at least daily and adjust dosage to achieve
aPTT in the therapeutic range of 1.5 to 2.5 times the mean laboratory Women who go into spontaneous labor may be
control value instructed to discontinue heparin at the onset of
SQ regimen108 regular uterine contractions. An epidural or spinal
10,000 or more IU SQ every 12 hours should not be placed for 24 hours after the last
Monitor aPTT and adjust SQ dose to achieve aPTT of 1.5 to 2.5 at six dose of therapeutic LMWH, 12 hours after SQ
hours after injection UFH and six hours after IV UFH.117

Prophylaxis
Table 12. Prophylactic Dosage for LMWH 108,112 Prophylaxis against VTE in pregnancy may be
required antenatally for women with a history of
Enoxaparin DVT or PE and for those with a known his-
(100 units/mg) Dalteparin Tinzaparin tory of thrombophilia. While better studies are
needed, LMWH appear to be the safest and most
Prophylactic 40 mg SQ daily 5,000 units 4,500 units SQ
dose SQ daily daily effective form of thromboprophylaxis in preg-
Obesity 60 mg SQ daily 7,500 units 75 U/kg SQ daily
nancy.141,144,159 Prophylactic doses of LMWH are
SQ daily listed in Table 12. Subcutaneous UFH may be
used as a lower cost alternative to LMWH; doses

20  Chapter B —
Medical Complications of Pregnancy

the prothrombin 20210A mutation or who have


Table 13. Prophylactic Dosage for UFH108 other risk factors for VTE.162 The authors of the
ACCP guidelines defend their recommendations
First trimester 5,000–7,500 IU SQ daily but acknowledge that evidence is lacking in many
Second trimester 7,500–10,000 IU SQ daily areas leaving room for variations in recommenda-
Third trimester 10,000 IU SQ daily, unless tions and clinical judgment.163
aPTT is elevated Women with mechanical heart valves should
be transferred to a high-risk specialist or man-
aged with close consultation. The manufacturer
are listed in Table 13. Some experts recommend of enoxaparin issued a warning against its use for
adjusting prophylactic LMWH doses for obesity,
but there are no evidence-based guidelines for this
practice.108 SOGC recommndations for dosing Table 14. Clinical Indications for Anticoagulant
with obesity are included in Table 12.112 Prophylaxis per 2012 ACCP Guidelines115
Antenatal low dose aspirin (75 to 100 mg) is
Indication 1: Personal history of DVT or PE, no known thrombophilia
recommended in combination with LMWH or
UFH for women with antiphospholipid anti- 0.A. DVT or PE with thrombogenic event (such as a hip fracture or a
prolonged surgery)
body syndrome and a history of three or more
Antenatal: no prophylaxis
pregnancy losses.115 Adding aspirin is also recom- Postpartum: six weeks LMWH or warfarin
mended for women with prosthetic heart valves at
high risk for thromboembolism.115 0.B. DVT or PE with no thrombogenic event, pregnancy-related or
estrogen-related VTE, history of multiple VTE but not on chronic
Aspirin appears to be relatively safe in preg- anticoagulation
nancy, although it has been associated with an Antenatal: anticoagulation with prophylactic or intermediate dose
increased risk of gastroschisis when taken in the of LMWH
first trimester. A meta-analysis of eight stud- Postpartum: six weeks LMWH or warfarin
ies – only one of which was an RCT -found no Indication 2: Women on chronic anticoagulation prior to pregnancy
overall risk of congenital anomalies for women Antenatal: anticoagulation with adjusted dose LMWH or 75 percent
taking aspirin in pregnancy (OR 1.33; 95 percent therapeutic dose of LMWH
CI 0.94 to 1.89), but did find an increased risk Postpartum: resumption of chornic anticoagulation
of gastroschisis after first trimester exposure (OR Indication 3: Women with no history of VTE but known thrombophilia
2.37; 95 percent CI 1.44 to 3.88).160 A meta- 2.A. H
 omozygous for factor V Leiden or the prothrombin 20210A
analysis of thirty-eight studies of variable quality mutation and positive family history for VTE
found that aspirin in pregnancy compared with Antenatal: prophylactic- or intermediate-dose of LMWH
placebo was not associated with an increased risk Postpartum: six weeks anticoagulation wiht prophylactic- or
of miscarriage (7 studies; RR 0.92; 95 percent CI intermediate-dose LMWH or warfarin
0.71 to 119), perinatal mortality (20 studies; RR 2.B. H
 omozygous for factor V Leiden or the prothrombin 20210A
0.92; 95 percent CI 0.81 to 1.05), or small for mutation and no family history for VTE
gestational age infants (12 studies; RR 0.96; 95 Antenatal: no prophylaxis
Postpartum: six weeks LMWH or warfarin
percent CI 0.87 to 1.07).161
Clinical indications for anticoagulant prophy- 2.C. Thrombophilia other than homozygous for factor V Leiden or the
prothrombin 20210A mutation and positive family history for VTE
laxis and recommendations for when to initi-
Antenatal: no prophylaxis
ate and discontinue therapy are summarized in
Postpartum: six weeks LMWH or warfarin
Table 14 (ACCP) and Table 15 (ACOG). ACCP
recommendations have been criticized for only 2.D. Thrombophilia other than homozygous for factor V Leiden or the
prothrombin 20210A mutation and no family history for VTE
recommending antenatal prophylaxis for women Antenatal: no prophylaxis
with no history of VTE who are homozygous Postpartum: no prophylaxis
for factor V Leiden or the prothrombin 20210A
2.E. A
 ntiphospholipid antibody syndrome by laboratory and clinical criteria
mutation and for not recommending postpar-
Antenatal: prophylactic LWMH and low-dose aspirin (75 to 100 mg
tum prophylaxis in women with thrombophilia per day)
who have a family history of thrombophilia Postpartum: six weeks LMWH or warfarin
other than homozygous for factor V Leiden or

— Chapter B  21
Chapter B

the treatment of pregnant patients with mechani- suggest that VTE prophylaxis after cesarean should
cal heart valves because of an undisclosed number be based on risk factors and states women with-
of post-marketing reports of thrombosed valves in out other risk factors for VTE prophylaxis other
patients receiving enoxaparin.164 than cesarean delivery only need early ambulation
Recommendations regarding post-cesarean however ACOG currently recommends pneu-
thromboprophylaxis vary. 2012 ACCP guidelines matic compression devices during cesarean for

Table 15. Clinical Indications for Anticoagulant Prophylaxis per 2013 ACOG Practice
Bulletin108

Indication 1: Personal history of single DVT or PE, no known thrombophilia


0.A. Single DVT or PE with thrombogenic event (such as a hip fracture or a prolonged surgery) not pregnancy- or
estrogen-related
Antenatal: no prophylaxis
Postpartum: 4-6 weeks LMWH or warfarin (no prophylaxis per some experts)
0.B. S
 ingle DVT or PE with no thrombogenic event (idiopathic), pregnancy-related or estrogen-related
Antenatal: prophylactic dose of LMWH or UFH (no prophylaxis per some experts)
Postpartum: 4-6 weeks LMWH or warfarin
Indication 2: Personal history of multiple DVT or PE, thrombophilia or no thrombophilia
1.A. Not on chronic anti-coagulation
Antenatal: prophylactic or therapeutic dose LMWH or UFH
Postpartum: 4-6 weeks LMWH or warfarin OR prophylactic or therapeutic dose LMWH or UFH for 6 weeks
1.B. On chronic anti-coagulation
Antenatal: therapeutic dose LMWH or UFH
Postpartum: resume chronic anticoagulation
Indication 3: Women with no history of VTE and known thrombophilia
2.A. F
 actor V Leiden heterozygous; prothrombin G20210A heterozygous; protein C or protein S deficiency (low-
risk thrombophilias)
Antenatal: no prophylaxis
Postpartum: no prophylaxis or 4-6 weeks LMWH or warfarin if additional risk factors
2.B. A
 ntithrombin deficiency; double heterozygous for prothrombin G20210A mutation and factor V Leiden;
factor V Leiden homozygous or prothrombin G20210A mutation homozygous (high-risk thrombophilias) and
no family history (first-degree relative) of VTE
Antenatal: no prophylaxis or prophylaxis with LWMH or UFH
Postpartum: 4-6 weeks LMWH or warfarin
2.C. A ntithrombin deficiency; double heterozygous for prothrombin G20210A mutation and factor V Leiden;
factor V Leiden homozygous or prothrombin G20210A mutation homozygous (high-risk thrombophilias) AND
family history (first-degree relative) of VTE – not on chronic anticoagulation
Antenatal: prophylactic, intermediate-dose or adjusted-dose LMWH or UFH
Postpartum: 4-6 weeks LMWH or warfarin OR intermediate or adjusted-dose LMWH or UFH for 6 weeks
(therapy level at least as high as antepartum treatment)
Indication 4: Women with history of single VTE and known thrombophilia — not on chronic anticoagulation
3.A. F
 actor V Leiden heterozygous; prothrombin G20210A heterozygous; protein C or protein S deficiency
(low-risk thrombophilias)
Antenatal: no prophylaxis or prophylactic or intermediate-dose LWMH or UFH
Postpartum: 4-6 weeks LMWH or warfarin OR intermediate-dose LMWH or UFH
3.B. A
 ntithrombin deficiency; double heterozygous for prothrombin G20210A mutation and factor V Leiden;
factor V Leiden homozygous or prothrombin G20210A mutation homozygous (high-risk thrombophilias)
Antenatal: prophylactic, intermediate-dose or adjusted-dose LMWH or UFH
Postpartum: 4-6 weeks LMWH or warfarin OR intermediate or adjusted-dose LMWH or UFH for 6 weeks
(therapy level at least as high as antepartum treatment)

22  Chapter B —
Medical Complications of Pregnancy

all women not already receiving pharmacological cal challenges can evolve during pregnancy. This
thromboprophylaxis.103,115 chapter attempts to better participants’ under-
Due to concern for increasing maternal mortality standing of the risk factors, diagnosis and man-
in the United States the Joint Commission in 2010 agement of hypertensive disorders of pregnancy,
recommended that pregnant women who are at high AFLP, peripartum cardiomyopathy and VTE.
risk for VTE should be given postpartum LMWH. The key to diagnosis of these problems is clinical
Cesarean delivery is a major risk factor for VTE, vigilance coupled with appropriate lab or imaging
however a 2010 Cochrane Review concluded that studies. A common clinical challenge is balancing
there is not enough evidence to recommend for or maternal and fetal well-being in diagnostic and
against the routine use of LMWH prophylaxis after treatment decisions.
cesarean delivery.139 There are many different guide-
lines regarding which post-cesarean patients to start
on heparin; an example is shown in Table 16.115,165 Table 16. Post Cesarean Prophylaxis
There is a paucity of data and a lack of consistent
recommendations regarding which patients should Risk factors
have pharmacologic prophylaxis initiated after Age > 35 yr
a vagina delivery. The decision whether to give Obesity (BMI > 30)
pharmacologic thromboprophylaxis after a vaginal Parity > 3
delivery to women with risk factors (Table 7) may Gross varicose veins
be made on an individual basis.112,113 The RCOG Current infection
recommends heparin prophylaxis after vaginal Preeclampsia
Immobility for >4 days before operation
delivery for women with a BMI > 40.113
Major current illness
Evidence is also lacking regarding the timing for
Emergency cesarean section during labor
initiation of post-delivery pharmacologic throm-
Prophylaxis recommendation:
boprophylaxis. Based on ACOG and RCOG rec-
Women with at least two of these risk factors should receive LMWH OR
ommendations, it is reasonable to start or resume mechanical prophylaxis (elastic compression stockings or intermittent
prophylactic heparin dosing four to six hours after pneumatic compression) until hospital discharge.
a vaginal delivery, 6 to 12 hours after a cesarean Women with three or more risk factors should receive mechanical
and four hours after epidural removal.103,113 There prophylaxis until ambulation and LMWH at least until hospital
discharge. Continuation of LMWH beyond hospital discharge for up to
is less than one percent risk of wound hematoma six weeks may be considered in selected patients
with LMWH prophylaxis.146
Major risk factors
Given the complexity of management decisions Morbid obesity (BMI > 40)
and variety of consensus guideline recommenda- Cesarean hysterectomy
tions regarding postpartum thromboprophy- Previous deep-vein thrombosis or known thrombophilia
laxis, hospitals and systems may want to develop Prophylaxis recommendation:
consistent local guidelines and standards of care to Women with any of these risk factors should receive LMWH and
ensure consistent practice and reduce the morbid- mechanical prophylaxis at least until discharged from hospital and in
ity and mortality from VTE in pregnancy. the case of c-hysterectomy, prior DVT or some thombophilias for six
or more weeks postpartum (see Tables 14 and 15).
Summary
Table adapted from Marik PE, Plante LA. Venous thromboembolic disease and
Pregnancy is a natural process that involves many pregnancy. N Engl J Med. 2008 Nov 6; 359(19): 2025-2033.
complex physiologic changes. Multiple medi-

— Chapter B  23
Chapter B

SORT: Key Recommendations for Practice


Evidence
Clinical Recommendation Rating References

Magnesium sulfate is the treatment of choice for women with preeclampsia A 56


with severe features to prevent eclamptic seizures (NNT=100) and placental
abruption (NNT=100).
Magnesium sulfate is more effective in preventing recurrent eclamptic seizures A 51,52
and decreasing maternal mortality than diazepam or phenytoin.
Low dose aspirin (75 to 81 mg daily) has small to moderate benefits for B 38
prevention of preeclampsia (NNT=72), preterm delivery and fetal death in
women at risk for developing preeclampsia.For women at greater risk of
developing preeclampsia the NNT falls to 19.
Calcium supplementation may decrease the incidence of hypertension, B 36
preeclampsia and mortality among women at high risk of developing
hypertension in pregnancy and women with low calcium intakes, however
women in the United States or other high resource countries are unlikely to
benefit from calcium supplementation.
Women with gestational hypertension or preeclampsia without severe features B 41
should have planned delivery at 37 weeks gestational age.
For managing preeclampsia with severe features between 24 and 34 weeks B 70
gestation, limited data suggests that, compared with an interventionist
approach (induction or cesarean delivery 12 to 24 hours after corticosteroid
administration), expectant management, with close monitoring of the mother
and fetus, reduces neonatal complications and neonatal stay in the newborn
intensive care nursery.
Either intravenous labetalol or hydralazine or oral nifedipine may be used for B 1,2,64
treating severe hypertension in pregnancy.
When possible, a woman should be involved in choosing what study to order B 126-128
when a V/Q scan or CTPA is indicated. Fetal radiation is higher with a V/Q
scan than a CTPA but absolute risk is low. Maternal radiation from CTPA
increases lifetime risk of breast cancer.
LMWHs are the agents of choice for antenatal thromboprophylaxis. Heparin B 146-150
and warfarin are safe with breastfeeding.

Strength of Recommendation Taxonomy (SORT)

Strength of Recommendation Definition


A • Recommendation based on consistent and good-quality patient-oriented evidence.
B • Recommendation based on inconsistent or limited-quality patient-oriented evidence.
C • Recommendation based on consensus, usual practice, opinion, disease-oriented evidence, or case series for studies
of diagnosis, treatment, prevention, or screening.
Patient-oriented evidence measures outcomes that matter to patients: morbidity, mortality, symptom improvement, cost
reduction, and quality of life. Disease-oriented evidence measures intermediate, physiologic, or surrogate end points
that might not reflect improvement in patient outcomes (eg, blood pressure, blood chemistry, physiologic function,
pathologic findings).
From Ebell MH, Siwek J, Weiss BD, et al. Strength of recommendation taxonomy (SORT): a patient-centered approach to
grading evidence in the medical literature. Am Fam Physician. 2004;69(3):548-556.

24  Chapter B —
Medical Complications of Pregnancy

References
1. National High Blood Pressure Education Program Working Group 18. Lin J, August P. Genetic Thrombophilias and Preeclampsia: A
on High Blood Pressure in Pregnancy. Report of the National High meta-analysis. Obstet Gynecol. 2005;105:182-192.
Blood Pressure Education Program Working Group on High Blood 19. Colucci F, Boulenouar S, Kieckbusch J, Moffett A. How does vari-
Pressure in Pregnancy. Am J Obstet Gynecol. 2000;183:S1-S22. ability of immune system genes affect placentation? Placenta. Aug
2. American College of Obstetricians and Gynecologists. Hyperten- 2011;32(8):539-545.
sion in pregnancy. Report of the American College of Obstetricians 20. McMaster MT, Zhou Y, Fisher SJ. Abnormal placentation and the
and Gynecologists’ Task Force on Hypertension in Pregnancy. syndrome of preeclampsia. Semin Nephrol. 2004;24:540-547.
Obstet Gynecol. 2013;122(5):1122-1131.
21. Merviel P, Carbillon L, Challier JC, Rabreau M, Beaufils M, Uzan
3. Ankumah N-A, Cantu J, Jauk V, et al. Risk of Adverse Pregnancy S. Pathophysiology of preeclampsia: links with implantation disor-
Outcomes in Women With Mild Chronic Hypertension Before 20 ders. Eur J Obstet Gynecol Reprod Biol. 2004;115:134-147.
Weeks of Gestation. Obstetrics & Gynecology. 2014.
22. Migini LE, Latthe PM, Villar J, Kilby MD, Carroli G, Khan KS. Map-
4. Magee LA, Duley L. Oral beta blockers for mild to moderate hyper- ping the theories of preeclampsia: the role of homocysteine.
tension during pregnancy. Cochrane Database Syst Rev. 2003;3. Obstet Gynecol. 2005;105:411-425.
5. Abalos E, Duley L, Steyn DW. Antihypertensive drug therapy for 23. Nelissen EC, van Montfoort AP, Dumoulin JC, Evers JL. Epi-
mild to moderate hypertension during pregnancy. Cochrane Data- genetics and the placenta. Human Reproduction Update. May-Jun
base Syst Rev. 2014;2:CD002252. 2011;17(3):397-417.
6. Von Dadelszen P, Ornstein MP, Bull SB, Logan AG, Koren G, 24. George EM, Granger JP. Endothelin: key mediator of hyperten-
Magee LA. Fall in mean arterial pressure and fetal growth sion in preeclampsia. American Journal of Hypertension. Sep
restriction in pregnancy hypertension: a meta-analysis. Lancet. 2011;24(9):964-969.
2000;355:87-92.
25. Levine RJ, Thadhani R, Quian C, et al. Urinary placental growth
7. National Institute for Health and Clinical Excellence: Guidance. factor and the risk of preeclampsia. JAMA. 2005;293:77-85.
Hypertension in Pregnancy: The Management of Hypertensive
Disorders During Pregnancy. National Collaborating Centre for 26. Wang A, Rana S, Karumanchi SA. Preeclampsia: the role of angio-
Women’s and Children’s Health (UK). 2010. genic factors in its pathogenesis. Physiology (Bethesda, Md.). Jun
2009;24:147-158.
8. Churchill D, Beevers GDG, Meher S, Rhodes C. Diuretics for
preventing pre-eclampsia. Cochrane Database of Systematic 27. Milne F, Redman C, Walker J, et al. The pre-eclampsia community
Reviews. 2007(1). guideline (PRECOG): how to screen for and detect onset of pre-
eclampsia in the community. BMJ. 2005;330:576-580.
9. American College of Obstetricians and Gynecologists. Committee
opinion no. 623: emergent therapy for acute- onset, severe hyper- 28. Spinnato JAn, Freire S, Pinto E, et al. Antioxidant therapy to pre-
tension during pregnancy and the post-partum period. Obstet vent preeclampsia: a randomized controlled trial. Obstet Gynecol.
Gynecol. 2015;125:521-525. 2007;110(6):1311-1318.
10. Shekhar S, Sharma C, Thakur S, Verma S. Oral nifedipine or intra- 29. Duckitt K, Harrington D. Risk factors for pre-eclampsia at antena-
venous labetalol for hypertensive emergency in pregnancy: a ran- tal booking: systematic review of controlled studies. BMJ. Mar 12
domized controlled trial. Obstet Gynecol. 2013;112(5):1057-1063. 2005;330(7491):565.
11. Barton JR, O’Brien JM, Bergauer NK, Jacques DL, Sibai BM. Mild 30. Villar J, Purwar M, Merialdi M, et al. World Health Organisation
gestational hypertension remote from term: Progression and out- multicentre randomised trial of supplementation with vitamins C
come. Am J Obstet Gynecol. 2001;184:979-983. and E among pregnant women at high risk for pre-eclampsia in
populations of low nutritional status from developing countries.
12. Gofton EN, Capewell V, Natale R, Gratton RJ. Obstetrical interven- BJOG. 2009;116(6):780-788.
tion rates and maternal and neonatal outcomes of women with
gestational hypertension. Am J Obstet Gynecol. 2001;185:798-803. 31. Levine RJ, Hauth JC, Curet LB, et al. Trial of calcium to prevent
pre-eclampsia. N Engl J Med. 1997;337:69-76.
13. Buchbinder A, Sibai BM, Caritis S, et al. Adverse perinatal out-
comes are significantly higher in severe gestational hypertension 32. Salvig JD, Olsen SF, Secher NJ. Effects of fish oil supplementation
than in mild preeclampsia. Am J Obstet Gynecol. 2002;186:66-71. in late pregnancy on blood pressure: a randomized controlled trial.
Br J Obstet Gynaecol. 1996;103:529-533.
14. Cruz MO, Gao W, Hibbard JU. Obstetrical and perinatal out-
comes among women with gestational hypertension, mild pre- 33. Poston L, Briley AL, Seed PT, Kelly FJ, Shennan AH. Vitamin
eclampsia, and mild chronic hypertension. Am J Obstet Gynecol. C and vitamin E in pregnant women at risk for pre-eclampsia
2011;205(3):260.e261-269. (VIP trial): randomised placebo-controlled trial. Lancet. Apr 8
2006;367(9517):1145-1154.
15. Rodriguez-Thompson D, Lieberman ES. Use of a random urinary
protein-to-creatinine ratio for the diagnosis of significant protein- 34. Roberts JM, Myatt L, Spong CY, et al. Vitamins C and E to Prevent
uria during pregnancy. Am J Obstet Gynecol. 2001;185:808-811. Complications of Pregnancy-Associated Hypertension. New Eng-
land Journal of Medicine. 2010;362(14):1282-1291.
16. Davison JM, Homuth V, Jeyabalan A, et al. New Aspects in
the Pathophysiology of Preeclampsia. J Am Soc Nephrol. 35. Rumbold A, Duley L, Crowther CA, Haslam RR. Antioxidants for
2004;15:2440-2448. preventing pre-eclampsia. Cochrane Database of Systematic
Reviews. 2008(1).
17. Kenny LC, Black MA, Poston L, et al. Early pregnancy prediction
of preeclampsia in nulliparous women, combining clinical risk and 36. Hofmeyr GJ, Lawrie TA, Atallah AN, Duley L, Torloni MR. Calcium
biomarkers: the Screening for Pregnancy Endpoints (SCOPE) inter- supplementation during pregnancy for preventing hypertensive
national cohort study. Hypertension. Sep 2014;64(3):644-652. disorders and related problems. Cochrane Database Syst Rev.
2014;6:CD001059.

— Chapter B  25
Chapter B

37. Guideline: Calcium Supplementation in Pregnant Women. Geneva: 56. Duley L, Gulmezoglu AM, Henderson-Smart DJ, Chou D. Magne-
World Health Organization; 2013. sium sulfate and other anticonvulsants for women with pre-eclamp-
38. Duley L, Henderson-Smart DJ, Meher S, King JF. Antiplatelet sia. Cochrane Database Syst Rev. 2010.
agents for preventing pre-eclampsia and its complications 57. ACOG Committee on Practice Bulletins--Obstetrics. ACOG Prac-
(Review). Cochrane Database of Systematic Reviews. 2007(2):121. tice Bulletin No. 33: Diagnosis and Mangement of Preeclampsia
39. Sibai BM. Diagnosis and Management of Gestational Hyperten- and Eclampsia. Obstet Gynecol. 2002;99:159-167.
sion and Preeclampsia. Obstet Gynecol. 2003;102:181-192. 58. Euser AG, Cipolla MJ. Magnesium sulfate for the treatment of
40. Spong CY, Mercer BM, D’Alton M, Kilpatrick S, Blackwell S, Saade eclampsia: a brief review. Stroke; a journal of cerebral circulation.
G. Timing of Indicated Late-Preterm and Early-Term Birth. Obstet Apr 2009;40(4):1169-1175.
Gynecol. 2011;118(2):323-333. 59. Lu J, Nightingale CH. Magnesium sulfate in eclampsia and pre-
41. Koopmans CM, Bijlenga D, Groen H, et al. Induction of labour eclampsia: pharmacokinetic principles. Clin Pharmacokinet. 2000;
versus expectant monitoring for gestational hypertension or mild 38(4):305-314.
pre-eclampsia after 36 weeks’ gestation (HYPITAT): a multicentre, 60. Sibai BM. Magnesium sulfate prophylaxis in preeclampsia:
open-label randomised controlled trial. Lancet. 2009;374(9694): Lessons learned from recent trials. Am J Obstet Gynecol.
979-988. 2004;190:1520-1526.
42. Tajik P, van der Tuuk K, Koopmans CM, et al. Should cervical 61. Abalos E, Duley L, Steyn DW, Henderson-Smart DJ. Antihyperten-
favourability play a role in the decision for labour induction in sive drug therapy for mild to moderate hypertension during preg-
gestational hypertension or mild pre-eclampsia at term? An explor- nancy. Cochrane Database Syst Rev. 2001;2.
atory analysis of the HYPITAT trial. BJOG. 2012;119(9):1123-1130. 62. Von Dadelszen P, Magee L. Antihypertensive Medications in Man-
43. Vijgen SM, Koopmans CM, Opmeer BC, et al. An economic analy- agement of Gestational Hypertension-Preeclampsia. Clin Obstet
sis of induction of labour and expectant monitoring in women with Gynecol. 2005;48(2):441-459.
gestational hypertension or pre-eclampsia at term (HYPITAT trial). 63. Martin JN, Jr., Thigpen BD, Moore RC, Rose CH, Cushman J,
BJOG. Dec 2010;117(13):1577-1585. May W. Stroke and severe preeclampsia and eclampsia: A para-
44. Dildy III GA. Complications of preeclampsia. In: Dildy III GA, Belfort digm shift focusing on systolic blood pressure. Obstet Gynecol.
MA, Saade GR, Phelan JP, Hankins GDV, Clark SL, eds. Critical 2005;105:246-254.
Care Obstetrics. 4th ed. Malden, Massachusetts: Blackwell Science; 64. Duley L, Meher S, Jones L. Drugs for treatment of very high blood
2004. pressure during pregnancy. Cochrane Database Syst Rev. 2013(7).
45. Magann EF, Martin JN. Twelve steps to optimal management of 65. Vermillion ST, Scardo JA, Newman RB, Chauhan SP. A random-
HELLP syndrome. Clinical Obstet Gynecol. 1999;42:532-550. ized, double-blind trial of oral nifedipine and intravenous labetalol
46. Duley L, Williams J, Henderson-Smart DJ. Plasma volume expan- in hypertensive emergencies of pregnancy. Am J Obstet Gynecol.
sion for treatment of pre-eclampsia. Cochrane Database Syst Rev. 1999;181(4):858-861.
2000;2. 66. Scardo JA, Vermillion ST, Newman RB, Chauhan SP, Hogg BB. A
47. The Magpie Trial Collaboratve Group. Do women with pre- randomized, double-blind, hemodynamic evaluation of nifedipine
eclampsia, and their babies, benefit from magnesium sulphate? and labetalol in preeclamptic hypertensive emergencies. Am J
The Magpie Trial: a randomized placebo-controlled trial. Lancet. Obstet Gynecol. 1999;181(4):862-866.
2002;359:1877-1890. 67. Odendaal HJ, Pattinson RC, Bam R, Grove D, Kotze TJ. Aggressive
48. Lucas MJ, Leveno KJ, Cunningham FG. A comparison of mag- or expectant mangement of patients with severe pre-eclampsia
nesium sulphate with phenytoin for the prevention of eclampsia. between 28-34 weeks’ gestation: a randomized controlled trial.
N Engl J Med. 1995;333:201-205. Obstet Gynecol. 1990;76:1070-1075.
49. Belfort MA, Anthony J, Saade GR, Allen JC, Jr. A Comparison 68. Sibai BM, Medicine SoM-F. Evaluation and management of severe
of Magnesium Sulphate and Nimodipine for the Prevention of preeclampsia before 34 weeks’ gestation. American Journal of
Eclampsia. N Engl J Med. 2003;348:304-311. Obstetrics and Gynecology. 2011;205(3):191-198.
50. Eclampsia trial collaborative group. Which anti-convulsant for 69. Sibai BM, Mercer BM, Schiff E, Friedman SA. Aggressive versus
women with eclampsia? Evidence from the collabortive trial. Lan- expectant management of severe preeclampsia at 28 to 32 weeks’
cet. 1995;345:1455-1463. gestation: a randomized controlled trial. Am J Obstet Gynecol.
51. Duley L, Henderson-Smart DJ, Chou D. Magnesium sulphate ver- 1994;171:818-822.
sus phenytoin for eclampsia. Cochrane Database of Systematic 70. Churchill D, Duley L, Thornton JG, Jones L. Interventionist ver-
Reviews. 2010(10). sus expectant care for severe pre-eclampsia between 24 and 34
52. Duley L, Henderson-Smart DJ, Walker GJA, Chou D. Magnesium weeks’ gestation. Cochrane Database of Systematic Reviews.
sulphate versus diazepam for eclampsia. Cochrane Database of 2013(7).
Systematic Reviews. 2010(12). 71. Vigil-De Gracia P, Tejada OR, Minaca AC, et al. Expectant manage-
53. Duley L, Gulmezoglu AM, Chou D. Magnesium sulphate ver- ment of severe preeclampsia remote from term: the MEXPRE Latin
sus lytic cocktail for eclampsia. Cochrane Database Syst Rev. Study, a randomized, multicenter clinical trial. Am J Obstet Gyne-
2010(9):CD002960. col. 2013;209(5):8.

54. Sibai B. Magnesium sulfate prophylaxis in preeclampsia: evidence 72. Sibai BM. What to expect from expectant management in severe
from randomized trails. Clin Obstet Gynecol. 2005;48(2):478-488. preeclampsia at <34 weeks gestation: pregnancy outcomes in
developed vs developing countries. Am J Obstet Gynecol. Nov
55. Sibai BM. Diagnosis, Prevention, and Management of Eclampsia. 2013;209(5):400-401.
Obstet Gynecol. 2005;105:402-410.

26  Chapter B —
Medical Complications of Pregnancy

73. Maia SB, Katz L, Neto CN, Caiado BV, Azevedo AP, Amorim MM. 90. O’Brien JM, Shumate SA, Satchwell SL, Milligan DA, Barton JR.
Abbreviated (12-hour) versus traditional (24-hour) post-partum Maternal benefit of corticosteriod therapy in patients with HELLP
magnesium sulfate therapy in severe pre-eclampsia. International (hemolysis, elevated liver enzymes, and low platelet count) syn-
journal of gynaecology and obstetrics: the official organ of the drome: impact on the rate of regional anesthesia. Am J Obstet
International Federation of Gynaecology and Obstetrics. Sep Gynecol. 2002;186:475-479.
2014;126(3):260-264. 91. Martin JN, Blake PG, Perry KG, Jr., McCaul JF, Hell LW, Martin RW.
74. Magee LA, von Dadelszen P. Prevention and treatment of postpar- The natural history of HELLP syndrome: patterns of disease pro-
tum hypertension. Cochrane Database of Systematic Reviews. gression and regression. Am J Obstet Gynecol. 1991;164:1500-1509.
2013(4). 92. Castro MA, Fasset MJ, Reynolds TB, Shaw KJ, Goodwin TM.
75. Mattar F, Sibai BM. Eclampsia. VIII. Risk factors for maternal mor- Reversible peripartum liver failure: a new perspective on the diag-
bidity. Am J Obstet Gynecol. 2000;182:307-312. nosis, treatment, and cause of acute fatty liver of pregnancy based
76. Cooray SD, Edmonds SM, Tong S, Samarasekera SP, Whitehead on 28 cases. Am J Obstet Gynecol. 2000;181:389-395.
CL. Characterization of symptoms immediately preceding eclamp- 93. Fesenmeir MF, Coppage KH, Lambers DS, Barton JR, Sibai BM.
sia. Obstet Gynecol. Nov 2011;118(5):995-999. Acute fatty liver of pregnancy in 3 tertiary care centers. Am J
77. Aagaard-Tillery KM, Belfort MA. Eclampsia: Morbidity, Mortality, Obstet Gynecol. 2005;192:1416-1419.
and Management. Clinical Obstet Gynecol. 2005;48:12-23. 94. Ibdah JA, Bennett MJ, Rinaldo P, et al. A fetal fatty-acid oxidation
78. Circulation. Adult Basic Life Supprt. 2000; http://circ.ahajournals. disorder as a cause of liver disease in pregnant women. N Engl J
org/content/102/suppl_1/I-22. Accessed 10/19/2014. Med. 1999;340:1723-1731.

79. Miguil M, Chekairi A. Eclampsia, study of 342 cases. Hypertension 95. Steingrub JS. Pregnancy-associated severe liver dysfunction. Crit
in pregnancy : official journal of the International Society for the Care Clin. 2004;20:763-776.
Study of Hypertension in Pregnancy. 2008;27(2):103-111. 96. Nelson DB, Yost NP, Cunningham FG. Acute fatty liver of preg-
80. Eke AC, Ezebialu IU, Okafor C. Presentation and outcome of nancy: clinical outcomes and expected duration of recovery. Am J
eclampsia at a tertiary center in South East Nigeria--a 6-year Obstet Gynecol. 2013;209(5):456.e451-457.
review. Hypertension in pregnancy: official journal of the Inter- 97. Amon E, Allen SR, Petrie RH, Belew JE. Acute fatty liver of preg-
national Society for the Study of Hypertension in Pregnancy. nancy associated with pre-eclampsia: management of hepatic
2011;30(2):125-132. failure with postpartum liver transplantation. Am J Perinatol.
81. Prevention CfDCa. Pregnancy Mortality Surveillance System. 2014; 1991;8:278-279.
http://www.cdc.gov/reproductivehealth/ MaternalInfantHealth/ 98. Jin FL, Cao MB, Bai YQ, Zhang YR, Yang YX, Zhang BY. Thera-
PMSS.html. Accessed Oct. 11, 2014. peutic Effects of Plasma Exchange for the Treatment of 39
82. MacKay AP, Berg CJ, Atrash HK. Pregnancy-Related Mor- Patients with Acute Fatty Liver of Pregnancy. Discovery Medicine.
tality From Preeclampsia and Eclampsia. Obstet Gynecol. 2012;13(72):369-373.
2001;97:533-538. 99. Pearson GD, Veille JC, Rahimtoola S. Peripartum Cardiomyopathy:
83. Weinstein L. Syndrome of hemolysis, elevated liver enzymes and National Heart, Lung, and Blood Institute and Office of Rare Dis-
low platelet count: a severe consequence of hypertension in preg- eases (National Institute of Health) Workshop Recommendations
nancy. Am J Obstet Gynecol. 1982;142:159-167. and Review. JAMA. 2000;283:1183-1188.

84. Sibai BM, Ramadan MK, Usta I, Salama M, Mercer BM, Friedman 100. Felker GM, Thompson RE, Hare JM, et al. Underlying causes and
SA. Maternal morbidity and mortality in 442 pregnancies with long-term survival in patients with initially unexplained cardiomy-
hemolysis, elevated liver enzymes, and low platelets (HELLP syn- opathy. N Engl J Med. 2000;342:1077-1084.
drome). Am J Obstet Gynecol. 1993;169:1000-1006. 101. Harper MA, Meyer RE, Berg CJ. Peripartum Cardiomyopathy
85. Sibai BM. Diagnosis, Controversies, and Mangement of the Syn- Population-Based Birth Prevalence and 7-Year Mortality. Obstet
drome of Hemolysis, Elevated Liver Enzymes, and Low Platelet Gynecol. 2012;120(5):1013-1019.
Count. Obstet Gynecol. 2004;103:981-991. 102. Bozkurt B, Villanueva FS, Holubkuv R, et al. Intravenous Immune
86. Silver RM, Berkowitz RL, Bussel J. ACOG Practice Bulletin #6: Globulin in the Therapy of Peripartum Cardiomyopathy. J Am Coll
thrombocytopenia in pregnancy. Chicago: American College of Cardiol. 1999;34:177-180.
Obstetricians and Gynecologists; 1999. 103. James A. Practice bulletin no. 123: thromboembolism in preg-
87. Barton JR, Sibai BM. Diagnosis and mangement of hemolysis, nancy. Obstet Gynecol. Sep 2011;118(3):718-729.
elevated liver enzymes, and low platelets syndrome. Clin Perinatol. 104. Heit JA, Kobbervig CE, James AH, Petterson TM, Bailey KR,
2004;31:807-833. Melton LJ, 3rd. Trends in the incidence of venous thromboembo-
88. Woudstra DM, Chandra S, Hofmeyr GJ, Dowswell T. Corticoste- lism during pregnancy or postpartum: a 30-year population-based
roids for HELLP (hemolysis, elevated liver enzymes, low platelets) study. Annals of internal medicine. Nov 15 2005;143(10):697-706.
syndrome in pregnancy. Cochrane Database of Systematic 105. Doyle NM, Ramirez MM, Mastrobattista JM, Monga M, Wagner LK,
Reviews. 2010(9). Gardner MO. Diagnosis of pulmonary embolism: a cost-effective
89. Fonseca JE, Mendez F, Catano C, Arias F. Dexamethasone treat- analysis. Am J Obstet Gynecol. Sep 2004;191(3):1019-1023.
ment does not improve the outcome of women with HELLP syn- 106. McColl MD, Ellison J, Greer IA. Tait RC. Walker ID. Prevalence
drome: a double-blind, placebo-controlled, randomized clinical of the post-thrombotic syndrome in young women with previous
trial. Am J Obstet Gynecol. 2005;193:1591-1598. venous thromboembolism. British Journal of Haematology. Feb
2000;108(2):272-274.

— Chapter B  27
Chapter B

107. Gerhardt A, Scharf RF, Zotz RB. Effect of hemostatic risk factors on 124. Cahill AG, Stout MJ, Macones GA, Bhalla S. Diagnosing pul-
the individual probability of thrombosis during pregnancy and the monary embolism in pregnancy using computed-tomographic
puerperium. Thrombosis and Haemostasis. Jul 2003;90(1):77-85. angiography or ventilation-perfusion. Obstet Gynecol. Jul
108. ACOG Practice Bulletin No. 138: Inherited thrombophilias in preg- 2009;114(1):124-129.
nancy. Obstet Gynecol. Sep 2013:122(3):706-717. 125. Lapner ST, Kearon C. Diagnosis and management of pulmonary
109. Zotz RB, Gerhardt A, Scharf RE. Prediction, prevention, and treat- embolism. BMJ. 2013;346:f757.
ment of venous thromboembolic disease in pregnancy. Seminars 126. Cook JV, Kyriou J. Radiation from CT and perfusion scanning in
in Thrombosis and Hemostasis. Apr 2003;92(2):143-154. pregnancy. BMJ. Aug 6 2005;331(7512):350.
110. Pabinger I, Grafenhofer H, Kyrle PA, et al. Temporary increase in 127. Remy-Jardin M, Remy J. Spiral CT angiography of the pulmonary
the risk for recurrence during pregnancy in women with a history of circulation. Radiology. Sep 1999;212(3):615-636.
venous thromboembolism. Blood. Aug 1 2002;100(3):1060-1062. 128. Allen C, Demetriades T. Radiation risk overestimated. Radiology.
111. Greer IA. Prevention of venous thromboembolism in preg- Aug 2006;240(2):613-614; discussion 614.
nancy. Best practice & research. Clinical haematology. Jun 129. I G. The acute management of thrombosis and embolism during
2003;16(2):261-278. pregnancy and the puerperium. Green-top Guideline No. 37b.
112. Chan WS, Rey E, Kent NE, et al. Venous thromboembolism and 2010.
antithrombotic therapy in pregnancy. Journal of obstetrics and gyn- 130. Kearon C. Diagnosis of pulmonary embolism. CMAJ: Canadian
aecology Canada : JOGC = Journal d’obstetrique et gynecologie Medical Association journal = journal de l’Association medicale
du Canada : JOGC. Jun 2014;36(6):527-553. canadienne. Jan 21 2003;168(2): 183-194.
113. Royal College of Obstetricians and Gynaecologists. Reducing the 131. Perrier A, Roy PM, Sanchez O, et al. Multidetector-row computed
risk of thrombosis and embolism during pregnancy and the puer- tomography in suspected pulmonary embolism. N Engl J Med. Apr
perium. RCOG. 2009. 28 2005;352(17):1760-1768.
114. Friederich PW, Sanson BJ, Simioni P, et al. Frequency of preg- 132. Quiroz R, Kucher N, Zou KH, et al. Clinical validity of a nega-
nancy-related venous thromboembolism in anticoagulant factor- tive computed tomography scan in patients with suspected
deficient women: implications for prophylaxis. Annals of Internal pulmonary embolism: a systematic review. JAMA. Apr 27
Medicine. Dec 15 1996;125(12):955-960. 2005;293(16):2012-2017.
115. Bates SM, Greer IA, Middeldorp S, Veenstra DL, Prabulos AM, 133. Schoepf UJ, Goldhaber SZ, Costello P. Spiral computed
Vandvik PO. VTE, thrombophilia, antithrombotic therapy, and preg- tomography for acute pulmonary embolism. Circulation. May 11
nancy: Antithrombotic Therapy and Prevention of Thrombosis, 9th 2004;109(18): 2160-2167.
ed: American College of Chest Physicians Evidence-Based Clini-
cal Practice Guidelines. Chest. Feb 2012;141(2 Suppl):e691S-736S. 134. Ramzi DW, Leeper KV. DVT and pulmonary embolism: Part I. Diag-
nosis. American family physician. Jun 15 2004;69(12):2829-2836.
116. Walker MC, Garner PR, Keely EJ, Rock GA, Reis MD. Changes
in activated protein C resistance during normal pregnancy. Am J 135. Kline JA, Johns KL, Colucciello SA, Israel EG. New diagnostic
Obstet Gynecol. Jul 1997;177(1):162-169. tests for pulmonary embolism. Annals of emergency medicine.
Feb 2000;35(2):168-180.
117. Greer IA, Thomas A. Thromboemboli disease in pregnancy and
the puerperium. Guidelines and Audit Committee of the Royal Col- 136. Committee on Practice Bulletins: Gynecology. ACOG Practice Bul-
lege of Obstetricians and Gynaecologists. 2010. letin No. 84: Prevention of deep vein thrombosis and pulmonary
embolism. Obstet Gynecol. Aug 2007;110(2 Pt 1):429-440.
118. Committee on Practice Bulletins: Obstetrics. Practice Bulletin No.
132: Antiphospholipid syndrome. Obstet Gynecol. Dec 2012;120(6): 137. Pillny M, Sandmann W, Luther B, et al. Deep venous throm-
1514-1521. bosis during pregnancy and after delivery: indications for and
results of thrombectomy. Journal of Vascular Surgery. Mar
119. Chan WS, Spencer FA, Ginsberg JS. Anatomic distribution of deep 2003;37(3):528-532.
vein thrombosis in pregnancy. CMAJ : Canadian Medical Associa-
tion journal = journal de l’Association medicale canadienne. Apr 138. Dong BR, Hao Q, Yue J, Wu T, Liu GJ. Thrombolytic therapy
20 2010;182(7):657-660. for pulmonary embolism. Cochrane Database Syst Rev.
2009(3):CD004437.
120. Chan WS, Lee A, Spencer FA, et al. Predicting deep venous throm-
bosis in pregnancy: out in “LEFt” field? Annals of Internal Medi- 139. Tooher R, Gates S, Dowswell T, Davis LJ. Prophylaxis for venous
cine. Jul 21 2009;151(2):85-92. thromboembolic disease in pregnancy and the early postnatal
period. Cochrane Database Syst Rev. 2010(5):CD001689.
121. Righini M, Jobic C, Boehlen F, et al. Predicting deep venous throm-
bosis in pregnancy: external validation of the LEFT clinical predic- 140. Che Yaakob CA, Dzarr AA, Ismail AA, Zuky Nik Lah NA, Ho JJ.
tion rule. Haematologica. Apr 2013;98(4):545-548. Anticoagulant therapy for deep vein thrombosis (DVT) in preg-
nancy. Cochrane Database Syst Rev. 2010(6):CD007801.
122. Nijkeuter M, Ginsberg JS, Huisman MV. Diagnosis of deep vein
thrombosis and pulmonary embolism in pregnancy: a systematic 141. Laurent P, Dussarat GV, Bonal J, et al. Low molecular weight hepa-
review. Journal of Thrombosis and Haemostasis: JTH. Mar 2006; rins: a guide to their optimum use in pregnancy. Drugs. 2002;62(3):
4(3):496-500. 463-477.
123. Kearon C, Akl EA, Comerota AJ, et al. Antithrombotic therapy 142. Carlin AJ, Farquharson RG, Quenby SM, Topping J, Fraser WD.
for VTE disease: Antithrombotic Therapy and Prevention of Prospective observational study of bone mineral density during
Thrombosis, 9th ed: American College of Chest Physicians Evi- pregnancy: low molecular weight heparin versus control. Human
dence-Based Clinical Practice Guidelines. Chest. Feb 2012;141(2 reproduction (Oxford, England). May 2004;19(5):1211-1214.
Suppl):e419S-494S.

28  Chapter B —
Medical Complications of Pregnancy

143. Casele H, Haney EI, James A, Rosene-Montella K, Carson M. 156. Hull RD, Raskob GE, Rosenbloom D, et al. Heparin for 5 days as
Bone density changes in women who receive thromboprophylaxis compared with 10 days in the initial treatment of proximal venous
in pregnancy. Am J Obstet Gynecol. Oct 2006;195(4):1109-1113. thrombosis. N Engl J Med. May 3 1990;322(18):1260-1264.
144. McColl MD, Greer IA. Low-molecular-weight heparin for the pre- 157. Ageno W, Steidl L, Ultori C, et al. The initial phase of oral anticoag-
vention and treatment of venous thromboembolism in pregnancy. ulation with warfarin in outpatients with deep venous thrombosis.
Current opinion in pulmonary medicine. Sep 2004;10(5):371-375. Blood coagulation & fibrinolysis : an international journal in hae-
145. Gouin-Thibault I, Pautas E, Siguret V. Safety profile of different low- mostasis and thrombosis. Jan 2003;14(1):11-14.
molecular weight heparins used at therapeutic dose. Drug safety 158. Horlocker TT, Wedel DJ, Rowlingson JC, Enneking FK. Executive
: an international journal of medical toxicology and drug experi- summary: regional anesthesia in the patient receiving antithrom-
ence. 2005;28(4):333-349. botic or thrombolytic therapy: American Society of Regional
146. Greer IA, Nelson-Piercy C. Low-molecular-weight heparins for Anesthesia and Pain Medicine Evidence-Based Guidelines
thromboprophylaxis and treatment of venous thromboembolism in (Third Edition). Regional anesthesia and pain medicine. Jan-Feb
pregnancy: a systematic review of safety and efficacy. Blood. Jul 15 2010;35(1):102-105.
2005;106(2):401-407. 159. Pettila V, Leinonen P, Markkola A, Hiilesmaa V, Kaaja R. Postpar-
147. Richter C HA, Huch R. Transfer of low molecular weight heparin tum bone mineral density in women treated for thromboprophy-
during breast feeding. 1997;78:734. laxis with unfractionated heparin or LMW heparin. Thrombosis and
haemostasis. Feb 2002;87(2):182-186.
148. Richter C, Sitzmann J, Lang P, Weitzel H, Huch A, Huch R. Excre-
tion of low molecular weight heparin in human milk. British journal 160. Kozer E, Nikfar S, Costei A, Boskovic R, Nulman I, Koren G.
of clinical pharmacology. Dec 2001;52(6):708-710. Aspirin consumption during the first trimester of pregnancy and
congenital anomalies: a meta-analysis. Am J Obstet Gynecol. Dec
149. Orme ML, Lewis PJ, de Swiet M, et al. May mothers given warfa- 2002;187(6):1623-1630.
rin breast-feed their infants? British medical journal. Jun 18 1977;
1(6076):1564-1565. 161. Kozer E, Costei AM, Boskovic R, Nulman I, Nikfar S, Koren G.
Effects of aspirin consumption during pregnancy on pregnancy
150. McKenna R, Cole ER, Vasan U. Is warfarin sodium contraindicated outcomes: meta-analysis. Birth defects research. Part B, Develop-
in the lactating mother? The Journal of pediatrics. Aug 1983;103(2): mental and reproductive toxicology. Feb 2003;68(1):70-84.
325-327.
162. De Stefano V, Grandone E, Martinelli I. Recommendations for
151. Bhutia S, Wong PF. Once versus twice daily low molecular weight prophylaxis of pregnancy-related venous thromboembolism in
heparin for the initial treatment of venous thromboembolism. carriers of inherited thrombophilia. Comment on the 2012 ACCP
Cochrane Database Syst Rev. 2013;7:CD003074. guidelines. Journal of thrombosis and haemostasis : JTH. Sep
152. Voke J, Keidan J, Pavord S, Spencer NH, Hunt BJ. The manage- 2013;11(9):1779-1781.
ment of antenatal venous thromboembolism in the UK and Ireland: 163. Bates SM, Greer IA, Middeldorp S, Veenstra DL, Prabulos AM,
a prospective multicentre observational survey. British Journal of Vandvik PO. Recommendations for prophylaxis of pregnancy-
Haematology. Nov 2007;139(4):545-558. related venous thromboembolism in carriers of inherited thrombo-
153. Shiach CR. Monitoring of low molecular weight heparin in philia. Comment on the 2012 ACCP guidelines: a rebuttal. Journal
pregnancy. Hematology (Amsterdam, Netherlands). Feb of thrombosis and haemostasis: JTH. Sep 2013;11(9):1782-1784.
2003;8(1):47-52. 164. Seshadri N, Goldhaber SZ, Elkayam U, et al. The clinical challenge
154. Linkins LA, Dans AL, Moores LK, et al. Treatment and prevention of bridging anticoagulation with low-molecular-weight heparin in
of heparin-induced thrombocytopenia: Antithrombotic Therapy patients with mechanical prosthetic heart valves: an evidence-
and Prevention of Thrombosis, 9th ed: American College of Chest based comparative review focusing on anticoagulation options in
Physicians Evidence-Based Clinical Practice Guidelines. Chest. pregnant and nonpregnant patients. American heart journal. Jul
Feb 2012;141(2 Suppl):e495S-530S. 2005;150(1):27-34.
155. Clouse LH, Comp PC. The regulation of hemostasis: the protein C 165. Marik PE, Plante LA. Venous thromboembolic disease and preg-
system. N Engl J Med. May 15 1986;314(20):1298-1304. nancy. N Engl J Med. Nov 6 2008;359(19):2025-2033.

— Chapter B  29
Objectives
Medical Complications of 
• Describe the following potentially life‐threatening
Pregnancy medical complications of pregnancy:
 Hypertensive disorders 
Revised January 2017  Acute fatty liver of pregnancy 
 Peripartum cardiomyopathy
 Deep venous thrombosis 
 Pulmonary embolism 
• Formulate a plan for diagnosis and treatment

Hypertensive Disorders of Pregnancy Hypertensive Disorders of Pregnancy
• Present in up to 10% of pregnancies • Classifications:
• The 6th leading cause of maternal mortality – Chronic hypertension (HTN)
– In the United States from 2006‐2009, hypertensive – Gestational HTN
disorders accounted for 9.9% of maternal deaths – Preeclampsia‐eclampsia
• Leading cause of neonatal mortality/morbidity, – Preeclampsia superimposed on chronic HTN
primarily due to the effects of preterm delivery
Information from the National High Blood Pressure Education Program: Working Group Report on High Blood Pressure in Pregnancy, 2000. Classification schema Information from National High Blood Pressure Education Program: Working Group Report on High Blood Pressure in Pregnancy, 2000. Classification schema
affirmed by 2013 ACOG Taskforce on Hypertension in Pregnancy. affirmed by 2013 ACOG Taskforce on Hypertension in Pregnancy.

Chronic Hypertension Gestational Hypertension
• Blood pressure (BP) is elevated >140/90 mm Hg prior • Non‐proteinuric HTN after 20 weeks gestation
to 20 weeks gestation  • Category includes:
• No proteinuria (unless proteinuria was pre‐existing – Women who go on to develop preeclampsia
prior to the pregnancy) – Women with previously undiagnosed chronic HTN
• BP remains elevated beyond 12 weeks postpartum – Transient gestational HTN
• Specific diagnosis is made postpartum, in retrospect

Copyright 2017© American Academy of Family Physicians. All rights reserved. 1


Preeclampsia‐Eclampsia Spectrum Preeclampsia: A Multi‐system Disorder
• Greatest cause of maternal/perinatal morbidity and Eyes CNS
Arteriolar spasm Seizures
mortality Retinal hemorrhage Intracranial hemorrhage
• Includes: Papilledema
Transient scotomata
CVA
Encephalopathy
– Preeclampsia‐specify with or without severe features Respiratory System Pancreas
Pulmonary edema Ischemic pancreatitis
– Terminology change: mild preeclampsia is no longer used! ARDS Kidneys
– Preeclampsia superimposed on chronic HTN Liver Acute renal failure
– Eclampsia Subcapsular hemorrhage Uteroplacental Circulation
Hepatic rupture IUGR
– HELLP syndrome Abruption
Hematopoietic System
HELLP syndrome Fetal compromise
DIC Fetal demise

Risk Factors for Preeclampsia Prevention of Preeclampsia 
• Primiparity  • Chronic renal disease  • Fish oils, Vitamins (D, C, or E), salt restriction, and
• First degree relative with  • Multiple gestation pregnancy bed rest do not prevent preeclampsia
history of preeclampsia =  • Diabetes  • Calcium may have some benefit in women at high
3x risk
• Systemic lupus erythematosus risk with low calcium diets (rare in the United States)
• Previous preeclamptic 
• Maternal age older than 40  • Aspirin has some benefit, but it is modest with a high
pregnancy = 7X risk
years 
• Chronic HTN  number needed to treat (NNT) for low‐risk women
• Obesity

Recommendations for Low‐Dose Aspirin Diagnosis of Preeclampsia
US Preventive Services Task Force (USPSTF) 2014 recommends low‐dose 
• New onset HTN after 20 weeks with proteinuria: 
aspirin for pregnant women with  – BP ≥140/90 mm Hg two times, taken 4 hours apart
• Any high‐risk factors: History of preeclampsia, multiple gestation, chronic – BP ≥60/110 mm Hg once
hypertension, antenatal diabetes, renal disease, autoimmune disease (systemic  • Proteinuria is defined by:
lupus erythematosus, antiphospholipid syndrome) – 24 hour protein ≥300 mg
• Multiple moderate risk factors: nulliparity, obesity (BMI >30 kg/m2), family hx – Timed urine protein level extrapolated out to 24 hour value 
preeclampsia, African American , low socioeconomic  – Protein/creatinine ratio of ≥0.3
status, age > 35 years – Urine dip + 1 or more (only if other methods are not available)
• ACOG has endorsed these recommendations – Proteinuria is NOT required if the patient has new onset HTN with 
specified findings (next slide)
Suggested dose: 60 to 81 mg daily started late in the first trimester
Information from US Preventive  Services Task Force, 2014.

2
Diagnosing Preeclampsia  New onset headache or
visual disturbances
Without Proteinuria Pulmonary edema
• New onset HTN with any of these findings:
– Platelets <100,000/uL Hepatic dysfunction
– Creatinine >1.1 mg/dL or doubled from baseline
-transaminases 2X normal
-right upper quadrant (RUQ) or
– Transaminases twice the normal levels epigastric pain
– Pulmonary edema
– Cerebral or visual symptoms Elevated creatinine
(>1.1 mg/dL or 2X baseline)
– Blood pressure >160/110
Systolic BP ≥160 mm Hg
Diastolic BP ≥110 mm Hg
Platelets <100,000/uL

Management of Preeclampsia  Management: Preeclampsia 
WITHOUT Severe Features WITH Severe Features
• Expectant management before 37 weeks
• Closely watch for development of severe features
• Admit to hospital and monitor closely on bedrest
• Establish baseline complete blood count (CBC), transaminases, creatinine,  • Treatment goals:
LDH, and uric acid
• Antepartum surveillance  – Prevent seizures
– Non‐stress tests (NST’s), amniotic fluid measurement, biophysical profile
– Growth ultrasound every 3 to 4 weeks
– Control BP to prevent cerebral hemorrhage
• Weekly labs – Expedite delivery, balancing maternal condition and fetal
– CBC, transaminases (AST, ALT) maturity
– No need to follow urine protein, as even >5 g is not considered a “severe feature”
• Delivery at 37 weeks

Maternal Evaluation Magnesium Sulfate
• Symptom assessment at least every 8 hours • Preferred anticonvulsant
– Assessing for headache, visual changes, RUQ or epigastric pain, – Slows neuromuscular conduction and decreases CNS irritability
retrosternal pain or pressure
• Vital signs, neuro checks and deep tendon reflexes every 15 to 60  – No significant effect on BP
minutes until stable – Prevents seizures (NNT = 100 for all women with preeclampsia; 
• Monitor intake and output; Insert foley catheter if needed NNT = 400 for preeclampsia without severe features )
– Notify delivering clinician for <30 mL urine per hour
– Decreases risk of placental abruption 
• Labs daily; may decrease to every other day when stable
– CBC, transaminases, creatinine, uric acid, LDH  (NNT = 100)
– Consider peripheral blood smear and coagulation profile
– Type and screen in labor

3
Indications for Magnesium Sulfate Magnesium Dosage and Monitoring
• Traditionally, all patients with preeclampsia received • 4 to 6 g intravenous IV loading dose over 15 to 20 
MgSO4 in labor minutes, followed by continuous infusion of 2 g/hour
• Current recommendation: • MgSO4 toxicity is uncommon with normal renal function
– Only treat women with MgSO4 who exhibit severe features • Check magnesium level if:
– Urine output <30 mL/hour
– Do NOT universally administer MgSO4 to all women with 
preeclampsia in labor – Elevated serum creatinine 
– Symptoms of MgSO4 toxicity
– Loss of patellar reflexes 

Antihypertensive Drugs for 
Magnesium Levels
mg/dL
Severe Preeclampsia
Normal 1.3 to 2.6 • Indication: Sustained BP elevation >160 mm Hg systolic or
Therapeutic 4 to 8 >110 diastolic (retaken within minutes is sufficient for 
Loss of patellar reflex 8 to 10 diagnosis)
Somnolence 10 to 12 • Timely treatment is essential
Respiratory depression 12 to 17
• Intravenous labetalol or hydralazine
Paralysis 15 to 17
Cardiac arrest • Oral nifedipine 
30 to 35
Antidote for MgSO4 is calcium gluconate 1 g IV over 3 minutes

Treatment of Severe Blood Pressures Delivery Decisions for Severe Preeclampsia
Labetalol • Vaginal delivery is preferred due to:
• Initial dose: 20 mg IV bolus over 2 minutes
– Lower risk of bleeding, infection, anesthesia reaction, and surgical 
• If BP remains ≥160/110 mm Hg, then repeat 10 minutes later with 40 mg IV, and 10 minutes 
later at 80 mg IV. If BP remains ≥160/110 mm Hg, switch to hydralazine complications 
• Maximum daily IV dose of 300 mg – Quicker recovery
• Cesarean delivery is indicated for:
Hydralazine – Continuous seizures or other emergent signs and symptoms
• Initial dose: 5 mg or 10 mg IV over 2 minutes 
• After 20 minutes, if BP remains ≥160/110 mm Hg, may repeat with 10 mg IV – Fetal distress
– Unfavorable cervix with severe prematurity (<30 weeks)
Nifedipine • Anesthesia methods:
• 10 to 20 mg PO; repeat in 30 minutes if BP remains elevated – Epidural/spinal preferred versus general
• Can also administer 10 to 20 mg PO every 2 to 6 hours for management
– General anesthesia is indicated if platelet count <50,000

4
Eclampsia Seizure Management
• Defined as the onset of seizures in a patient with preeclampsia • Avoid anticonvulsant polypharmacy
• Etiology of eclampsia is uncertain  • Protect airway to minimize aspiration
– cerebral edema, cerebral ischemia are possible causes • Prevent maternal injury
• BP is often significantly elevated, but in 15% of cases BP may be 
normal (diastolic ≤90 mm Hg) • Administer MgSO4 to control convulsions
• Can occur before, during, or after delivery • When stable, plan for delivery

HELLP Syndrome Clinical Presentation of HELLP
• A presentation of preeclampsia with severe features • Extremely variable presentation of symptoms
• Common findings: 
• HELLP acronym: – RUQ pain, epigastric pain, nausea and vomiting
– Hemolysis – 85% hypertensive
– Elevated Liver enzymes • Differential diagnosis can include biliary colic, pancreatitis, fatty 
liver of pregnancy, GERD, hepatitis, idiopathic thrombocytopenic 
– Low Platelets
purpura (ITP), thrombotic thrombocytopenic purpura (TTP) 
• Timing of diagnosis
– 30% of women are postpartum

Laboratory Findings in HELLP Management of HELLP
• Hemolysis • Similar to preeclampsia with severe features
– Abnormal peripheral smear – Stabilize mother
– LDH elevated – Evaluate fetus for compromise
– Determine optimal timing/route of delivery
• Liver enzymes – Use continuous fetal monitoring 
– Transaminases > twice the normal – Manage BP and fluid status

• Platelet count • All women should receive MgSO4 while symptomatic
– <100,000/uL or in labor

5
Postpartum Preeclampsia Management Initiation of Postpartum Magnesium?
• Improvement is usually rapid after delivery Women who have not required magnesium prior to delivery 
– Risk of seizures are greatest in the first 24 hours after birth may require postpartum initiation of MgSO4 if the following 
– Diuresis signals resolving of the disease process occurs:
• Monitor BP and urine output • New onset of HTN with cerebral symptoms (headache or 
blurred vision) 
• When already on MgSO4, continue until clinically stable • Preeclampsia with severe HTN 
– Usually 24 hours postpartum (≥160/110 mm Hg)
– If blood pressure remains elevated 24 hours postpartum, do not  • Eclampsia
use NSAID’s for pain control

Postpartum Management: In Hospital Postpartum Management: at Home
• 24 hours  • Educate women with preeclampsia/gestational 
– continue magnesium if severe preeclampsia HTN:
– watch I&O – Perform daily self‐assessment for symptoms
– Follow‐up for blood pressure check in 7 to 10 days
• 72 hours
– BP monitoring in hospital or equivalent at home • All women should:
• Treat persistent HTN – Receive information about signs/symptoms of 
postpartum preeclampsia at the time of discharge
– If 2 values >150/100 mm Hg at least 4 to 6 hours apart
– Understand preeclampsia may occur up to 4 weeks 
– Persistent BP >160/110 mm Hg (goal is to treat within 1 hour) postpartum
– Use PO nifedipine or labetalol (good safety profiles and fast onset) Reprinted with permission from The Preeclampsia Foundation. Improving understanding of preeclampsia. 
Am J Obstet Gynecol. 2012.

Acute Fatty Liver of Pregnancy (AFLP)  Diagnosis of AFLP
• Occurs in 1/7,000 to 16,000 pregnancies • Hypoglycemia
• Presents in the third trimester as: – Present in 50% of AFLP patients
– Vomiting, abdominal pain, anorexia, jaundice  • AST elevated, but usually <500 IU/L
– May progress to liver failure, ascites, renal failure, 
encephalopathy, or death • Bilirubin elevated, but usually <5 mg/dL
• Differential diagnosis  • PT and PTT prolonged, fibrinogen decreased
– HELLP syndrome, hepatitis, toxin‐induced liver damage • Liver biopsy diagnostic but rarely needed
• Delivery is the most important form of treatment

6
Peripartum Cardiomyopathy Peripartum Cardiomyopathy Management
• Heart failure developing in the last month of • Document with echocardiography
pregnancy or within 5 months after delivery • Avoid ACE inhibitors and excess diuresis in the
– Incidence 1/3,000 to 4,000 births antepartum period
– Mortality rate is 5% to 20%; fifth leading cause in the US
• Counsel regarding risk in subsequent pregnancies
• Presents with dyspnea, fatigue, edema
• Differential diagnosis: pulmonary embolism (PE),
amniotic fluid embolism, pneumonia, other causes of
congestive heart failure

Venous Thromboembolism (VTE) Risk Factors for VTE
• Includes deep vein thrombosis (DVT) and PE • Personal or family history of VTE • Severe varicose veins
• Thrombophilic disorders • Hyperemesis
• Occurs in 1/500 to 2,000 pregnancies • Multiparity (more than four  • Hypertensive disorder of pregnancy
deliveries) • Prolonged bed rest or immobility
• Leading cause of maternal mortality in developed • Age greater than 35 years • Major medical problems 
countries • Obesity (mechanical heart valve, 
• Smoking inflammatory bowel disease, 
• 25% of patients with untreated DVT develop PE • Infection/sepsis nephrotic syndrome, sickle cell 
disease, myeloproliferative 
• Undiagnosed PE has a 30% mortality rate • Cesarean delivery disorders)
• Dehydration • Postpartum hemorrhage

Thrombophilic Disorders Clinical Presentation of DVT
Inherited thrombophilia Acquired thrombophilia • 88% present in left leg • 88% present in the left leg
• Factor V Leiden mutation • Antiphospholipid antibody  • 71% in ileofemoral veins,  • 71% in ileofemoral veins, 
• Prothrombin G20210A  syndrome not involving calf veins not involving calf veins
mutation • Unilateral leg pain and  • Unilateral leg pain and 
• Methylene tetrahydrofolate swelling swelling
reductase mutation • Calf circumference  • Calf circumference 
• Antithrombin deficiency difference >2 cm difference >2 cm
• Protein C deficiency • Wells clinical criteria for VTE  • Wells clinical criteria for VTE 
prediction is not validated  prediction is not validated 
• Protein S deficiency 
in pregnancy in pregnancy

7
LEFt Criteria DVT Diagnosis – Ultrasound
Ultrasound
Left leg symptoms 
Edema (leg circumference discrepancy of ≥2 cm)  Meets diagnostic
Negative
criteria for DVT
First trimester at the time symptoms present

Low clinical suspicion


• DVT in pregnancy occurs in 12% of women with at least one of LEFt criteria High clinical suspicion
Begin
• 0% occurrence with none  anticoagulation
therapy No anticoagulation
needed

Anti-coagulate
AND
Repeat ultrasound in one week
Information from Chan W, Lee A, Spencer F, et al. Predicting deep venous thrombosis in pregnancy: out in "LEFt" field? Ann Intern Med. 2009;151:85‐92. OR Perform MRI

Pulmonary Embolism (PE) Pulmonary Embolism (PE)
• Two‐thirds occur postpartum • Two‐thirds occur postpartum
• Presentation varies from mild dyspnea • Presentation varies from mild dyspnea
and tachycardia to cardiopulmonary and tachycardia to cardiopulmonary
collapse collapse
• Treat (stabilization,oxygen,hemodynamic • Treat (stabilization,oxygen,hemodynamic
support) and evaluate simultaneously support) and evaluate simultaneously

PE Diagnosis V/Q or CT  Scan to Diagnose PE
Clinical suspicion of PE
• V/Q scan is preferable to CT Pulmonary Angiogram 
Stabilization and consideration of anticoagulation (CTPA) for diagnosis of PE in women who have a negative 
Clinical surveillance Venous Doppler lower extremities
Therapeutic
anticoagulation
chest x‐ray 
Negative and low  (Optional) Thrombosis
suspicion • CTPA involves 2.5 to 3.0 rads of radiation
Chest x-ray
Normal and ongoing
– 1 rad is estimated to increase the lifetime risk of breast cancer 
suspicion of PE  Abnormal
by 13.6% for a woman younger than 35 years
CTPA or V/Q scan CTPA
– Some feel that this is an overestimation and that 1 rad would 
Normal and  increase a 30‐year‐old woman’s lifetime risk of breast cancer by 
minimal
suspicion of PE 
Normal and ongoing 
suspicion of PE
PE 0.2%
Anticoagulation Consider anticoagulation and Therapeutic anticoagulation
not indicated repeat imaging

CTPA = CT pulmonary angiogram; V/Q scan = ventilation-perfusion scan

8
Direct Thrombin and 
Anticoagulation Options in Pregnancy
Factor Xa Inhibitors
• Low molecular weight heparin (LMWH) is the treatment
of choice for PE  • Limited evidence in pregnancy
• Unfractionated heparin (UFH) is an alternative if LMWH • Category B and C, but not recommended until more
is not available or is contraindicated evidence can be obtained
• Warfarin
– Contraindicated during pregnancy – crosses the placenta; risk of • May be a good alternative in cases of severe heparin
embryopathy, miscarriage, and stillbirth allergy or heparin induced thrombocytopenia
– May be used postpartum
• Both heparin and warfarin are safe with breastfeeding

Initial Treatment for DVT and PE VTE Prophylaxis
• LMWH • LMWH is the drug of choice
– Enoxaparin 1 mg/kg SQ every 12 hours • Antepartum prophylaxis is indicated for
– Dalteparin 100 units/kg SQ every 12 hours or 200 units/kg
– history of DVT/PE 
SQ every 24 hours
– Tinzaparin 175 units/kg SQ every 24 hours – some thrombophilias
• UFH • When to start and stop prophylaxis is dependent
– 10,000 or more IU SQ every 12 hours upon clinical situation
– Adjust dose to aPTT 1.5× to 2× control (60 to 80 seconds) – usually continued for 6 weeks postpartum

Postcesarean DVT Prophylaxis Precautions with Anticoagulation


• Sequential compression devices (SCD’s) for all cesarean  • Discontinue heparin 
deliveries – 24 hours prior to scheduled inductions and cesarean deliveries
• Consider addition of low molecular weight heparin based on  – At the onset of labor
risk factors: • Epidural 
• Obesity
– With therapeutic LMWH, avoid epidural for 24 hours after last 
• Age dose 
• Parity
– With prophylactic once‐daily LMWH, avoid epidural for 10 to 12 
• Prolonged labor hours after last dose
• Preeclampsia
– With unfractionated heparins, obtaining an aPTT can help guide 
• Chorioamnionitis decision for safe epidural placement

9
Summary
• Multiple medical challenges can evolve during 
pregnancy
• Key to diagnosis is clinical vigilance, and 
obtaining appropriate laboratory or imaging 
studies
• Balancing maternal and fetal well‐being can be 
challenging clinically
• Consultation is of value in difficult cases

10
Chapter C
Vaginal Bleeding in Late Pregnancy

Jennifer Frank, MD, Zachary Baeseman, MD, Larry Leeman, MD, MPH
Revised January 2017

Learning Objectives
At the end of this activity, learners will be able to:
1. Identify serious causes of vaginal bleeding in the second half of pregnancy.
2. Describe a systematic approach to identify the cause of bleeding.
3. Describe specific treatment options based on diagnosis.

Introduction placental abruption, uterine scar disruption (uterine


A 22-year-old multiparous woman presents to labor rupture), and vasa previa. Non-emergent causes of
and delivery at 28 weeks’ gestation after having an epi- bleeding include cervical dilatation during normal labor,
sode of vaginal bleeding. Initially, she noticed a small which is commonly accompanied by a small amount
amount of blood-tinged mucus, but 1 hour before of blood or blood-tinged mucus (bloody show). Many
admission, she experienced a gush of bright red blood. pregnant women experience spotting or minor bleeding
Her first delivery was a cesarean delivery for failure to after sexual intercourse or a digital vaginal examination.
progress at term and her second was an elective repeat Cervicitis, cervical ectropion, cervical polyps, and cervi-
cesarean delivery at 38 weeks’ gestation. cal cancer are other possible causes of minor bleeding.
A 30 year-old nulliparous woman is undergoing oxyto- The history, physical examination, ultrasonography
cin augmentation at 36 weeks’ gestation for preeclampsia for placental location, and a brief period of observa-
with severe features. Her blood pressure has been as high tion typically differentiate minor from serious causes
as 170/110 mm Hg, and she is receiving an intravenous of vaginal bleeding. Evaluation with a sterile speculum
(IV) infusion of magnesium sulfate. Her cervix has been examination may be performed safely before ultra-
dilated 6 cm for 1 hour, and she has begun to report sonographic evaluation; however, digital examina-
pain between contractions. A large amount of vaginal tion should not be performed until ultrasonography
bleeding follows her next cervical check. excludes a placenta previa.2
Both of these cases raise important questions. What
is the cause of the vaginal bleeding? Is it life-threatening Management of Antepartum Hemorrhage
for the mother and/or infant? What steps should be Major causes of vaginal bleeding in pregnancy and their
taken for diagnosis and management? risk factors are listed in Table 1. The initial manage-
Vaginal bleeding after mid-pregnancy is associated with ment of significant bleeding in late pregnancy is similar
maternal and fetal risks. In addition to maternal morbidity regardless of the etiology and is focused on assessing
secondary to acute hemorrhage and operative delivery, the maternal and fetal well-being. Assessment of vital signs
fetus may be compromised by uteroplacental insufficiency, and circulatory stability are the first steps along with
premature birth, and perinatal mortality.1,2 Optimal man- establishing IV access and beginning prompt fluid
agement of late pregnancy bleeding depends on accurate resuscitation, as indicated. Mild tachycardia is normal
identification of the cause and timely intervention. in pregnancy, but orthostatic changes in blood pressure
or pulse may indicate hypovolemia. Signs and symp-
Causes of Late Pregnancy Bleeding toms of shock are late findings in pregnant women,
The four conditions that account for most cases of seri- and the earliest signs of hypovolemic shock may not
ous or life-threatening hemorrhage are placenta previa, appear until there has been greater than 1 L of maternal

Copyright 2017© American Academy of Family Physicians. All rights reserved. — Chapter C  1
Chapter C

blood loss. Hypotension, tachycardia, and mater- All patients with bleeding should at least have a
nal symptoms of hemodynamic instability are complete blood count, and blood type and anti-
ominous indicators, and women with these signs body screen. With major hemorrhage, additional
require immediate fluid resuscitation, activation of testing should include a blood type and cross,
a massive transfusion protocol, and preparation for coagulation studies, blood urea nitrogen, creati-
a potential urgent cesarean delivery.3 nine, and liver function studies.2 The interpreta-
The history should guide a physical examination tion of laboratory results requires understanding
and survey for trauma. Examination of the mater- of how laboratory values may differ in pregnancy.
nal abdomen should include assessment of fundal Initial laboratory testing may not indicate a
height, estimated fetal weight, fetal presentation, coagulopathy; therefore testing should be repeated
location of tenderness (if present), and palpation if clinical suspicion remains. Thrombocytopenia
for uterine contractions. Visual estimates of blood is the most common laboratory abnormality in
loss should be recorded but may be inaccurate or disseminated intravascular coagulation (DIC),
fail to account for concealed hemorrhage. but a low platelet count can be confused with
gestational thrombocytopenia.4 Fibrinogen levels
are higher in pregnancy, often above 400 mg/
Table 1. Risk Factors for Major Causes of Vaginal dL. Therefore a normal result in a nonpregnant
Bleeding in Late Pregnancy patient of 250 to 350 mg/dL may be abnormal
in pregnancy; fibrinogen levels less than 200 mg/
Placenta Previa Uterine Rupture dL may indicate DIC.5 Normal plasma thrombo-
Advanced maternal age (>40 years) Abnormal placentation plastin and partial thromboplastin time values are
Chronic hypertension History of uterine surgery shorter in pregnancy. D-dimer is typically elevated
Multiparity Labor induction (especially in pregnancy, which reduces its clinical relevance,
Multiple gestations prostaglandins) but D-dimer can be monitored serially to detect
Previous cesarean delivery Maternal connective tissue
disease
a rising value. The laboratory findings consistent
Tobacco use with a diagnosis of DIC in order of importance
Uterine curettage Non-European ethnic descent
Trauma are platelets (decreasing value), PT (prolongation),
Previous uterine surgery, including
uterine curettage TOLAC fibrin-related marker (increasing), and fibrinogen
Cocaine use Uterine anomalies (decreasing).4 If coagulation studies are not readily
History of placenta previa available, a serum sample may be drawn and taped
Vasa Previa
Chronic hypertension In vitro fertilization
to the wall for a simple and inexpensive clot test. If
In vitro fertilization Low-lying and second trimester no clot or a poor quality clot is present after 7 to
Placental Abruption placenta previa 10 minutes, then coagulopathy is present. Women
Chronic hypertension Multiple gestation who are Rh negative should receive Rho (D)
Multiparity Succenturiate-lobed and immune globulin; a Kleihauer-Betke test can be
bilobed placenta performed to determine the appropriate dose.5,6
Preeclampsia
Velamentous cord insertion
Previous abruption Continuous fetal monitoring is recommended
Short umbilical cord to determine if there is a fetal indication for
Sudden decompression of an urgent operative delivery.7 Fetal heart rate (FHR)
overdistended uterus decelerations, tachycardia, or loss of variability
Thrombophilias
may resolve with adequate maternal resuscitation.
Tobacco, cocaine, or
methamphetamine use However, a persistently concerning FHR tracing
Trauma: blunt abdominal or sudden (Category III or Category II without variability)
deceleration may require delivery before the etiology of the
Unexplained elevated maternal hemorrhage is established.
alpha fetoprotein level
Uterine fibroids Placenta Previa
Definitions and Pathophysiology
TOLAC = Trial of labor after cesarean delivery.
Adapted from Sakornbut E, Leeman L, Fontaine P. Late pregnancy bleeding.
Placenta previa occurs when the placenta implants
Am Fam Physician. 2007;75:119-206. in a location overlying or in close proximity to
the internal cervical os.8 The pathophysiology of

2  Chapter C —
Vaginal Bleeding in Late Pregnancy

placenta previa is not fully understood. Normally, deliveries are associated with increasing risk of
placental implantation favors a fundal location. As placenta previa.17
pregnancy progresses, the apparent migration of
the placenta away from the lower uterine segment Morbidity
is caused by the growth of placental trophoblasts Maternal morbidity associated with placenta pre-
toward the fundus (with its richer blood supply) via can result from maternal hemorrhage, cesar-
and by the development or elongation of the lower ean delivery, or abnormal placental attachment,
uterine segment. Abnormal implantation of the specifically placenta accreta, increta, or percreta.
placenta may occur when there is disruption or Complete placenta previa is associated with higher
scarring of the uterine cavity, most commonly as a morbidity than low-lying or marginal placenta
result of a previous cesarean delivery. previa.18,19
Transvaginal ultrasonography allows precise Women who have had a prior cesarean delivery
assessment of the distance between the internal os and have a placenta previa or low anterior placenta
and the placental edge. The placenta is described in a subsequent pregnancy are at increased risk
as a complete previa when it covers the internal os of abnormal placental attachment.2,20 The risk
and as a marginal previa when the edge lies within of placenta accreta, increta, or percreta reaches
2 cm of the os. When the edge is 2 to 3.5 cm from approximately 10% when placenta previa coex-
the os, the placenta may be described as low-lying ists with previous cesarean delivery.21,22 The risk
(Figure 1).9 of abnormal placental attachment also increases
with the number of previous cesarean deliveries.20
Epidemiology Women with a history of cesarean delivery who
Placenta previa is a common incidental finding present with placenta previa or a placenta located at
on second trimester ultrasonography, present on the site of the previous incision should be evaluated
approximately 4% of ultrasound studies per- for potential placenta accreta with color flow Dop-
formed at 20 to 25 weeks’ gestation but only 0.5% pler ultrasonography by an experienced sonogra-
of pregnancies at term.10 The likelihood of a previa pher.23,24 Magnetic resonance imaging of the pelvis
persisting until term increases if the previa is may help confirm the diagnosis of an invasive pla-
complete, if it is present at a later gestational age, centa and delineate organ involvement in women
or if there is a history of cesarean delivery.11,12 The with a placenta percreta.20 A suspected placenta
extent to which the placenta overlaps the internal accreta necessitates preparation for possible cesar-
os at 18 to 23 weeks is highly predictive for the ean hysterectomy, including appropriate surgical
persistence of placenta previa.13,14 If the overlap expertise and availability of blood products.
is less than 1.5 cm at 18 to 23 weeks, placenta Perinatal morbidity and mortality associated
previa typically resolves.14 If the overlap is 2.5 cm with placenta previa are mainly related to the com-
or greater at 20 to 23 weeks, persistence to term
is likely.13 When a placenta previa or low-lying
placenta is identified during the second trimester, Figure 1. Abnormal Placental Implantation
ultrasound should be repeated at approximately
32 weeks to determine if it has regressed. Consider
a repeat scan at 36 weeks for patients where the
previa has persisted to determine the optimal route
and timing of delivery.10 Routine late pregnancy
ultrasound in low-risk or unselected populations
does not confer benefit to the mother or infant
and may increase cesarean delivery rates.15
Risk factors associated with placenta previa
include chronic hypertension, multiparity, mul-
tiple gestations, increasing maternal age, previous
cesarean delivery, cocaine use, tobacco use, uter- Low-Lying Marginal Complete
ine curettage, previous placenta previa, and male 2 to 3.5 cm from os Within 2 cm of os Covers os
fetal sex.2,10,12,16 Increasing numbers of cesarean

— Chapter C  3
Chapter C

plications of prematurity because the blood loss a speculum to view the vaginal vault and cervix
comes from the maternal circulation.25 Therefore should not result in disruption of a placenta previa.
the management of placenta previa and timing of
delivery is influenced by gestational age and fetal Management
lung maturity balanced against the degree of hem- A Cochrane review showed few randomized trials
orrhage and urgency of the maternal condition. A of interventions for placenta previa.26 Outpatient
cohort study of American women with placenta management of placenta previa in stable women
previa showed that greater than half delivered at who do not have active bleeding can be considered,
term (more than 37 weeks’ gestation), approxi- but the data is limited to make a firm recommen-
mately 28% delivered between 34 and 37 weeks’ dation.2 If selected, outpatient management carries
gestation, and approximately 17% delivered before the important criteria of close proximity to the
34 weeks’ gestation.10 hospital of delivery during the third trimester and
having someone available to assist in the event of
Clinical Presentation bleeding or onset of labor. Women with asymp-
Symptomatic placenta previa typically manifests tomatic previa in the second trimester can continue
as vaginal bleeding in the late second or third tri- normal activities until follow-up ultrasonography is
mester, often after sexual intercourse. The bleed- performed. Women with persistent placenta previa
ing is painless unless labor or placental abruption in the third trimester should report any bleeding
occurs. A large central previa will typically present and abstain from intercourse and use of tampons.
with bleeding at 26 to 28 weeks’ gestation—the When bleeding occurs, women with placenta pre-
so-called sentinel bleed. This initial sentinel bleed via should be evaluated in the hospital.2
typically is not sufficient to produce hemodynamic Because most neonatal morbidity and mortal-
instability or to threaten the fetus in the absence of ity associated with placenta previa results from
cervical instrumentation or digital examination. complications of prematurity, the main thera-
peutic strategy is to prolong pregnancy until
Diagnosis fetal lung maturity is achieved. Tocolytic agents
Placenta previa should be suspected in patients may be used safely to prolong gestation if vagi-
who have a persistent malpresentation. A cephalic nal bleeding occurs with preterm contractions.
presentation may be impossible because of the One prospective study showed that the use of
presence of a large placenta filling the pelvis. tocolytics in women with symptomatic placenta
Regardless of previous imaging results, vaginal previa was associated with prolonged gestation
bleeding (particularly if it is painless or provoked and increased birth weight without increased
by intercourse) in the setting of a high presenting risk of maternal hemorrhage or fetal complica-
part or abnormal lie should prompt clinical suspi- tions.27 Corticosteroids should be administered to
cion for placenta previa.2 women with bleeding from the placenta previa at
The diagnosis of placenta previa is confirmed by 24 to 34 weeks’ estimated gestation to promote
ultrasound localization of the placenta. When fetal lung maturity.28 A recent randomized trial
placenta previa is suspected on transabdominal showed benefit of antenatal steroid administra-
ultrasonography, transvaginal ultrasonography tion between 34 and 36 5/7 weeks gestational age
should be performed.2 Transvaginal ultrasonogra- in reducing the risk of respiratory complications
phy is safe and more accurate than transabdominal compared with no treatment.29
ultrasonography for localizing the placental edge Cervical cerclage has been proposed as a
and the internal os. Transvaginal ultrasound iden- means of prolonging pregnancies complicated
tifies that 26% to 60% of placentas diagnosed as by placenta previa because bleeding occurs in
low-lying on transabdominal scan are not actually late pregnancy as the placenta is sheared from
low-lying and do not require further monitoring, a lengthening lower uterine segment and dilat-
and the addition of transvaginal ultrasonography ing cervix.30 Although a Cochrane meta-analysis
clarifies the diagnosis in 26% of cases.2 showed that cerclage decreased the risk of pre-
Preoperative ultrasound can give information mature birth before 34 weeks, it is recommended
on fetal lie and placental location to determine the that additional studies of cerclage be performed
location of uterine incision.10 Gentle insertion of before this clinical practice is introduced.2,26

4  Chapter C —
Vaginal Bleeding in Late Pregnancy

The need for emergency cesarean delivery is Abruption is the most common cause of serious
more common in women with three or more epi- vaginal bleeding, occurring in 1% of pregnan-
sodes of antepartum bleeding and first episode of cies.38 The incidence of abruption increased
bleeding before 29 weeks’ gestation.31 In women between 1979 and 2001, possibly as a result of
with a complete placenta previa, cervical length rising rates of hypertension, increased stimulant
measured on transvaginal ultrasound may pre- abuse, and increased surveillance bias by diagnosis
dict risk of preterm emergency cesarean delivery with ultrasonography.39
secondary to massive hemorrhage.32 For women Risk factors associated with abruption include
with a marginal previa, it is recommended that abdominal trauma; cocaine, amphetamine, or
decision on mode of delivery be deferred until tobacco use; chronic hypertension; preeclampsia;
ultrasonography is performed at 35 to 36 weeks.2 thrombophilias; chorioamnionitis; oligohydram-
Women whose placental edge is 2 cm or more nios; iron deficiency anemia; premature rupture
from the internal os at term can expect to deliver of membranes; and abruption in a previous
vaginally unless heavy bleeding ensues.9 Women pregnancy.38,40-43
whose placenta is located 1 to 2 cm from the os
may attempt vaginal delivery in a facility capable Pathophysiology
of moving rapidly to cesarean delivery if neces- Placental abruption can result from several dif-
sary, with the majority of women achieving vagi- ferent pathophysiological processes. In some
nal delivery without increased hemorrhage.8,9,33 cases, abnormalities in placental development and
The optimal time for delivery for a stable patient implantation that start in the first trimester lead
with placenta previa is not firmly established, to specific pathologic changes that in turn lead to
but delivery between 36 and 37 weeks has been abruption.38 With blunt trauma to the abdomen,
recommended to maximize maternal and neona- shearing of the uterine-placenta interface leads to
tal outcomes.34-36 Amniocentesis for assessment placental detachment and hemorrhage that can be
of fetal lung maturity is not recommended for overt or retroplacental.44 In one large retrospective
determining the optimal timing of delivery.36 In a study of all injured pregnant women at Level I and
retrospective study evaluating the optimal timing Level II trauma centers, 84% experienced blunt
of delivery, infants born at 35 to 37 weeks were trauma and 16% had penetrating injuries. Placen-
no more likely to have fetal anemia, fetal distress, tal abruption was the most common complication,
neonatal seizures, increased ventilator needs, or occurring in 3.5% of injured pregnant women and
infant mortality compared with infants born at resulting in a rate of intrauterine demise greater
38 weeks. Infants born at 35 to 36 weeks were at than 50%.45 Other etiologies of abruption include
increased risk of 5-minute Apgar scores less than 7 vasoconstriction associated with cocaine use and
and neonatal intensive care unit admission.37 sudden uterine decompression after rupture of
Indications for operative delivery include the membranes or delivery of a first twin.38,46
presence of persistent, brisk vaginal bleeding,
which poses a threat to the stability of the mater- Prevention
nal-fetal dyad, or any vaginal bleeding in a preg- The incidence of placental abruption may be
nancy where the fetus is sufficiently mature to be decreased by cessation of tobacco, cocaine, or
delivered with safety. General anesthesia has been amphetamine use and appropriate care for hyper-
associated with increased intraoperative blood tensive disorders of pregnancy.38 In patients with
loss and need for blood transfusion. Regional preeclampsia, treatment with magnesium sulfate is
anesthesia appears to be a safe alternative, but it associated with reduced risk of placental abruption
may need to be converted to general anesthesia if (relative risk 0.64; 95% confidence interval [CI] =
surgery is prolonged.24 0.5 to 0.83).47
Pregnant women involved in severe motor vehi-
Placental Abruption cle accidents have an increased risk of abruption;
Epidemiology proper restraints are frequently not used because of
Placental abruption is the separation of the discomfort.48 Appropriate use of seat belts during
placenta from the uterine wall before delivery. It pregnancy should be routinely encouraged during
can be partial or complete and can vary in degree. prenatal care.

— Chapter C  5
Chapter C

Clinical Presentation may be of assistance in planning care. Ultrasound


Placental abruption typically manifests as vaginal findings, if present, may include a retroplacental
bleeding associated with abdominal pain, which echolucency, abnormal thickening of the placenta,
may vary from mild cramping to severe pain. or an abnormally round torn up edge of the pla-
Women with posterior placental abruption may centa. Computerized tomography (CT) is also
report back pain rather than abdominal pain, and able to identify placental abruption, and pregnant
patients with abruption because of trauma may patients undergoing CT evaluation after trauma
experience pain without bleeding (concealed hem- should have careful evaluation of the placenta.55
orrhage). Concealed placental abruption has been
shown to occur in 20% to 63% of abruptions.49-52 Management
Although the rate of concealed hemorrhage varies Because the unpredictable nature of abruption
widely and has not been studied in large numbers does not allow for controlled trials, management
of women, it is important for the clinician to be remains empiric. A Cochrane review found no
aware of a possible significant abruption even in randomized controlled trials assessing interven-
the absence of vaginal bleeding.53 tions for placental abruption that met inclusion
The history should include questions regarding criteria.56 However, a recent large cohort study
trauma (including domestic violence), presence of has indicated the risk of placental abruption in a
pain and contractions, rupture of membranes, and subsequent pregnancy in women with a history of
assessment of risk factors, including a history of abruption in their first pregnancy is significantly
hypertension (or symptoms of preeclampsia) and increased (adjusted odds ratio [OR] = 93). They
stimulant (cocaine, amphetamines) use. propose medical induction at 37 weeks in women
The bleeding may be bright, dark, or inter- with a history of placental abruption.41
mixed with amniotic fluid. Blood from concealed
hemorrhage is typically dark, having been seques- Mild Abruption
tered behind the membranes. The amount of A stable patient with a small partial abruption
vaginal bleeding is not indicative of the severity and a preterm fetus may be managed successfully
of abruption.38 It may be difficult to determine in a conservative manner. Tocolysis is typically
whether bleeding represents exuberant bloody contraindicated, except in mild abruption before
show or abruption. If bleeding is noted at the 34 weeks’ gestation when it may be used to allow
time of rupture of membranes, vasa previa should administration of corticosteroids.57 It is impor-
also be considered. tant to note that IV magnesium sulfate may be
indicated before 32 weeks’ gestation, but not as
Physical Examination and Diagnostic Testing a tocolytic, but rather for fetal neuroprotection
Fetal heart tones and uterine activity should be to reduce the incidence of cerebral palsy during
documented by continuous monitoring.7 Tetanic premature delivery.
contractions may be present and, if measured by Women experiencing recurrent bleeding attrib-
an intrauterine pressure catheter, are typically uted to placental separation may be diagnosed as
recorded as a high resting tone with superimposed having a chronic abruption. Management will be
small frequent contractions. The presence of based on the degree of bleeding and gestational
this finding is significant because it will often be age. When expectant management occurs in the
accompanied by a concerning (Category II or III) setting of chronic abruption, serial ultrasonogra-
fetal tracing. phy for fetal growth and antepartum surveillance
Ultrasound examination may show retroplacental are indicated in the third trimester because of the
clot or hemorrhage. However, acute blood clots potential for uteroplacental insufficiency.39
and the placenta are hyperechoic on ultrasonog-
raphy and can be difficult to distinguish from one Severe Abruption
another.54 The diagnosis of abruption is largely a clini- Initial management includes rapid stabilization
cal diagnosis and urgent management should never be of maternal cardiopulmonary status and assess-
delayed for ultrasound confirmation. If the mother ment of fetal well-being. Delay can be fatal to the
and fetus are stable, the placental location and fetus; 30% of perinatal mortalities in one case
appearance and fetal lie and fetal weight estimation series occurred within 2 hours of admission.58

6  Chapter C —
Vaginal Bleeding in Late Pregnancy

In a study of placental abruption in the United efficacy. In addition, cryoprecipitate or factor VIII
States, perinatal mortality was 119/1,000 births in may be of specific benefit in severe coagulopathy.
pregnancies complicated by placental abruption Consideration of maternal transfer from a rural
in comparison with 8.2/1,000 births among other site is based on many factors. Patients present-
births; the majority of perinatal mortality was ing with abruption and a live fetus are typically
associated with preterm delivery.59 not stable for transfer because operative delivery
Maternal stabilization requires monitoring of may be needed on an immediate basis at any time
vital signs and urine output along with serial eval- during labor. In this instance, neonatal transfer
uation of the hematocrit and coagulation studies (rather than maternal-fetal) may be a necessary
to determine whether disseminated intravascular intervention for the premature or sick newborn.
coagulation is present.40 The circulatory status of If fetal demise has occurred, a patient who does
the patient with abruption should be maintained not have coagulopathy and is hemodynamically
to permit a margin of reserve. Hourly urine output stable may be cared for with appropriate resources.
should be maintained at 30 mL/hour or greater. Blood bank supply may determine whether or not
Hematocrit should be maintained above 30%. In a patient needs transport to a referral facility.
patients with preeclampsia or other confounding Fetomaternal hemorrhage may occur with rup-
factors, central blood pressure monitoring may ture of fetal vessels in the placenta. The Kleihauer-
assist in the fluid management. Betke test is useful to determine dosage of Rho
A concerning FHR tracing (eg, persistent (D) immune globulin in Rh-negative patients, but
Category III or Category II without variability) is not useful for the diagnosis of abruption.63,64
in the setting of placental abruption necessitates
rapid, typically cesarean, delivery.60,61 A decision- Uterine Rupture
to-delivery interval of 20 minutes or less resulted Epidemiology and Pathophysiology
in improved neonatal outcomes in a case-control Uterine scar disruption spans a spectrum from
study of severe abruption.60 Occasionally, abrup- occult dehiscence discovered at repeat cesarean
tion occurs during the second stage and an opera- delivery to complete uterine rupture requiring
tive vaginal delivery may be attempted. Neonatal emergency laparotomy. In uncommon instances,
resuscitation personnel should be available for all uterine rupture can be spontaneous and occur in
deliveries, vaginal or operative. the absence of risk factors. In complete rupture,
When fetal mortality occurs secondary to the fetus or placenta may be partially or com-
abruption, vaginal delivery should be the goal.62 pletely extruded from the uterus. This chapter
Labor should be permitted as long as adequate focuses only on uterine rupture presenting with
progress is made and maternal status can be sup- third trimester bleeding.
ported. Although labor is often hypertonic with Spontaneous uterine rupture is found in only
abruption, it may also be hypotonic. Oxytocin 0.03% to 0.08% of all delivering women, but in
augmentation is not contraindicated, but should approximately 0.8% of women with a uterine scar
be used judiciously with intrauterine pressure from prior surgery.65 Previous cesarean incision
monitoring. Indications for operative delivery is the most common etiology for uterine rupture.
with fetal demise include other maternal indica- Classic or T-shaped uterine incisions are associ-
tions for cesarean delivery, failure of labor pro- ated with a higher likelihood of uterine rupture
gression, and brisk hemorrhage that cannot be compared with a low transverse incision.66 Two or
compensated for by transfusion. more previous cesarean deliveries and labor induc-
Approximately one-third of patients with tion each increase the incidence of uterine rupture
placental abruption with fetal demise will develop from approximately 0.9% to 1.8% and 0.77% to
coagulopathy. Coagulopathy is typically not seen 2.24% respectively.65 Although uncommon, there
in the patient presenting with abruption and a is a concomitant rise in the incidence of uterine
live fetus. Coagulopathy occurring with abrup- rupture with increasing rates of cesarean deliver-
tion may be related to two etiologies: consumptive ies.67 Other causes include inappropriate oxytocin
coagulopathy and DIC. Replacement of platelets usage and trauma.66
and fresh-frozen plasma should be administered Conditions that predispose to uterine scar
just before operative delivery to provide maximum disruption include previous uterine surgery

— Chapter C  7
Chapter C

(eg, myomectomy) that involves the full thickness scar should be advised to come to the hospital for
of the myometrium, congenital uterine anomaly, evaluation of new onset of contractions, abdomi-
uterine overdistension, intra-amniotic installa- nal pain, or vaginal bleeding as soon as possible.73
tion, gestational trophoblastic neoplasia, previous Spontaneous antepartum rupture in nonlaboring
uterine rupture, maternal obesity, and adenomyo- women is extremely rare and is typically associ-
sis.66,68 Other risk factors include labor induction ated with identifiable risk factors. In a case series of
(OR 12.60; 95% CI = 4.4 to 36.4), labor induc- women experiencing spontaneous uterine rupture
tion with prostaglandins (OR 2.72; 95% CI = 1.6 in the second or third trimester, six of seven events
to 4.7), and non-European ethnicity (OR 2.87; (during 13 years) involved placenta previa or per-
95% CI = 1.8 to 4.7).66 Conditions present during creta and five of the seven uterine ruptures occurred
delivery that predispose to uterine rupture include in women with prior cesarean deliveries.76 These
fetal anomaly, vigorous uterine pressure, difficult findings suggest that prior uterine scar and abnor-
manual removal of the placenta, or abnormalities mal placenta both play a role in uterine rupture.77
of placental implantation.68
The most common maternal morbidity asso- Management
ciated with uterine rupture is hemorrhage and In the case of a sudden change in fetal baseline
subsequent anemia, requiring blood transfusion.69 or the onset of repetitive FHR decelerations, the
Other morbidities include bladder injury (8.8%) clinician should institute intrauterine resuscitation
and hysterectomy, which accompanies 14% with maternal position change, IV fluids, discon-
to 33% of uterine ruptures.70,71 Although rare, tinuation of oxytocin, oxygen administration,
maternal mortality from DIC and sepsis has been and consideration for subcutaneous terbutaline.
reported.69 The incidence of perinatal mortal- If these measures are not effective, then emergent
ity associated with uterine rupture is variably cesarean or operative vaginal delivery may be
reported, ranging from 0% to 60%.72 Fetal and indicated. Asymptomatic scar disruption may be
maternal morbidity are higher in cases of uterine found at the time of cesarean delivery or palpation
rupture of an unscarred uterus.70 of the uterine cavity after vaginal delivery. Routine
inspection of the uterine scar after vaginal deliv-
Clinical Presentation ery is not recommended and asymptomatic scar
The classic presentation for symptomatic, sig- disruption is expectantly managed.
nificant uterine rupture includes vaginal bleed-
ing, pain, cessation of contractions, absence of Vasa Previa
fetal heart tones, loss of station, easily palpable Vasa previa occurs when fetal blood vessels,
fetal parts through the maternal abdomen, and unprotected by the umbilical cord or placenta,
profound maternal tachycardia and hypotension. run through the membranes and across or within
However, most cases (67% to 70%) of uterine 2 cm of the cervix. Although uncommon, with an
rupture initially present with abnormal fetal incidence of 1/2,500 deliveries, it is important to
monitoring.65,70 In a review of 159,456 deliveries, be familiar with vasa previa because rapid inter-
the most frequent finding associated with uterine vention is essential for fetal survival.10,78
rupture was a sudden deterioration of the FHR
pattern.73 In one case-control study, markedly Epidemiology and Pathophysiology
abnormal FHR tracings starting 1 hour before This uncommon cause of severe obstetric hemor-
birth were significantly associated with uterine rhage typically occurs in pregnancies with a low-
rupture in patients undergoing a trial of labor after lying placenta and velamentous insertion, bipartite
cesarean delivery.74 There may be a progression of placenta, or a placenta with a succenturiate lobe.79
signs from nonspecific, severe variable decelera- During rupture of the membranes, the fetal vessels
tions to the characteristic recession of the fetal are at risk of rupture, which can lead to significant
head or suprapubic bulging. Contractions may fetal blood loss. Historical studies showed a 33%
show a stair-step appearance of gradually decreas- to 100% rate of perinatal mortality secondary to
ing amplitude on tocodynamometry.75 vasa previa,80 but antenatal diagnosis is associated
Thirteen percent of uterine ruptures occur out- with a reduction in the rate of neonatal morbid-
side of the hospital. Patients with a prior uterine ity and mortality.79 Risk factors for vasa previa

8  Chapter C —
Vaginal Bleeding in Late Pregnancy

include in vitro fertilization, multiple gestation, amount of tap water to lyse red blood cells. After
placenta previa, and bilobed or succenturiate- 5 minutes of centrifugation, the supernatant is
lobed placentas.80,81 removed and mixed with 1 mL of 1% sodium
hydroxide (NaOH) for every 5 mL of supernatant.
Clinical Presentation and Diagnosis A pink color indicates fetal hemoglobin. Adult
The goal of diagnosis is antenatal detection and hemoglobin is brown. Additionally, a Wright stain
delivery before membrane rupture.82 Prenatal diag- of blood collected from the vagina can be evalu-
nosis using ultrasound with color flow Doppler ated for the presence of nucleated red blood cells,
significantly affects fetal survival. Prenatal diagno- which are common in fetal blood but uncommon
sis is associated with a 98% survival rate compared in adult blood. This test can be performed without
with a 44% survival rate with intrapartum or post- delay, assuming a stable FHR.
partum diagnosis.78 Because of increased prenatal
diagnosis, contemporary case series show perinatal Management
mortality rates less than 10%.78 Prenatal ultra- In a woman with antenatally detected vasa pre-
sound has a 93% detection rate and a specificity of via who presents with premature rupture of fetal
99% with an optimal detection window between membranes or labor, cesarean delivery should be
18 and 26 weeks’ gestation; detection is less likely performed.78 Delivery should not be deferred for
on ultrasound performed in the third trimester.78 confirmation of fetal blood in women with severe
Clinically, vasa previa is suspected when vaginal hemorrhage or when fetal heart tones are concern-
bleeding occurs with membrane rupture, clas- ing. If the onset of vaginal bleeding occurred with
sically in the setting of a FHR tracing showing rupture of membranes and the FHR is concerning,
bradycardia or a sinusoidal pattern.78 cesarean delivery should be performed immedi-
The hemorrhage is fetal blood, and exsanguina- ately. Because fetal exsanguination is the cause of
tion can occur rapidly because the average blood neonatal mortality in this disorder, preparation for
volume of a term fetus is approximately 250 mL.83 resuscitation at the delivery includes availability
Transabdominal combined with transvaginal of normal saline for a 10 to 20 mL/kg bolus to
ultrasound may improve diagnostic accuracy. administer if the newborn is in shock.
Power Doppler and three-dimensional ultraso- In the presence of antenatal diagnosis of vasa
nography are reported to assist with diagnosis, but previa, serial ultrasounds are recommended to
superiority to two-dimensional ultrasound has not evaluate for regression of vessels, which can occur
been shown.10 In addition to diagnosis by ultra- in approximately 20% of women.78 With per-
sound or classic clinical presentation, vasa previa sistent vasa previa, antenatal steroids should be
may be diagnosed by MRI, and intrapartum iden- administered between 28 and 32 weeks’ gesta-
tification of fetal blood intermixed with vaginal tion. Hospitalization at 30 to 34 weeks should
blood.79 Rarely, vessels are palpated in the pre- be considered, allowing for closer observation for
senting membranes, prohibiting artificial rupture signs of labor onset and proximity to operative
and vaginal delivery. FHR tracing may show fetal delivery if membranes rupture. However, data to
tachycardia or a sinusoidal pattern.10 support this approach compared with outpatient
If fetal heart tones are not concerning, a blood management is lacking.78 Outpatient manage-
sample from the vaginal vault may be obtained ment can be considered for asymptomatic women
to check for fetal blood cells or fetal hemoglobin. with no uterine activity and a long closed cervix
The Kleihauer-Betke test and hemoglobin electro- on transvaginal ultrasound.82 More than half of
phoresis measuring the presence of fetal hemoglo- women managed as outpatients will eventually
bin are sensitive but are too slow to use clinically.79 require hospitalization for a complication.78 The
The Apt test (alkali denaturation test) is faster to optimal gestational age for delivery is not deter-
perform but is insensitive.79 The Apt test is based mined, but cesarean delivery between 34 and
on the resistance of fetal hemoglobin to denatur- 36 weeks has been recommended to balance the
ation by alkaline agents and can be performed risk of respiratory distress syndrome with risk of
on the labor and delivery unit.84 Performance of membrane rupture and fetal exsanguination.10,78,82
the Apt test involves obtaining a sample of blood Amniocentesis to assess for fetal lung maturity is
from the vagina and then mixing it with a small not indicated because timing of delivery would

— Chapter C  9
Chapter C

not be influenced by the result.78 Delaying deliv- recommended for women who had a placenta pre-
ery beyond 37 weeks is not recommended. via or low-lying placenta during a second trimester
ultrasound with subsequent resolution.78
Prevention
There are no strategies for primary prevention of Summary
vasa previa. However, hemorrhage theoretically is Vaginal bleeding in late pregnancy may occur
preventable with antenatal screening for women at because of potentially life-threatening conditions
high risk and cesarean delivery at 34 to 36 weeks for mother and newborn. Clinicians must be able to
when vasa previa is present. Screening is carried out distinguish emergent causes of bleeding from those
with transvaginal color flow Doppler to identify the that are less urgent and be prepared to act quickly
presence of vessels in the fetal membranes. Screen- and decisively with severe maternal hemorrhage or
ing in a general population has not been recom- suspected vasa previa. Vaginal examination should
mended because the condition is rare (one diagnosis be avoided until placental location is known. The
per 5,215 screenings).85 Screening is recommended timely diagnosis of vaginal bleeding in late preg-
for women at increased risk, which includes women nancy, including antenatal diagnosis with color flow
after the detection of a low-lying, bilobed, multi- ultrasound, can reduce perinatal mortality. This
lobed, or succenturiate-lobed placenta, in women chapter identifies major causes of vaginal bleeding
with velamentous cord insertion on a routine ultra- in the second half of pregnancy, describes a system-
sound, or women who are pregnant after in vitro atic approach to identifying the cause of bleeding,
fertilization.10,79 Careful evaluation of the placenta and identifies the appropriate management of the
including site of cord insertion on routine ultra- woman with late vaginal bleeding. Institutional
sound may facilitate identifying women at greater policies should be in place to ensure adequate blood
risk of vasa previa. A transvaginal ultrasound with bank response to massive hemorrhage and to mobi-
color and pulsed Doppler at 32 weeks has been lize resources for emergent cesarean delivery.

10  Chapter C —
Vaginal Bleeding in Late Pregnancy

SORT: Key Recommendations for Practice


Evidence
Clinical Recommendation Rating References

Placenta previa is a common incidental finding on second trimester ultrasonography and should A 10,11
be confirmed in the third trimester.
Corticosteroids should be administered to women with bleeding from placenta previa at 24 to A 28
34 weeks’ gestation.
Treatment of preeclampsia with magnesium sulfate decreases the risk of placental abruption and A 47
improves maternal outcomes.
Antenatal diagnosis of vasa previa is associated with significant reduction in perinatal mortality. A 78
Delivery in pregnancies complicated by placenta previa may be safely delayed by the use of tocolytics. B 29
Outpatient management of placenta previa is appropriate in selected patients who do not have active B 2
bleeding and who can rapidly access a hospital with operative labor and delivery services.
Transvaginal ultrasonography may be safely performed in women with a placenta previa and is more B 2
accurate in detecting placental location than transabdominal ultrasonography.
In patients presenting with abruption, a decision-to-delivery interval of 20 minutes or less results in B 60
improved neonatal outcomes.
A sterile speculum examination can be performed safely in women with second or third trimester C 2
vaginal bleeding before ultrasonographic evaluation of placental localization, but a digital
examination should be avoided until placenta previa is excluded by ultrasonography.
Women with bleeding in late pregnancy who are Rh negative should receive Rho (D) immune globulin C 5,6
after performance of a Kleihauer-Betke test to determine appropriate dose.
In women with placenta previa and previous cesarean delivery, imaging with color flow Doppler C 2
ultrasonography should be performed to evaluate for placenta accreta.
Ultrasound and color flow Doppler screening for vasa previa is recommended in women at increased C 2,85
risk including women who are pregnant after in vitro fertilization, when a low-lying placenta is
detected on second trimester ultrasound, and in the presence of low or velamentous cord insertion,
bilobate, or succenturiate placenta.
Magnetic resonance imaging of the pelvis may help confirm a diagnosis of invasive placenta and C 20
identify organ involvement associated with placenta percreta.

Strength of Recommendation Taxonomy (SORT)

Strength of Recommendation Definition


A • Recommendation based on consistent and good-quality patient-oriented evidence.
B • Recommendation based on inconsistent or limited-quality patient-oriented evidence.
C • Recommendation based on consensus, usual practice, opinion, disease-oriented evidence, or case series for studies of diagnosis, treat-
ment, prevention, or screening.
Patient-oriented evidence measures outcomes that matter to patients: morbidity, mortality, symptom improvement, cost reduction, and quality
of life. Disease-oriented evidence measures intermediate, physiologic, or surrogate end points that might not reflect improvement in patient
outcomes (eg, blood pressure, blood chemistry, physiologic function, pathologic findings).
From Ebell MH, Siwek J, Weiss BD, et al. Strength of recommendation taxonomy (SORT): a patient-centered approach to grading evidence in
the medical literature. Am Fam Physician. 2004;69(3):548-556.

— Chapter C  11
Chapter C

References
1. McCormack RA, Doherty DA, Magann EF, Hutchinson 16. Yang Q, Wen SW, Phillips K, Oppenheimer L, Black
M, Newnham JP. Antepartum bleeding of unknown D, Walker MC. Comparison of maternal risk factors
origin in the second half of pregnancy and pregnancy between placental abruption and placenta previa.
outcomes. BJOG. 2008;115(11):1451-1457. Am J Perinatol. 2009;26(4):279-286.
2. Royal College of Obstetricians and Gynaecologists. 17. Silver RM, Landon MB, Rouse DJ, et al; National Insti-
Green-top Guideline No. 63. Antepartum Haemorrhage. tute of Child Health and Human Development Maternal-
London, England: Royal College of Obstetricians and Fetal Medicine Units Network. Maternal morbidity
Gynaecologists; 2011. associated with multiple repeat cesarean deliveries.
3. De Kock J, Heyns T, Van Rensburg GH. The ABC of Obstet Gynecol. 2006;107(6):1226-1232.
haemorrhagic shock in the pregnant woman. Profes- 18. Bahar A, Abusham A, Eskandar M, Sobande A, Alsuna-
sional Nursing Today. 2008;12(5):54-57. idi M. Risk factors and pregnancy outcome in differ-
4. Erez O, Mastrolia SA, Thachil J. Disseminated intra- ent types of placenta previa. J Obstet Gynaecol Can.
vascular coagulation in pregnancy: insights in patho- 2009;31(2):126-131.
physiology, diagnosis and management. Am J Obstet 19. Tuzovic L. Complete versus incomplete placenta
Gynecol. 2015;213(4):452-463. previa and obstetric outcome. Int J Gynaecol Obstet.
5. Jain V, Chari R, Maslovitz S, et al; Maternal Fetal Medi- 2006;93(2):110-117.
cine Committee. Guidelines for the management of a 20. Committee on Obstetric Practice. Committee opinion
pregnant trauma patient. J Obstet Gynaecol Can. 2015 no. 529: placenta accreta. Obstet Gynecol. 2012;120(1):
;37(6):553-574. 207-211.
6. American College of Obstetrics and Gynecology. 21. Miller DA, Chollet JA, Goodwin TM. Clinical risk factors
ACOG practice bulletin. Prevention of Rh D alloimmuni- for placenta previa-placenta accreta. Am J Obstet Gyne-
zation. Number 4, May 1999 (replaces educational bul- col. 1997;177(1):210-214.
letin Number 147, October 1990). Clinical management 22. Zaki ZM, Bahar AM, Ali ME, Albar HA, Gerais MA. Risk
guidelines for obstetrician-gynecologists. Int J Gynae- factors and morbidity in patients with placenta previa
col Obstet. 1999;66(1):63-70. accreta compared to placenta previa non-accreta. Acta
7. American College of Obstetricians and Gynecolo- Obstet Gynecol Scand. 1998;77(4):391-394.
gists. ACOG Practice Bulletin No. 106: Intrapartum fetal 23. Comstock CH, Love JJ Jr, Bronsteen RA, et al. Sono-
heart rate monitoring: nomenclature, interpretation, graphic detection of placenta accreta in the second and
and general management principles. Obstet Gynecol. third trimesters of pregnancy. Am J Obstet Gynecol.
2009;114(1):192-202. 2004;190(4):1135-1140.
8. Vergani P, Ornaghi S, Pozzi I, et al. Placenta previa: dis- 24. Oppenheimer L; Society of Obstetricians and Gyn-
tance to internal os and mode of delivery. Am J Obstet aecologists of Canada. Diagnosis and manage-
Gynecol. 2009;201(3):266.e1-266.e5. ment of placenta previa. J Obstet Gynaecol Can.
9. Bhide A, Thilaganathan B. Recent advances in the man- 2007;29(3):261-273.
agement of placenta previa. Curr Opin Obstet Gynecol. 25. Zlatnik MG, Cheng YW, Norton ME, Thiet MP, Caughey
2004;16(6):447-451. AB. Placenta previa and the risk of preterm delivery. J
10. Silver RM. Abnormal Placentation: Placenta Previa, Matern Fetal Neonatal Med. 2007;20(10):719-723.
Vasa Previa, and Placenta Accreta. Obstet Gynecol. 26. Neilson JP. Interventions for suspected placenta prae-
2015;126(3):654-668. via. Cochrane Database Syst Rev. 2003;(2):CD001998.
11. Mustafá SA, Brizot ML, Carvalho MH, Watanabe L, Kah- 27. Sharma A, Suri V, Gupta I. Tocolytic therapy in conser-
hale S, Zugaib M. Transvaginal ultrasonography in pre- vative management of symptomatic placenta previa. Int
dicting placenta previa at delivery: a longitudinal study. J Gynaecol Obstet. 2004;84(2):109-113.
Ultrasound Obstet Gynecol. 2002;20(4):356-359.
28. Roberts D, Dalziel S. Antenatal corticosteroids for
12. Dashe JS, McIntire DD, Ramus RM, Santos-Ramos R, accelerating fetal lung maturation for women at
Twickler DM. Persistence of placenta previa according risk of preterm birth. Cochrane Database Syst Rev.
to gestational age at ultrasound detection. Obstet Gyne- 2006;(3):CD004454.
col. 2002;99(5 Pt 1):692-697.
29. Gyamfi-Bannerman C, Thom EA, Blackwell SC, et al;
13. Becker RH, Vonk R, Mende BC, Ragosch V, Entezami NICHD Maternal–Fetal Medicine Units Network. Antena-
M. The relevance of placental location at 20-23 ges- tal Betamethasone for Women at Risk for Late Preterm
tational weeks for prediction of placenta previa at Delivery. N Engl J Med. 2016;374(14):1311-1320.
delivery: evaluation of 8650 cases. Ultrasound Obstet
Gynecol. 2001;17(6):496-501. 30. Cobo E, Conde-Agudelo A, Delgado J, Canaval H,
Congote A. Cervical cerclage: an alternative for the
14. Taipale P, Hiilesmaa V, Ylöstalo P. Transvaginal ultraso- management of placenta previa? Am J Obstet Gynecol.
nography at 18-23 weeks in predicting placenta previa at 1998;179(1):122-125.
delivery. Ultrasound Obstet Gynecol. 1998;12(6):422-425.
31. Pivano A, Alessandrini M, Desbriere R, et al. A score
15. Bricker L, Neilson JP, Dowswell T. Routine ultrasound to predict the risk of emergency caesarean delivery in
in late pregnancy (after 24 weeks’ gestation). Cochrane women with antepartum bleeding and placenta praevia.
Database Syst Rev. 2008;(4):CD001451. Eur J Obstet Gynecol Reprod Biol. 2015;195:173-176.

12  Chapter C —
Vaginal Bleeding in Late Pregnancy

32. Ghi T, Contro E, Martina T, et al. Cervical length and 50. Leunen K, Hall DR, Odendaal HJ, Grové D. The
risk of antepartum bleeding in women with com- profile and complications of women with placental
plete placenta previa. Ultrasound Obstet Gynecol. abruption and intrauterine death. J Trop Pediatr.
2009;33(2):209-212. 2003;49(4):231-234.
33. Al Wadi K, Schneider C, Burym C, Reid G, Hunt J, Men- 51. Pepperell RJ, Rubinstein E, MacIsaac IA. Motor-car acci-
ticoglou S. Evaluating the safety of labour in women dents during pregnancy. Med J Aust. 1977;1(7):203-205.
with a placental edge 11 to 20 mm from the internal cer- 52. Sinha P, Kuruba N. Ante-partum haemorrhage: an
vical Os. J Obstet Gynaecol Can. 2014;36(8):674-677. update. J Obstet Gynaecol. 2008;28(4):377-381.
34. Blackwell SC. Timing of delivery for women with stable 53. Doyle C, Angelotti T. Diagnosis of an unsuspected
placenta previa. Semin Perinatol. 2011;35(5):249-251. maternal hemorrhage via fetal heart rate tracing. J Clin
35. Zlatnik MG, Little SE, Kohli P, Kaimal AJ, Stotland NE, Anesth. 2004;16(6):465-468.
Caughey AB. When should women with placenta pre- 54. Glantz C, Purnell L. Clinical utility of sonography in the
via be delivered? A decision analysis. J Reprod Med. diagnosis and treatment of placental abruption. J Ultra-
2010;55(9-10):373-381. sound Med. 2002;21(8):837-840.
36. American College of Obstetricians and Gynecologists. 55. Manriquez M, Srinivas G, Bollepalli S, Britt L, Drachman
ACOG committee opinion no. 560: Medically indicated D. Is computed tomography a reliable diagnostic modal-
late-preterm and early-term deliveries. Obstet Gynecol. ity in detecting placental injuries in the setting of acute
2013;121(4):908-910. trauma? Am J Obstet Gynecol. 2010;202(6):611.e1-611.e5.
37. Balayla J, Wo BL, Bédard MJ. A late-preterm, early- 56. Neilson JP. Interventions for treating placental abrup-
term stratified analysis of neonatal outcomes by ges- tion. Cochrane Database Syst Rev. 2003;(1):CD003247.
tational age in placenta previa: defining the optimal
timing for delivery. J Matern Fetal Neonatal Med. 57. Towers CV, Pircon RA, Heppard M. Is tocolysis safe
2015;28(15):1756-1761. in the management of third-trimester bleeding? Am J
Obstet Gynecol. 1999;180(6 Pt 1):1572-1578.
38. Oyelese Y, Ananth CV. Placental abruption. Obstet
Gynecol. 2006;108(4):1005-1016. 58. Knab DR. Abruptio placentae. An assessment of the
time and method of delivery. Obstet Gynecol. 1978;
39. Ananth CV, Oyelese Y, Yeo L, Pradhan A, Vintzileos AM. 52(5):625-629.
Placental abruption in the United States, 1979 through
2001: temporal trends and potential determinants. Am J 59. Ananth CV, Wilcox AJ. Placental abruption and peri-
Obstet Gynecol. 2005;192(1):191-198. natal mortality in the United States. Am J Epidemiol.
2001;153(4):332-337.
40. Hladky K, Yankowitz J, Hansen WF. Placental abruption.
Obstet Gynecol Surv. 2002;57(5):299-305. 60. Kayani SI, Walkinshaw SA, Preston C. Pregnancy out-
come in severe placental abruption. BJOG. 2003;110(7):
41. Ruiter L, Ravelli ACJ, de Graaf IM, Mol BWJ, Pajkrt E. 679-683.
Incidence and recurrence rate of placental abruption: a
longitudinal linked national cohort study in the Nether- 61. Witlin AG, Sibai BM. Perinatal and maternal outcome
lands. Am J Obstet Gynecol. 2015;213(4):573.e1-573.e8. following abruptio placentae. Hypertens Pregnancy.
2001;20(2):195-203.
42. Arnold DL, Williams MA, Miller RS, Qiu C, Sorensen
TK. Iron deficiency anemia, cigarette smoking and 62. Boisramé T, Sananès N, Fritz G, et al. Placental abrup-
risk of abruptio placentae. J Obstet Gynaecol Res. tion: risk factors, management and maternal-fetal prog-
2009;35(3):446-452. nosis. Cohort study over 10 years. Eur J Obstet Gynecol
Reprod Biol. 2014;179:100-104.
43. Brenner B, Aharon A. Thrombophilia and adverse preg-
nancy outcome. [v.]. Clin Perinatol. 2007;34(4):527-541, v. 63. Emery CL, Morway LF, Chung-Park M, Wyatt-Ashmead
J, Sawady J, Beddow TD. The Kleihauer-Betke test.
44. Brown HL. Trauma in pregnancy. Obstet Gynecol. Clinical utility, indication, and correlation in patients with
2009;114(1):147-160. placental abruption and cocaine use. Arch Pathol Lab
45. Rogers FB, Rozycki GS, Osler TM, et al. A multi- Med. 1995;119(11):1032-1037.
institutional study of factors associated with fetal death 64. Update ACOG. Major and minor trauma in pregnancy.
in injured pregnant patients. Arch Surg. 1999;134(11): Vol. 35, No. 6. December 2009. Medical Information
1274-1277. Systems Inc. 2 Seaview Blvd. Port Washington, New
46. Fleming AD. Abruptio placentae. Crit Care Clin. 1991; York. Available at http://www.acogupdate.com/.
7(4):865-875. 65. American College of Obstetricians and Gynecologists.
47. Duley L, Gülmezoglu AM, Henderson-Smart DJ, Chou ACOG Practice bulletin no. 115: Vaginal birth after previ-
D. Magnesium sulphate and other anticonvulsants for ous cesarean delivery. Obstet Gynecol. 2010;116(2 Pt 1):
women with pre-eclampsia. Cochrane Database Syst 450-463.
Rev. 2010;(11):CD000025. 66. Mirza FG, Gaddipati S. Obstetric emergencies. Semin
48. Reis PM, Sander CM, Pearlman MD. Abruptio placen- Perinatol. 2009;33(2):97-103.
tae after auto accidents. A case-control study. J Reprod 67. Al-Zirqi I, Stray-Pedersen B, Forsén L, Vangen S. Uter-
Med. 2000;45(1):6-10. ine rupture after previous caesarean section. BJOG.
49. Golan A, Sandbank O, Teare AJ. Trauma in late preg- 2010;117(7):809-820.
nancy. A report of 15 cases. S Afr Med J. 1980;57(5):
161-165.

— Chapter C  13
Chapter C

68. Smith JG, Mertz HL, Merrill DC. Identifying risk factors 77. Jauregui I, Kirkendall C, Ahn MO, Phelan J. Uterine
for uterine rupture. [viii.]. Clin Perinatol. 2008;35(1):85- rupture: a placentally mediated event? Obstet Gynecol.
99, viii. 2000;95(4)(Suppl 1):S75.
69. Aziz N, Yousfani S. Analysis of uterine rupture at 78. Sinkey RG, Odibo AO, Dashe JS; Society of Maternal-
university teaching hospital Pakistan. Pak J Med Sci. Fetal (SMFM) Publications Committee. #37: Diagnosis
2015;31(4):920-924. and management of vasa previa. Am J Obstet Gynecol.
70. Zwart JJ, Richters JM, Ory F, de Vries JIP, Bloemenkamp 2015;213(5):615-619.
KWM, van Roosmalen J. Uterine rupture in The Neth- 79. Gagnon R, Morin L, Bly S, et al; Diagnostic Imaging
erlands: a nationwide population-based cohort study. Committee; Maternal Fetal Medicine Committee. Guide-
BJOG. 2009;116(8):1069-1078, discussion 1078-1080. lines for the management of vasa previa. J Obstet Gyn-
71. National Institutes of Health Consensus Development aecol Can. 2009;31(8):748-760.
Conference Panel. National Institutes of Health Consen- 80. Oyelese KO, Turner M, Lees C, Campbell S. Vasa pre-
sus Development conference statement: vaginal birth via: an avoidable obstetric tragedy. Obstet Gynecol
after cesarean: new insights March 8-10, 2010. Obstet Surv. 1999;54(2):138-145.
Gynecol. 2010;115(6):1279-1295. 81. Baulies S, Maiz N, Muñoz A, Torrents M, Echevarría M,
72. Guise JM, McDonagh MS, Osterweil P, Nygren P, Chan Serra B. Prenatal ultrasound diagnosis of vasa praevia
BK, Helfand M. Systematic review of the incidence and and analysis of risk factors. Prenat Diagn. 2007;27(7):
consequences of uterine rupture in women with previ- 595-599.
ous caesarean section. BMJ. 2004;329(7456):19-25. 82. Hasegawa J, Arakaki T, Ichizuka K, Sekizawa A. Man-
73. Leung AS, Leung EK, Paul RH. Uterine rupture after agement of vasa previa during pregnancy. J Perinat
previous cesarean delivery: maternal and fetal conse- Med. 2015;43(6):783-784.
quences. Am J Obstet Gynecol. 1993;169(4):945-950. 83. Nelle M, Zilow EP, Kraus M, Bastert G, Linderkamp O.
74. Desseauve D, Bonifazi-Grenouilleau M, Fritel X, Lathél- The effect of Leboyer delivery on blood viscosity and
ize J, Sarreau M, Pierre F. Fetal heart rate abnormali- other hemorheologic parameters in term neonates. Am
ties associated with uterine rupture: a case-control J Obstet Gynecol. 1993;169(1):189-193.
study: A new time-lapse approach using a standard- 84. Apt L, Downey WS Jr. Melena neonatorum: the swal-
ized classification. Eur J Obstet Gynecol Reprod Biol. lowed blood syndrome; a simple test for the differentia-
2016;197:16-21. tion of adult and fetal hemoglobin in bloody stools. J
75. Matsuo K, Scanlon JT, Atlas RO, Kopelman JN. Stair- Pediatr. 1955;47(1):6-12.
case sign: a newly described uterine contraction pattern 85. Lee W, Lee VL, Kirk JS, Sloan CT, Smith RS, Com-
seen in rupture of unscarred gravid uterus. J Obstet stock CH. Vasa previa: prenatal diagnosis, natural
Gynaecol Res. 2008;34(1):100-104. evolution, and clinical outcome. Obstet Gynecol.
76. Vaknin Z, Maymon R, Mendlovic S, Barel O, Herman A, 2000;95(4):572-576.
Sherman D. Clinical, sonographic, and epidemiologic
features of second- and early third-trimester spontane-
ous antepartum uterine rupture: a cohort study. Prenat
Diagn. 2008;28(6):478-484.

14  Chapter C —
Objectives
Vaginal Bleeding in 
• Identify serious causes of vaginal bleeding in the
Late Pregnancy second half of pregnancy
Revised January 2017 • Describe a systematic approach to identify the cause
of bleeding
• Describe specific treatment options based on
diagnosis

Causes of Late Pregnancy Bleeding Initial Management 
• Placenta previa • Same initial steps regardless of etiology
• Placental abruption 
Life-Threatening • Assess vital signs, circulatory stability
• Uterine scar disruption 
• Ruptured vasa previa • Secure intravenous access, administer fluids
• Cervical polyp
• Bloody show/cervical change
• Cervicitis or cervical ectropion
• Vaginal trauma
• Cervical cancer

History and Examination Diagnostic Evaluation
• Targeted history and physical • Continuous electronic fetal monitoring and tocodynamometry
• Abdominal examination for estimated fetal weight (EFW),  • Ultrasound for placental location, presence of clots, and fetal
estimated gestational age (EGA), and fetal presentation presentation
 Localize tenderness • Obtain baseline laboratory tests
 Complete blood count
 Palpate for uterine contractions
 Blood type and antibody screen
• Gentle speculum examination is safe  Consider: coagulation studies, blood urea nitrogen (BUN), creatinine, 
liver function testing, and type and cross
• NO digital vaginal exam unless placental location is known
• Prepare for possible emergent cesarean delivery

Copyright 2017© American Academy of Family Physicians. All rights reserved. 1


Placenta Previa Prevalence of Placenta Previa
• Common finding on second trimester ultrasound, 
most regress
 Previa found on 4% of ultrasounds at 20 to 25 weeks, only 
0.5% by term
• More likely to persist with previous cesarean delivery, 
complete previa
• No benefit to routine screening ultrasound in 
Low-Lying: Marginal: Complete: late pregnancy
2 to 3.5 cm from os Within 2 cm of os Completely covers os

Risk Factors for Placenta Previa  Morbidity with Placenta Previa
• Advanced maternal age • Maternal
• Chronic hypertension  Hemorrhage
• Multiparity  Blood transfusion
• Multiple gestation  Cesarean delivery
• Previous cesarean delivery  Hysterectomy
• Previous placenta previa  Invasive placenta (accreta, increta, or percreta)
• Previous uterine curettage • Fetal
• Smoking  Prematurity

Invasive Placenta Placenta Previa with Increta
• Placenta accreta, increta, and percreta are more 
common with placenta previa
• Increased risk with previous cesarean delivery
 Risk increases with increasing number of 
cesarean deliveries
• Evaluate with color flow Doppler

2
Clinical Presentation ‐ Previa Ultrasound – Placenta Previa
• Classic presentation is painless bleeding  • When a previa is seen on transabdominal ultrasound,
 late second or third trimester transvaginal scan should be performed
 provoked by intercourse  More accurate visualization of placental edge and 
 may be accompanied by preterm contractions  internal os
• Suspect previa with persistent malpresentation
• Full bladder can create false appearance of
• Diagnosis confirmed with ultrasound localization 
of placenta anterior previa
 Confirm with transvaginal ultrasound • Presenting part may overshadow posterior previa

Ultrasound – Placenta Previa Management of Placenta Previa


• When active bleeding is assessed in the hospital
 If preterm, consider maternal transfer to appropriate level of care
 Consider antenatal corticosteroids and tocolysis
 Full dose of Rho (D) immune globulin if Rh negative
• Absence of active bleeding can be managed expectantly
• No intercourse or tampons in the third trimester
• Avoid digital examinations 
• Confirm unresolved previa with ultrasound at 32 weeks
• Ultrasound at 36 weeks can help determine mode of delivery

Ongoing Management of  Neonatal Morbidity from 
Placenta Previa Placenta Previa
• If bleeding stops, can consider outpatient management if • Greatest morbidity and mortality is related to prematurity
 Assurance of current maternal and fetal well‐being • Tocolysis can increase the duration of pregnancy and birth 
 Patient lives in close proximity to hospital weight without increased maternal/fetal complications
 Immediate evaluation can occur if new bleeding episode starts
or with onset of labor • Antenatal steroids should be administered to patients with 
symptomatic previa between 24 and 34 weeks gestation
• Need for emergency cesarean delivery increases with
 Three or more episodes of antepartum bleeding
 Initial episode of bleeding occurring at <29 weeks
 Shortened cervical length on serial ultrasounds

3
Delivery Decisions – Placenta Previa Placental Abruption
• For known low‐lying placenta or marginal previa with no bleeding,  • Premature separation of placenta from uterine wall
perform ultrasound at 36 weeks
 If placenta is located ≥2 cm from internal os, expect vaginal delivery  Partial or complete abruption can occur
 If 1 to 2 cm from os, may attempt vaginal delivery in setting with  • Occurs in 1% of pregnancies
immediate surgical backup
• Perform cesarean delivery if   • Apparent increase in incidence
 Ultrasound indicates complete previa 
 Fetal head is not engaged  Increased diagnosis (ultrasound) vs. Increased risk factors 
 Concerning fetal heart rate tracing (hypertension)
 Brisk or persistent bleeding 
• Regional anesthesia is safe and results in less blood loss   

Risk Factors for Abruption Clinical Presentation ‐ Abruption


• Hypertensive disorders of pregnancy • Abdominal pain is the hallmark symptom
 Treating preeclampsia with magnesium sulfate (MgSO4)   Varies from mild cramping to severe pain
decreases risk 
• Abdominal trauma  With back pain – think posterior abruption 
• Tobacco, cocaine, stimulants • Vaginal bleeding or bloody amniotic fluid
• Thrombophilias  Differentiate from exuberant bloody show
• Chorioamnionitis  • Concealed bleeding occurs in 20% to 63% of cases
• Oligohydramnios with PROM
 Visible bleeding may not accurately reflect blood loss
• History of abruption in a previous pregnancy

Evaluation ‐ Abruption Abruption is a Clinical Diagnosis


• Check vital signs to assess for circulatory instability • Diagnosis of placental abruption is highly dependent 
• Palpate maternal abdomen to assess for upon clinical assessment
 Location of tenderness 
 Tetanic contractions  • Do not delay definitive management to obtain 
• Monitor fetal heart rate (FHR) and contractions an ultrasound
 Continuous fetal electrocardiogram
 Consider amniotomy and intrauterine pressure catheter • Ultrasound may show characteristic features, but has 
• Ultrasound to determine  low sensitivity for detecting placental abruption
 Estimated fetal weight, fetal lie, and presentation
 Evaluate the placenta 

4
Diagnosis ‐ Abruption Abruption Severity
• Ultrasound findings  • Mild 
may indicate:  Often identified at delivery with retroplacental clot 
 Retroplacental present upon inspection of placenta
echolucency • Severe 
 Abnormal thickening   Symptomatic, tender abdomen 
of placenta • Severe with fetal demise
 Torn edge of placenta  without coagulopathy (two‐thirds)  
 with coagulopathy (one‐third)

Management of Severe Abruption Coagulopathy with Abruption
• Expeditious operative or vaginal delivery • Occurs in one‐third of fetal demises
 Decision‐to‐delivery interval >20 minutes increases incidence of fetal 
mortality or cerebral palsy  • Usually not seen with delivery of live fetus
• Maintain maternal circulation • Etiologies: consumption, disseminated intravascular 
 Urine output >30 mL/hour
 Hematocrit >30% coagulation (DIC) 
• Prepare for neonatal resuscitation • Administer platelets, fresh frozen plasma (FFP) 
• If fetal demise before operative delivery
 Vaginal delivery preferred, unless severely bleeding
 Check for coagulopathy

Uterine Rupture Risk Factors – Uterine Rupture


• Occult dehiscence versus symptomatic rupture • Previous cesarean incision (most common)
• Rare in an unscarred uterus • Other uterine surgery involving full thickness myometrium
• Inappropriate oxytocin usage
 <0.1% overall incidence • Labor induction
 0.8% of women with uterine scar • Uterine over‐distention 
• Previous cesarean incision is the most common  • Abnormal placental attachment, difficult removal
etiology • Uterine or fetal anomaly
• Gestational trophoblastic neoplasia
• Adenomyosis

5
Morbidity with Uterine Rupture Clinical Findings – Uterine Rupture
• Maternal • Sudden deterioration of FHR pattern is the most common  
 Hemorrhage with anemia (most common) initial sign 
 Bladder rupture • Vaginal bleeding
 Hysterectomy
 Maternal mortality 
• Pain, sudden onset
• Fetal  • Stair step decrease or cessation of contractions 
 Respiratory distress • Loss of fetal station
 Hypoxia • Palpable fetal parts through maternal abdomen
 Acidemia • Profound maternal tachycardia and hypotension
 Neonatal mortality

Management – Uterine Rupture Vasa Previa


• Symptomatic rupture • Fetal vessels run in 
– Emergent cesarean delivery membranes between 
cervix and presenting part
– Outcome good if decision to incision delivery time occurs 
• Rarest cause of 
in under 18 minutes in one study hemorrhage
• Can result in fetal 
blood loss
• Rapid intervention is 
essential for fetal survival

Vasa Previa – Associated Conditions Vasa Previa – Clinical Presentation


• In vitro fertilization • Can be detected antenatally
• Low‐lying placenta or   Will be able to palpate vessels during cervical examination
previa present in second 
 Color flow Doppler ultrasound 
or third trimester
• Bilobed and  • First sign is typically bleeding with membrane rupture
succenturiate lobed  • Important to remember blood loss is fetal
placentas
 Fetal blood circulation = 250 mL total
• Velamentous insertion of 
the umbilical cord
Bilobed placenta with velamentous insertion

6
Vasa Previa– Intrapartum Management Vasa Previa– Antepartum Diagnosis
• Tests for fetal hemoglobin as source of hemorrhage • Critical for fetal survival as allows for planned cesarean delivery
have limited usefulness  97% survival when detected antenatally
 Apt test insensitive  44% survival without antenatal diagnosis
 Kleihauer‐Betke test or hemoglobin electrophoresis test • If placenta previa present on second trimester ultrasound, 
results are too slow  perform follow‐up ultrasound with color flow Doppler to rule 
out vasa previa
• Immediate cesarean delivery if FHR concerning
• If persistent vasa previa is present 
• Administer normal saline 10 to 20 mL/kg bolus to  Administer corticosteroids
newborn if in shock due to blood loss after delivery  Deliver at 35 to 36 weeks

Summary
• Late pregnancy bleeding may herald diagnoses with significant 
potential for maternal morbidity/mortality
• Rapid clinical diagnosis is imperative
• In the setting of maternal hemorrhage and concerning FHR 
tracing, do not delay management to do diagnostic testing
• No vaginal digital examination until placental location is known
• Ultrasound can help determine the cause of bleeding
• When antenatal ultrasound identifies placenta previa, color flow 
Doppler study is recommended to evaluate for placenta accreta 
and vasa previa

7
Chapter D
Preterm Labor and Premature
Rupture of Membranes

William Sayres, MD, Karen Wildman, MD, FAAFP, Colleen Zimmermann, MD


Revised January 2016

Learning Objectives
At the end of this activity, learners will be able to:
1. Discuss risk factors associated with preterm labor (PTL) and premature
rupture of membranes (PROM).
2. Identify patients who may benefit from antenatal progesterone.
3. Define PTL and PROM, and describe their significance.
4. Outline initial evaluation of PTL and PROM.
5. Describe management of PTL and PROM.
6. Discuss neonatal group B streptococcal prevention strategies.

Introduction mately 40% to 45% of PTDs are the result of spon-


Previous preterm delivery (PTD) is the most impor- taneous PTL with intact membranes. Another 25%
tant historical risk factor for subsequent PTD. Other to 30% are associated with PPROM. The remaining
important antenatal risk factors include multiple 30% to 35% of PTDs are the result of delivery by labor
gestation, bacterial vaginosis (BV), and a short cervical induction or cesarean delivery for medical indications
length (CL) on ultrasound. Progesterone may be used such as preeclampsia with severe features, placental
to decrease the likelihood of PTD in patients with a abruption, or intrauterine growth restriction.2
prior PTD or a short CL. In a patient presenting with From 1981 through 2006, the incidence of PTD
preterm contractions, CL on transvaginal ultrasound increased by more than 30%, and peaked at 12.80% in
or a fetal fibronectin (fFN) test can be used to help 2006.1 Since that time, the rate of PTD has gradually
assess the risk of PTD. Administration of antenatal decreased each year. The rate of early PTD (delivery
corticosteroids (ACS) remains our most important before 34 weeks) has declined 3.7% and the rate of late
intervention to improve neonatal outcomes in women PTD (delivery at 34 to 36 weeks) has declined 9.2%
presenting with preterm labor (PTL). Tocolytic agents since 2006.3 The declining rate of PTD may be attrib-
may delay PTD, which allows time to administer ste- uted to multiple factors. Since 2006, both the teenage
roids or transfer to a facility with a neonatal intensive birth rate and the rate of higher order multiples (trip-
care unit (NICU). Management of preterm premature lets or higher gestations) have declined.4 These groups
rupture of membranes (PPROM) may include admin- are at higher risk for PTD. There is some evidence
istering antibiotics or ACS, or inducing labor based that public policy recommendations to reduce elective
on the gestational age at presentation. Management early term (37 to 38 weeks) deliveries have also reduced
of premature rupture of membranes (PROM) at term PTDs.4,5 Finally, interventions such as vaginal proges-
includes early induction/augmentation of labor. terone, intramuscular 17 alpha-hydroxyprogesterone
caproate (17P), and use of cerclage in selected popula-
Epidemiology tions are likely contributing to the decreased rate.4
The incidence of PTD, defined as occurring before The PTD rate in the United States still remains
37 weeks’ gestation, was 11.4% in 2013.1 Approxi- among the highest in the developed world and signifi-

Copyright 2017© American Academy of Family Physicians. All rights reserved. — Chapter D  1
Chapter D

cant disparities exist in the rates between racial weeks’ gestation, a subsequent pregnancy has a 26%
ethnic groups. In 2012, the PTD rate for non- risk of PTD (95% CI = 10.8 to 15.9).11 Multiple
Hispanic white infants was 10.3% compared to prior PTDs further increase risk. Women with a
16.5% for non-Hispanic black infants, and 11.6% preceding, medically indicated PTD are at increased
for Hispanic infants.6 risk for a subsequent medically indicated PTD, and
are also at risk for a subsequent spontaneous PTD.12
Risk Factors for PTD Multiple gestation is a strong predictor for PTD
Although many risk factors for PTD have been with the majority of twin pregnancies deliver-
identified, up to 50% of PTDs occur in pregnan- ing at less than 37 weeks because of spontaneous
cies with no known risk factors, and over half of PTL or medical indications for delivery.13 Higher
women with identifiable risk factors for PTD will order multiples further increase the risk for PTD.
ultimately deliver at term.7,8 Furthermore, the In 2011, 11% of twin, 36% of triplet, and more
causality of identified risk factors has been diffi- than 67% of quadruplet pregnancies delivered
cult to prove. Many risk-scoring systems for PTD before 32 weeks.14
have been developed, but none have been shown Cervical shortening in the second trimester is
to reduce preterm birth.9 Despite this, identifying associated with increased risk for PTD.15 When
women at risk is of growing importance as treat- measuring CL, transvaginal ultrasound (TVU)
ment options such as progesterone and cerclage should be used because transabdominal ultra-
become available. Common risk factors for PTD sound may miss up to 57% of cervices shorter
are outlined in Table 1. than 25 mm.16 The gestational age at which TVU
Prior PTD is the single most important identifi- CL is most predictive of preterm birth is 14 to
able risk factor for recurrent PTD. Women carrying 34 weeks,17,18 with most TVU CL measurements
a singleton after a previous singleton PTD have a performed at 18 to 24 weeks. The cutoff to define
20% risk of recurrence (95% confidence interval a short cervix varies between studies, with 20 mm
[CI] = 19.9 to 20.6).10 In contrast, after a term to 25 mm most commonly used. The shorter the
delivery, women have a 2.7% risk of PTD in the cervix and the earlier in gestation the shortening is
subsequent pregnancy. To break this down further, detected, the higher the risk of PTD.19 In women
after delivery between 32 to 36 weeks, a subse- without vaginal bleeding, the rate of change in cer-
quent pregnancy has a 14.7% risk of PTD (95% vical shortening when measured by sequential cervi-
CI = 5.84 to 6.42), and after delivery before 28 cal ultrasounds is associated with increased risk of
PTD (odds ratio [OR] 1.2; 95% CI = 1.1 to 1.4).20
Universal screening of TVU to measure CL of
Table 1. Risk Factors for PTL women with a singleton gestation and no prior
PTD is a subject of debate. Two large randomized
Maternal Characteristics Pregnancy Characteristics controlled trials (RCTs) showed that universal
Non-Hispanic black race6 Shortened cervix 15 screening of CL and treatment with daily vaginal
BMI <19 kg/m2 31 Multiple gestation13 progesterone in women with a short CL reduces
Low economic status2 Use of assisted reproductive technology2
the occurrence of preterm birth and improves
Stressful life events2 Interpregnancy interval <6 months30
neonatal outcomes.21,22 Universal CL screening
Maternal Medical History Cocaine or heroin use2
Alcohol use (>10 drinks/week)144
at 18 to 24 6/7 weeks would require 400 to 600
Previous PTD10
Tobacco use36 screenings to prevent one early preterm birth at
Previous abortions
(>1 versus none)143 Maternal abdominal surgery during less than 34 weeks’ gestation.23 The Society for
Uterine anomalies2 pregnancy 2 Maternal Fetal Medicine in 2012 concluded that
Infection
Maternal depression during pregnancy 145 CL screening is a reasonable approach but can-
Presence of thyroid autoantibodies146 not be universally recommended at this time, and
Bacterial vaginosis25
Polyhydramnios or oligohydramnios2 practitioners who elect to implement universal CL
Intrauterine infection27
Vaginal bleeding from placental screening should follow published guidelines.24
Periodontal infection28 abruption or placenta previa
Low vitamin D level147
Genitourinary infection is a risk factor for
PTD. BV increases the risk of PTD (OR 2.19;
BMI = body mass index; PTD = preterm delivery; PTL = preterm labor. 95% CI = 1.54 to 3.12) and spontaneous abor-
tion (OR 9.91; 95% CI = 1.99 to 49.34).21,25

2  Chapter D —
Table 2. Progesterone Formulation and Dosage for the Prevention of PTD24,39,40

Indication Progestogen Dosing

Prior PTD 17-alpha-hydroxyprogesteron caproate 250 mg IM weekly from 16 to 36 weeks


No prior PTD, CL <20 mm Vaginal progesterone gel 90 mg Daily from diagnosis of short CL until
at <24 weeks Vaginal progesterone capsule 200 mg 36 weeks

CL = cervical length; IM = intramuscularly; PTD = preterm delivery.

Women screened and found positive for BV at increase risk of PTD remains a subject of debate.
less than 16 weeks’ gestation had an even higher A recent meta-analysis demonstrated that women
likelihood of PTD (OR 7.55; 95% CI = 1.80 to with a prior loop electrosurgical excision have a
31.65).25 Other infections such as trichomoniasis, similar risk for PTD as women with cervical dys-
chlamydia, and/or gonorrhea are also associated plasia without an excisional procedure, suggesting
with increased risk for PTD, although evidence a shared underlying risk factor.32,33
is conflicting about whether treatment for these Maternal toxin exposure, including pollution
infections reduces the risk of PTD.26 Pyelone- and ozone exposure, have been linked with PTD.34
phritis, symptomatic lower urinary tract infec- The association of maternal cigarette smoking
tion, and asymptomatic bacteriuria have all been with increased risk of PTD and small-for-gesta-
associated with increased risk for PTD.26 tional-age infants has been recognized for more
Among women who experience PTL, Myco- than 50 years.35 Maternal cigarette smoking has a
plasma species and Ureaplasma urealyticum are dose dependent association with PTD; smoking
the most commonly identified microorganisms 10 to 20 cigarettes per day has a relative risk (RR)
in the amniotic cavity.2 These vaginal flora likely of 1.2 to 1.5 and smoking more than 20 cigarettes
ascend from the vagina and have been impli- per day has a RR of 1.5 to 2.0.36 A prospective
cated in chorioamnionitis, PTL, and PPROM.27 study of 2,504 nulliparous women found the risk
Inflammatory mediators (such as cytokines and for PTD and small-for-gestational-age infants was
TNF-alpha), and cellular immune activation likely similar for women who quit smoking before 15
play a role in initiating labor, and are major areas weeks’ gestation and non-smokers, emphasizing
of ongoing research. Non-genitourinary infec- the importance of counseling and intervention on
tion such as pneumonia has also been associated this modifiable risk factor.37
with PTD. Periodontal infection doubles the risk
for PTD, however antenatal treatment does not Prevention of PTD
appear to alter outcomes.28 Prevention of PTD has been the focus of intense
The presence of uterine contractions is associ- research for the past several decades. Antenatal
ated with an increased likelihood of PTD. In progesterone has shown promise in recent tri-
a study of 306 women with 34,908 hours of als. Screening and treatment of BV has been less
monitoring, increased uterine contractions were successful historically, although better results are
associated with PTD; however, there was not a apparent with earlier screening, focus on high
threshold frequency of contractions that effectively risk women, and appropriate choice of antibiotic.
identified a high-risk group.29 Additionally, cervical cerclage continues to be used
Other maternal characteristics are associated for specific clinical indications.
with PTD. Non-Hispanic black race is signifi-
cantly associated with increased risk for PTD.6 Antenatal Progesterone
An inter-pregnancy interval of less than 6 months The mechanisms by which progesterone prevents
increases the risk of PTD by 1.4 (95% CI = 1.24 PTL include reduction of gap junction formation,
to 1.58).30 Maternal nutritional status and, in oxytocin antagonism, maintenance of cervical
particular, pre-pregnancy body mass index of integrity and anti-inflammation.38
less than 19 kg/m2 increases the odds of PTD by Intramuscular 17P should be used to treat
3.96.31 Whether cervical excisional procedures women with a history of spontaneous PTD
such as cone biopsy or loop electrosurgical excision (Table 2).24,39,40 Women with a history of preterm

— Chapter D  3
Chapter D

birth who are treated with weekly 17P injections trials21,22 and three meta-analyses have demon-
experience lower incidences of perinatal mortal- strated decreased incidence of PTD and neonatal
ity, PTD before 34 and 37 weeks’ gestations.41 In morbidity and mortality with this intervention
contrast, the largest vaginal progesterone trial in (Table 3).41,47,48 In contrast, a large randomized
women with previous PTD recorded no improve- trial of intramuscular 17P in women with a CL
ment in PTD rates at 32 weeks or less compared less than 30 mm failed to reduce the frequency of
to placebo; therefore vaginal progesterone is not PTD or improve neonatal outcome.49 The indica-
recommended in this demographic group.42 After tions and benefit for cerclage in women with no
17P is started, it should not be discontinued history of PTD and a short cervix on endovaginal
because that increases the risk of recurrent PTD.43 ultrasound are unclear.39,50
Cervical length should be evaluated by TVU In the special case of multiple gestation, neither
every 2 weeks from 16 to 23 6/7 weeks’ gesta- progesterone nor cerclage improves outcomes.41,51,52
tion in women with a history of PTD (who are Cervical pessary is actively being studied for the
also being treated with 17P). If CL is less than prevention of PTD.53 The Arabin pessary may
25 mm, cervical cerclage should be offered.24,39,40 delay onset of labor by sealing the endocervi-
Although premature cervical change might be cal canal or by changing the angle of the cervi-
the result of a structurally weak cervix, shorten- cal canal. A trial of cervical pessary in women
ing might also be the result of endocrine, para- with short cervix resulted in decreased PTD and
crine, or inflammatory processes.38 For example, improved neonatal outcomes.54 However, a large
infection is associated with up to 51% of patients Dutch trial of pessaries in twin pregnancies did
presenting with cervical insufficiency.44 Cerclage not result in improved outcomes.55
specifically addresses structural deficits in the
cervix and would not necessarily be as effective for Management of Infection
other processes such as infection. Meta-analysis Microbe-induced inflammation is associated
of five trials of cervical cerclage for CL less than with PTL.56 Periodontal disease is associated with
25 mm demonstrated reduced PTD, and reduced PTD, but antenatal treatment does not affect
composite perinatal mortality and morbidity.45 A pregnancy outcome.57 Screening for and treating
large (1,014 patients) randomized trial demon- asymptomatic bacteriuria, possibly by preventing
strated reduced previable birth, perinatal mortal- pyelonephritis, is associated with a reduction in
ity, and incidence of birth at less than 35 weeks the incidence of low-birth-weight newborns but
if CL was less than 15 mm at time of cerclage.46 not in PTD.58
In an adjusted indirect meta-analysis of 9 stud- Asymptomatic BV is associated with PTD (less
ies, vaginal progesterone and cerclage reduced the than 33 weeks [OR 1.9; CI = 0.94 to 3.84]) and
incidence of PTD at less than 32 weeks as well as late miscarriage (OR 2.77; CI = 0.94 to 8.16).59
composite perinatal morbidity and mortality.47 Three studies screened asymptomatic, low-risk
Women without a history of PTD but with women for BV early in the second trimester and
a shortened cervix (less than 20 mm by ultra- treated with clindamycin (vaginally in two and
sound) should receive vaginal progesterone from orally in one). In all three studies, the treated
time of diagnosis until 36 weeks.24,39,40 Two large women had lower rates of PTD and fewer late

Table 3. Progesterone and Cervical Cerclage for the Prevention of PTD

Indication PTD RR (CI) Neonatal Morbidity and Mortality

Vaginal progesterone for short cervix48 Delivery <33 weeks: 0.58 (0.42 to 0.80) Composite: 0.57 (0.40 to 0.81)
17P for previous PTD41 Delivery <34 weeks: 0.31 (0.14 to 0.69) Mortality: 0.50 (0.33 to 0.75)
Cerclage for short cervix and previous PTD 47 Delivery <32 weeks: 0.63 (0.45 to 0.88) Mortality: 0.58 (0.35 to 0.98)

17P = 17 alpha-hydroxyprogesterone caproate; CI = confidence interval; PTD = preterm delivery; RR = relative risk.

4  Chapter D —
Preterm Labor and Premature Rupture of Membranes

second-trimester miscarriages.60-62 Even so, meta-


analysis of studies of screening and managing BV Table 4. Amsel’s Criteria for Diagnosis
in pregnancy did not show improved pregnancy of BV66
outcomes.63 The US Preventive Services Task Force
recommends against screening for BV in low-risk Diagnosis requires three of four findings:
women and concludes that evidence is insufficient Homogenous, white, non-inflammatory discharge
that smoothly coats the vaginal walls
to recommend for or against screening for and
managing BV in high-risk patients.64 Specific atten- Presence of clue cells on microscopic examination
tion to screening and management before 22 weeks pH of vaginal fluid >4.5
and use of clindamycin instead of metronidazole Fishy odor of vaginal discharge before or after
seem to be associated with improved pregnancy addition of 10% KOH
outcomes.65 Although most studies use criteria
based on gram stain, most clinicians will rely on BV = bacterial vaginosis; KOH = potassium hydroxide.

Amsel’s criteria for diagnosis of BV (Table 4).66


Clindamycin was used for the management of
BV in the three studies cited above with either 300
mg orally twice daily for 5 to 7 days or 2% vaginal Table 5. CDC Recommendations for Clindamycin
cream nightly for 3 to 6 days.60-62 Recommended Treatment of BV in Pregnancy148
treatment regimens are listed in Table 5. Treatment Dosage

Smoking Cessation and Other Clindamycin cream 2% One full applicator (5 g) intravaginally at
Interventions for the Prevention of PTD bedtime for 7 days
In a large observational study in Sweden, women Alternative Regimens
with a previous PTD and who smoked during Clindamycin 300 mg Orally twice daily for 7 days
that pregnancy had a decreased risk of subsequent Clindamycin ovules 100 mg Intravaginally once at bedtime for 3 days
PTD if they had discontinued smoking.67 Smok-
ing cessation programs reduce the RR of PTD BV = bacterial vaginosis; CDC = Centers for Disease Control and Prevention.
(0.84; CI = 0.72 to 0.98).68 Interventions for
smoking cessation are not equally effective, nor
are they always transferrable to different settings.
Programs using rewards plus social support were Table 6. Assessment of the Patient Presenting with
most effective.68 Risk of PTD is not improved Premature Contractions72,77
with regular nursing contact,69 periodontal care,70
or nutritional supplementation.71 Question Assessment

Assessment of the Symptomatic Patient Are the membranes History


ruptured? Leakage of fluid from cervical os during
When evaluating a woman presenting with pre- sterile speculum examination
mature contractions, goals of assessment include Nitrazine reaction of fluid
determining if membranes are ruptured, if infec- Ferning of fluid
tion is present, and stratifying her risk of PTD Placental alpha microglobulin-1 test
(Table 6). The diagnosis of PTL is typically based (AmniSure)
on the clinical criteria of regular uterine contrac- Ultrasound for oligohydramnios
Amnioinfusion of indigo carmine (if above
tions accompanied by change of cervical dilation tests are non-diagnostic)
or effacement or both in a woman at less than
Is infections present? Sexually transmitted infection status
37 weeks’ gestational age. Initial presentation with
Urinary tract infection
regular uterine contractions and cervical dila- Chorioamnionitis, possibly subclinical
tion of at least 2 cm may also be used to diagnose
What is the likelihood History
PTL. Assessment of fetal status and evaluation that the patient will Fetal fibronectin level (if nothing per vagina
for medical complications such as preeclampsia delivery prematurely? for 24 hours)
or abruption are always warranted. A protocol for Transvaginal cervical length ultrasound
evaluation is listed below.

— Chapter D  5
Chapter D

Are the Membranes Ruptured? What is the Likelihood of PTD?


A sterile speculum examination assists in evaluation Diagnosing PTL remains challenging despite the
of membrane integrity and facilitates collection of multitude of clinical tools available to the clinician.
fluid for an fFN test. Digital cervical examinations Fewer than 15% of women presenting to triage
should be avoided to minimize risk of infection. with preterm contractions will go on to deliver pre-
Direct observation of amniotic fluid leaking from maturely.76 In one observational study of women
the cervical os and pooling in the vaginal vault is hospitalized for threatened PTL, 38% went on to
diagnostic of ruptured membranes. Gentle fundal delivery during their first admission, but greater
pressure or having the patient cough during the than half who were discharged to home continued
examination may facilitate leakage. Amniotic fluid, their pregnancy until term.77,78 Although frequency
when allowed to air dry on a slide, will demon- of uterine contractions is significantly related to
strate ferning or arborization under microscopic risk of PTD, contraction frequency alone is not
evaluation. A false-positive test for ferning is pos- sensitive and has a low positive predictive value for
sible if cervical mucus is inadvertently tested. The PTD.79 Assessment of the cervix by digital exami-
normal vaginal environment has a pH of 4.5 to nation is subjective and also has a low predictive
6.0, whereas the pH of amniotic fluid is 7.1 to 7.3. value for PTD.80 Early, accurate diagnosis of PTL
Amniotic fluid will therefore change the color of increases the ability to transfer a laboring mother
nitrazine paper from orange to blue.72 A false-posi- to a facility with an NICU, administer glucocorti-
tive nitrazine test is possible if blood, semen, or BV coids, consider magnesium sulfate for neuroprotec-
is present.72 In equivocal cases, rupture of mem- tion, and initiate prophylactic treatment for GBS.
branes may be diagnosed with a test for placental Fetal fibronectin testing may aid in identify-
alpha microglobulin-1 protein (AmniSure) in cervi- ing women at low risk for PTD who present with
covaginal fluid. In a large meta-analysis, the test for symptoms of PTL. fFN is an extracellular matrix
placental alpha macroglobulin-1 was 96% sensitive glycoprotein found at the maternal-fetal interface,
and 98.9% specific for rupture of membranes.73 In and in normal pregnancies it is almost undetect-
some cases of prolonged rupture of membranes, able in vaginal secretions. With disruption of this
very little fluid may be present for analysis making interface, which occurs before the onset of labor,
diagnosis difficult. Oligohydramnios on ultrasound fFN is released into vaginal secretions. fFN testing
supports the diagnosis of ruptured membranes, may be done between 24 and 34 weeks’ gesta-
but is not diagnostic. If the diagnosis of membrane tion and is most useful to identify women who
rupture remains uncertain after a complete evalua- are at low risk for delivery in the following 10
tion, ultrasound guided transabdominal instillation to 14 days. The negative predictive value of the
of indigo carmine into the amniotic fluid may be fFN assay is greater than 99% for delivery within
used. The passage of blue-dyed fluid through the 14 days, as confirmed by multiple studies.81 How-
vagina, staining a pad or tampon, is diagnostic of ever, the positive predictive value is only 13% to
membrane rupture.72 30% for delivery in 7 to 10 days for symptomatic
patients.82 The test should not be performed when
Is Infection Present? there is active vaginal bleeding, or when inter-
A high index of suspicion for infection should be course, digital vaginal examination, or endovaginal
used when evaluating a woman with preterm con- ultrasound has occurred in the preceding 24 hours
tractions. Testing for sexually transmitted infec- because these can yield a false positive test result.83
tions, urinary tract infection, and BV is warranted.
Subclinical chorioamnionitis, infection without The Role of CL Measurement in
the classic findings of fever, uterine tenderness, PTL Management
foul-smelling discharge, and maternal tachycardia, The presence of a shortened CL has been asso-
may be present.74 Unless group B Streptococcus ciated with preterm birth, although the utility
(GBS) testing has been done in the past 5 weeks, of CL measurement by ultrasound in assessing
a vaginal/rectal culture should be collected. If threatened PTL is not as clear. A retrospective
delivery appears likely and GBS culture is either study of 1,077 women presenting with preterm
unavailable or positive, GBS prophylaxis should be contractions found that although cervical shorten-
administered.75 ing was associated with increased risk of delivery

6  Chapter D —
Preterm Labor and Premature Rupture of Membranes

in the following 7 to 14 days, the overall accuracy


of CL alone to predict PTD was relatively poor.76 Figure 1. Risk of Preterm Delivery Within 7 Days
Another prospective study of 665 women com- Using Fetal Fibronectin and Cervical Length for
bined both CL and fFN testing to risk stratify Risk Stratification
women presenting for PTL. This study found Positive fFN Negative fFN
that women with CL of at least 30 mm had a low 60
risk of delivery within 7 days (0.7%), regardless 52.0

Preterm Delivery Within 7 Days (%)


of the fFN result. Women with a CL less than 50
15 mm had a high risk of delivery within 7 days
(27% if fFN was negative, and 52% if fFN was 40
positive). For women with an intermediate CL
27.0
between 15 mm and 30 mm, a negative fFN result 30
correctly identified women at low risk (less than
5%) for delivery within 7 days (Figure 1).77 These 20
14.0
results are consistent with results from a similar
study conducted in 2005.84 By using tests such 10
as fFN and CL measurements, which have high 3.2 2.6
0.0
negative predictive values, it may be possible to 0
Cervical length Cervical length Cervical length
identify women at low risk for PTD. However, >30 mm between 15 and 30 mm <15 mm
there is no test for early PTL with a high positive
predictive value, and accurate determination of fFN = fetal fibronectin.
PTL remains a challenging diagnosis. Information from van Baaren GJ, Vis JY, Wilms FF, et al. Predictive value of cer-
vical length measurement and fibronectin testing in threatened preterm labor.
Management of PTL Obstet Gynecol. 2014;123(6):1185-1192.
After PTL has been diagnosed, several interven-
tions should be implemented to improve neonatal
outcomes, including maternal transfer to a facility less than 33 weeks’ gestation who remain at risk
with a higher level nursery if indicated, adminis- of delivery within 7 days, and who received their
tration of corticosteroids, antibiotic prophylaxis first ACS dose at least 14 days prior, receive a
of neonatal GBS infection, consideration of single repeat course of ACS.85,86 However, further
magnesium for neuroprotection, and preparation doses of ACS do not show additional benefit and
for a preterm birth. Each of these interventions is are not recommended.88
discussed in more detail below.

Antenatal Corticosteroid (ACS) Treatment Table 7. Effect of ACS on Fetal


Treatment with ACS decreases neonatal mortality Outcomes in PTL87
and morbidity as listed in Table 7.
Treatment with ACS does not increase the Outcome RR (95% CI)
risk of maternal mortality, chorioamnionitis, or
Neonatal mortality 0.69 (0.58 to 0.81)
puerperal sepsis.85 ACS use is effective in women
Respiratory distress 0.66 (0.59 to 0.73)
with PROM and pregnancy-related hypertension syndrome
syndromes.72,85 A course of ACS is recommended
Intraventricular hemorrhage 0.54 (0.43 to 0.69)
for women between 24 and 34 weeks’ gestation
who are at risk of PTD within 7 days. Steroids Necrotizing enterocolitis 0.46 (0.29 to 0.74)
may be considered for women between 23 and Respiratory support, 0.80 (0.65 to 0.99)
24 weeks.86 The optimal effects of ACS appear to intensive care admissions
wane after a week.87 For women who remain at Systemic infections in the 0.56 (0.38 to 0.85)
first 48 hours of life
risk of PTD, a single repeat dose of ACS improves
the outcomes of respiratory distress syndrome ACS = antenatal corticosteroids; CI = confidence inter-
and poor infant outcomes without affecting long val; PTL = preterm labor; RR = relative risk.
term outcomes. It is recommended that women at

— Chapter D  7
Chapter D

A 2016 study suggests that betamethasone men used in magnesium administration. A 2012
reduces respiratory complications when given to Cochrane review did not show superiority of any
women with threatened preterm labor (at least regimen over any other and called for further stud-
3cm dilated or 75% effaced), rupture of mem- ies to define the optimal dosing.91,92 Two com-
branes, or an indication for planned late preterm monly used regimens are listed in Table 9, and are
delivery between 34 0/7 and 36 6/7 weeks. applicable to women between 24 and 32 weeks at
Doses of corticosteroids are noted in Table 8. In high risk of delivery within 24 hours.91,93
general, dexamethasone and betamethasone show
similar results in respiratory distress and perinatal Tocolysis
mortality, but one meta-analysis showed a decrease Tocolytic drugs are used for short-term pregnancy
in intraventricular hemorrhage and length of prolongation (up to 48 hours), with the goal of
NICU stay with use of dexamethasone compared allowing time for administration of ACS, magne-
with betamethasone.89 There is evidence that even sium sulfate for neuroprotection, antibiotics for
a single dose of either agent shows benefit, so it GBS prophylaxis, and maternal transfer if neces-
is recommended that a first dose be given even sary. They may be useful in pregnancies between
if it is unlikely that the patient will receive the viability and 34 weeks’ estimated gestational age
subsequent doses. However, there is no evidence with established PTL, and in the absence of evi-
of improved outcomes with accelerated dosing dence of maternal or fetal compromise (ie, cho-
(ie, shortening the dosage interval to complete the rioamnionitis, severe preeclampsia, maternal
course before an imminent delivery).85,88,89 instability, fetal demise or lethal anomaly, worri-
some fetal status). After cessation of labor, there
Magnesium Sulfate for Neuroprotection is no evidence of benefit of long term tocolysis for
Magnesium sulfate administered immediately further prolongation of pregnancy.85 Pharmaceuti-
before and at the time of delivery of a preterm cal treatment options are listed in Table 10.
infant decreases the rate of cerebral palsy (RR Nifedipine decreases the likelihood of deliver-
0.68; 95% CI = 0.54 to 0.87, number needed to ing within 48 hours (RR 0.30; 95% CI = 0.21 to
treat is 63).90 Various trials have shown this clear 0.43). Nifedipine may show advantages over other
benefit, but have varied in regard to the regi- tocolytics. Compared with betamimetics, nifedip-
ine increased time before birth, decreased maternal
adverse events, and improved neonatal outcomes
Table 8. ACS for Fetal Maturation (very preterm birth, respiratory distress syndrome,
necrotizing enterocolitis, intraventricular hemor-
Corticosteroid Dosage rhage, neonatal jaundice, and NICU admissions).
Compared with magnesium sulfate, nifedipine
Betamethasone Two doses of 12 mg IM twice showed decreased maternal adverse events and
administered 24 hours apart
decreased NICU admissions.94
Dexamethasone Four doses of 6 mg IM
administered every 6 hours
Betamimetics (eg, terbutaline, ritodrine) are
effective in delaying delivery for 48 hours.95 Stud-
ACS = antenatal corticosteroids; IM = intramuscularly. ies have not shown an improvement in measures
of fetal outcomes, but maternal adverse effects and

Table 9. Magnesium Sulfate for Neuroprotection 91,93

Magnesium Sulfate
Loading Dose Maintenance Dose Repeat Treatment

4 g over 20 to 30 minutes 1 g/hour continued until No immediate repeat doses


birth or for 24 hours
6 g over 20 to 30 minutes 2 g/hour continued until If less than 6 hours have elapsed since cessation,
birth or for 12 hours restart maintenance dose. If more than 6 hours have
elapsed, rebolus and then start maintenance dose.

8  Chapter D —
Preterm Labor and Premature Rupture of Membranes

adverse events are significant.95 Terbutaline is the tocolysis longer than 48 to 72 hours because of
most commonly used betamimetic for tocolysis. the lack of demonstrated efficacy and the poten-
In 2011, the US Food and Drug Administration tial for serious maternal cardiac complications
issued a warning against the use of terbutaline for and mortality.96 Injectable terbutaline may have

Table 10. Pharmaceutical Agents for Tocolysis

Drug (Class) Dosage Comments Contraindications and Adverse Effects

Magnesium sulfate 4 to 6 g bolus over In widespread use in the Contraindication: myasthenia gravis
15 to 20 minutes, United States, meta-analysis Maternal adverse effects: flushing, lethargy,
then 1 to 2 g/hour fails to show improvement headache, muscle weakness, diplopia,
(3 g/hour maximum) in outcomes. Comparison dry mouth, pulmonary edema, cardiac
studies show similar arrest. Toxicity rare with serum level
effectiveness to other <10 mg/dL. Respiratory depression and
agents in delay of delivery. subsequent arrest can occur at levels
May be used for fetal >10 to 12 mg/dL.
neuroprotection at less than Newborn adverse effects: lethargy,
32 weeks. Dosing regimens hypotonia, respiratory depression,
vary; use hospital protocols. demineralization with prolonged use
Nifedipine (Calcium 30 mg oral loading Nifedipine increases the Contraindication: maternal hypotension
channel blocker) dose, then 10 to incidence of delivery Maternal adverse effects: flushing,
20 mg every 4 to before 48 hours. Neonatal headache, dizziness, nausea, transient
6 hours mortality not affected. hypotension
Decreased incidence of No fetal adverse effects noted.
neonatal respiratory distress
syndrome, necrotizing
enterocolitis, intraventricular
hemorrhage, and jaundice.
Terbutaline 0.25 mg subcutaneously Betamimetic drugs may delay Maternal contraindications: heart disease,
(Betamimetic) every 20 minutes for delivery for 48 hours, but poorly controlled diabetes, thyrotoxicosis
up to three doses neonatal outcomes are Maternal adverse effects:
variable and maternal Cardiac arrhythmias, pulmonary edema,
adverse effects are myocardial ischemia, hypotension,
common. tachycardia
Injectable terbutaline may Hyperglycemia, hyperinsulinemia,
have a narrow role in hypokalemia, antidiuresis, altered
treatment of tachysystole or thyroid function
in emergent situations.
Physiologic tremor, palpitations,
US FDA warning against long- nervousness, nausea/vomiting, fever,
term or oral use because hallucinations
of maternal adverse events
and lack of efficacy. Fetal and neonatal adverse effects:
tachycardia, hypoglycemia,
hypocalcemia, hyperbilirubinemia,
hypotension, intraventricular hemorrhage
Indomethacin Loading dose: 50 mg NSAIDs theoretically intervene Contraindications: maternal renal or hepatic
(NSAID) rectally or 50 to more proximally in the labor impairment, active peptic ulcer disease,
100 mg orally cascade than the other oligohydramnios
Maintenance dose: agents. Efficacy appears Maternal adverse effects: nausea, heartburn
25 to 50 mg orally similar to other agents. Fetal adverse effects: constriction of the
every 4 hours for Maternal adverse-effect profile ductus arteriosus (not recommended
48 hours is favorable. after 32 weeks’ gestation), pulmonary
Other NSAIDs (sulindac, hypertension, reversible decrease in
ketorolac) may be used. renal function with oligohydramnios,
intraventricular hemorrhage,
hyperbilirubinemia, necrotizing enterocolitis

FDA = Food and Drug Administration; NSAID = nonsteroidal anti-inflammatory drug.

— Chapter D  9
Chapter D

a narrow role for tocolysis in emergent settings or infection among neonates.75 Several large retro-
with uterine tachysystole.85 spective record reviews show gaps in adherence
Indomethacin, a nonsteroidal anti-inflammatory to current recommendations, especially in PTL,
drug (NSAID), used as a tocolytic agent increases PPROM, and the use of clindamycin as opposed
the likelihood of delivery at greater than 37 weeks to cefazolin in penicillin-allergic patients.105,106
and average gestational age at delivery (weighted A vaginal-rectal swab for GBS culture should
mean difference 3.53 weeks) with low maternal be obtained when women present with PTL or
adverse effects.97 Because NSAIDs can interfere PPROM if results from prior testing done within
with fetal prostaglandin synthesis, concerns have the last 5 weeks are not available. Intrapartum
been raised regarding fetal safety. Meta-analyses antibiotics (penicillin or ampicillin) should be
of observational studies have shown conflicting started on admission and continued until birth,
results in regard to fetal safety (mostly no effects), or until it is determined that the woman is not in
but have raised concerns about severe intraven- true PTL or a negative GBS culture result becomes
tricular hemorrhage, necrotizing enterocolitis, and available. In cases of PPROM when antibiotics are
periventricular leukomalacia.98-100 More research used for prolonging latency, antibiotic coverage
and RCTs are needed in this area. According to should include coverage for GBS.
one decision analysis, prostaglandin inhibitors may Women who are allergic to penicillin should
be the optimal first-line agent for PTL before 32 receive cefazolin unless the allergic response was
weeks’ gestation.101 Because of the risk of pre- anaphylaxis, angioedema, respiratory distress, or
mature closure of the ductus arteriosus, NSAIDs urticaria (Figure 2).75 Clindamycin and vancomy-
should not be used for more than 48 hours or cin are the antibiotics of last resort for women with
beyond 32 weeks’ gestation. life threatening penicillin allergies. GBS isolates
Magnesium sulfate used for tocolysis has not show increasing resistance to clindamycin, and
been shown to prolong pregnancy or improve clindamycin has poor penetration into amniotic
neonatal outcomes when compared with placebo fluid.105 It should be used only when the GBS iso-
or other tocolytics.102,103 In addition, a Cochrane late can be shown to be sensitive to both clindamy-
meta-analysis shows an increased risk of fetal and cin and erythromycin. Vancomycin should be
neonatal mortality of borderline significance (RR used in women with the serious penicillin aller-
4.56; 95% CI = 1.00 to 20.86) and an increase gies described and unknown GBS status, or GBS
in length of NICU admission when compared to strains resistant to clindamycin. CDC algorithms
calcium channel blockers (but not in comparison for screening and GBS prophylaxis treatment of
to NSAIDs).102,103 women with threatened PTD are presented below:
In women receiving magnesium sulfate for neu-
roprotection who continue to labor after admin- Preterm Premature Rupture of the Fetal
istration of magnesium sulfate, consideration Membranes (PPROM)
may be given to addition of another tocolytic. Twenty-five percent to 30% of PTDs are pre-
However, a Cochrane meta-analysis was unable to ceded by PPROM.2 The biochemical events
reach any conclusions regarding combinations of leading to PPROM appear to be different from
tocolytics in regards to safety and efficacy, cit- those leading to PTL. Collagen fibers contribute
ing a lack of large well-designed trials.104 Caution to the tensile strength of the fetal membranes.
should be used if using calcium channel blockers Matrix metalloproteinase (a collagenase) activ-
and magnesium because of theoretical maternal ity is increased, possibly the result of infection or
cardiac complications.85 other inflammation, and may be the final com-
mon pathway leading to membrane rupture. In
Neonatal GBS Prophylaxis the context of placental abruption, thrombin may
The incidence of neonatal GBS infection and be involved as well.107
mortality has dropped significantly since the wide The earlier in pregnancy PROM (and PTL
adoption of the guidelines from the Centers for in general) occurs, the more likely that its etiol-
Disease Control and Prevention (CDC) for the ogy is associated with infection.72 Risk factors for
prevention of neonatal GBS disease. However, PPROM are similar to those for PTL with intact
GBS remains the leading cause of mortality due to membranes.108 Delivery is likely within a week of

10  Chapter D —
Preterm Labor and Premature Rupture of Membranes

rupture. However, the earlier in pregnancy the are complications of prematurity. Intrauterine
rupture occurs, the greater the potential latency complications include umbilical cord compression,
period.109 Clinically evident, intra-amniotic infec- abruption of the placenta, infection and pulmo-
tion will develop in 13% to 60% of cases, the nary developmental abnormalities.111 Infection
likelihood of which is increased by digital vaginal may lead to maternal morbidity and likely plays a
examination.72,110 Principal threats to the fetus role in initiation of labor.

Figure 2. Algorithm for Screening for Group B Streptococcal (GBS) Colonization and
Use of Intrapartum Prophylaxis for Women With Preterm Labor (PTL)75

Patient admitted with signs and


symptoms of preterm labor*

Obtain vaginal-rectal swab for GBS culture†

Patient entering true labor?¶

No Yes

Continue GBS prophylaxis Discontinue GBS prophylaxis


until delivery**

Obtain GBS culture results

Positive Not available prior to labor Negative


onset and patient still preterm

No GBS prophylaxis at onset of true


labor;†† repeat vaginal-rectal culture
if patient reaches 35-37 weeks’ gesta-
GBS prophylaxis at
tion and has not yet delivered§§
onset of true labor

*At < 37 weeks and 0 days’ gestation.


†If patient has undergone vaginal-rectal GBS culture within the preceding 5 weeks, the results of that culture should
guide management. GBS-colonized women should receive intrapartum antibiotic prophylaxis. No antibiotics are indicated
for GBS prophylaxis if a vaginal-rectal screen within 5 weeks was negative.
§See Figure 3 below for recommended antibiotic regimens.
**Patient should be regularly assessed for progression to true labor; if the patient is considered not to be in true labor,
discontinue GBS prophylaxis.
***If GBS culture results become available prior to delivery and are negative, then discontinue GBS prophylaxis.
††Unless subsequent GBS culture prior to delivery is positive.

§§A negative GBS screen is considered valid for 5 weeks. If a patient with a history of PTL is re-admitted with signs and

symptoms of PTL and had a negative GBS screen >5 weeks prior, she should be rescreened and managed according to
this algorithm at that time.
Reprinted from Verani JR, McGee L, Schrag SJ; Division of Bacterial Diseases, National Center for Immunization and
Respiratory Diseases, Centers for Disease Control and Prevention (CDC). Prevention of perinatal group B streptococcal
disease—revised guidelines from CDC, 2010. MMWR Recomm Rep. 2010;59(RR-10):1-36.

— Chapter D  11
Chapter D

Figure 3. Recommended Regimens for Intrapartum Antibiotic Prophylaxis for


Prevention of Early-onset Group B Streptococcal (GBS) Disease

Patient allergic to penicillin?

No Yes

Penicillin G, 5 million units IV Patient with a history of any of


initial dose, then 2.5-3.0 million the following after receiving
units† every 4 hours until delivery penicillin or a cephalosporin?§
or • Anaphylaxis
Ampicillin, 2 g IV initial dose, then • Angioedema
1 g IV every 4 hours until delivery • Respiratory distress
• Urticaria

No Yes

Cefazolin, 2 g IV initial dose, then Isolate susceptible to


1 g IV every 8 hours until delivery clindamycin§ and erythromycin?**

No Yes

Vancomycin, 1 g IV every Clindamycin, 900 mg IV


12 hours until delivery every 8 hours until delivery

*Broader spectrum agents, including an agent active against GBS, might be necessary for treatment of chorioamnionitis.
†Doses ranging from 2.5 to 3.0 million units are acceptable for the doses administered every 4 hours following the initial
dose. The choice of dose within that range should be guided by which formulations of penicillin G are readily available to
reduce the need for pharmacies to specially prepare doses.
§Penicillin-allergic patients with a history of anaphylaxis, angioedema, respiratory distress, or urticaria following adminis-
tration of penicillin or a cephalosporin are considered to be at high risk for anaphylaxis and should not receive penicillin,
ampicillin, or cefazolin for GBS intrapartum prophylaxis. For penicillin-allergic patients who do not have a history of those
reactions, cefazolin is the preferred agent because pharmacologic data suggest it achieves effective intraamniotic con-
centrations. Vancomycin and clindamycin should be reserved for penicillin-allergic women at high risk for anaphylaxis.
¶If laboratory facilities are adequate, clindamycin and erythromycin susceptibility testing should be performed on pre-
natal GBS isolates from penicillin-allergic women at high risk for anaphylaxis. If no susceptibility testing is performed, or
the results are not available at the time of labor, vancomycin is the preferred agent for GBS intrapartum prophylaxis for
penicillin-allergic women at high risk for anaphylaxis.
**Resistance to erythromycin is often but not always associated with clindamycin resistance. If an isolate is resistant to
erythromycin, it might have inducible resistance to clindamycin, even if it appears susceptible to clindamycin. If a GBS
isolate is susceptible to clindamycin, resistant to erythromycin, and testing for inducible clindamycin resistance has
been performed and is negative (no inducible resistance), then clindamycin can be used for GBS intrapartum prophylaxis
instead of vancomycin.
IV = intravenously.
Reprinted from Verani JR, McGee L, Schrag SJ; Division of Bacterial Diseases, National Center for Immunization and
Respiratory Diseases, Centers for Disease Control and Prevention (CDC). Prevention of perinatal group B streptococcal
disease—revised guidelines from CDC, 2010. MMWR Recomm Rep. 2010;59(RR-10):1-36.

Initial Evaluation of the Patient with Sterile speculum examination: If rupture


• 
Suspected PPROM of membranes is suspected, digital examina-
Accurate dating is critical: Review dating
•  tion should be avoided because it shortens
criteria as management choices are determined the latency period before onset of labor and
by gestational age. increases the risk of infection. Diagnosis of

12  Chapter D —
Preterm Labor and Premature Rupture of Membranes

rupture of membranes was previously PPROM antibiotic regimens that include


discussed. ampicillin 1 g intravenously every 6 hours are
• Ultrasound
  evaluation: Oligohydramnios adequate for GBS prophylaxis, even though
supports the diagnosis of membrane rupture. a 4-hour dosing interval is typical for routine
Oligohydramnios will also decrease the accuracy GBS intrapartum prophylaxis.75 One gram oral
of fetal weight and gestational assessment. Low dose of azithromycin has been substituted in
amniotic volume increases the likelihood of cord some protocols for erythromycin with no appar-
compression and other complications. ent change in outcomes.119,120 The antibiotics
• Assessment
  of fetal lung maturity: Vaginal and dosages used in a large National Institute
amniotic fluid may be tested for lamellar body of Child Health and Human Development trial
count and phosphatidyl glycerol to evaluate are listed in Table 11.121
fetal lung maturity between 32 and 34 weeks’ • Corticosteroids:
  ACS administration in the
gestation.112,113 Amniocentesis allows collection setting of PPROM reduces the risk of neonatal
of fluid for fetal pulmonary maturity testing and respiratory distress syndrome, intraventricular
for evaluation of infection.108 hemorrhage, and necrotizing enterocolitis.87
• Screen
  for infection: Infection is associated There is a trend to decreased neonatal mortality.
with PPROM. If GBS status is unknown, then Incidence of maternal and neonatal infection is
culture should be obtained. Consider screening not increased.87,122 Corticosteroid dosages are
for sexually transmitted infections. listed in Table 10.
• Fetal
  monitoring: Electronic fetal heart rate • Tocolysis:
  A common argument for the use of
and uterine contraction monitoring during tocolytics in PTL is that delaying delivery allows
initial assessment may identify fetal compromise for the administration of corticosteroids. As
and asymptomatic contractions opposed to antibiotic and corticosteroid treat-
ment, tocolysis in the context of PPROM lacks
Management of PPROM evidence of benefit.123 Association of infection
As in the case of PTL with intact membranes, the with onset of labor would be expected to be
management of PPROM necessitates a balance stronger in PPROM as opposed to PTL with
between the advantages of delaying delivery and intact membranes indicating circumspection in
risks of prolonging fetal exposure to a poten- choosing tocolysis.
tially hostile environment. Initial management is • Magnesium
  sulfate for neuroprotection: In a
predicated on gestational age with the expected secondary analysis of a RCT, magnesium sulfate
benefits of expectant management and admin- did not change neonatal outcomes or deliv-
istration of corticosteroids increasing for earlier ery latency.124 However, magnesium sulfate is
gestational ages. The potential for development of
infection and the decreasing likelihood of neonatal
morbidity leads to the current standard practice Table 11. Antibiotic Therapy in PPROM121
of induction of labor at 34 weeks.114 Aside from Antibiotic Dosage
that standard, clinical trials have not demonstrated
clear advantages to elective delivery.115,116 Initial Therapy
• Monitor
  for clinical infection: Maternal fever, Ampicillin 2 g IV every 6 hours for
uterine tenderness, and fetal tachycardia are 48 hours
indicators of infection. Erythromycin 250 mg IV every 6 hours
• Antepartum
  fetal testing: Recommendations for 48 hours
for antenatal surveillance are based primar- Follow-Up Therapy
ily on expert opinion. The non-stress test and Amoxicillin 250 mg orally every
biophysical profile are used to assess for fetal 8 hours for 5 days
compromise.108,117 Erythromycin base 333 mg orally every
8 hours for 5 days
• Antibiotic
  therapy: At gestations between 24
and 32 weeks, management with antibiotics IV = intravenously; PPROM = preterm premature rupture
prolongs pregnancy and decreases fetal morbid- of membranes.
ity, chorioamnionitis, and maternal infection.118

— Chapter D  13
Chapter D

typically administered according to one of the 28 hours after PROM.72,126 Steps to diagnose rup-
several protocols for fetal neuroprotection. ture of membrane were discussed previously.
Maternal and neonatal infection are the primary
Gestational Age and Management concerns when PROM is not followed quickly
of PPROM by spontaneous labor. Antibiotic prophylaxis and
• 37
  0/7 or more weeks: Induce labor. If early induction have shown benefit in reducing
unknown GBS and no other indication for infection in infants of mothers with PROM who
GBS prophylaxis, then there is no need to start are colonized with GBS.127 Planned induction
antibiotics unless there has been rupture of (compared with expectant management) shows
membranes greater than 18 hours or tempera- a reduction in chorioamnionitis and endometri-
ture ≥38°C (100.4°F). tis without affecting risk of operative delivery.
• 34
  0/7 to 36 6/7 weeks: Offer expectant man- Planned induction had no effect on the number
agement versus induction. If GBS unknown, of neonatal infections but did reduce the number
start antibiotics pending results. See the Ante- of NICU admissions.128 Routine administration
natal Corticosteroid (ACS) Treatment section for of antibiotics in PROM does not reduce neonatal
2016 evidence regarding benefit of betametha- sepsis, maternal infections, stillbirth or neonatal
sone in this time frame. A 2016 study demon- mortality, although the quality of the studies
strated that expectant management of PPROM included are judged to be low.129
between 34 0/7 and 36 6/7 weeks’ gestation
(late preterm) resulted in significantly lower Planned Delivery Versus Expectant
rates of respiratory distress, mechanical ventila- Management in Patient Not in Labor
tion, days spent in NICU, and cesarean delivery Labor induction with oxytocin is recommended
but significantly higher rates of antepartum or when term PROM occurs and labor does not
intrapartum hemorrhage, intrapartum fever, use follow.128 In one trial women preferred active to
of postpartum antibiotics, and longer hospital expectant management.126 However, the differ-
stay.125 The authors concluded that offering ences in maternal and neonatal outcomes are
expectant management for women with late minimal for induction versus expectant manage-
preterm PPROM is reasonable.125 ment. If mother and fetus are well, then expect-
• 24
  0/7 to 33 6/7 weeks: Administer antibiot- ant management of PROM would be acceptable
ics and corticosteroids. Add magnesium sulfate along with appropriate counseling regarding
for neuroprotection when in labor if under 32 potential risk.128 Women who are colonized
weeks gestational age. Monitor for infection and with GBS should receive antibiotic prophylaxis
other intrauterine fetal complications. If there is and be encouraged to proceed with induction to
no evidence of fetal compromise and labor does reduce neonatal GBS infection.75,127 Other routine
not begin spontaneously, these pregnancies are administration of antibiotics does not appear to
managed expectantly until they reach 34 weeks. affect maternal or neonatal outcomes.129
• 23
  0/7 to 23 6/7 weeks: Consider administering
antibiotics and corticosteroids based on parental Delivery of the Preterm Infant
choice only after counseling with neonatology Despite prevention efforts, PTD affected 1 in
and maternal-fetal medicine clinicians regarding 9 pregnancies in the United States in 2013 and
outcomes of interventions including cesarean remains a major factor in infant morbidity and
delivery and resuscitation at periviable gestational mortality.1 A few interventions can improve out-
age. If resuscitation is planned, add magnesium comes for those infants who are born prematurely.
sulfate for neuroprotection when in labor.86 Premature delivery at a facility with a high-
volume level III NICU results in better neonatal
PROM at Term outcomes. If delivery is not imminent and level
Rupture of the membranes before the onset of labor III services are unavailable, then maternal trans-
occurs in 8% of pregnancies beyond 37 weeks’ ges- fer is indicated.130,131 For fetuses between 24 to
tation. In most women, PROM at term is followed 34 weeks’ gestation, administration of ACS, as
quickly by spontaneous labor. Ninety-five percent discussed previously, decreases the rate of respira-
of women managed expectantly will deliver within tory distress syndrome, intracranial hemorrhage,

14  Chapter D —
Preterm Labor and Premature Rupture of Membranes

and necrotizing enterocolitis. Administration of developed a web-based tool for describing out-
magnesium for neuroprotection can decrease the comes of these neonates based on gestational age,
rate of cerebral palsy. weight, sex, and administration of ACS, which
is available at http://www.nichd.nih.gov/about/
Delayed Umbilical Cord Clamping org/cdbpm/pp/prog_epbo/epbo_case.cfm. The
For the preterm infant, delayed clamping of the physician counseling the parents should also know
umbilical cord has been shown to decrease intra- the most recent outcome data for their own NICU
ventricular hemorrhage and the need for neonatal facility to assist with decision making.
transfusion.132,133 To perform delayed cord clamp-
ing, the infant should be held at or below the level Global Implications
of the placenta while waiting 30 to 120 seconds Low-income countries often do not have the
before clamping the cord. Required neonatal resources needed for the prevention and manage-
resuscitation efforts should not be delayed to allow ment of PTL. Infants born at 32 weeks, who may
for delayed cord clamping. Delayed cord clamp- have had a normal life in other parts of the world,
ing is recommended as a standard practice in the may die of respiratory failure because a respirator
delivery of the preterm infant. However, further and surfactant are unavailable, as is an infrastructure
research and larger studies are indicated to confirm for rapid transport to a tertiary care facility. Com-
and refine these findings.133,134 plex social and medical infrastructure necessary for
technology-based care and follow-up of preterm
Route of Delivery neonates may not exist in these settings. In 2012, a
Retrospective observational studies have not coalition of experts released Born Too Soon: The
shown a clear benefit to any particular route of Global Action Report on Preterm Birth. It lists the
delivery for the preterm infant.135 The choice of first country-level estimates of preterm birth and
vaginal or cesarean birth should be made based revealed that there are about 15 million preterm
on standard obstetrical indications. The rate of births per year137 with over 85% occurring in Asia
cesarean delivery is higher for preterm than for and Africa.138 In 2014, the stakeholders who created
term deliveries because the indications for surgery that report also released Every Newborn: An Action
are more common in prematurity. Preterm fetuses Plan to Avoid Preventable Deaths. The compo-
are more likely to have a malpresentation and are nents of the action plan for preterm birth included
less able to handle the potential stresses of labor. worldwide access to newborn resuscitation, low
Continuous fetal monitoring is important to costs corticosteroids,139 and kangaroo care in which
detect signs of fetal intolerance of labor. Prophy- maternal skin-to-skin care is used in place of an
lactic episiotomy or forceps delivery have not been incubator. Others have urged caution with intro-
shown to have any benefit to the preterm fetus. ducing corticosteroids until there is evidence sup-
Vacuum assisted delivery should not be performed porting benefit in low-resource settings.140 An RCT
at less than 34 weeks because of the risk of intra- on ACS treatment was conducted in six countries:
cranial hemorrhage.135,136 Argentina, Guatemala, India, Kenya, Pakistan, and
Zambia.141 The study showed no benefit from ACS
Infants at the Threshold of Viability treatment and increased rates of maternal infection.
Infants born at 22 to 25 weeks’ gestation are con- Additional research is needed to clarify the role
sidered to be at the threshold of viability. They are of ACS treatment in low-resource settings. More
at a high risk of neonatal mortality and severe long information on PTL in developing countries can be
term disability, and the difference in maturity of found in the Global ALSO Program (available at
even a few days can significantly affect these risks. www.aafp.org/globalalso).
Parents of these infants should receive careful
counseling and accurate information to assist them Conclusion
in making decisions regarding tocolysis, labor It is possible to identify some patients at high risk
management, resuscitation, and NICU interven- for PTD, and a subset of these may benefit from
tions.130 Based on outcome information from preventive interventions such as antenatal proges-
1997 to 2003, the National Institute of Child terone. In the triage of patients presenting with
Health and Human Development (NICHD) has preterm contractions, it is possible to stratify their

— Chapter D  15
Chapter D

risk of subsequent PTD. The major role of tocoly- aged expectantly, there is little research to support
sis is delay of delivery during the 48 hours neces- that approach and some research supports imme-
sary for full therapeutic effect of antenatal steroids. diate induction of labor or induction after a short
Management of PPROM presents its own set of interval of expectant management. PTL remains
challenges and is primarily based on gestational an area of intense research activity and therapeutic
age. Antenatal steroids and antibiotics are useful in evolution. Addressing the role of infection in PTL
some cases. Although term PROM is often man- is a particular area of interest.

SORT: Key Recommendations for Practice


Evidence
Clinical Recommendation Rating References

Antenatal progesterone supplementation decreases the incidence of preterm A 41


delivery in high-risk patients.
Administration of corticosteroids in women with preterm labor between 26 and A 87
34 weeks’ gestation reduces the incidence of neonatal mortality, respiratory
distress syndrome, and intraventricular hemorrhage.
Magnesium sulfate before delivery of infants between 24 to 32 weeks’ gestation A 90
reduces the rate of cerebral palsy.
Betamethasone reduces respiratory complications when given to women with B 142
threatened preterm labor (at least 3 cm dilated or 75% effaced) or ruptures
of membranes or an indication for planned late preterm delivery between
34 0/7 and 36 6/7 weeks.
Expectant management with PPROM between 34 0/7 and 36 6/7 weeks is B 125
associated with significantly lower rates respiratory distress, mechanical
ventilation, days spent in NICU, and cesarean delivery but significantly higher
rates of antepartum or intrapartum hemorrhage, intrapartum fever, use of
postpartum antibiotics, and longer hospital stay.

Strength of Recommendation Taxonomy (SORT)

Strength of Recommendation Definition


A • Recommendation based on consistent and good-quality patient-oriented evidence.
B • Recommendation based on inconsistent or limited-quality patient-oriented evidence.
C • Recommendation based on consensus, usual practice, opinion, disease-oriented evidence, or case series for studies
of diagnosis, treatment, prevention, or screening.
Patient-oriented evidence measures outcomes that matter to patients: morbidity, mortality, symptom improvement, cost
reduction, and quality of life. Disease-oriented evidence measures intermediate, physiologic, or surrogate end points
that might not reflect improvement in patient outcomes (eg, blood pressure, blood chemistry, physiologic function,
pathologic findings).
NICU = neonatal intensive care unit; PPROM = preterm premature rupture of membranes.
From Ebell MH, Siwek J, Weiss BD, et al. Strength of recommendation taxonomy (SORT): a patient-centered approach to
grading evidence in the medical literature. Am Fam Physician. 2004;69(3):548-556.

16  Chapter D —
Preterm Labor and Premature Rupture of Membranes

References
1. Martin JA, Hamilton BE, Osterman MJK. Births in the 17. Berghella V, Tolosa JE, Kuhlman K, Weiner S, Bolognese
United States, 2013. NCHS data brief, no 175. Hyattsville, RJ, Wapner RJ. Cervical ultrasonography compared with
MD: National Center for Health Statistics. Dec 2014. manual examination as a predictor of preterm delivery.
2. Goldenberg RL, Culhane JF, Iams JD, Romero R. Epi- Am J Obstet Gynecol. 1997;177(4):723-730.
demiology and causes of preterm birth. Lancet. 2008; 18. Berghella V, Baxter JK, Hendrix NW. Cervical assess-
371(9606):75-84. ment by ultrasound for preventing preterm delivery.
3. Hamilton BE, Martin JA, Osterman MJK, Curtin SC. Cochrane Database Syst Rev. 2013;1(1):CD007235.
Births: Prelimnary data for 2013. National vital statistics 19. Berghella V, Roman A, Daskalakis C, Ness A, Baxter
reports; vol 63 no 2. Hyattsville, MD: National Center for JK. Gestational age at cervical length measurement
Health Statistics. May 2014. and incidence of preterm birth. Obstet Gynecol. 2007;
4. Schoen CN, Tabbah S, Iams JD, Caughey AB, Berghella 110(2 Pt 1):311-317.
V, Why the United States preterm birth rate is declining. 20. Behrendt N, Gibbs RS, Lynch A, Hart J, West NA, Iams
Am J Obstet Gynecol. 2015;213(2):175-180. JD. Rate of change in cervical length in women with
5. Chang HH, Larson J, Blencowe H, et al; Born Too Soon vaginal bleeding during pregnancy. Obstet Gynecol.
preterm prevention analysis group. Preventing preterm 2013;121(2 Pt 1):260-264.
births: analysis of trends and potential reductions with 21. Fonseca EB, Celik E, Parra M, Singh M, Nicolaides KH;
interventions in 39 countries with very high human Fetal Medicine Foundation Second Trimester Screen-
development index. Lancet. 2013;381(9862):223-234. ing Group. Progesterone and the risk of preterm birth
6. Martin JA, Hamilton BE, Osterman MJ, Curtin SC, Mat- among women with a short cervix. N Engl J Med. 2007;
thews TJ. Births: final data for 2012. Natl Vital Stat Rep. 357(5):462-469.
2013;62(9):1-68. 22. Hassan SS, Romero R, Vidyadhari D, et al; PREGNANT
7. Muglia LJ, Katz M. The enigma of spontaneous preterm Trial. Vaginal progesterone reduces the rate of preterm
birth. N Engl J Med. 2010;362(6):529-535. birth in women with a sonographic short cervix: a mul-
ticenter, randomized, double-blind, placebo-controlled
8. Liong S, Di Quinzio MK, Fleming G, Permezel M, Rice trial. Ultrasound Obstet Gynecol. 2011;38(1):18-31.
GE, Georgiou HM. Prediction of spontaneous preterm
labour in at-risk pregnant women. Reproduction. 2013; 23. Berghella V. Universal cervical length screening for pre-
146(4):335-345. diction and prevention of preterm birth. Obstet Gynecol
Surv. 2012;67(10):653-658.
9. Davey MA, Watson L, Rayner JA, Rowlands S. Risk
scoring systems for predicting preterm birth with 24. Society for Maternal-Fetal Medicine Publications Com-
the aim of reducing associated adverse outcomes. mittee, with assistance of Vincenzo Berghella. Proges-
Cochrane Database Syst Rev. 2011;(11):CD004902. terone and preterm birth prevention: translating clinical
trials data into clinical practice. Am J Obstet Gynecol.
10. Kazemier BM, Buijs PE, Mignini L, Limpens J, de Groot 2012;206(5):376-386.
CJ, Mol BW; EBM CONNECT. Impact of obstetric history
on the risk of spontaneous preterm birth in singleton 25. Leitich H, Bodner-Adler B, Brunbauer M, Kaider A,
and multiple pregnancies: a systematic review. BJOG. Egarter C, Husslein P. Bacterial vaginosis as a risk fac-
2014;121(10):1197-1208, discussion 1209. tor for preterm delivery: a meta-analysis. Am J Obstet
Gynecol. 2003;189(1):139-147.
11. Lykke JA, Paidas MJ, Langhoff-Roos J. Recurring com-
plications in second pregnancy. Obstet Gynecol. 2009; 26. Cunnington M, Kortsalioudaki C, Heath P. Genitourinary
113(6):1217-1224. pathogens and preterm birth. Curr Opin Infect Dis.
2013;26(3):219-230.
12. Ananth CV, Vintzileos AM. Medically indicated preterm
birth: recognizing the importance of the problem. [viii.]. 27. Murtha AP, Edwards JM. The role of Mycoplasma and
Clin Perinatol. 2008;35(1):53-67, viii. Ureaplasma in adverse pregnancy outcomes. Obstet
Gynecol Clin North Am. 2014;41(4):615-627.
13. Stock S, Norman J. Preterm and term labour in multiple
pregnancies. Semin Fetal Neonatal Med. 2010;15(6): 28. Offenbacher S, Boggess KA, Murtha AP, et al. Progres-
336-341. sive periodontal disease and risk of very preterm deliv-
ery. Obstet Gynecol. 2006;107(1):29-36.
14. Martin JA, Hamilton BE, Ventura SJ, Osterman MJ,
Mathews TJ. Births: final data for 2011. Natl Vital Stat 29. Iams JD, Newman RB, Thom EA, et al; National Institute
Rep. 2013;62(1):1-69, 72. of Child Health and Human Development Network of
Maternal-Fetal Medicine Units. Frequency of uterine
15. Iams JD, Goldenberg RL, Meis PJ, et al; National Insti- contractions and the risk of spontaneous preterm deliv-
tute of Child Health and Human Development Maternal ery. N Engl J Med. 2002;346(4):250-255.
Fetal Medicine Unit Network. The length of the cervix
and the risk of spontaneous premature delivery. N Engl 30. Conde-Agudelo A, Rosas-Bermúdez A, Kafury-Goeta
J Med. 1996;334(9):567-572. AC. Birth spacing and risk of adverse perinatal out-
comes: a meta-analysis. JAMA. 2006;295(15):1809-1823.
16. Hernandez-Andrade E, Romero R, Ahn H, et al. Trans-
abdominal evaluation of uterine cervical length during 31. Moutquin JM. Classification and heterogeneity of pre-
pregnancy fails to identify a substantial number of term birth. BJOG. 2003;110(Suppl 20):30-33.
women with a short cervix. J Matern Fetal Neonatal
Med. 2012;25(9):1682-1689.

— Chapter D  17
Chapter D

32. Conner SN, Frey HA, Cahill AG, Macones GA, Colditz 47. Conde-Agudelo A, Romero R, Nicolaides K, et al. Vagi-
GA, Tuuli MG. Loop electrosurgical excision procedure nal progesterone vs. cervical cerclage for the preven-
and risk of preterm birth: a systematic review and meta- tion of preterm birth in women with a sonographic short
analysis. Obstet Gynecol. 2014;123(4):752-761. cervix, previous preterm birth, and singleton gestation:
33. McIntosh J, Feltovich H, Berghella V, Manuck T; Soci- a systematic review and indirect comparison metaanaly-
ety for Maternal-Fetal Medicine (SMFM). Electronic sis. Am J Obstet Gynecol. 2013;208(1):42.e1-42.e18.
address: pubs@smfm.org. The role of routine cervical 48. Romero R, Nicolaides K, Conde-Agudelo A, et al.
length screening in selected high- and low-risk women Vaginal progesterone in women with an asymptomatic
for preterm birth prevention. Am J Obstet Gynecol. sonographic short cervix in the midtrimester decreases
2016;215(3):B2-B7. preterm delivery and neonatal morbidity: a systematic
34. Hansen C, Neller A, Williams G, Simpson R. Maternal review and metaanalysis of individual patient data. Am
exposure to low levels of ambient air pollution and J Obstet Gynecol. 2012;206(2):124.e1-124.e19.
preterm birth in Brisbane, Australia. BJOG. 2006;113(8): 49. Grobman WA, Thom EA, Spong CY, et al; Eunice Ken-
935-941. nedy Shriver National Institute of Child Health and
35. Simpson WJ. A preliminary report on cigarette smoking Human Development Maternal-Fetal Medicine Units
and the incidence of prematurity. Am J Obstet Gynecol. (MFMU) Network. 17 alpha-hydroxyprogesterone capro-
1957;73(4):807-815. ate to prevent prematurity in nulliparas with cervical
length less than 30 mm. Am J Obstet Gynecol. 2012;207
36. Behrman RE, Butler AS, eds. Preterm Birth: Causes, (5):390.e1-390.e8.
Consequences, and Prevention. Washington, DC:
National Academies Press; 2007. 50. Alfirevic Z, Stampalija T, Roberts D, Jorgensen AL.
Cervical stitch (cerclage) for preventing preterm birth
37. McCowan LM, Dekker GA, Chane E, et al;SCOPE in singleton pregnancy. Cochrane Database Syst Rev.
consortium. Spontaneous preterm birth and small for 2012;4(4):CD008991.
gestational age infants in women who stop smoking
early in pregnancy: prospective cohort study. BMJ. 51. Rafael TJ, Berghella V, Alfirevic Z. Cervical stitch (cer-
2009;338:b1081. Erratum in BMJ. 2009;338:b1558. clage) for preventing preterm birth in multiple pregnancy.
Cochrane Database Syst Rev. 2014;9(9):CD009166.
38. Iams JD, Romero R, Culhane JF, Goldenberg RL. Pri-
mary, secondary, and tertiary interventions to reduce 52. Rode L, Tabor A. Prevention of preterm delivery in twin
the morbidity and mortality of preterm birth. Lancet. pregnancy. Best Pract Res Clin Obstet Gynaecol. 2014;
2008;371(9607):164-175. 28(2):273-283.

39. Iams JD. Clinical practice. Prevention of preterm parturi- 53. Arabin B, Alfirevic Z. Cervical pessaries for prevention
tion. N Engl J Med. 2014;370(3):254-261. of spontaneous preterm birth: past, present and future.
Ultrasound Obstet Gynecol. 2013;42(4):390-399.
40. Committee on Practice Bulletins—Obstetrics, The
American College of Obstetricians and Gynecologists. 54. Goya M, Pratcorona L, Merced C, et al; Pesario Cervi-
Practice bulletin no. 130: prediction and prevention of cal para Evitar Prematuridad (PECEP) Trial Group.
preterm birth. Obstet Gynecol. 2012;120(4):964-973. Cervical pessary in pregnant women with a short cervix
(PECEP): an open-label randomised controlled trial.
41. Dodd JM, Jones L, Flenady V, Cincotta R, Crowther CA. Lancet. 2012;379(9828):1800-1806.
Prenatal administration of progesterone for prevent-
ing preterm birth in women considered to be at risk of 55. Liem S, Schuit E, Hegeman M, et al. Cervical pessaries
preterm birth. Cochrane Database Syst Rev. 2013;7(7): for prevention of preterm birth in women with a mul-
CD004947. tiple pregnancy (ProTWIN): a multicentre, open-label
randomised controlled trial. Lancet. 2013;382(9901):
42. O’Brien JM, Adair CD, Lewis DF, et al. Progesterone 1341-1349.
vaginal gel for the reduction of recurrent preterm birth:
primary results from a randomized, double-blind, pla- 56. Romero R, Dey SK, Fisher SJ. Preterm labor: one syn-
cebo-controlled trial. Ultrasound Obstet Gynecol. 2007; drome, many causes. Science. 2014;345(6198):760-765.
30(5):687-696. 57. Macones GA, Parry S, Nelson DB, et al. Treatment of
43. Rebarber A, Ferrara LA, Hanley ML, et al. Increased localized periodontal disease in pregnancy does not
recurrence of preterm delivery with early cessation of reduce the occurrence of preterm birth: results from the
17-alpha-hydroxyprogesterone caproate. Am J Obstet Periodontal Infections and Prematurity Study (PIPS). Am
Gynecol. 2007;196(3):224.e1-224.e4. J Obstet Gynecol. 2010;202(2):147.e1-147.e8.

44. Romero R, Espinoza J, Kusanovic JP, et al. The preterm 58. Smaill F, Vazquez JC. Antibiotics for asymptomatic
parturition syndrome. BJOG. 2006;113(Suppl 3):17-42. bacteriuria in pregnancy. Cochrane Database Syst Rev.
2007;(2):CD000490.
45. Berghella V, Rafael TJ, Szychowski JM, Rust OA, Owen J.
Cerclage for short cervix on ultrasonography in women 59. Leitich H, Kiss H. Asymptomatic bacterial vaginosis
with singleton gestations and previous preterm birth: a and intermediate flora as risk factors for adverse preg-
meta-analysis. Obstet Gynecol. 2011;117(3):663-671. nancy outcome. Best Pract Res Clin Obstet Gynaecol.
2007;21(3):375-390.
46. Owen J, Hankins G, Iams JD, et al. Multicenter random-
ized trial of cerclage for preterm birth prevention in 60. Lamont RF, Duncan SLB, Mandal D, Bassett P. Intra-
high-risk women with shortened midtrimester cervical vaginal clindamycin to reduce preterm birth in women
length. Am J Obstet Gynecol. 2009;201(4):375.e1-375.e8. with abnormal genital tract flora. Obstet Gynecol.
2003;101(3):516-522.

18  Chapter D —
Preterm Labor and Premature Rupture of Membranes

61. Ugwumadu A, Manyonda I, Reid F, Hay P. Effect of early 77. van Baaren GJ, Vis JY, Wilms FF, et al. Predictive value
oral clindamycin on late miscarriage and preterm deliv- of cervical length measurement and fibronectin testing
ery in asymptomatic women with abnormal vaginal flora in threatened preterm labor. Obstet Gynecol. 2014;123
and bacterial vaginosis: a randomised controlled trial. (6):1185-1192.
Lancet. 2003;361(9362):983-988. 78. McPheeters ML, Miller WC, Hartmann KE, et al. The epi-
62. Kiss H, Petricevic L, Husslein P. Prospective ran- demiology of threatened preterm labor: a prospective
domised controlled trial of an infection screening pro- cohort study. Am J Obstet Gynecol. 2005;192(4):1325-
gramme to reduce the rate of preterm delivery. BMJ. 1329, discussion 1329-1330.
2004;329(7462):371-374. 79. Berghella V, Iams JD, Newman RB, et al; National Insti-
63. Brocklehurst P, Gordon A, Heatley E, Milan SJ. Anti- tute of Child Health and Human Development Network
biotics for treating bacterial vaginosis in pregnancy. of Maternal-Fetal Medicine Units. Frequency of uterine
Cochrane Database Syst Rev. 2013;1(1):CD000262. contractions in asymptomatic pregnant women with or
64. U.S. Preventive Services Task Force. Screening for bac- without a short cervix on transvaginal ultrasound scan.
terial vaginosis in pregnancy to prevent preterm delivery: Am J Obstet Gynecol. 2004;191(4):1253-1256.
U.S. Preventive Services Task Force recommendation 80. Gomez R, Galasso M, Romero R, et al. Ultrasonographic
statement. Ann Intern Med. 2008;148(3):214-219. examination of the uterine cervix is better than cervical
65. Oliver RS, Lamont RF. Infection and antibiotics in the digital examination as a predictor of the likelihood of pre-
aetiology, prediction and prevention of preterm birth. mature delivery in patients with preterm labor and intact
J Obstet Gynaecol. 2013;33(8):768-775. membranes. Am J Obstet Gynecol. 1994;171(4):956-964.

66. Amsel R, Totten PA, Spiegel CA, Chen KC, Eschenbach 81. Anwar A, Lindow SW, Greaves L, Hall S, Jha R. The
D, Holmes KK. Nonspecific vaginitis. Diagnostic criteria use of fetal fibronectin in suspected pre-term labour.
and microbial and epidemiologic associations. Am J J Obstet Gynaecol. 2014;34(1):45-47.
Med. 1983;74(1):14-22. 82. Honest H, Bachmann LM, Gupta JK, Kleijnen J, Khan
67. Cnattingius S, Granath F, Petersson G, Harlow BL. The KS. Accuracy of cervicovaginal fetal fibronectin test in
influence of gestational age and smoking habits on the predicting risk of spontaneous preterm birth: systematic
risk of subsequent preterm deliveries. N Engl J Med. review. BMJ. 2002;325(7359):301.
1999;341(13):943-948. 83. Ramsey PS, Andrews WW. Biochemical predictors of
68. Coleman T, Chamberlain C, Davey MA, Cooper SE, preterm labor: fetal fibronectin and salivary estriol. Clin
Leonardi-Bee J. Pharmacological interventions for pro- Perinatol. 2003;30(4):701-733.
moting smoking cessation during pregnancy. Cochrane 84. Gomez R, Romero R, Medina L, et al. Cervicovaginal
Database Syst Rev. 2012;9(9):CD010078. fibronectin improves the prediction of preterm delivery
69. Dyson DC, Danbe KH, Bamber JA, et al. Monitoring based on sonographic cervical length in patients with
women at risk for preterm labor. N Engl J Med. 1998; preterm uterine contractions and intact membranes.
338(1):15-19. Am J Obstet Gynecol. 2005;192(2):350-359.

70. Michalowicz BS, Hodges JS, DiAngelis AJ, et al; OPT 85. American College of Obstetricians and Gynecologists;
Study. Treatment of periodontal disease and the risk of Committee on Practice Bulletins—Obstetrics. ACOG
preterm birth. N Engl J Med. 2006;355(18):1885-1894. practice bulletin no. 127: Management of preterm labor.
Obstet Gynecol. 2012;119(6):1308-1317.
71. Kramer MS, Kakuma R. Energy and protein intake in
pregnancy. Cochrane Database Syst Rev. 2003;(4): 86. American College of Obstetricians and Gynecologists.
CD000032. Practice Bulletin No. 159: Management of Preterm
Labor. Obstet Gynecol. 2016;127(1):e29-e38.
72. Practice bulletins No. 139: premature rupture of mem-
branes. Obstet Gynecol. 2013;122(4):918-930. 87. Roberts D, Dalziel S. Antenatal corticosteroids for
accelerating fetal lung maturation for women at risk of
73. Ramsauer B, Vidaeff AC, Hösli I, et al. The diagnosis preterm birth. Cochrane Database Syst Rev. 2006;(3):
of rupture of fetal membranes (ROM): a meta-analysis. CD004454.
J Perinat Med. 2013;41(3):233-240.
88. Crowther CA, McKinlay CJD, Middleton P, Harding JE.
74. Ratcliffe SD, Baxley EG, Cline MK, Sakornbut EL, eds. Repeat doses of prenatal corticosteroids for women at
Family Medicine Obstetrics. 3rd ed. Philadelphia, PA: risk of preterm birth for improving neonatal health out-
Elsevier, 2008. 448-457. comes. Cochrane Database Syst Rev. 2011;(6):CD003935.
75. Verani JR, McGee L, Schrag SJ; Division of Bacte- 89. Brownfoot FC, Gagliardi DI, Bain E, Middleton P, Crowther
rial Diseases, National Center for Immunization and CA. Different corticosteroids and regimens for accelerat-
Respiratory Diseases, Centers for Disease Control and ing fetal lung maturation for women at risk of preterm
Prevention (CDC). Prevention of perinatal group B strep- birth. Cochrane Database Syst Rev. 2013;8(8):CD006764.
tococcal disease—revised guidelines from CDC, 2010.
MMWR Recomm Rep. 2010;59(RR-10):1-36. 90. Doyle LW, Crowther CA, Middleton P, Marret S, Rouse
D. Magnesium sulphate for women at risk of preterm
76. Melamed N, Hiersch L, Domniz N, Maresky A, Bardin birth for neuroprotection of the fetus. Cochrane Data-
R, Yogev Y. Predictive value of cervical length in women base Syst Rev. 2009;(1):CD004661.
with threatened preterm labor. Obstet Gynecol. 2013;
122(6):1279-1287. 91. Reeves SA, Gibbs RS, Clark SL. Magnesium for fetal
neuroprotection. Am J Obstet Gynecol. 2011;204(3):202.
e1-202.e4.

— Chapter D  19
Chapter D

92. Bain E, Middleton P, Crowther CA. Different magnesium 107. Thomson A, Norman J. Biology of preterm labour. In:
sulphate regimens for neuroprotection of the fetus for Norman J, Greer I, eds. Preterm Labour, managing risk
women at risk of preterm birth. Cochrane Database in clinical practice. Cambridge, UK: Cambridge Univer-
Syst Rev. 2012;2(2):CD009302. sity Press; 2005. 26-75.
93. Magee L, Sawchuck D, Synnes A, von Dadelszen P. 108. Simhan HN, Canavan TP. Preterm premature rupture
SOGC Clinical Practice Guideline. Magnesium sulphate of membranes: diagnosis, evaluation and management
for fetal neuroprotection. J Obstet Gynaecol Can. 2011; strategies. BJOG. 2005;112(Suppl 1):32-37.
33(5):516-529. 109. Melamed N, Hadar E, Ben-Haroush A, Kaplan B, Yogev
94. Flenady V, Wojcieszek AM, Papatsonis DNM, et al. Cal- Y. Factors affecting the duration of the latency period
cium channel blockers for inhibiting preterm labour and in preterm premature rupture of membranes. J Matern
birth. Cochrane Database Syst Rev. 2014;6(6):CD002255. Fetal Neonatal Med. 2009;22(11):1051-1056.
95. Neilson JP, West HM, Dowswell T. Betamimetics for 110. Alexander JM, Mercer BM, Miodovnik M, et al. The
inhibiting preterm labour. Cochrane Database Syst Rev. impact of digital cervical examination on expectantly
2014;2(2):CD004352. managed preterm rupture of membranes. Am J Obstet
96. US Food and Drug Administration. FDA Drug Safety Gynecol. 2000;183(4):1003-1007.
Communication: New warnings against use of terbuta- 111. Mercer BM, Crocker LG, Boe NM, Sibai BM. Induction
line to treat preterm labor. Available at http://www.fda. versus expectant management in premature rupture
gov/Drugs/DrugSafety/ucm243539.htm. of the membranes with mature amniotic fluid at 32 to
97. Khanprakob T, Laopaiboon M, Lumbiganon P, Sang- 36 weeks: a randomized trial. Am J Obstet Gynecol.
komkamhang US. Cyclo-oxygenase (COX) inhibitors 1993;169(4):775-782.
for preventing preterm labour. Cochrane Database Syst 112. Lewis DF, Futayyeh S, Towers CV, Asrat T, Edwards MS,
Rev. 2012;10(10):CD007748. Brooks GG. Preterm delivery from 34 to 37 weeks of
98. Loe SM, Sanchez-Ramos L, Kaunitz AM. Assessing the gestation: is respiratory distress syndrome a problem?
neonatal safety of indomethacin tocolysis: a system- Am J Obstet Gynecol. 1996;174(2):525-528.
atic review with meta-analysis. Obstet Gynecol. 2005; 113. Besnard AE, Wirjosoekarto SA, Broeze KA, Opmeer
106(1):173-179. BC, Mol BW. Lecithin/sphingomyelin ratio and lamellar
99. Hammers AL, Sanchez-Ramos L, Kaunitz AM. Ante- body count for fetal lung maturity: a meta-analysis. Eur
natal exposure to indomethacin increases the risk J Obstet Gynecol Reprod Biol. 2013;169(2):177-183.
of severe intraventricular hemorrhage, necrotizing 114. Naef RW III, Allbert JR, Ross EL, Weber BM, Martin RW,
enterocolitis, and periventricular leukomalacia: a sys- Morrison JC. Premature rupture of membranes at 34
tematic review with metaanalysis. Am J Obstet Gynecol. to 37 weeks’ gestation: aggressive versus conservative
2015;212(4):505.e1-505.e13. management. Am J Obstet Gynecol. 1998;178(1 Pt 1):
100. Amin SB, Sinkin RA, Glantz JC. Metaanalysis of the 126-130.
effect of antenatal indomethacin on neonatal outcomes. 115. van der Ham DP, van der Heyden JL, Opmeer BC, et al.
Am J Obstet Gynecol. 2007;197(5):486.e1-486.e10. Management of late-preterm premature rupture of mem-
101. Haas DM, Imperiale TF, Kirkpatrick PR, Klein RW, branes: the PPROMEXIL-2 trial. Am J Obstet Gynecol.
Zollinger TW, Golichowski AM. Tocolytic therapy: 2012;207(4):276.e1-276.e10.
a meta-analysis and decision analysis. Obstet Gynecol. 116. Buchanan SL, Crowther CA, Levett KM, Middleton P,
2009;113(3):585-594. Morris J. Planned early birth versus expectant man-
102. Crowther CA, Brown J, McKinlay CJD, Middleton P. agement for women with preterm prelabour rupture of
Magnesium sulphate for preventing preterm birth in membranes prior to 37 weeks’ gestation for improving
threatened preterm labour. Cochrane Database Syst pregnancy outcome. Cochrane Database Syst Rev.
Rev. 2014;8(8):CD001060. 2010;(3):CD004735.

103. Mercer BM, Merlino AA; Society for Maternal-Fetal 117. Sharp GC, Stock SJ, Norman JE. Fetal assessment
Medicine. Magnesium sulfate for preterm labor and pre- methods for improving neonatal and maternal out-
term birth. Obstet Gynecol. 2009;114(3):650-668. comes in preterm prelabour rupture of membranes.
Cochrane Database Syst Rev. 2014;10(10):CD010209.
104. Vogel JP, Nardin JM, Dowswell T, West HM, Oladapo
OT. Combination of tocolytic agents for inhibiting pre- 118. Kenyon S, Boulvain M, Neilson JP. Antibiotics for pre-
term labour. Cochrane Database Syst Rev. 2014;7(7): term rupture of membranes. Cochrane Database Syst
CD006169. Rev. 2013;12(12):CD001058.

105. Fairlie T, Zell ER, Schrag S. Effectiveness of intrapar- 119. Pierson RC, Gordon SS, Haas DM. A retrospective com-
tum antibiotic prophylaxis for prevention of early-onset parison of antibiotic regimens for preterm premature rup-
group B streptococcal disease. Obstet Gynecol. 2013; ture of membranes. Obstet Gynecol. 2014;124(3):515-519.
121(3):570-577. 120. Haas D. Preterm Premature Rupture of Membranes:
106. American College of Obstetricians and Gynecologists Erythromycin Versus Azithromycin a Randomized
Committee on Obstetric Practice. ACOG Committee Trial Comparing Their Efficacy to Prolong Latency
Opinion No. 485: Prevention of early-onset group B (PEACE Trial). Available at http://clinicaltrials.gov/show/
streptococcal disease in newborns. Obstet Gynecol. NCT01556334.
2011;117(4):1019-1027.

20  Chapter D —
Preterm Labor and Premature Rupture of Membranes

121. Mercer BM, Miodovnik M, Thurnau GR, et al; National 134. Tarnow-Mordi WO, Duley L, Field D, et al. Timing of
Institute of Child Health and Human Development cord clamping in very preterm infants: more evidence is
Maternal-Fetal Medicine Units Network. Antibiotic needed. Am J Obstet Gynecol. 2014;211(2):118-123.
therapy for reduction of infant morbidity after preterm 135. American College of Obstetrics and Gynecology. Oper-
premature rupture of the membranes. A randomized ative vaginal delivery. Clinical management guidelines
controlled trial. JAMA. 1997;278(12):989-995. for obstetrician-gynecologists. Int J Gynaecol Obstet.
122. Harding JE, Pang J, Knight DB, Liggins GC. Do 2001;74(1):69-76.
antenatal corticosteroids help in the setting of pre- 136. Cargill YM, MacKinnon CJ, Arsenault MY, et al; Clinical
term rupture of membranes? Am J Obstet Gynecol. Practice Obstetrics Committee. Guidelines for operative
2001;184(2):131-139. vaginal birth. J Obstet Gynaecol Can. 2004;26(8):747-761.
123. Mackeen AD, Seibel-Seamon J, Muhammad J, Bax- 137. Lawn JE, Kinney MV, Belizan JM, et al; Born Too Soon
ter JK, Berghella V. Tocolytics for preterm premature Preterm Birth Action Group. Born too soon: accelerating
rupture of membranes. Cochrane Database Syst Rev. actions for prevention and care of 15 million newborns
2014;2(2):CD007062. born too soon. Reprod Health. 2013;10(Suppl 1):S6.
124. Horton AL, Lai Y, Rouse DJ, et al; Eunice Kennedy 138. March of Dimes. White Paper on Preterm Birth: The
Shriver National Institute of Child Health and Human Global and Regional Toll. 2009. Available at http://www.
Development Maternal-Fetal Medicine Units Net- marchofdimes.org/materials/white-paper-on-preterm-
work. Effect of magnesium sulfate administration for birth.pdf.
neuroprotection on latency in women with preterm
premature rupture of membranes. Am J Perinatol. 139. Lawn JE, Blencowe H, Oza S, et al; Lancet Every New-
2015;32(4):387-392. born Study Group. Every Newborn: progress, priorities,
and potential beyond survival. Lancet. 2014;384(9938):
125. Morris JM, Roberts CL, Bowen JR, et al; PPROMT Col- 189-205.
laboration. Immediate delivery compared with expect-
ant management after preterm pre-labour rupture of the 140. Azad K, Costello A. Extreme caution is needed before
membranes close to term (PPROMT trial): a randomised scale-up of antenatal corticosteroids to reduce preterm
controlled trial. Lancet. 2016;387(10017):444-452. deaths in low-income settings. Lancet Glob Health.
2014;2(4):e191-e192.
126. Hannah ME, Ohlsson A, Farine D, et al; TERMPROM
Study Group. Induction of labor compared with expect- 141. Althabe F, Belizán JM, McClure EM, et al. A population-
ant management for prelabor rupture of the membranes based, multifaceted strategy to implement antenatal
at term. N Engl J Med. 1996;334(16):1005-1010. corticosteroid treatment versus standard care for the
reduction of neonatal mortality due to preterm birth in
127. Hannah ME, Ohlsson A, Wang EE, et al. Maternal low-income and middle-income countries: the ACT clus-
colonization with group B Streptococcus and prelabor ter-randomised trial. Lancet. 2015;385(9968):629-639.
rupture of membranes at term: the role of induction of
labor. TermPROM Study Group. Am J Obstet Gynecol. 142. Gyamfi-Bannerman C, Thom EA, Blackwell SC, et al;
1997;177(4):780-785. NICHD Maternal–Fetal Medicine Units Network. Antena-
tal Betamethasone for Women at Risk for Late Preterm
128. Dare MR, Middleton P, Crowther CA, Flenady VJ, Delivery. N Engl J Med. 2016;374(14):1311-1320.
Varatharaju B. Planned early birth versus expectant
management (waiting) for prelabour rupture of mem- 143. Makhlouf MA, Clifton RG, Roberts JM, et al; Eunice
branes at term (37 weeks or more). Cochrane Database Kennedy Shriver National Institute of Child Health
Syst Rev. 2006;(1):CD005302. Human Development Maternal-Fetal Medicine Units
Network. Adverse pregnancy outcomes among women
129. Wojcieszek AM, Stock OM, Flenady V. Antibiotics with prior spontaneous or induced abortions. Am J Peri-
for prelabour rupture of membranes at or near term. natol. 2014;31(9):765-772.
Cochrane Database Syst Rev. 2014;10(10):CD001807.
144. Kesmodel U, Olsen SF, Secher NJ. Does alcohol
130. American College of Obstetricians and Gynecologists. increase the risk of preterm delivery? Epidemiology.
ACOG Practice Bulletin: Clinical Management Guide- 2000;11(5):512-518.
lines for Obstetrcian-Gynecologists: Number 38, Sep-
tember 2002. Perinatal care at the threshold of viability. 145. Li D, Liu L, Odouli R. Presence of depressive symptoms
Obstet Gynecol. 2002;100(3):617-624. during early pregnancy and the risk of preterm deliv-
ery: a prospective cohort study. Hum Reprod. 2009;
131. Warner B, Musial MJ, Chenier T, Donovan E. The effect 24(1):146-153.
of birth hospital type on the outcome of very low birth
weight infants. Pediatrics. 2004;113(1 Pt 1):35-41. 146. Negro R, Stagnaro-Green A. Thyroid autoantibodies,
preterm birth, and miscarriage. BMJ. 2011;342:d2260.
132. Rabe H, Diaz-Rossello JL, Duley L, Dowswell T. Effect
of timing of umbilical cord clamping and other strate- 147. Bodnar LM, Platt RW, Simhan HN. Early-pregnancy
gies to influence placental transfusion at preterm birth vitamin D deficiency and risk of preterm birth subtypes.
on maternal and infant outcomes. Cochrane Database Obstet Gynecol. 2015;125(2):439-447.
Syst Rev. 2012;8(8):CD003248. 148. Workowski KA, Berman S; Centers for Disease Control
133. Committee on Obstetric Practice, American College of and Prevention (CDC). Sexually transmitted diseases
Obstetricians and Gynecologists. Committee Opinion treatment guidelines, 2010. MMWR Recomm Rep.
No.543: Timing of umbilical cord clamping after birth. 2010;59(RR-12):1-110.
Obstet Gynecol. 2012;120(6):1522-1526.

— Chapter D  21
Objectives
Preterm Labor and Premature 
• Discuss risk factors associated with preterm labor (PTL) 
Rupture of Membranes and premature rupture of fetal membranes (PROM) 
• Identify patients who may benefit from antenatal 
progesterone
Revised January 2017 • Define PTL and PROM, and describe their significance
• Outline initial evaluation of PTL and PROM
• Describe management of PTL and PROM
• Discuss neonatal group B streptococcal (GBS) prevention 
strategies

Preterm Birth Rates in United States by 
Epidemiology of Preterm Births
Race from 1990‐2013
• Incidence 11.4% • Spontaneous preterm
labor intact membranes
45%
• PPROM 25%
• Electively induced 30%

Prevention of Preterm Delivery: 
Risk Factors History of PTD
• History of preterm delivery • Interpregnancy interval • Management
<6 months – 17 alpha‐hydroxyprogesterone caproate (17P) 250 mg IM weekly from 16 to
• Multiple gestation 20 weeks through 36 weeks gestation 
• Short cervical length on • Tobacco use – Check cervical length via ultrasound every 2 weeks from 16‐22 weeks
– Consider cerclage for cervical length <25 mm
endovaginal ultrasound • Infection
• Benefit of 17P for women with history of preterm delivery
– Bacterial vaginosis – 0.31 RR of birth at <34 weeks [CI = 0.14 to 0.69]
– <25 mm at 18 to 25 weeks
– Bacteriuria – 0.50 RR of neonatal mortality [CI = 0.33 to 0.75]
• Non‐Hispanic black race – Urinary tract infection 
(UTI) Information from Dodd JM, Jones L, Flenady V, Cincotta R, Crowther CA. Prenatal administration of progesterone for preventing preterm birth in women considered to be at 
risk of preterm birth. Cochrane Database Syst Rev. 2013;7(7):CD004947.

Copyright 2017© American Academy of Family Physicians. All rights reserved. 1


Prevention of PTD: Ultrasound Short Cervix Assessment of the Symptomatic Patient
• Cervical length ≤20 mm at ≤24 weeks • Maternal and fetal well‐being
• Vaginal progesterone 90 mg gel, or 200 mg capsule daily from  • Is she in labor?
diagnosis until 36 weeks
• What is the likelihood of delivery?
• Benefit
– Delivery <33 weeks RR 0.58 [CI = 0.42 to 0.80] • Are the membranes ruptured?
– Reduced neonatal morbidity/mortality RR 0.57 [CI = 0.40 to 0.81] • Is there infection present?

Information from Romero R, Nikolaides K, Conde‐Agudelo A, et al. Vaginal progesterone in women with an asymptomatic sonographic short cervix in the 


midtrimester decreases preterm delivery and neonatal morbidity: a systematic review and metaanalysis of individual patient data. Am J Obstet Gynecol. 
2012;206(2):124.e1‐124.e19.

Defining Preterm Labor Assessing Likelihood of Preterm Delivery
• Preterm • Fetal fibronectin (fFN)
– Clearly establish gestational dating • Cervical length by ultrasound
• Labor
– Regular uterine contractions with: 
• Cervical effacement ≥80%

• Cervical dilation ≥2 cm

Fetal Fibronectin (fFN) Cervical Length Measurement
• Placental glycoprotein
• Negative predictive value (NPV) >99% for delivery in 7 to 
14 days
• Positive predictive value (PPV) 13% to 30% for delivery in 
7 to 10 days
– Test inaccurate and not to be performed in presence of vaginal 
bleeding or if within 24 hours of:
• Intercourse
• Digital vaginal examination • Measurement is on the residual length 
• Endovaginal ultrasound • Funnel length is not included in cervical length

2
Risk of Preterm Delivery Within 7 Days Using Fetal  Is Infection Present?
Fibronectin and Cervical Length for Risk Stratification
60.0%
52.0%
• Group B Streptococcus carrier status
Preterm Delivery Within 7 days (%)

50.0%
– If unknown, obtain swab during evaluation

40.0%

30.0% 27.0% Sexually transmitted infection (STI) status


20.0%
14.0% • Bacterial vaginosis
10.0%

0.0%
3.2%
0.0%
2.6%
• Trichomonas
Cervical length  >30mm Cervical length between 15 and 30 mm Cervical length   <15 mm

Positive fFN Negative fFN • Urinary infection


• Chorioamnionitis
Information from van Baaren GJ, Vis JY, Wilms FF, et al. Predictive Value of cervical Length Measurement and Fibronectin Testing in Threatened Preterm
Labor. Obstet Gynecol. 2014;123(6):1185-1192.

Are the Membranes Ruptured? Management of Preterm Labor
• Sterile speculum  • Antenatal corticosteroids
examination
– Pool of fluid in vaginal vault
• Tocolysis
– Nitrazine testing • Transfer to higher level of care
– Ferning of amniotic fluid
• GBS prophylaxis
• Oligohydramnios by 
ultrasound
• Amnisure (Placental alpha 
microglobulin‐1 test) Ferning pattern of dried amniotic fluid

Tocolysis Antenatal Corticosteroid Treatment
• Tocolysis is used to delay delivery up to 48 hours for: • 24 to 34 weeks (consider after 23 weeks)
– Antenatal steroids • Effective: intact or ruptured membranes
– GBS prophylaxis • Betamethasone: 12 mg IM every 24 hours X two doses
– Magnesium infusion for neuroprotection • Dexamethasone: 6 mg IM every 12 hours X four doses
– Transport to a higher level of care if needed • A single repeat course of steroids is indicated if: 
• No indication for long‐term tocolysis – There is continued risk of PTD and EGA is <33 weeks
– AND it has been >14 days from the last steroid administration

3
Betamethasone Between 
Drugs for Tocolysis
34 0/7 to 36 6/7 weeks • Nifedipine
• 2016 study supports betamethasone use between 34 0/7 to 36 6/7
• Decreased respiratory complications • Terbutaline
• Only administer if at least 3cm dilated OR 75% effaced OR spontaneous 
rupture of membranes • Indomethacin
• Don’t administer if already received course of steroids for threatened 
preterm labor within the last 14 days • Magnesium sulfate
• Don’t administer if chorioamnionitis
• Don’t administer if tocolysis
• Don’t use in setting of multiple gestation, diabetes, previous exposure to 
steroids during pregnancy, or pregnancies with major non‐lethal fetal 
malformations

Nifedipine Terbutaline
• Decreased likelihood of  • Typically fewer maternal  • Decreased likelihood of delivery within 48 hours
delivery within 48 hours adverse effects than other  • May be useful in triage setting when patients have uterine 
tocolytics tachysystole: fast administration and onset of action
• Improved neonatal  • O.25 mg SQ every 20 to 30 minutes for up to three doses
outcomes – Flushing
• Contraindications
– Headache
• Contraindication: maternal  – Heart disease
– Edema – Poorly controlled diabetes
hypotension
– Dizziness – Thyrotoxicosis
• 30 mg loading dose • Maternal adverse effects: multiple and significant US FDA warnings 
– Nausea
– Then 10 to 20 mg orally every  against use for longer than 48 to 72 hours because of proven harm 
4 to 6 hours and lack of evidence of long term benefits

Indomethacin Indomethacin
• Effective in increasing EGA at delivery by average of  • 50 mg oral or rectal loading dose
3.5 weeks – Then 25 to 50 mg orally every 4 to 6 hours
– 200 mg/24 hours maximum 
• Niches
– Limit use to 48 hours due to effect on fetal ductus arteriosus and renal 
– PTL <32 weeks
function
– PTL associated with polyhydramnios
• Fetal adverse effects
– Closure of ductus arteriosus
– Oligohydramnios

4
Magnesium Sulfate Delivery of the Premature Infant
• Not an effective tocolytic (no prolongation of pregnancy, no improvement  • Interventions to improve neonatal outcomes
in outcomes)
• Still used frequently around United States – Delivery at facility with NICU
• Contraindication: myasthenia gravis – Antenatal corticosteroids
• Maternal adverse effects are numerous including: – Magnesium for neuroprotection
– Flushing, lethargy, headache, weakness, diplopia, pulmonary edema, cardiac or 
respiratory arrest – Delayed cord clamping
• Magnesium sulfate used for fetal neuroprotection may coincide with tocolysis
– In this setting, magnesium may be considered as tocolytic for up to 48 hours if contractions  – Appropriate route of delivery
have stopped
– If contractions continue while magnesium is being used for neuroprotection, a second drug 
– Skilled neonatal resuscitation including avoidance of heat 
may be needed for tocolysis. Studies are inconclusive regarding which tocolytic to use in  loss
this setting.  

Magnesium Sulfate: 
Magnesium Sulfate: Neuroprotection
Neonatal Neuroprotection
• Decreases cerebral palsy when given immediately  Magnesium sulfate Maintenance dose Repeat treatment
loading dose
prior to and up through the time of delivery 4 g over 20 to 30 minutes 1 g/hour continued until birth  No immediate repeat doses

• Indicated for 24 to 32 weeks EGA or for 24 hours

• Several published protocols have been shown to 
6 g over 20 to 30 minutes 2 g/hour continued until birth  If less than 6 hours have elapsed since cessation, 
or for 12 hours restart maintenance dose.  If more than 6 hours 
be effective have elapsed, rebolus and then start maintenance 

dose.

Delayed Cord Clamping Delivery Route
• Decreases intraventricular hemorrhage and need for  • Gestational age does not dictate delivery route
transfusion • Premature fetuses have a higher rate of malpresentation
• After delivery, hold the infant at or below the level of  and less tolerance of labor stresses, resulting in a higher 
the placenta for 1 to 3 minutes prior to clamping and  rate of cesarean delivery
cutting the umbilical cord • Vacuum delivery is contraindicated under 34 weeks due 
• In severely depressed infants, do not delay neonatal  to risk of intracranial hemorrhage
resuscitation efforts in order to allow for delayed  • Forceps delivery or an episiotomy does not improve or 
cord clamping worsen fetal outcomes

5
GBS Prophylaxis GBS Prophylaxis
• Universal screening • Women who should receive intrapartum antimicrobial 
– Vaginal and rectal GBS colonization prophylaxis include:
– At 35 to 37 weeks gestation, or at the time patient  – GBS bacteriuria in any concentration during current pregnancy
presents with PTL – Previous birth of an infant with GBS disease
• Women with negative vaginal and rectal GBS – Positive GBS vaginal‐rectal screening culture
– Unknown GBS status at the onset of labor AND GBS risk factors
screening cultures done at 35 weeks or later do not
• ROM >18 hours
require intrapartum antimicrobial prophylaxis for • T >100.4°F (38°C)
GBS, even if obstetric risk factors develop • EGA <37 weeks

GBS Prophylaxis in PTL GBS Antibiotic Choice
• Obtain GBS swab at presentation • Penicillin (PCN) or ampicillin first‐line
• Start antibiotics if <37 weeks EGA and if either: • Cefazolin second‐line for PCN allergy (unless allergy was 
– GBS status is unknown  anaphylaxis or urticaria)
– GBS status is positive • Clindamycin only if severe PCN allergy AND culture 
• Discontinue if not progressing to true labor proven sensitivity to clindamycin and erythromycin
• Vancomycin for severe PCN allergy and either unknown 
• Restart according to GBS culture status and risk
sensitivity OR clindamycin or erythromycin resistance
factors if labor restarts

Preterm Premature Rupture of Fetal 
PPROM: 24 to 34 weeks
Membranes (PPROM)
• Risks similar to PTL with intact membranes
• Corticosteroids
• Clinically evident intra‐amniotic infection develops in 13% to 60%  • Antibiotics 24 to 32 weeks: ampicillin, erythromycin, and 
of cases amoxicillin
• Intrapartum complications • Antibiotics 32 to 37 weeks: if GBS is unknown and no 
– Umbilical cord compression other reason to give prophylaxis, start antibiotics 
– Abruption of placenta pending culture
– Pulmonary developmental abnormalities
• Neonatal complications similar to PTD with intact membranes
• Tocolysis: Controversial? If used, usually only so that a 
course of antenatal corticosteroids can be given and/or 
• Likelihood of delivery in short term increases with gestational age
at presentation to allow transport to a facility with a higher level NICU

6
2016 study Supporting Expectant 
ROM Without Labor
Management of PPROM at 34 to 37 weeks
• <37 weeks 
– GBS prophylaxis if GBS status is unknown • Found lower rates of respiratory distress, mechanical 
• ≥37 weeks  ventilation, days spent in NICU, and Cesarean 
– Expedite delivery in setting that is able to care for preterm infant delivery 
• <34 weeks • Higher rates of antepartum or intrapartum 
– Admit to hospital with NICU for expectant management if no evidence 
of infection or fetal concerns hemorrhage, intrapartum fever, use of postpartum 
– Give antenatal corticosteroids antibiotics, and longer hospital stays
– Administer ampicillin and erythromycin to prolong latent labor

Prolonged Rupture of Membranes
Summary
(PROM) at Term
• Identify patients at risk for PTD and offer 
• Consider risk of infection
• GBS prophylaxis
progesterone if appropriate early in pregnancy
– If positive culture on rapid test or GBS unknown with intrapartum risk     • Fetal fibronectin and cervical length can be used to 
factors including: risk stratify patients with threatened preterm labor
• Rupture of membranes ≥18 hours
• Temperature ≥100.4°F (38.0°C) • The major role of tocolysis is to delay delivery for 48 
• Elective induction of labor is associated with decreased NICU  hours to allow for the full effect of antenatal steroids 
admissions
– For GBS colonized mothers, rates of neonatal infection decreased

Summary
• Improved outcomes in PTD with
– Antenatal corticosteroids
– GBS prophylaxis
– Magnesium for neuroprotection
– Delivery at facility with NICU
• Gestational age determines PPROM management
• Screen all women for GBS at 35 to 37 weeks EGA, earlier if 
they present with PTL
• Antibiotic prophylaxis as indicated
• Treat term PROM with expeditious delivery

7
Chapter E
Intrapartum Fetal Surveillance

Barbara A. True, MN, CNS, RNC-OB, C-EFM, R. Eugene Bailey, MD, FAAFP
Revised January 2016

Learning Objectives
After completing this activity, learners will be able to:
1. Summarize fetal monitoring techniques. 4. Develop an overall assessment and management
2. Explain the concepts of structured intermittent plan for CEFM and SIA, especially for Category II
auscultation (SIA) and continuous electronic fetal tracings, using the mnemonic DR C BRAVADO
monitoring (CEFM). and published algorithms.
3. Apply standardized National Institute of Child 5. Discuss future research in fetal monitoring.
Health and Human Development (NICHD)
terminology when interpreting CEFM (NICHD
1997, revised 2008).

Introduction tages and disadvantages, and providers should be aware


Fetal monitoring is essential during the labor and deliv- of these.
ery process. Use of continuous electronic fetal moni- Structured intermittent auscultation is a noninvasive
toring (CEFM) remains the mainstay technique used technique. While this technique sounds simple, provid-
during labor and delivery,1 but structured intermittent ers need to be sure they are obtaining the FHR and not
auscultation (SIA) can be applied to low-risk pregnan- the maternal heart rate.6-8 This can be done by simulta-
cies. Overall, CEFM has resulted in a reduction in neously palpating or electronically reading the maternal
neonatal seizures but no significant decrease in cerebral radial pulse and listening for the FHR. Auscultation also
palsy, infant mortality, or other standard measures of can be a difficult technique to perform in obese patients.
infant well-being.2 CEFM has resulted in increased use Continuous electronic fetal monitoring can be exter-
of cesarean delivery and operative vaginal deliveries.2 nal or internal. External CEFM consists of a Doppler
Use of CEFM is limited by interobserver and intrao- ultrasound to capture the FHR and a tocotransducer
bserver variability in interpretation, mainly as a result (tocodynamometer) to detect uterine activity. External
of lack of standardization.3 Management guidelines monitors require frequent readjusting to obtain an
based on standardized interpretation have recently adequate tracing and signal quality, and they may be
been developed to provide a framework for applying difficult to use in patients who are obese, ambulating,
to clinical practice the information obtained through active in bed, or pushing in varied birth positions.
fetal monitoring. This includes a five-tier classification Internal CEFM consists of an FSE and an intrauter-
of tracings and a separate algorithm for interpreting ine pressure catheter (IUPC) to record contractions.
Category II tracings.4,5 Rupture of membranes and cervical dilatation are
required for placement. Morbidity, although uncom-
Fetal Heart Rate Surveillance Techniques mon, includes maternal and fetal soft tissue injuries
There are two techniques for assessing fetal heart rate and intrauterine infections. Relative contraindications
(FHR): (1) SIA using a handheld Doppler device or include active maternal genital herpes, HIV infection,
fetal stethoscope and (2) CEFM via external electronic hepatitis C virus infection, and any presentation in
fetal monitor or internal electronic fetal monitor (fetal which placement would not be possible, such as pla-
spiral electrode [FSE]). Both techniques have advan- centa previa.9 In addition, FSEs should not be placed

Copyright 2017© American Academy of Family Physicians. All rights reserved. — Chapter E  1
Chapter E

in the setting of undiagnosed vaginal bleeding The method of subsequent fetal monitoring will
until placenta previa is ruled out by ultrasound. then be determined based on the interpretation of
The electrode may be placed on the buttocks in this 20-minute tracing and other factors including
breech presentations; however, care must be taken risk factors, institutional policies, and patient and
to avoid placement on the fetal genitalia. The FSE provider preferences. In some cases, women will
captures the FHR using an electrocardiogram remain on the fetal monitor longer if the tracing is
signal. The IUPC allows for the accurate calibra- suspicious. A 2012 systematic review of more than
tion of uterine activity using Montevideo units 13,000 women demonstrated that compared with
(MVUs), measured in mm Hg. Adequate uterine SIA, an EFM tracing in triage for low-risk women
activity is generally MVUs of 180 to 240 mm Hg showed no benefit and actually increased the risk
with 91% of women delivering successfully with of cesarean delivery by approximately 20%.11 In
MVUs of 200 to 224 mm Hg.10 another systematic review of three randomized
controlled trials including 11,259 women, and
Considerations in Selecting Fetal 11 observational studies including 5,831 women,
Surveillance Technique no significant difference was found in outcomes
The ability to use SIA is dependent upon an when comparing women who underwent EFM on
adequate nurse-to-patient ratio and training in admission with those who did not.12
SIA. In some settings, CEFM is the only option.
Selection of monitoring technique depends on Indications for CEFM
risk factors present, patient and medical/nursing Indications for CEFM include maternal medi-
care provider preferences, available resources, and cal complications (ie, type 1 diabetes), maternal
departmental policies. obstetrical complications (ie, preeclampsia), intra-
partum complications, use of uterine stimulants,
Risk for Woman and Fetus and known fetal conditions (ie, anomalies, anemia,
The decision to choose SIA or CEFM begins with intrauterine growth restriction [IUGR]). If a preg-
assessing maternal medical and obstetrical risk nancy is considered low-risk, the American Col-
factors, and fetal risks for uteroplacental insuf- lege of Obstetricians and Gynecologists (ACOG)
ficiency. After assessing the risk factors, a decision and the Association of Women’s Health, Obstetric
can be made regarding optimal fetal surveillance. and Neonatal Nurses (AWHONN) consider inter-
mittent auscultation an acceptable choice.6,9,13,14
Staff Availability and Comfort Level ACOG recommends that women with high-risk
One of the critical steps in choosing a fetal surveil- conditions should undergo CEFM.13
lance technique is consideration of the personnel
available in the maternity care unit, especially in Effect of Fetal Surveillance on Patient,
the case of SIA. The nurses’ comfort levels with Support Personnel, and Staff
SIA should be assessed. In-service training can be The effect that fetal surveillance has on all indi-
offered to familiarize nurses with the auscultative viduals present during labor and delivery must be
monitoring technique or enhance their skills and considered. Use of CEFM may decrease maternal
comfort level.6,9 mobility and reduce contact between the woman
and her labor partner. In addition, CEFM may dis-
Informed Consent of the Patient tract maternity providers and nurses if they focus on
A discussion of labor monitoring should occur the monitor rather than on the care and attention
before labor onset so options can be explored and to the laboring woman. CEFM should not be used
questions answered. Advantages and disadvantages as a substitute for continuous care during labor.
of CEFM and SIA should be reviewed, and patient
preferences can be determined. CEFM Outcomes
The only clinically significant benefit shown
Electronic Fetal Monitoring at Admission with routine CEFM is the reduction of neonatal
In the United States most, if not all, women in seizures in the immediate newborn period. This is
labor in a hospital maternity care unit will have an a short-term benefit because at the end of 1 year,
EFM placed continuously for at least 20 minutes. these infants did not have permanent sequelae.2

2  Chapter E —
Intrapartum Fetal Surveillance

An unrealistic expectation persists that an abnor- Successful implementation of SIA can be


mal FHR tracing will predict and allow for the achieved keeping in mind the following guidelines:8
prevention of cerebral palsy.13 The incidence of 1. The presence of nurses and providers experi-
cerebral palsy has been stable since the introduc- enced in auscultation, palpation of contractions,
tion of CEFM. Cerebral palsy is attributed to and auditory recognition of FHR changes are
events that occur before labor in approximately necessary.
70% of cases. Only 4% of cases caused by 2. Institutional policy should be developed
hypoxic ischemic encephalopathy can be directly to address the technique and frequency of
linked to intrapartum events.13 In newborns with assessment.
an estimated fetal weight greater than or equal to 3. Clinical interventions should follow when con-
2,500 g (5.5 lb), it has been estimated that the cerning findings are present.
positive predictive value of an abnormal FHR 4. Nurse-to-laboring woman ratio is 1:1.6,9,15
tracing for cerebral palsy is 0.14%,13 and the false- Obtaining the FHR via auscultation can be a
positive rate of EFM for cerebral palsy is greater time-consuming and challenging process when
than 99%.13 Considering there are no clear ben- occurring at every 5- to 30-minute intervals. In
efits to CEFM but there are associated increases in addition, the nurse actually has a 1:2 ratio in
obstetrical interventions, providers should allow that he/she is caring for both the woman and
for the use of SIA in low-risk pregnancies. the fetus(s).
Any potential benefit of CEFM should be 5. User-friendly documentation tools for record-
evaluated based on a woman’s risk status. A joint ing SIA findings.
decision between the pregnant woman and her 6. Ready availability of auscultation devices.
provider should then be made regarding use of 7. Culture embracing normalcy of childbirth and
CEFM versus SIA during labor. In maternity minimization of unnecessary interventions.
centers that offer SIA, appropriately selected
patients should be educated and encouraged to Interpretation of SIA Findings
use this modality. To date, no studies have been Normal findings for SIA include a normal FHR
conducted to assess the optimal frequency for SIA baseline between 110 to 160 bpm regular rhythm,
in the absence of risk factors. Professional organi- presence of FHR increases from baseline, and the
zations differ slightly as to their recommendations absence of FHR decreases from baseline. These
for SIA frequency (Table 1). findings indicate a fetus that is well-oxygenated
General guidelines for performing SIA have been and non-acidotic, which can be followed in a
set forth by AWHONN and ACNM (Table 2). routine manner. SIA findings of FHR baselines

Table 1. Professional Organization Recommendations for SIA

Latent Phase Active First Stage Active Second Stage

ACOG13 Every 15 min Every 5 min


ACOG/AAP Guidelines Every 15 min Every 5 min
for Perinatal Care14
ACNM15 Every 15 to 30 min Every 15 min, every 5 min
with initiation of pushing
AWHONN9 Every 15 to 30 min Every 5 to 15 min
RCOG16 Every 15 min Every 5 min
SOGC 17 At time of assessment Every 15 to 30 min Every 5 min
and approximately
every hour thereafter

AAP = American Academy of Pediatrics; ACNM = American College of Nurse-Midwives; ACOG = American College of
Obstetricians and Gynecologists: AWHONN = Association of Women’s Health, Neonatal and Obstetric Nurses; RCOG =
Royal College of Obstetricians and Gynaecologists; SOGC = Society of Obstetricians and Gynaecologists of Canada.

— Chapter E  3
Chapter E

describes FHR tracings as visual patterns that


Table 2. Guidelines for Auscultation6,9,15 should be adaptable to computerized interpreta-
tion, and notes that definitions and categorizations
Procedure for Auscultation
should be applied to intrapartum tracings but also
1. P
 alpate the abdomen to determine the position can be used in the antepartum period.13,19
of the fetus (Leopold maneuvers).
When performing CEFM, it is recommended
2. Place the Doppler over the area of maximum
intensity of fetal heart tones, generally over the that the monitor strips be reviewed frequently and
fetal back. that healthcare providers periodically document
3. Differentiate maternal heart rate from FHR by their assessments.13 In uncomplicated patients, fetal
palpating maternal radial artery. monitor strips should be reviewed every 30 min-
4. P
 alpate for uterine contraction during period of utes during the first stage of labor, and every
FHR auscultation to determine relationship. 15 minutes during the second stage. If the preg-
5. Count FHR between contractions for at least nancy is complicated (ie, IUGR, preeclampsia), the
30 to 60 seconds to determine the average
baseline rate.
monitor strip should be reviewed more frequently:
every 15 minutes during the first stage of labor and
6. C
 ount FHR after uterine contraction using multiple
consecutive 6- to 10-second intervals for 30 to every 5 minutes during the second stage.13
60 seconds to determine differences between
baseline FHR and fetal response to contractions DR C BRAVADO
(this may be subject to local protocols).
The mnemonic DR C BRAVADO—(DR = deter-
FHR = fetal heart rate.
mine risk), contractions (C), the fetal monitor strip
(BRA = baseline rate, V = variability, A = accelera-
tions, and D = decelerations), and interpretation
greater than or equal to 160 bpm or less than or (O = overall assessment)—is a systematic approach
equal to 110 bpm, irregular rhythms, and the to the interpretation of FHR tracings for CEFM;
presence of either gradual or abrupt decreases from it was developed specifically for the ALSO pro-
the baseline may be indicative of a fetus who is not gram. When using this mnemonic, the tracing of
optimally oxygenated. These SIA findings require the FHR and uterine contractions should be of
increased surveillance and follow-up depending on adequate quality for visual interpretation; if using
maternal and fetal risk factors and labor progress.9 SIA, adequate quality is required of the assessment
of FHR sounds or a composite of intermittent aus-
Interpretation of CEFM FHR Tracings cultation between contractions (baseline) and dur-
Continuous electronic fetal monitoring has been ing 6- to 10-second intervals for 60 seconds during
under close scrutiny because of the lack of con- and after palpated contractions.9 Definitions for
sistent interpretation of FHR tracings.3,4,18,19 In decelerations and variability apply to either internal
1997, the National Institute of Child Health or external monitoring.19 Definitions for late decel-
and Human Development (NICHD) developed erations, early decelerations, variable decelerations,
guidelines to “develop standardized and unam- and variability are reserved for electronic tracings
biguous definitions for FHR tracings.”18 The goal alone and should not be used with SIA tech-
of this standardization was to set forth research niques.6,9 However, increases and decreases from
guidelines to enable future studies to determine baseline can be accurately determined using SIA.6,9
the predictive value of EFM, which could lead
to an evidence-based approach for management Determine Risk
of EFM tracings.18 In 2008, the NICHD revised Before any FHR tracing can be interpreted, the
their definitions, interpretation, and research history of the patient and the clinical context
guidelines.19 ACOG incorporated these guidelines of the situation need to be evaluated in order to
into a 2009 practice bulletin on EFM definitions determine risk.19 A number of FHR character-
and the three-tiered FHR interpretation system.13 istics are dependent on gestational age and the
In 2010, ACOG released a second practice bul- physiologic status of the woman. Consideration
letin on management of intrapartum fetal heart must be given to medications, prior antepartum
tracings based on the three-tier category system testing, maternal status, and fetal medical condi-
and management of uterine tachysystole.20 ACOG tions, such as anomalies, growth restriction, and

4  Chapter E —
Intrapartum Fetal Surveillance

arrhythmias.19 In this context, risk primarily refers When performing SIA, the average baseline rate
to the risk of having or developing uteroplacental should be determined between contractions.
insufficiency or the risk of a sudden event such as Causes of a change in baseline rate may include
placental abruption or cord prolapse. The ability change in fetal status, chorioamnionitis, drugs,
of the fetus to respond to hypoxic events, based on maternal fever, position, and prematurity.9,20,24
these risks, must be considered in the overall man-
agement of fetal monitoring (SIA or CEFM). For Bradycardia
example, a tracing with late decelerations might be Bradycardia is defined as a baseline heart rate
interpreted and managed differently in a woman less than 110 bpm for greater than or equal to
with preeclampsia and long labor compared with 10 minutes.19 Mild bradycardia (90 to 110 bpm)
that of a woman with normal pregnancy and a with moderate variability may represent normal
normal previous tracing who just received epidural physiologic variation with increased vagal tone,
analgesia and is hypotensive. and typically is not associated with hypoxemia.9,23
Rates less than 70 bpm may be seen in fetuses with
Contractions congenital heart disease or myocardial conduc-
Uterine monitoring can be performed internally tion defects.24 Maternal causes of fetal bradycardia
using an IUPC, externally using a tocodynamom- include supine positioning, hypotension, hypogly-
eter (tocotransducer), or by palpation to deter- cemia, tachysystole, or hypothermia. Fetal causes
mine the duration and frequency of contractions. include prolonged cord occlusion, prolapsed cord,
Strength of a contraction cannot be determined rapid descent, or fetal decompensation.9,20,24
with the external tocodynamometer and requires
placement of an IUPC or palpation of the abdom- Tachycardia
inal wall. Uterine contraction frequency can be Tachycardia is defined as a baseline heart rate
quantified as the number present over a 10-minute greater than 160 bpm for greater than or equal to
period, averaged over 30 minutes. Routine use 10 minutes.13,19,20 Fetal movement and maternal
of an IUPC is discouraged due to lack of clinical anxiety, fever, dehydration, ketosis, and beta-
benefit and increased risk of fever.21-23 adrenergic agent use all can cause fetal tachycardia
Contractions are classified as normal (5 or fewer unassociated with hypoxia.9,20 Fetal immaturity,
contractions in a 10-minute period averaged over thyrotoxicosis, and anemia also can cause fetal
30 minutes) or tachysystole (more than 5 con- tachycardia.9,20 Fetal tachycardia can also be caused
tractions in a 10-minute period averaged over by illicit drug use9,20 and, in some instances, mater-
30 minutes).13,19 Tachysystole is qualified as to the nal drug screening is indicated. Persistent tachy-
presence or absence of decelerations. Tachysystole cardia greater than 180 bpm, especially if maternal
applies to both spontaneous and stimulated labor, fever is present, suggests chorioamnionitis.25 An
but management may differ if the uterine activ- FHR baseline greater than 200 bpm is frequently
ity is induced rather than spontaneous.13 Hyper- due to fetal arrhythmia or other congenital
stimulation and hypercontractility are poorly anomaly.20 “In isolation, fetal tachycardia is poorly
defined, and therefore use of these terms should predictive for fetal hypoxemia or acidemia unless
be discontinued.13,20 accompanied by minimal or absent FHR variability
or recurrent decelerations or both.”20 If tachycardia
Baseline Rate persists in the preterm fetus, close surveillance and
The baseline heart rate is calculated by averaging assessment for other causes is warranted.24
the rate rounded to 5 bpm intervals over a 10-min-
ute segment. Segments that should be excluded are Variability
those that have marked variability (more than 25 The FHR normally exhibits fluctuations in base-
bpm), are greater than or equal to 25 bpm above line heart rate activity that is irregular in ampli-
or below the baseline, or contain accelerations or tude and frequency. The presence of variability
decelerations. There must be at least a 2-minute represents an intact nervous pathway through the
identifiable segment within any 10-minute period. cerebral cortex, the midbrain, the vagus nerve,
This 2-minute segment does not need to be con- and the normal cardiac conduction system. When
tiguous. The normal range is 110 to 160 bpm.19 the fetus is well oxygenated, the central nervous

— Chapter E  5
Chapter E

system responds with moderate variability.9,13,19 ability.20 Drugs including analgesics, anesthetics,
However, minimal or absent variability alone is barbiturates, tranquilizers, atropine, and magne-
not necessarily predictive of hypoxemia or meta- sium sulfate may also induce quiet periods in the
bolic acidemia.19 Fetal sleep cycles, narcotics, FHR tracing without fetal compromise. In addi-
cardiac conduction defects, and congenital central tion, steroid administration to induce fetal lung
nervous system anomalies can cause a decrease in maturation may reduce variability.13
variability that is not hypoxic in origin.19,24 Vari-
ability is considered the most important predictor Accelerations
of fetal oxygenation and is, therefore, a vital ele- Accelerations are visually apparent, abrupt increases
ment of CEFM assessment.19 in FHR above the most recent baseline with an
The finding most strongly associated with fetal onset to peak of less than 30 seconds. The peak of
acidemia is absent baseline variability accompanied the acceleration is 15 bpm or greater (10 bpm or
by recurrent late decelerations, recurrent variable greater if less than 32 weeks’ gestation) and lasts for
decelerations, or bradycardia.19 The presence of 15 seconds or more (10 seconds or more if less than
moderate variability and/or the presence of accelera- 32 weeks’ gestation).13,19 The return to baseline is
tions is highly predictive of a non-acidotic fetus.13,19 within 2 minutes. If the acceleration lasts 2 minutes
The significance of marked variability is unclear but or more but less than 10 minutes, it is defined as a
may be indicative of a fetus that is under hypoxic prolonged acceleration. The absence of accelerations
stress yet retains the ability to maintain central does not necessarily indicate fetal acidemia, but may
oxygenation. The presence of minimal, absent, warrant the need for further evaluation. When used
or marked variability should be further evaluated in antenatal testing, a contraction stress test (CST),
within the context in which it occurs. Although the or biophysical profile (BPP) may be used to clarify
NICHD definitions require absent variability for fetal status in the presence of a nonreactive non-
classification as Category III, other fetal monitoring stress test (ie, less than two FHR accelerations in 20
experts have advocated for combining persistent minutes that do not meet 15 bpm × 15 seconds
absent and minimal variability due to the difficulty or 10 bpm × 10 seconds, per above criteria). The
in separating these two categories.5 presence of spontaneous or stimulated accelerations
National Institute of Child Health and Human is highly predictive of a non-acidotic fetus.13,19
Development guidelines state that variability should The presence of accelerations, whether sponta-
no longer be described as short-term (beat-to-beat) neous or stimulated, is strongly predictive of nor-
or long-term, nor should the terms good, increased, mal acid-base status at the time of observation.13,19
decreased, or average be used. Definitions to charac- When accelerations are seen in association with
terize variability are specifically classified as absent, contractions and variable decelerations, the latter
minimal, moderate, or marked (Table 3).19 generally indicate partial cord compression and
The degree of FHR variability is affected by may be referred to as shoulders. This occurs when
the fetal state and by multiple causes other than the vein is compressed in the umbilical cord but
uteroplacental insufficiency or acidosis. Normal the thicker-walled arteries remain patent. These
fetuses may have decreased variability with no accelerations are actually part of the variable
known cause. Sleep cycles of 20 to 40 minutes deceleration and should not be used to assess fetal
or longer may cause a normal decrease in vari- acid-base status.
While the presence of accelerations assures the
provider of normal acid-base status, the disappear-
Table 3. Definitions of FHR Variability ance of FHR accelerations does not necessarily
mean the fetus is hypoxic.19 Drugs and sleep cycles
Variability Amplitude Range may cause accelerations to disappear. Therefore, if
accelerations disappear, providers should look for
Absent Undetectable other indicators of compromise such as decelera-
Minimal Detectable to 5 bpm or fewer tions worsening in depth, duration, or frequency,
Moderate 6 to 25 bpm decreased baseline variability or baseline tachy-
Marked >25 bpm cardia, or bradycardia. In addition, providers
can attempt to elicit accelerations using scalp or

6  Chapter E —
Intrapartum Fetal Surveillance

vibroacoustic stimulation. Of note, FHR accelera- Variable decelerations may be accompanied by


tions should not be elicited during FHR decelera- other characteristics, the clinical significance of
tions or bradycardia (ie, use of scalp stimulation).9 which requires further investigation. Some exam-
Such actions only delay initiation of appropriate ples include a slow return of the FHR after the end
intrauterine resuscitation measures and further of the contraction, biphasic decelerations, tachycar-
compromise fetal status. dia after variable deceleration(s), accelerations pre-
ceding and/or following a deceleration (sometimes
Decelerations called shoulders or overshoots), and fluctuations in
Decelerations are defined in terms of the rate of the FHR in the trough of the deceleration.19
onset (abrupt versus gradual) and the relationship Variables are the most common FHR decelera-
to uterine contractions. If decelerations occur with tion that occur during labor and are generally
50% or greater of contractions during 20-minutes, associated with normal perinatal outcomes.20
they are considered to be recurrent decelerations. Assessment of the tracing should include changes
If they occur with less than 50% of contractions in the overall baseline rate and variability, and
during 20 minutes, they are called intermit- the recurrence, depth, and duration of the decel-
tent decelerations.13,19 Use of the word repetitive, eration. The presence of moderate variability or
although used in the past, should be avoided accelerations suggests the absence of acidemia.20
because it implies that decelerations are occurring Management generally involves relieving cord com-
with every contraction rather than with the major- pression by turning the woman or amnioinfusion.
ity (50% or greater). Decelerations are classified as Administering oxygen and reducing or discontinu-
early, variable, late, or prolonged.13,19 ing uterine stimulants may also be helpful.20

Early Decelerations Late Decelerations


Early decelerations are visually apparent gradual Late decelerations are visually apparent gradual
decreases in FHR with return-to-baseline in decreases in FHR with return to baseline with the
association with a uterine contraction. The onset onset to nadir of 30 seconds or more. The onset,
to nadir is 30 seconds or more and the nadir nadir, and recovery of the deceleration generally
occurs at the same time as the peak of the contrac- occur after the beginning, peak, and ending of
tion.13,19 They are almost always benign if no other the contraction, respectively.19 Late decelerations
abnormalities of the FHR tracing are noted, and are associated with uteroplacental insufficiency
represent transient local changes in blood flow as a and fetal hypoxemia. If uncorrected they can lead
result of stimulus of the vagal nerve centers due to to acidemia and myocardial depression. When
head compression. combined with absent or minimal variability or
other FHR abnormalities, there is an increased
Variable Decelerations likelihood of significant fetal compromise; imme-
Variable decelerations are visually apparent abrupt diate evaluation and intervention are indicated.
decreases in FHR below the baseline with onset Subtle, shallow late decelerations are easily missed
to nadir of 30 seconds or less. The decrease in but clinically significant. They can be detected by
FHR is 15 bpm or greater from the baseline with holding a straight edge along the baseline.
duration of 15 seconds or more but less than 2
minutes, and may or may not be associated with Prolonged Decelerations
contractions.13,19 Variable decelerations are most Prolonged decelerations are visually apparent
commonly the result of cord compression result- decreases in FHR baseline of 15 bpm or more, last-
ing in a rise in peripheral resistance and change ing 2 minutes or more, but less than 10 minutes.19
in oxygenation. This causes sudden fetal hyper- A sudden deterioration in the FHR tracing can
tension, increased parasympathetic outflow, and be seen after vaginal examination, placement of
slowing of the fetal pacemaker. However, inter- fetal spiral electrode, amniotomy, uterine tachy-
pretation is complicated because variable decelera- systole secondary to administration of oxytocin or
tions also can result from decreased arterial oxygen a cervical ripening agent, maternal hypotension
concentration secondary to uteroplacental insuf- (eg, secondary to regional anesthesia), maternal
ficiency from other causes. seizures, or fetal movement producing transient

— Chapter E  7
Chapter E

cord compression. If the fetus was not previously tive of abnormal fetal pH status. These tracings
compromised, recovery will typically occur with require prompt evaluation and implementation
discontinuation of the inciting event or agent, of interventions to address the tracing. Category
position change, increased intravenous fluids, III tracings are clearly abnormal and predictive of
maternal oxygen supplementation, or a combina- abnormal acid-base status.19 Prompt evaluation and
tion of these measures. When accompanied by consideration of immediate delivery is required.
change in variability or baseline, decelerations
are more likely to be associated with fetal acid- NICHD FHR Classification System19
base abnormalities. Factors known to cause these Category I FHR Tracings
changes should be identified and corrected. Category I tracings are normal tracings that are
strongly predictive of normal fetal acid-base status
Sinusoidal Fetal Heart Rate Tracings at the time of observation and must include all of
A sinusoidal FHR tracing is a specific pattern that the following:
is not consistent with any of the above defini- • Baseline of 110 to 160 bpm
tions. It is generally thought to be due to severe • Moderate baseline variability
fetal anemia.24-26 It is described as a visually appar- • Absence of late or variable decelerations
ent, regular smooth sine wave-like undulating • Absence or presence of early decelerations
tracing seen within the FHR baseline. Providers • Absence or presence of accelerations (spontane-
are cautioned not to confuse this tracing with ous or elicited)
FHR variability. The sine waves undulate slowly
and regularly, generally with a cycle frequency of Category II FHR Tracings
3 to 5 cycles (waves)/minute. The tracing must con- Category II tracings are indeterminate tracings
tinue for at least 20 minutes,19 but providers may that are not predictive of fetal acid base status and
need to intervene sooner in emergencies (eg, bleed- cannot be classified as Category I or Category III.
ing from a ruptured vasa previa). Of note, the term The presence of moderate variability and/or accel-
pseudosinusoidal is not recognized by the NICHD. erations is highly predictive of normal fetal acid
In the past, this term was used to denote a tracing base status.13,19,20 These tracings require prompt
that frequently occurred following narcotic admin- evaluation and implementation of interventions
istration or sometimes during ultrasound, and was to address the tracings. Category II tracings occur
associated with rhythmic fetal movements such as in the majority of fetuses during labor.13,19 Some
rapid breathing, sucking movements of the mouth, examples of causes of Category II tracings include:
hiccoughing, and thumb sucking.26 • Tachycardia
• Bradycardia not accompanied by absent
Overall Assessment variability
Having assessed the contraction pattern and FHR • Baseline with minimal or marked variability
tracing and defined the risk, an overall assessment • Baseline with absent variability not accompanied
of the situation and management plan should be by recurrent decelerations
made. The terms fetal distress and birth asphyxia • Recurrent variable decelerations with minimal to
are inappropriate and have no place in the assess- moderate variability
ment. In the past, terms describing the FHR trac- • Recurrent late decelerations with moderate
ing were reassuring and non-reassuring, but since variability
the report from the 2008 NICHD workshop, the • Variable decelerations with slow return, overshoot
assessment of fetal status has been organized into a or shoulders
three-tiered system: Category I, II, or III. Manage- • Prolonged deceleration lasting more than
ment of the woman must be based on the clinical 2 minutes but less than 10 minutes
context and the FHR tracing category, and must • No acceleration after fetal stimulation
include a plan for further fetal surveillance if labor
is allowed to continue. Category III FHR Tracings
Typically, Category I tracings are considered Category III tracings are predictive of abnormal
normal and can be followed routinely. Category fetal acid-base status at the time they are observed.
II tracings are indeterminate and not predic- These require prompt evaluation and expedient

8  Chapter E —
Intrapartum Fetal Surveillance

interventions to address the pattern. Interventions Categorization of SIA Findings


may include, as appropriate, administration of AWHONN and the American College of Nurse
oxygen, administration of an IV fluid bolus, repo- Midwives have recently proposed a categorization
sitioning, discontinuation of uterine stimulants, for interpretation of SIA (Table 4).6,9 Standard-
and treatment of maternal hypotension. If these izing language for both of these FHR assessment
fail, immediate delivery should be considered. techniques may decrease communication errors
Category III tracings include: and provide for a common standardized language.
• Sinusoidal pattern There is not a Category III for SIA because this
OR category requires the ability to assess variability,
• Absent FHR variability with any of the following: which is not reliably and consistently possible
– Recurrent late decelerations when using SIA.6,9 In addition, the terms for decel-
– Recurrent variable decelerations eration tracings (ie, late, variable, early) are reserved
– Bradycardia for CEFM and should not be used for SIA.
When using CEFM during labor, providers
and nurses should review tracings regularly.5,9,20 Suggested FHR Management
This periodic review includes maintaining an The 2010 ACOG Practice Bulletin on manage-
adequate quality tracing, identifying and respond- ment of intrapartum FHR tracings presents a stan-
ing to FHR abnormalities, and appropriately dardized approach to FHR management.20 This
communicating findings. Some institutions now was based in part on the framework developed by
use tools for risk management and patient safety Parer and colleagues who analyzed studies of FHR
including communication strategies. An example tracings to develop four hypotheses:28,29
is the Situation-Background-Assessment-Recom- 1. Moderate variability, even in the presence
mendation (SBAR) technique developed by Kaiser of decelerations, is strongly associated with an
Permanente of Colorado. This technique provides absence of acidemia (pH greater than 7.15) and a
a framework for communication among members nondepressed fetus (5-minute Apgar score greater
of the health care team about a patient’s status.27 than or equal to 7).
Adequate documentation is necessary and many
institutions now use computerized charting, flow 2. Minimal or absent (undetectable) variability
sheets, clinical pathways, or fetal tracing archival in the presence of late or variable decelerations is
processes. Any written information on the trac- associated with acidemia (pH less than 7.15) and a
ing (ie, emergent situations during labor) should depressed fetus (5-minute Apgar score less than 7).
coincide with these automated processes to avoid
confusion and minimize litigation risk.9 Docu-
mentation may occur at different intervals than Table 4. Interpretation of Auscultation
the actual assessment and may take the form of Findings6,9,15
summary notes.9
Documentation of the fetal heart tracing and Category I - Normal: Includes ALL of the following:
categorization during labor should include: Normal FHR baseline 110 to 160 bpm
Regular rhythm
1. FHR data (ie, baseline rate, variability, periodic
Presence of FHR increases, or accelerations, from
changes, categorization) the baseline
2. Uterine activity characteristics obtained by Absence of FHR decreases, or decelerations, from
palpation or pressure transducer (ie, frequency, the baseline
duration, intensity, presence or absence of Category II - Indeterminate: May include any of
tachysystole) the following:
3. Specific actions taken when changes occur in Irregular rhythm
FHR or uterine activity Presence of FHR decreases or decelerations from
the baseline
4. Other maternal observations and assessments
Tachycardia (baseline FHR >160 bpm >10 min)
5. Maternal and fetal responses to interventions Bradycardia (baseline FHR <110 bpm >10 min)
6. Subsequent return to normal findings
7. Pertinent communication with other health FHR = fetal heart rate.
care providers

— Chapter E  9
Chapter E

3. The depth of the deceleration or bradycardia The standard rule of 30 minutes from decision-
is directly proportional to fetal depression and to-incision, although used frequently, has not
acidemia, especially with late decelerations and in been supported in the literature to reduce adverse
tracings with minimal or absent FHR variability. neonatal outcomes.20,30 In addition, immediate
delivery of a fetus with an unknown duration of a
4. After an initial normal FHR tracing, progres- Category III tracing may not improve outcomes
sive decelerations, in the absence of catastrophic if the fetus has experienced a pre-existing hypoxic
events, results in acidemia that develops over a insult.20
significant amount of time, generally 1 hour.
Parer and colleagues concluded that the presence Category II EFM Tracings
of moderate variability had a 98% negative predic- Category II tracings include all tracings that are
tive value for fetal acidosis or an Apgar score less not classified as Category I or III; these occur at
than 7.28,29 Minimal or absent variability with late some time in over 80% of labors.31 Because these
or variable decelerations was predictive of neona- tracings may represent fetal compromise, recom-
tal acidosis or neonatal depression, with 23% of mendations are to evaluate the tracing, perform
fetuses having these adverse findings; there was appropriate corrective measures when indicated,
some evidence that absent variability is more pre- and then re-evaluate. Because Category II tracings
dictive. The depth of decelerations has a stronger represent a wide variety of concerns, the pres-
association with fetal acidosis for late decelerations ence of accelerations (spontaneous or induced) or
than for variable decelerations. In the fetus with moderate variability are useful in assessing normal
decreased variability and decelerations, acidosis fetal acid-base status. If neither of these charac-
was shown to develop slowly over time, except in teristics is present after appropriate intrauterine
the setting of sudden bradycardia as may occur resuscitative measures, or if the tracing progresses
with placental abruption or cord prolapse.28,29 to Category III, consideration should be given
to delivery of the fetus. If the tracing reverts to
Category I EFM Tracings Category I after appropriate intervention, then
Category I EFM tracings are considered normal previous monitoring may be resumed.
and are not associated with fetal acidemia.13,19,20 Category II tracings are frequently encountered
Recommendations are to continue the current at some point during labor, and their manage-
monitoring (whether SIA or EFM); periodically ment can be challenging without defined man-
evaluate and document the clinical status, under- agement protocols. Category II tracings can
lying risk factors, and tracings; and change the range from almost normal to tracings in which
management strategy only if the tracing changes to acidemia is either present or developing rapidly.
a Category II or III. Without rapid intervention, these tracings can
rapidly evolve into Category III tracings. Clini-
Category III EFM Tracings cal considerations when managing these tracings
Category III EFM tracings are considered abnor- must include gestational age, fetal growth status
mal and predictive of abnormal fetal acid-base (ie, IUGR), maternal medical and obstetrical con-
status at the time of observation.13,19,20 Category ditions, comorbidities, labor progress, and avail-
III tracings require prompt evaluation. Recom- able resources and personnel to respond.
mendations are to correct fetal acidemia to reduce
outcomes of neonatal encephalopathy, cerebral Algorithm for Category II Management
palsy, and neonatal acidosis. Preparation for deliv- Clark and colleagues have suggested an algorithm
ery, development of a time frame for delivery, and for managing Category II tracings (Figure 1 and
performance of intrauterine resuscitative measures Table 5).5 This algorithm follows the defini-
are essential. If tracings do not improve with tions set forth by the NICHD and is supported
appropriate corrective maneuvers, prompt delivery by a growing body of evidence. The goal of this
of the fetus is indicated.13,19,20 algorithm is to present a standardized way to man-
Considerations in preparing for an operative age Category II tracings that encourages vaginal
delivery in the presence of a Category III tracing delivery in fetuses whose FHR tracings demon-
should be made judiciously and expeditiously. strate minimal risk for progression to clinically

10  Chapter E —
Intrapartum Fetal Surveillance

significant acidemia. Use of this algorithm requires If delivery is indicated by the algorithm, ideally it
application of specific definitions for significant should be initiated within 30 minutes of the deci-
decelerations and takes into account labor phase sion. The algorithm can be discontinued at any
and labor progress (Table 5). This algorithm is time the provider feels more rapid intervention
only applicable to Category II tracings and is not is required.
to be used in extremely premature fetuses. While this algorithm is somewhat complex, a
When a Category II tracing is identified, intra- web-based app has simplified its use for clinical
uterine resuscitation measures are instituted and practice (available at http://cat2.perigen.com/cat2/).
the algorithm is delayed for 30 minutes to allow
these interventions to improve the tracing. After Five-Tier Classification
30 minutes, if these measures do not alleviate the In addition to the above approach for manag-
Category II tracing, the algorithm is begun. The ing Category II tracings, a five-tiered classifica-
algorithm is initiated by an assessment of moder- tion scheme has also been proposed by Parer and
ate variability or FHR accelerations because this colleagues (Table 6).29,32 Similar to the algorithm
rules out clinically significant acidemia. From in Table 5, the purpose of this classification system
there, the assessment includes the presence or is to classify FHR monitor tracings according
absence of significant decelerations, the stage of to risk of fetal acidemia, determine the risk of
labor, and whether labor is progressing normally. evolution to a more serious tracing, and construct

Figure 1. Algorithm for Management of Category II Fetal Heart Rate Tracings

Moderate variability or accelerations

Yes No

Significant decelerations with Significant decelerations with


>50% of contractions for 1 hour* >50% of contractions for 30 minutes*

Yes No Yes No

Observe Cesarean Observe for 1 hour


or OVD
Latent phase Active phase Second phase

Persistent pattern

Cesarean Normal labor progress Normal progress


Yes No

Yes No Yes No
Cesarean Manage per
or OVD algorithm
Observe Cesarean Observe Cesarean
or OVD

*That have not resolved with appropriate conservative corrective measures, which may include supplemental oxygen, maternal position
changes, intravenous fluid administration, correction of hypotension, reduction or discontinuation of uterine stimulation, administration of uter-
ine relaxant, amnioinfusion, and/or changes in second stage breathing and pushing techniques.
OVD = operative vaginal delivery.
Reprinted from 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(2):89-97.

— Chapter E  11
Chapter E

a standard process for FHR tracing management definitions are according to the NICHD state-
with the ultimate aim of minimizing newborn ment on the nomenclature of FHR tracings.
infant acidemia without excessive obstetric In the five-tier approach, each FHR tracing has
intervention. been color-coded to represent no threat of acide-
A grid categorizes all possible heart rate patterns mia (green, no intervention required) to severe
based on baseline rate (ie, normal, tachycardia, threat for acidemia (red, emergent delivery recom-
bradycardia), type of decelerations (ie, early, late, mended). Three intermediate categories—blue,
variable, prolonged), and quantity of variability yellow, orange—are NICHD Category II tracings
(ie, undetectable, minimal, moderate, marked). All and represent increasing concern and response for
evolving acidemia (Table 7).29,32
In 2012 Coletta and colleagues compared the
Table 5. Management of Category II FHR Tracings: NICHD three-tier system to the proposed five-
Clarifications for Use of Algorithm tier system in Table 6.32 For tracings categorized
by either orange or red, there was 79% sensitivity
1. V
 ariability refers to predominant baseline FHR pattern (marked,
moderate, minimal, absent) during a 30-minute evaluation period,
and 100% specificity for a pH less than 7 with no
as defined by NICHD. false positives. By using the five-tier system 79%
2. Marked variability is considered same as moderate variability for of fetal acidemia was correctly identified in the
purposes of this algorithm. orange and red tracings, compared with only 12%
3. Significant decelerations are defined as any of the following: in the NICHD Category III. All tracings with
• Variable decelerations lasting longer than 60 seconds and pH greater than 720 were correctly categorized as
reaching a nadir more than 60 bpm below baseline.
• Variable decelerations lasting longer than 60 seconds and
blue, green, or yellow. The five-tier system also
reaching a nadir less than 60 bpm regardless of the baseline. better identified tracings that resulted in lower
• Any late decelerations of any depth. Apgar scores, admission to the NICU, and need
• A ny prolonged deceleration, as defined by the NICHD. Due to for oxygen supplementation.32 The complexity of
the broad heterogeneity inherent in this definition, identification this five-tier system has made practical application
of a prolonged deceleration should prompt discontinuation of the
algorithm until the deceleration is resolved. difficult, however, this has changed recently due to
4. A pplication of algorithm may be initially delayed for up to 30 the availability of a mobile app (available at http://
minutes while attempts are made to alleviate category II pattern www.obapps.org/), making the application to the
with conservative therapeutic interventions (eg, correction of clinical setting more user-friendly.33 The Japan
hypotension, position change, amnioinfusion, tocolysis, reduction or
discontinuation of oxytocin). Society of Obstetrician and Gynecologists also
5. Once a category II FHR pattern is identified, FHR is evaluated and recommended the use of this five-tier system based
algorithm applied every 30 minutes. on its validity with acid-base balance.34,35 How-
6. A ny significant change in FHR parameters should result in ever, another study reports no difference between
reapplication of algorithm. very normal or very abnormal tracings using this
7. For category II FHR patterns in which algorithm suggests delivery is
indicated, such delivery should ideally be initiated within 30 minutes
system. They conclude that it is yet to be deter-
of decision for cesarean. mined whether one system is superior in predict-
8. If at any time tracing reverts to category I status, or deteriorates ing fetal acidemia.36
for even a short time to category III status, the algorithm no longer
applies. However, algorithm should be reinstituted if category I Intrauterine Resuscitative Measures
pattern again reverts to category II.
9. In fetus with extreme prematurity, neither significance of certain FHR Various intrauterine resuscitative measures are to
patterns of concern in more mature fetus (eg, minimal variability) or be undertaken for any FHR tracing that is con-
ability of such fetuses to tolerate intrapartum events leading to certain cerning regardless of classification scheme used
types of category II patterns are well defined. This algorithm is not
intended as guide to management of fetus with extreme prematurity.
(Table 8).20,37 The goals of these corrective mea-
10. A lgorithm may be overridden at any time if, after evaluation of patient, sures are aimed at the underlying suspected cause
physician believes it is best interest of the fetus to intervene sooner. of the abnormality in the FHR tracing. They are
also dependent on the associated FHR abnormal-
FHR = fetal heart rate; NICHD = National Institute of Child Health and Human ity identified.
Development.
Possible interventions include:
Reprinted from Clark SL, Nageotte MP, Garite TJ, et al. Intrapartum manage-
ment of category II fetal heart rate tracings: towards standardization of care.
• Change in maternal position to lateral position-
Am J Obstet Gynecol. 2013;209(2):89-97. ing (left or right) or on hands and knees
• Administer maternal oxygen

12  Chapter E —
Intrapartum Fetal Surveillance
Table 6. Proposed Five-Tier FHR Classification System29

Proposed Five-tier FHR Classification

Category Risk of Acidemia Risk of Evolution Action

Green Mostly Cat I None Very low None

Blue Cat II No central fetal academia Low Conservative techniques, begin


preparation for delivery

Yellow Cat II No central fetal acidemia, but FHR suggestive Moderate Conservative techniques and
of intermittent reductions in oxygen which increased surveillance
may result in fetal oxygen debt

Orange Cat II Borderline/acceptably low; fetus potentially High Conservative techniques and
on verge of decompensation prepare for urgent delivery

Red Cat III Unacceptably high; evidence of actual or Already present Delivery
impending damaging fetal asphyxia

Table 7. Risk Categories for Fetal Acidemia Related to Variability, Baseline Rate, and Presence of
Recurrent Decelerations

Mild Moderate Severe Mild Moderate Severe Mild Moderate Severe


Variable No Early VD VD VD LD LD LD PD PD PD

Moderate (normal) variability


Tachycardia B B B Y O Y Y O Y Y Y

Normal G G G B Y B Y Y Y Y O

Mild bradycardia Y Y Y Y O Y Y O Y Y O

Moderate bradycardia Y Y O O O O

Severe bradycardia O O O O O
Minimal variability
Tachycardia B Y Y O O O O R O O O

Normal B B Y O O O O R O O R

Mild bradycardia O O R R R R R R R R R

Moderate bradycardia O O R R R R

Severe bradycardia R R R R R
Absent variability
Tachycardia R R R R R R R R R R R

Normal O R R R R R R R R R R

Mild bradycardia R R R R R R R R R R R

Moderate bradycardia R R R R R R

Severe bradycardia R R R R R
Sinusoidal R
Marked variability Y

B = blue; G = green; LD = late decelerations; O = orange; PD = prolonged decelerations; R = red; VD = variable decelerations; Y = yellow.
Reprinted from Parer JT, Ikeda T. A framework for standardized management of intrapartum fetal heart rate patterns. Am J Obstet Gynecol.
2007;197(1):26.e1-26.e6.
Chapter E

• Administer intravenous fluid bolus positioning on either side is superior when com-
• Reduce uterine contraction frequency pared to supine positioning.37
• Discontinue oxytocin or cervical ripening agents Administration of maternal oxygen with the
• Administer tocolytic drug goal of improving fetal oxygenation remains a
• Initiate amnioinfusion with recurrent variable common intervention, yet study results regarding
decelerations its benefit for the fetus remain controversial and
• If prolapsed umbilical cord is noted, continuous may even suggest a possible detrimental effect.37-43
elevation of the presenting fetal part until opera- A recent Cochrane review found that there was
tive delivery occurs not enough evidence to support or refute the use
• Modify pushing efforts in second stage of labor of oxygen for intrauterine resuscitation.44 Due
Other important considerations are: to the controversy in the literature, a reasonable
• Assess maternal vital signs to assess for hypotension approach is to provide oxygen after other appro-
• Perform vaginal examination to assess progress of priate intrauterine resuscitation techniques have
labor and rule out prolapsed umbilical cord not been effective, and to discontinue its use once
• Change method of FHR monitoring if strip is the desired fetal response has been achieved.9,39
inadequate quality for assessment Administration of an intravenous fluid bolus is
• If recent epidural and hypotension with new late another common technique thought to increase
decelerations, prepare for possible administration intravascular volume, uteroplacental perfusion
of ephedrine and, thus, fetal oxygenation.37,39 Simpson and
Lateral positioning reduces compression of James found that fetal oxygenation saturation
the inferior vena cava and aorta. Repositioning improved with a 500 mL bolus of lactated
the woman may also reduce compression of the Ringer solution and increased greatest when a
umbilical cord. Several studies indicate that lateral 1,000 mL bolus was administered. The positive
effect

Table 8. Potential Intrauterine Resuscitative Measures for Category II or Category III


Tracings20

Goal Associated FHR Changes Potential Interventions

Improve uteroplacental Recurrent late decelerations Lateral positioning


blood flow Prolonged decelerations Administer maternal oxygen
Minimal or absent FHR Administer intravenous fluid bolus
variability Discontinue or reduce uterine stimulants
Administer tocolytic drugs
Correct maternal hypotension
Modify maternal expulsive (pushing) efforts
Reduce uterine Tachysystole with Category II Lateral positioning
activity or Category III tracing Administer intravenous fluid bolus
Discontinue or reduce uterine stimulants
Administer tocolytic drugs
Alleviate/reduce Recurrent variable Reposition to where FHR is most improved
umbilical cord decelerations Discontinue uterine stimulants
compression Prolonged decelerations Initiate amnioinfusion if variable decelerations
Bradycardia recurrent
Modify maternal expulsive (pushing) efforts
Check for prolapsed cord. If identified,
continuously elevate presenting part until
operative delivery occurs.

FHR = fetal heart rate.


Always: check cervix, maternal vital signs

14  Chapter E —
Intrapartum Fetal Surveillance

of the bolus continued for 15 minutes after postpartum endometritis, and the incidence of
completion.39 neonatal and maternal hospital stays greater than
Modification of pushing efforts can have a 3 days.53 In addition, mean cord umbilical pH was
significant effect on the FHR tracing. Coached higher. No improvement in long-term neonatal
sustained Valsalva pushing (ie, closed glottis, “hold outcomes has been detected.
your breath and count to 10” for three times with Although typically considered safe, amnioin-
each contraction) can have a deleterious effect fusion carries a few precautions and potential
on maternal and fetal oxygenation37,45-47 with a complications. Amnioinfusion is indicated only
resulting increase in number and severity of FHR for recurrent variable decelerations and is not indi-
decelerations.46,48 A preventive measure would be cated for late decelerations, fetal bradycardia, thick
to avoid pushing until the woman feels the urge to meconium, or oligohydramnios with a normal
push, thereby minimizing the length of the active heart rate tracing.20,52-54
pushing phase and fetal exposure to the hypoxic Amnioinfusion should also not be attempted
stress of pushing. Modification of pushing efforts when cesarean delivery is indicated, such as in
includes temporarily discontinuing pushing to transverse lie or placenta previa. It should never be
allow the fetus to recover, or to push with every undertaken when doing so would result in a delay
second or third contraction.9,37,45,49,50 of an indicated definitive treatment. With breech
presentation, multiple gestation, or when placental
Ancillary Testing for Category II and abruption is suspected, caution should be taken in
III FHR Tracings performing amnioinfusion. Complications include
Fetal scalp pH testing is no longer commonly per- umbilical cord prolapse, rupture of a previous
formed in the United States and has been replaced cesarean scar, amniotic fluid embolism, acute uter-
with assessment of moderate FHR variability or ine hypertonus with a Category II or Category III
the presence of FHR accelerations. If accelerations fetal heart tracing, and acute polyhydramnios.
are not present, fetal stimulation may be used to In the past, amnioinfusion was used to dilute
elicit accelerations. Current evidence indicates the thick meconium as a prophylactic measure to pre-
presence of either moderate variability or accelera- vent newborn meconium aspiration. Randomized
tions reliably predicts the absence of fetal metabolic controlled trials did not confirm the efficacy of
acidemia at the time either of these characteristics this approach and the ACOG no longer considers
is observed.19 A meta-analysis showed that if there this an indication for amnioinfusion.4,54
is absent or minimal variability without spontane-
ous accelerations, the presence of an acceleration Guidelines for Performing Amnioinfusion
after scalp stimulation or fetal acoustic stimulation Amnioinfusion can be performed by continuous or
indicates that the fetal pH is greater than 7.20,51 intermittent techniques. A randomized controlled
Intrauterine resuscitation should be utilized trial showed there was no difference between the
appropriately for the clinical situation. If the fetal two techniques for resolving variable decelerations.55
heart tracing is a Category III and does not resolve For continuous infusion:
quickly, plans for an expedient delivery should 1. Perform a vaginal examination to determine
be undertaken. Category II tracings need to be presentation and dilation, and to rule out cord
assessed in light of the entire clinical picture. Some prolapse.
Category II tracings may require emergent delivery 2. Obtain informed consent.
if there is no response to interventions. 3. Place the patient in the left-lateral position.
Place an IUPC and consider placement of an
Amnioinfusion FSE. If available, use a double lumen catheter
Amnioinfusion should be considered for suspected for saline infusion.
cord compression to reduce the occurrence of 4. If a double lumen catheter is not available,
recurrent variable heart rate decelerations in the attach an 18-gauge needle to IV tubing con-
first stage of labor and to lower the use of cesarean nected to normal saline or lactated Ringer
delivery.52 Amnioinfusion has been associated with solution. Attach extension tubing filled with
a reduction in cesarean delivery rates, FHR decel- distilled water between the IUPR and the
erations, Apgar scores less than 7 at 5 minutes, transducer. Insert the 18-gauge needle into the

— Chapter E  15
Chapter E

side port of the extension tubing. Alternatively, indicated. Several studies have evaluated fECG
insert a second single lumen catheter (one for analysis documenting the effectiveness to reduce
IUPC and one for amnioinfusion). operative vaginal deliveries, fetal scalp sampling,
5. Infuse fluid, giving 250 to 500 mL initially, neonatal encephalopathy, and fetal acidosis (pH
followed by 50 to 60 mL/hour maintenance less than 7.05).62-65 One drawback to this technol-
infusion until FHR abnormalities resolve. ogy is that it requires rupture of the membranes
note: Resting tone will be increased while the and internal fetal scalp monitoring.
infusion is running, but elevated baseline tone before Another area of research is the use of computer
infusion is a contraindication. analysis of key components of the fetal tracing,66-68
or decision analysis, for the interpretation of the
Management of Tachysystole EFM tracing.69 These have not been shown to
Uterine contractions cause a cessation of uterine improve clinical outcomes.
blood flow and, thus, oxygen delivery to the fetus. Fetal pulse oximetry was developed using an
In most healthy fetuses, this temporary decline is internal monitoring device and required rupture of
well tolerated. However, uterine tachysystole has membranes to continuously monitor fetal oxy-
been shown to cause a progressive decline in fetal genation saturation during labor. Trials have not
oxygenation and arterial blood gases at birth,56-60 shown significance differences in reducing cesar-
increased risk for operative delivery, lower Apgar ean delivery rates or interventions with the use of
scores, as well as an increase in NICU admissions.59 fetal pulse oximetry, therefore use of fetal pulse
Tocolysis should be considered, especially in the oximetry is no longer recommended.70-72
setting of tachysystole associated with Category II
or Category III tracings when standard intrauter- International Context
ine resuscitative measures are not successful.20,60 A In many low-resource settings, EFM is not an
recent Cochrane review demonstrated that betami- option. SIA may be with a fetoscope rather than a
metic therapy reduced the number of FHR abnor- Doppler stethoscope. Although amnioinfusion is
malities.61 If tachysystole is induced and a Category no longer indicated for meconium in high-resource
II or III tracing is present, uterine stimulants settings where EFM is available, it does seem to be
should also be decreased or discontinued.20 Other indicated for meconium in low-resource settings
interventions for tachysystole may include maternal where monitoring capabilities are limited.73 More
repositioning and IV fluid bolus.9,20,60 Evaluation information on intrapartum fetal surveillance and
for placental abruption should occur; tocolysis other topics relevant to low-resource settings can
is contraindicated when abruption is suspected be found in the Global ALSO manual (available at
because it may worsen the abruption.39 www.aafp.org/globalalso).

Suggested Management of Tachysystole20 Summary


An algorithm for the management of uterine Initiation of fetal monitoring begins with assess-
tachysystole is shown in Figure 2. ment of maternal and fetal risk. Because CEFM
has a low positive predictive value and can result
Areas for Future Development in increased rates of cesarean delivery, intermittent
Although CEFM continues to be the standard for auscultation is recommended for low-risk preg-
fetal monitoring, research is being conducted to nancies. However, hospital policy as well as staff
enhance CEFM with computerized interpretation availability and experience must be considered
and to develop newer methodologies to monitor before deciding to use this technique. Clinicians
fetal well-being during labor. should be ready to change monitoring to CEFM if
Fetal hypoxemia results in biphasic changes in a high-risk situation develops or if the provider is
the ST segment of the fetal electrocardiogram unable to adequately assess fetal heart tones.
(fECG) waveform and an increase in the T/QRS If CEFM is selected for fetal surveillance, inter-
ratio. ST segment automated analysis software can pretation needs to be done with consideration of
record the frequency of ST events and, combined the clinical background, the overall tracing, and
with changes in CEFM, can be used to deter- stage of labor, and in conjunction with fetal scalp
mine if intervention during the labor process is or acoustic stimulation. Outcomes may still be

16  Chapter E —
Intrapartum Fetal Surveillance

Figure 2. Management Algorithm for Uterine Tachysystole

Uterine tachysystole

Spontaneous labor Labor induction or augmentation

Category I Category II or Category I Category II or


FHR tracing III FHR tracing FHR tracing III FHR tracing

No interventions Intrauterine resuscitative Decrease Decrease or stop


required measures* uterotonics uterotonics

If no resolution, Intrauterine resuscitative


consider tocolytic measures

If no resolution,
consider tocolytic

FHR = fetal heart rate.


Reprinted from American College of Obstetricians and Gynecologists. ACOG practice bulletin No 116: Management of
intrapartum fetal heart rate tracings. Obstet Gynecol. 2010;116(5):1232-1240.

unaffected using this technique, even in high-risk Regardless of the technology used, the patient/
pregnancies. Efforts have recently been undertaken support relationship is paramount during the labor
to standardize the definitions, interpretation, process. Clinicians should not allow any monitor-
and general management of FHR tracings. DR ing approach to substitute for personal attention
C BRAVADO is a helpful mnemonic for defin- to the woman and fetus throughout labor.
ing risk and EFM interpretation. It is critical that If an institution has a risk management or
institutions and hospitals ensure that all labor and patient safety committee, regular monitoring and
delivery personnel are trained in FHR surveil- compliance with all aspects of fetal surveillance
lance and interpretation, and in management of should be undertaken. The committee should
findings. Communication among team members be composed of physicians, nurses, administra-
is critical, and tools or strategies to maximize accu- tors, and other pertinent staff for successful
racy and completeness of transfer of information implementation.
should be used (eg, SBAR) to minimize medical
errors and maximize patient safety.

— Chapter E  17
Chapter E

SORT: Key Recommendations for Practice


Evidence
Clinical Recommendation Rating References

Structured intermittent auscultation should be offered to low-risk women as an alternative to CEFM. A 2


CEFM does not decrease perinatal mortality. CEFM reduced neonatal seizures (NNT= 661) but
does not reduce the incidence of cerebral palsy. CEFM increased cesarean delivery rates (overall)
(NNT = 20) and instrumental vaginal births (NNT = 33).
The presence of moderate FHR variability is predictive of normal fetal acid-base status. B 19,28
The presence of FHR accelerations, whether spontaneous or stimulated, is highly predictive of B 19,51
normal fetal acid-base status.
A Category III fetal monitoring tracing increases the likelihood of fetal acidosis and requires prompt B 19,20
measures for intrauterine resuscitation and consideration of the need for urgent delivery if these
measures do not result in improvement.
A period of CEFM on maternity unit admission for low-risk women compared to initiation of A 11,12
structured auscultation at time of admission results in significant increased interventions including
epidural analgesia (NNT = 19), CEFM (NNT = 7) and fetal blood scalp testing (NNT = 45), and is not
recommended.
Amnioinfusion for umbilical cord compression in the presence of decelerations reduces FHR A 53
decelerations (NNT = 3), cesarean delivery overall (NNT = 8), Apgar score <7 at 5 min (NNT = 33),
low cord arterial pH (<7.20) (NNT = 8), neonatal hospital stay >3 days (NNT = 5), and maternal
hospital stay greater than 3 days (NNT = 7). When the FHR tracing includes recurrent variable FHR
decelerations (Category II or Category III) amnioinfusion should be considered. Amnioinfusion is
not indicated for late FHR decelerations.
Tachysystole, whether spontaneous or stimulated, when accompanied by a Category II or Category B 57-60
III tracing, warrants evaluation and appropriate management.
IV fluid bolus, lateral positioning, and oxygen administration, when used together, may improve fetal B 39
oxygenation during labor.
Modifying pushing techniques improves fetal oxygenation and may be considered during the second B 37,46-48
stage of labor. Modifying pushing techniques includes discontinuation of pushing, pushing every
2nd or 3rd contraction, pushing with an open glottis, and/or pushing for 6- to 8-second intervals.

Strength of Recommendation Taxonomy (SORT)

Strength of Recommendation Definition


A • Recommendation based on consistent and good-quality patient-oriented evidence.
B • Recommendation based on inconsistent or limited-quality patient-oriented evidence.
C • Recommendation based on consensus, usual practice, opinion, disease-oriented evidence, or case series for studies of diagnosis, treat-
ment, prevention, or screening.
Patient-oriented evidence measures outcomes that matter to patients: morbidity, mortality, symptom improvement, cost reduction, and quality
of life. Disease-oriented evidence measures intermediate, physiologic, or surrogate end points that might not reflect improvement in patient
outcomes (eg, blood pressure, blood chemistry, physiologic function, pathologic findings).
CEFM = continuous electronic fetal monitoring; FHR = fetal heart rate; IV = intravenous; NNT = number needed to treat.
From Ebell MH, Siwek J, Weiss BD, et al. Strength of recommendation taxonomy (SORT): a patient-centered approach to grading evidence in
the medical literature. Am Fam Physician. 2004;69(3):548-556.

18  Chapter E —
Intrapartum Fetal Surveillance

References
1. Martin JA, Hamilton BE, Osterman MJ, Curtin SC, Mat- 15. American College of Nurse-Midwives. Intermittent Aus-
thews TJ. Births: final data for 2013. Natl Vital Stat Rep. cultation for Intrapartum Fetal Heart Rate Surveillance
2015;64(1):1-65. (replaces ACNM Clinical Bulletin #9, March 2007). J
2. Alfirevic Z, Devane D, Gyte GML. Continuous cardioto- Midwifery Womens Health. 2010;55(4):397-403.
cography (CTG) as a form of electronic fetal monitoring 16. Royal College of Obstetricians and Gynaecologists.
(EFM) for fetal assessment during labour. Cochrane The Use of Electronic Fetal Monitoring. The use and
Database Syst Rev. 2013;5(5):CD006066. interpretation of cardiotocography in intrapartum fetal
3. Chauhan SP, Klauser CK, Woodring TC, Sanderson M, surveillance. Evidence-based Clinical Guideline Num-
Magann EF, Morrison JC. Intrapartum nonreassuring ber 8. London: RCOG Press; 2001.
fetal heart rate tracing and prediction of adverse out- 17. Liston R, Sawchuck D, Young D; Society of Obstetrics
comes: interobserver variability. Am J Obstet Gynecol. and Gynaecologists of Canada; British Columbia Peri-
2008;199(6):623.e1-623.e5. natal Health Program. Fetal health surveillance: antepar-
4. Parer JT, Hamilton EF. Comparison of 5 experts and tum and intrapartum consensus guideline. [Erratum in J
computer analysis in rule-based fetal heart rate interpre- Obstet Gynaecol Can. 2007;29] [11]. J Obstet Gynaecol
tation. Am J Obstet Gynecol. 2010;203(5):451.e1-451.e7. Can. 2007;29(9)(Suppl 4):S3-S56.

5. Clark SL, Nageotte MP, Garite TJ, et al. Intrapartum 18. National Institute of Child Health and Human Develop-
management of category II fetal heart rate tracings: ment Research Planning Workshop. Electronic fetal
towards standardization of care. Am J Obstet Gynecol. heart rate monitoring: research guidelines for interpreta-
2013;209(2):89-97. tion. Am J Obstet Gynecol. 1997;177(6):1385-1390.

6. Feinstein N, Sprague A, Trepanier M, eds. Fetal Heart 19. Macones GA, Hankins GD, Spong CY, Hauth J,
Rate Auscultation. 2nd ed. Washington DC: Associa- Moore T. The 2008 National Institute of Child Health
tion of Women’s Health, Obstetric and Neonatal Nurses and Human Development workshop report on elec-
(AWHONN); 2008. tronic fetal monitoring: update on definitions, inter-
pretation, and research guidelines. Obstet Gynecol.
7. Murray ML. Maternal or fetal heart rate? Avoiding intra- 2008;112(3):661-666.
partum misidentification. J Obstet Gynecol Neonatal
Nurs. 2004;33(1):93-104. 20. American College of Obstetricians and Gynecolo-
gists. Practice bulletin no. 116: Management of intra-
8. Nurani R, Chandraharan E, Lowe V, Ugwumadu A, partum fetal heart rate tracings. Obstet Gynecol.
Arulkumaran S. Misidentification of maternal heart rate 2010;116(5):1232-1240.
as fetal on cardiotocography during the second stage
of labor: the role of the fetal electrocardiograph. Acta 21. Bakker JJ, Verhoeven CJM, Janssen PF, et al. Out-
Obstet Gynecol Scand. 2012;91(12):1428-1432. comes after internal versus external tocodynamometry
for monitoring labor. N Engl J Med. 2010;362(4):306-313.
9. Lyndon A, Ali L, eds. Association of Women’s Health
Obstetric and Neonatal Nursing. Fetal Heart Monitoring 22. Bakker JJ, Janssen PF, van Halem K, et al. Internal
Principles and Practices. 4th ed. Washington DC: Kend- versus external tocodynamometry during induced
all Hunt Professional; 2009. or augmented labour. Cochrane Database Syst Rev.
2013;8(8):CD006947.
10. Hauth JC, Hankins GD, Gilstrap LC III, Strickland
DM, Vance P. Uterine contraction pressures with 23. Harper LM, Shanks AL, Tuuli MG, Roehl KA, Cahill AG.
oxytocin induction/augmentation. Obstet Gynecol. The risks and benefits of internal monitors in laboring
1986;68(3):305-309. patients. Am J Obstet Gynecol. 2013;209(1):38.e1-38.e6.

11. Devane D, Lalor JG, Daly S, McGuire W, Smith V. 24. Freeman RK, Garite TJ, Nageote MP, Mille LA. Fetal
Cardiotocography versus intermittent auscultation of Heart Rate Monitoring, 4th ed. New York, NY: Wolters
fetal heart on admission to labour ward for assess- Kluwer, Lippincott Williams & Wilkins; 2012.
ment of fetal wellbeing. Cochrane Database Syst Rev. 25. Gabbe SG, Niebyl JR, Galan HL, et al. Intrapartum Fetal
2012;2(2):CD005122. Evaluation. In: Obstetrics: Normal and Problem Preg-
12. Bix E, Reiner LM, Klovning A, Oian P. Prognostic value nancies. 6th ed. Philadelphia, PA: Saunders; 2012.
of the labour admission test and its effectiveness 26. Modanlou HD, Murata Y. Sinusoidal heart rate pattern:
compared with auscultation only: a systematic review. Reappraisal of its definition and clinical significance. J
BJOG. 2005;112(12):1595-1604. Obstet Gynaecol Res. 2004;30(3):169-180.
13. American College of Obstetricians and Gynecolo- 27. SBAR Technique for Communication. A Situational
gists. ACOG Practice Bulletin No. 106: Intrapartum fetal Briefing Model. Available at http://www.ihi.org/
heart rate monitoring: nomenclature, interpretation, resources/Pages/Tools/SBARTechniqueforCommuni-
and general management principles. Obstet Gynecol. cationASituationalBriefingModel.aspx.
2009;114(1):192-202. 28. Parer JT, King T, Flanders S, Fox M, Kilpatrick SJ. Fetal
14. Riley LE, Stark AR, eds. Guidelines for Perinatal Care. acidemia and electronic fetal heart rate patterns: is
7th ed. Washington DC: American College of Obstetri- there evidence of an association? J Matern Fetal Neo-
cians and Gynecologists; 2012. natal Med. 2006;19(5):289-294.
29. Parer JT, Ikeda T. A framework for standardized man-
agement of intrapartum fetal heart rate patterns. Am J
Obstet Gynecol. 2007;197(1):26.e1-26.e6.

— Chapter E  19
Chapter E

30. Bloom SL, Leveno KJ, Spong CY, et al; National Insti- 45. Association of Women’s Health Obstetric and Neonatal
tute of Child Health and Human Development Maternal- Nursing. Nursing Care and Management of the Sec-
Fetal Medicine Units Network. Decision-to-incision ond Stage of Labor: Evidence-Based Clinical Practice
times and maternal and infant outcomes. Obstet Gyne- Guideline. 2nd ed. Washington DC; 2008.
col. 2006;108(1):6-11. 46. Simpson KR, James DC. Effects of immediate ver-
31. Jackson M, Holmgren CM, Esplin MS, Henry E, Var- sus delayed pushing during second-stage labor on
ner MW. Frequency of fetal heart rate categories fetal well-being: a randomized clinical trial. Nurs Res.
and short-term neonatal outcome. Obstet Gynecol. 2005;54(3):149-157.
2011;118(4):803-808. 47. Yildirim G, Beji NK. Effects of pushing techniques in
32. Coletta J, Murphy E, Rubeo Z, Gyamfi-Bannerman C. birth on mother and fetus: a randomized study. Birth.
The 5-tier system of assessing fetal heart rate tracings 2008;35(1):25-30.
is superior to the 3-tier system in identifying fetal acide- 48. Hansen SL, Clark SL, Foster JC. Active pushing
mia. Am J Obstet Gynecol. 2012;206(3):226.e1-226.e5. versus passive fetal descent in the second stage of
33. Apps OB. FHR 5-Tier: Fetal Heart Rate. Available at labor: a randomized controlled trial. Obstet Gynecol.
http://www.obapps.org/. 2002;99(1):29-34.
34. Sadaka A, Furuhashi M, Minami H, Miyazaki K, Yoshida 49. Brancato RM, Church S, Stone PW. A meta-analysis
K, Ishikawa K. Observation on validity of the five-tier sys- of passive descent versus immediate pushing in nul-
tem for fetal heart rate pattern interpretation proposed liparous women with epidural analgesia in the sec-
by Japan Society of Obstetricians and Gynecologists. ond stage of labor. J Obstet Gynecol Neonatal Nurs.
J Matern Fetal Neonatal Med. 2011;24(12):1465-1469. 2008;37(1):4-12.
35. Okai T, Ikeda T, Kawarabayashi T, et al; Perinatology 50. Hanson L. Second-stage labor care: challenges in
Committee of the Japan Society of Obstetrics and spontaneous bearing down. J Perinat Neonatal Nurs.
Gynecology. Intrapartum management guidelines 2009;23(1):31-39, quiz 40-41.
based on fetal heart rate pattern classification. J Obstet 51. Skupski DW, Rosenberg CR, Eglinton GS. Intrapartum
Gynaecol Res. 2010;36(5):925-928. fetal stimulation tests: a meta-analysis. Obstet Gynecol.
36. Gyamfi Bannerman C, Grobman WA, Antoniewicz L, 2002;99(1):129-134.
Hutchinson M, Blackwell S. Assessment of the con- 52. Hofmeyr GJ, Lawrie TA. Amnioinfusion for potential
cordance among 2-tier, 3-tier, and 5-tier fetal heart or suspected umbilical cord compression in labour.
rate classification systems. Am J Obstet Gynecol. Cochrane Database Syst Rev. 2012;1(1):CD000013.
2011;205(3):288.e1-288.e4.
53. American College of Obstetricians and Gynecologist
37. Garite TJ, Simpson KR. Intrauterine resuscitation during and American Academy of Pediatrics. Intrapartum Con-
labor. Clin Obstet Gynecol. 2011;54(1):28-39. siderations and Assessment. In Neonatal Encephalopa-
38. Haydon ML, Gorenberg DM, Nageotte MP, et al. The thy. 2nd ed. Washington DC;2014.
effect of maternal oxygen administration on fetal 54. ACOG Committee Obstetric Practice. ACOG Commit-
pulse oximetry during labor in fetuses with nonreas- tee Opinion Number 346, October 2006: amnioninfu-
suring fetal heart rate patterns. Am J Obstet Gynecol. sion does not prevent meconium aspiration syndrome.
2006;195(3):735-738. Obstet Gynecol. 2006;108(4):1053.
39. Simpson KR, James DC. Efficacy of intrauterine resus- 55. Rinehart BK, Terrone DA, Barrow JH, Isler CM, Barril-
citation techniques in improving fetal oxygen status dur- leaux PS, Roberts WE. Randomized trial of intermittent
ing labor. Obstet Gynecol. 2005;105(6):1362-1368. or continuous amnioinfusion for variable decelerations.
40. Hamel MS, Anderson BL, Rouse DJ. Oxygen for intra- Obstet Gynecol. 2000;96(4):571-574.
uterine resuscitation: of unproved benefit and poten- 56. Ross MG. Labor and fetal heart rate decelerations: rela-
tially harmful. Am J Obstet Gynecol. 2014;211(2):124-127. tion to fetal metabolic acidosis. Clin Obstet Gynecol.
41. Garite TJ, Nageotte MP, Parer JT. Should we really avoid 2011;54(1):74-82.
giving oxygen to mothers with concerning fetal heart 57. Bakker PCAM, Kurver PHJ, Kuik DJ, Van Geijn HP. Ele-
rate patterns? Am J Obstet Gynecol. 2015;212(4):459- vated uterine activity increases the risk of fetal acidosis
460, 459.e1. at birth. Am J Obstet Gynecol. 2007;196(4):313.e1-313.e6.
42. Haydon ML, Gorenberg DM, Nageotte MP, et al. The 58. Simpson KR, James DC. Effects of oxytocin-induced
effect of maternal oxygen administration on fetal uterine hyperstimulation during labor on fetal oxygen
pulse oximetry during labor in fetuses with nonreas- status and fetal heart rate patterns. Am J Obstet Gyne-
suring fetal heart rate patterns. Am J Obstet Gynecol. col. 2008;199(1):34.e1-34.e5.
2006;195(3):735-738.
59. Heuser CC, Knight S, Esplin MS, et al. Tachysystole in
43. Hamel MS, Hughes BL, Rouse DJ. Whither oxygen term labor: incidence, risk factors, outcomes, and effect
for intrauterine resuscitation? Am J Obstet Gynecol. on fetal heart tracings. [Erratum in Am J Obstet Gyne-
2015;212(4):461-462, 461.e1. col. 2014;210] [2]. Am J Obstet Gynecol. 2013;209(1):32.
44. Fawole B, Hofmeyr GJ. Maternal oxygen administra- e1-32.e6.
tion for fetal distress. Cochrane Database Syst Rev. 60. Simpson KR, Miller L. Assessment and optimization
2012;12(12):CD000136. of uterine activity during labor. Clin Obstet Gynecol.
2011;54(1):40-49.

20  Chapter E —
Intrapartum Fetal Surveillance

61. Kulier R, Hofmeyr GJ. Tocolytics for suspected intra- 69. Westgate JA, Wibbens B, Bennet L, Wassink G, Parer
partum fetal distress. Cochrane Database Syst Rev. JT, Gunn AJ. The intrapartum deceleration in center
2000;(2):CD000035. stage: a physiologic approach to the interpretation of
62. Neilson JP. Fetal electrocardiogram (ECG) for fetal fetal heart rate changes in labor. Am J Obstet Gynecol.
monitoring during labour. Cochrane Database Syst Rev. 2007;197(3):236.e1-236.e11.
2013;5(5):CD000116. 70. East CE, Brennecke SP, King JF, Chan FY, Colditz PB;
63. Devoe LD, Ross M, Wilde C, et al. United States FOREMOST Study Group. The effect of intrapartum
multicenter clinical usage study of the STAN 21 elec- fetal pulse oximetry, in the presence of a nonreassur-
tronic fetal monitoring system. Am J Obstet Gynecol. ing fetal heart rate pattern, on operative delivery rates:
2006;195(3):729-734. a multicenter, randomized, controlled trial (the FORE-
MOST trial). Am J Obstet Gynecol. 2006;194(3):606.
64. Vayssiere C, Haberstich R, Sebahoun V, David E, Roth e1-606.e16.
E, Langer B. Fetal electrocardiogram ST-segment analy-
sis and prediction of neonatal acidosis. Int J Gynaecol 71. Bloom SL, Spong CY, Thom E, et al; National Institute of
Obstet. 2007;97(2):110-114. Child Health and Human Development Maternal-Fetal
Medicine Units Network. Fetal pulse oximetry and cesar-
65. Ojala K, Vääräsmäki M, Mäkikallio K, Valkama M, ean delivery. N Engl J Med. 2006;355(21):2195-2202.
Tekay A. A comparison of intrapartum automated
fetal electrocardiography and conventional cardioto- 72. East CE, Begg L, Colditz PB, Lau R. Fetal pulse oxim-
cography—a randomised controlled study. BJOG. etry for fetal assessment in labour. Cochrane Database
2006;113(4):419-423. Syst Rev. 2014;10(10):CD004075.

66. Greene MF. Obstetricians still await a deus ex machina. 73. Hofmeyr GJ, Xu H, Eke AC. Amnioinfusion for meco-
N Engl J Med. 2006;355(21):2247-2248. nium-stained liquor in labour. Cochrane Database Syst
Rev. 2014;1(1):CD000014.
67. Salamalekis E, Hintipas E, Salloum I, et al. Computer-
ized analysis of fetal heart rate variability using the
acknowledgement: The ALSO® Program thanks
matching pursuit technique as an indicator of fetal
hypoxia during labor. J Matern Fetal Neonatal Med. and acknowledges Mr. Kim Hinshaw, MB, BS,
2006;19(3):165-169. FRCOB for the creation of the DR C BRAVADO
68. Giannubilo SR, Buscicchio G, Gentilucci L, Palla GP, mnemonic. Mr. Hinshaw is a consultant obstetri-
Tranquilli AL. Deceleration area of fetal heart rate trace cian and past chair of ALSO® United Kingdom.
and fetal acidemia at delivery: a case-control study.
J Matern Fetal Neonatal Med. 2007;20(2):141-144.

— Chapter E  21
Objectives
Intrapartum Fetal Surveillance • Summarize fetal monitoring techniques
• Explain the concepts of structured intermittent auscultation 
(SIA) and continuous electronic fetal monitoring (CEFM) 
• Apply standardized National Institute of Child Health and 
Revised January 2017 Human Development (NICHD) terminology when interpreting 
CEFM (NICHD 1997, revised 2008)
• Develop an overall assessment and management plan for 
CEFM and SIA, especially for Category II tracings, using the 
mnemonic DR C BRAVADO and published algorithms
• Discuss future research in fetal monitoring

CEFM Choice of Monitoring Method
• Introduced in the 1960’s  • Based on
• Goal to improve neonatal outcomes has been unfulfilled  – Risk status of mother and fetus
despite widespread use – Patient preference
• Limitations: – Staff
• Availability
– Low specificity
• Training
– Category III ‐ tracing not predictive of poor outcome • Comfort with intermittent auscultation
• Strengths: – Hospital protocols
– High sensitivity • Admission fetal monitoring not required to differentiate 
– Category I ‐ tracing predictive of good outcome between SIA and CEFM

Indications for Fetal Monitoring Indications for Fetal Monitoring
• High‐Risk Conditions – Requires CEFM • Low Risk
– Maternal medical complications (eg, diabetes, chronic  – Can choose CEFM or SIA
hypertension)
– Maternal obstetrical complications (eg, history of previous  – Discussion regarding method of monitoring should occur 
cesarean, post‐term, preeclampsia, gestational diabetes) before labor and address patient preference
– Intrapartum complications and use of oxytocin or prostaglandins – Choice of monitoring may depend on: 
for labor induction/augmentation
– Use of epidural analgesia  • Nursing staff availability and comfort/knowledge with SIA
– Known or suspected fetal conditions (eg, anomalies, fetal  • Hospital protocols
anemia, IUGR)

Copyright 2017© American Academy of Family Physicians. All rights reserved.


1
Structured Intermittent Auscultation: 
Outcomes with CEFM
An Alternative to CEFM
• Evidence from randomized controlled trials (2008) • Place Doppler over point of maximum fetal heart tone intensity 
• CEFM versus SIA • Differentiate maternal pulse from fetal pulse
– No difference in 1‐minute Apgar scores <7 • Palpate uterus for contractions
• Assess fetal heart rate (FHR) at two different intervals:
– No difference in NICU admission rates – Count FHR for 30 to 60 seconds between contractions to establish FHR baseline
– Decrease in neonatal seizures (NNT = 661) – Count FHR for multiple consecutive 6‐second intervals during and immediately 
following the contraction (multiply by 10 to get rate). Assess for differences from 
– Increase in cesarean deliveries, especially in low‐risk  prior baseline rate to determine potential for FHR decelerations.
pregnancies (NNH = 20) • Listen every 15 to 30 minutes in first stage of labor and every 5 to 15 
minutes in second stage of labor
– Increase in operative vaginal deliveries (NNH = 30) Information from Feinstein NF, et al. Fetal Heart Rate Auscultation. 2nd ed. Washington DC; 2008: Association of 
Women’s Health, Obstetric and Neonatal Nurses (AWHONN)

Frequency Review of CEFM Tracings CEFM Interpretation: DR C BRAVADO
• Patients without complications • Determine Risk
– Every 30 minutes in the active labor phase, and every 15 minutes in  • Contractions
the second labor stage after pushing begins
• Patients with complications (eg, IUGR, preeclampsia)
• Baseline RAte
– Every 15 minutes in the active labor phase, and every 5 minutes in the  • Variability Use NICHD definitions
second stage after pushing begins • Accelerations
• Clinicians should periodically document their assessment • Decelerations
Information from ACOG Practice Bulletin No. 106: Intrapartum fetal heart rate monitoring: nomenclature,  • Overall assessment
interpretation, and general management principles. Obstet Gynecol. 2009;114(1):192‐202. DR C BRAVADO was developed by Kim Hinshaw, MB BS, MRCOG 

DR = Determine Risk C = Contractions
• Consider: • Method of monitoring
– Prenatal risk factors – Palpation, external transducer, or intrauterine pressure catheter
• Pattern and intensity
– Intrapartum risk factors
– Adequate strength
– Fetal reserve – Normal: ≤5 contractions in 10 minutes averaged over 
– Labor progress 30 minutes
– Tachysystole: >5 contractions in 10 minutes averaged over 
30 minutes
• Is there risk of uteroplacental insufficiency? – NICHD ‐ terms such as hyperstimulation and hypercontractility
are poorly defined, and should not be used

2
Suggested Tachysystole Management B  RA= Baseline RAte
• Mean FHR rounded to increments of 5 bpm during a 10‐
minute segment.
– Excluding: 
• Segments of baseline that differ >25 bpm
• Periods of accelerations, decelerations and marked variability
• Baseline must be at least 2 minutes (not necessarily 
contiguous 2 minutes) in any 10‐minute window
• Baselines less than 2 minutes in any 10‐minute window are 
considered indeterminate. In this case, refer to prior 10‐
minute window for determination of baseline.
Reprinted from American College of Obstetricians and Gynecologists. Practice bulletin no. 116: Management of intrapartum fetal heart rate tracings. 
Obstet Gynecol. 2010;116(5):1232‐1240.

Normal Baseline Rate 110 to 160 bpm Baseline Bradycardia
• Baseline <110 bpm for greater than 10 minutes
Maternal Causes Fetal Causes
Supine positioning Prolonged cord occlusion/cord prolapse

Hypotension Fetal hypoxia
Connective tissue disease (ie, lupus) Cardiac conduction or anatomic defects

Hypothermia Post dates >42 0/7 weeks
Hypoglycemia
Excessive vagal stimulation (ie, pushing in 
second stage)
Tachysystole

Baseline Bradycardia Baseline Tachycardia
• Baseline >160 bpm for greater than 10 minutes
Maternal Causes     Fetal Causes
Fever/infection/chorioamnionitis Anemia
Dehydration Fetal cardiac abnormalities
Hyperthyroidism Tachyarrhythmias (SVT)
Anemia Chronic fetal hypoxia
Maternal anxiety Severe prematurity
Cocaine, amphetamines Prolonged fetal activity
Parasympatholytic or beta‐sympathomimetic  Vibroacoustic stimulation
drugs (terbutaline)

3
Baseline Tachycardia V = Variability
• Fluctuations in baseline fetal heart rate that are irregular 
in amplitude and frequency
• No longer described as
– Short‐term (beat‐to‐beat), long‐term, or “good”
• Now characterized as
– Absent: undetectable
– Minimal: amplitude range detectable, but 5 bpm
– Moderate: 6 to 25 bpm
– Marked: >25 bpm

Absent Variability Minimal Variability
• Amplitude range is undetectable • Amplitude range detectable but 5 bpm

Moderate Variability Marked Variability
• Amplitude range 6 to 25 bpm • Amplitude range >25 bpm

4
Causes of Decreased Variability A = Accelerations
• Visually apparent, abrupt increase in FHR above baseline
Maternal Causes Fetal Causes
– Onset to peak in <30 seconds
Fever Fetal sleep cycles (20 to 40 minute duration)
• Peak 15 bpm above baseline
CNS depressants (ie, opioids, benzodiazepines, 
magnesium sulfate)
Prematurity
– 10 bpm if <32 weeks gestation
General anesthesia Cardiac anomalies • Lasts for 15 seconds
Alcohol Fetal tachycardia
– 10 seconds if <32 weeks gestation
Corticosteroids Hypoxia/acidosis • Returns to baseline within 2 minutes
Anticholinergics/parasympatholycis CNS anomalies • Prolonged acceleration 2 minutes, but <10 minutes

Accelerations D = Decelerations
• Classification
– Early
– Variable 
– Late
• Decelerations are defined by their rate of onset and their 
timing related to the contraction
• Described as recurrent if decelerations occur with ≥50% of 
contractions in any 20‐minute period
• Described as intermittent if decelerations occur with <50% of 
contractions in any 20‐minute period

Early Deceleration Early Decelerations
• Visually apparent, gradual decrease in FHR with • Mirrors the
return to baseline in association with a uterine contraction
contraction • Onset to nadir 
– Onset to nadir 30 seconds is ≥30 seconds
– Nadir occurs at same time as peak of contraction
• Physiology of early deceleration
– Fetal head compression local changes in blood flow
stimulation of vagal centers in the fetus

5
Variable Deceleration Variable Decelerations
• Visually apparent, abrupt decrease in FHR below baseline
– Onset to nadir ≤30 seconds
– Decrease in FHR is 15 bpm, with duration of 15 seconds but 
2 minutes
– Not necessarily associated with contractions
• Physiology of variable deceleration
– Cord compression rise in fetal peripheral resistance sudden 
fetal hypertension parasympathetic outflow slows fetal 
atrial pacemaker

Amnioinfusion Amnioinfusion Technique
• Reduces cord compression • Check cervix for dilatation, cord prolapse
• Reduces variable decelerations • Obtain patient informed consent
• Place intrauterine pressure catheter (IUPC) and consider 
• Fewer cesarean deliveries when used need for fetal scalp electrode (FSE)
• Use with recurrent variable decelerations  • Infuse normal saline or lactated Ringer solution
• Not useful for late decelerations • Initial volume infused of 250 to 500 mL
• Maintenance rate of 50 to 60 mL/hour
Information from American College of Obstetricians and Gynecologists. Practice bulletin no. 116: Management of intrapartum fetal heart rate tracings. 
• Warming of fluids not needed
Obstet Gynecol. 2010;116(5):1232‐1240.

Late Deceleration Late Decelerations
• Visually apparent, gradual decrease in FHR with return 
to baseline
– Onset to nadir 30 seconds
– Onset, nadir, and recovery of the deceleration occur after the 
beginning, peak, and ending of the contraction respectively
• Physiology of a late deceleration
– Uteroplacental insufficiency fetal hypoxemia O2 
centralization via peripheral vasoconstriction hypertension
myocardial depression
– If hypoxia not corrected, may lead to fetal acidemia

6
Prolonged Deceleration Causes of Sudden Decrease in FHR 
• Any deceleration ≥2 minutes but <10 minutes • Amniotomy
• Cord prolapse
• Vaginal examination
• FSE/IUPC placement
• Tachysystole
• Maternal hypotension or position change
• Maternal vagal maneuvers (pushing, vomiting, 
breath‐holding)

Sinusoidal Sinusoidal
• Smooth, sine wave‐like, undulating pattern in FHR baseline
• Sine wave cycle frequency 3 to 5/minute
• Persists for ≥20 minutes (rules out narcotic induced)
• Etiology:
– Fetal anemia (fetal hemorrhage, RBC hemolysis, Rh sensitization)
– Evolving asphyxia
– May represent a terminal pattern 
• Requires rapid evaluation and response

O = Overall Assessment NICHD FHR Classification System
• Assessment of fetal status (NICHD) based on risk of fetal  • Category I: Normal
acidemia – Follow routinely
– Category I – No risk – Predictive of normal acid‐base status at time of observation
– Category II – Indeterminate risk (insufficient research) • Category II: Indeterminate
– Category III – High risk – Not predictive of abnormal fetal pH status
• Management plan (ACOG Tech Bulletin No. 116) – Unable to classify in Categories I or III due to insufficient data
– Based on clinical context – Requires prompt evaluation and efforts to resolve tracing
– Must include plan for further surveillance • Category III: Abnormal
– Appropriate corrective measures based on the cause of the FHR  – Predictive of abnormal fetal acid‐base status
tracing abnormality
– Prompt evaluation, intervention and consider immediate delivery

7
Category I: 
DR. C BRAVADO
Normal FHR Tracings
• Must have all of these characteristics:
– Baseline 110 to 160 bpm
– Moderate baseline variability
– Late or variable decelerations absent
– Early decelerations present or absent
– Accelerations present or absent

Category II:  Category III: 
Indeterminate FHR Tracings Abnormal FHR Tracings 
• Present at some point during >80% of labors • Sinusoidal pattern
• May show any of the following:
OR
– Tachycardia 
– Baseline with absent, minimal, or marked variability • Absent FHR variability with any of the following:
– Recurrent variable decelerations with minimal to moderate variability – Recurrent late decelerations
– Recurrent late decelerations with moderate variability
– Variable deceleration with slow return, overshoots or shoulders
– Recurrent variable decelerations
– Prolonged deceleration – Bradycardia
– No accelerations after fetal stimulation

Summary: Intrauterine Resuscitation Measures
Suggested FHR Management

Reprinted from American College of Obstetricians and Gynecologists. Practice bulletin no. 116: Management of intrapartum fetal heart rate tracings. Reprinted from American College of Obstetricians and Gynecologists. Practice bulletin no. 116: Management of intrapartum fetal heart rate tracings.
Obstet Gynecol. 2010;116(5):1232‐1240. Obstet Gynecol. 2010;116(5):1232‐1240.

8
Proposed Category II Management Category II Management: Clark Algorithm 
• Two recent proposals for Category II management:
– One proposes an algorithm with specific definitions
– One proposes a 5‐tier classification approach, rather than a 3‐
tiered approach
– Both incorporate NICHD terminology and have developing 
evidence to support
– Both are relatively complicated, but web‐based and mobile apps 
make for easier application
– See your textbook chapter for more information!
Reprinted from 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(2):89‐97.

Category II Management: Clark Algorithm 5‐Tier Management System
• Use of this proposed management scheme involves use of strict  • Uses NICHD criteria for variability 
definitions for significant decelerations • Bradycardia, prolonged, late and variable decelerations 
• Algorithm only used for category II tracings (not applicable for  classified as mild, moderate, and severe
Category I or III)
• Application of algorithm may be delayed for up to 30 minutes while  • Each pattern classified as to risk of evolving acidemia 
intrauterine resuscitation measures are being implemented using a color‐coding system
• Algorithm reapplied every 30 minutes • 3‐steps:
• Algorithm not applicable for extreme prematurity – What is baseline rate?
• Complexity decreased with web‐based app (NOT MOBILE) at  – What is variability?
http://cat2.perigen.com/cat2 – What is type of recurrent deceleration?
Parer JT, Ikeda T. A framework for standardized management of intrapartum fetal heart rate patterns. Am J Obstet Gynecol. 2007;197(1):26.e1‐26.e6.

Proposed 5‐Tier System Alternatives to Scalp pH
• Scalp pH sampling now uncommon
– There are noninvasive alternative methods to determine absence of 
fetal metabolic acidemia such as:
• Assessment for moderate baseline FHR variability
• Assessment for FHR accelerations (15 bpm x 15 seconds or 10 bpm 10 second 
for <32 weeks gestation)
– Spontaneous
– Stimulated (acoustic, scalp stimulation)
– If accelerations are present, fetal capillary pH >7.20
• Less data regarding interpretation as a method if no 
accelerations are present
Complexity decreased by use of an app at http://www.obapps.org/

9
Future Monitoring Research Summary
• Fetal electrocardiogram (ECG) wave analysis • CEFM is widely used
– STAN – SIA is acceptable and perhaps preferable in the low risk patient 
• Expensive • DR C BRAVADO and NICHD definitions provide systematic ways to define 
• Did reduce operative deliveries and fetal scalp sampling tracings as Category I, II, or III
• Recommendations for the management of Intrapartum FHR tracings and 
• Computerized cardiotocography (CTG) tachysystole have been published ‐ ACOG Technical Bulletin No. 116
– Awaiting further studies • Two newly proposed Category II management systems are promising but
• Continuous fetal oximetry complicated; apps are available
• Presence of moderate variability and/or accelerations is key in utilizing 
– No proven benefit in reducing cesarean delivery rates algorithms for management of Category II tracings
– Currently only utilized in research settings • New technologies such as fetal ECG analysis continue to be evaluated 

10
Chapter F
Labor Dystocia

Sara G. Shields, MD, MS, Stephen Ratcliffe, MD, MPH


Revised January 2017

Learning Objectives
At the end of this activity, learners will be able to:
1. Understand the difference between normal labor 3. Summarize prevention methods.
and labor dystocia. 4. Describe how obesity and induction may affect
2. Describe how to diagnose and treat dystocia. labor dystocia.

Introduction to avoid performing cesareans during a prolonged latent


Caring for women with dystocia, literally meaning diffi- phase that is mistaken for active labor. During latent
cult labor, is one of the greatest challenges of maternity labor, regular painful contractions result in minimal or
care. Although this particular challenge may not need slow cervical change. The latent phase of labor begins
the same emergent treatment as other clinical scenarios with maternal perception of painful regular contractions
in the ALSO course, labor dystocia is common, and and ends when the rate of dilation begins to accelerate.8
appropriate evidence-based care can improve clinical Recent studies suggest that normal labor may have
outcomes. This chapter reviews important concepts in a longer latent phase and thus a wider time range and
the diagnosis, treatment, and prevention of dystocia. a less clear-cut transition to active labor than previ-
Dystocia refers to prolonged or slowly-progressing ously defined by Friedman in the 1950s.9 Reanalysis
labor, which is a common situation for nulliparous of data from the large National Collaborative Perinatal
women as gauged by the number that require augmen- Project from the 1960s, when fewer obstetric interven-
tation, operative vaginal delivery, or cesarean delivery. tions were practiced, has allowed the normal course
In 2014, approximately 20% of women in the United of natural spontaneous labor to be more accurately
States received oxytocin augmentation,1 and, in 2013, defined. This reanalysis showed that the active phase
the primary cesarean delivery rate (cesarean delivery of labor, when rapid dilation begins, may not start for
in women without a prior cesarean) in reporting states multiparous women until at least 5 cm of dilation, and
was 22.4%.2 Dystocia is responsible for 30% to 50% it may not start for nulliparous women until an even
of primary cesarean deliveries in the United States.3,4 greater dilation that is harder to define.10 Similar data
Although the overall US cesarean delivery rate has from contemporary studies suggest that the active phase
declined slightly in recent years, it remains high at more of labor may not occur for multiparous women until
than 32% (Figure 1),5 and thus attention is focusing on 6 cm dilation.11 A 2013 study showed that more than
the two main drivers of the primary cesarean rate—dys- 40% of primiparous women and over 30% of multipa-
tocia and fetal heart rate (FHR) tracing interpretation.3,6 rous women who are undergoing cesarean delivery for
The wide variation in cesarean delivery rates for labor dystocia are actually less than 5 cm dilated and not in
dystocia suggests a need for more thoughtful, evidence- active labor.4
based care.3,4,7 All maternity care clinicians need exper- Not only is the start of active labor later and harder
tise in the care of women experiencing dystocia. to define than previously thought, the normal rate
of cervical change in active labor is slower. Table 1
Rethinking Latent Labor lists more accurate labor stage durations. In one large
Understanding normal and abnormal labor progress retrospective cohort study, after 6 cm dilation, nullipa-
requires understanding latent or prodromal labor, so as rous women took an average of 2.2 hours to reach full

Copyright 2017© American Academy of Family Physicians. All rights reserved. — Chapter F  1
Chapter F

Figure 1. Total and Primary Cesarean Rate and Vaginal Birth After Previous Cesarean
(VBAC): United States, 1989-2014
Total cesarean section Primary (initial) Cesarean section Vaginal birth after cesarean
section (VBAC)
34

30
Rate per 100 Live Births

26

22

18

14

10

6
1989 1991 1993 1995 1997 1999 2001 2003 2005 2007 2009 2011 2013
Year

Primary Cesarean and VBAC Rates From Revised (2003) US Birth Certificate, 2005-2012

Primary Number of states % of total


Year cesarean rate VBAC rate using certificate US births

2005 24.3% 10.1% 12 31%


2006 23.5% 8.5% 19 49%
2007 23.4% 8.3% 22 53%
2008 23.8% 8.4% 27 65%
2009 23.8% 8.4% 28 66%
2010 23.6% 9.2% 33 76%
2011 23.3% 9.7% 36 83%
2012 21.5% 10.2% 38 86%

Data are not available to delineate national trends in rates of primary cesarean section and vaginal birth after cesar-
ean (VBAC) from 2005 onward, due to differences in adoption of the revised (2003) birth certificate form. The above
data are provided to help understand recent patterns of use but should not be used to compare figures from year to
year due to the changing cohorts using the revised 2003 form.

dilation, with the 95th percentile standard devia- interventions that increases the risk of cesarean
tion at 10.1 hours, or less than 0.5 cm per hour.11 delivery. Clinicians need to recognize that cervical
According to Friedman, the outer limit of progress dilation is not linear, particularly for nulliparous
for nulliparas is 1.2 cm per hour in active phase. women in early labor.10
The clinical implication of using an inaccurate To prevent the misdiagnosis of labor dystocia in
labor curve is that many women are inadvertently women who are still in the latent phase, clinicians
admitted before progressive labor yet held to can avoid admitting women too early.13 This prac-
traditional expectations of the rate of progress tice reduces by more than half the risk of needing
of active labor,12 thus resulting in a misdiagno- augmentation of labor or epidural analgesia.14 In a
sis of labor dystocia with a resultant cascade of recent prospective cohort study, women admitted

2  Chapter F —
Labor Dystocia

Table 1. Labor Progress in Nulliparous and Multiparous Women

Nulliparous Multiparous

Active Labor starts >6 cm >6 cm


Rate of dilation in active Median 1.8 cm/hr (95% SD 0.4 cm/hr) Median 2.5 cm/hr (95% SD 0.4 cm/hr)
labor
2nd stage duration Median 0.9 hr (95% SD 3.1 hr) Median 0.3 hr (95% SD 1.7 hr)

Information from Laughon SK, Branch DW, Beaver J, Zhang J. Changes in labor patterns over 50 years. Am J Obstet
Gynecol. 2012;206(5):419.e1-419.e9; Spong CY, Berghella V, Wenstrom KD, Mercer BM, Saade GR. Preventing the first
cesarean delivery: summary of a joint Eunice Kennedy Shriver National Institute of Child Health and Human Develop-
ment, Society for Maternal-Fetal Medicine, and American College of Obstetricians and Gynecologists Workshop. Obstet
Gynecol. 2012;120(5):1181-1193.

before 6 cm dilation had a significantly higher risk ing on how dilated a woman is. For example,
of cesarean delivery compared to women admitted using Table 3, if she is 5 cm dilated, dystocia is
with more advanced dilation (13.2% versus 3.5%; the diagnosis if it takes longer than 3.2 hours to
relative risk [RR] 3.73; 95% confidence interval change to 6 cm, or therefore is progressing less
[CI] = 1.94 to 7.17).15 As alternatives to admission than 0.3 to 0.4 cm per hour. Similarly, if she is
in latent labor, clinicians can encourage adequate 8 cm dilated, dystocia occurs if she is progressing
hydration, rest, emotional and physical support, less than 0.7 cm per hour. Given the limited accu-
and if needed, pharmacologic sedation with anti- racy of cervical examinations for minor degrees
histamines or opiates such as morphine.16 of dilation and the desire to avoid increased risk
Patience is critical for women who are admit- of infection, examinations to assess progress are
ted before active labor. This is especially impor- commonly performed at 2- to 4-hour intervals. As
tant before 6 cm of dilation. Options for care of part of making this diagnosis, the clinician needs
women admitted between 3 and 6 cm of dilation
include an antepartum unit for rest and support,
including the availability of pharmacologic treat- Table 2. Definitions of Failed Induction and Arrest
ment for maternal exhaustion or pain, as previ- Disorders
ously mentioned. There should be clear maternal
or fetal indications to augment labor during the Failed Induction of Labor
Failure to generate regular (eg, every 3 min) contractions and cervical
latent phase to justify the risks associated with change after at least 24 hours of oxytocin administration, with artificial
oxytocin augmentation such as uterine tachysys- membrane rupture if feasible
tole, fetal intolerance of labor, and increased rates First-Stage Arrest
of operative intervention.8,17 Latent labor may be Six centimeters or greater dilation with membrane rupture and
longer in women undergoing labor induction than no cervical change for 4 hours or more of adequate contractions
in women in spontaneous labor. Cesarean delivery (eg, >200 Montevideo units) or 6 hours or more of oxytocin
administration if contractions inadequate
for labor dystocia should not be done in latent
labor unless certain criteria are met (Table 2).3 Second-Stage Arrest
No progress (descent or rotation) for:
Diagnosis of Labor Dystocia ≥4 hours in nulliparous women with an epidural
≥3 hours in nulliparous women without an epidural
Contemporary studies have led to new definitions ≥3 hours in multiparous women with an epidural
of arrested labor. Once the clinician ascertains that ≥2 hours in multiparous women without an epidural
a woman has reached active labor (6 cm of dila-
tion), the rate of change may accelerate (Table 3), Adapted from Spong CY, Berghella V, Wenstrom KD, Mercer BM, Saade GR.
but patience regarding the rate of dilation remains Preventing the first cesarean delivery: summary of a joint Eunice Kennedy
Shriver National Institute of Child Health and Human Development, Society for
important. 3,10 Maternal-Fetal Medicine, and American College of Obstetricians and Gyne-
Clinical dystocia can be defined as the 95th per- cologists Workshop. Obstet Gynecol. 2012;120(5):1181-1193.
centile of dilation rate at different points depend-

— Chapter F  3
Chapter F

0.95).18 Nulliparous women who had amniotomy


Table 3. Duration of Each Centimeter had a slightly shorter second stage of labor than
Change in Cervical Dilatation for those who did not (but only by 1 to 10 minutes).
Nulliparous Women with Spontaneous However, routine amniotomy alone in spontane-
Onset of Labor ous labor does not change overall first stage labor
length or cesarean delivery rates.
Cervical Median 95th Percentile
Change (cm) (hour) (hour) Pharmacologic augmentation. Oxytocin alone
does not improve vaginal delivery rates. Although
3 to 4 1.8 8.1 the overall duration of labor may shorten in
4 to 5 1.3 6.4 women who receive oxytocin early in labor for
5 to 6 0.8 3.2 slow labor, this intervention alone does not ulti-
6 to 7 0.6 2.2 mately change the cesarean delivery rate.19
7 to 8 0.5 1.6 Both amniotomy and pharmacologic aug-
8 to 9 0.5 1.4
mentation. Combining amniotomy with oxytocin
9 to 10 0.5 1.8
may be the most effective augmentation strategy.
Adapted from Zhang J, Landy HJ, Branch DW, et al;
In trials of prevention strategies for labor dystocia,
Consortium on Safe Labor. Contemporary patterns of early augmentation with both amniotomy and
spontaneous labor with normal neonatal outcomes. oxytocin was associated with a slight reduction in
Obstet Gynecol. 2010;116(6):1281-1287.
the number of cesarean births (RR 0.87; 95% CI
= 0.77 to 0.99) and a shortened duration of labor
(average mean difference [MD] -1.28 hours; 95%
to address the following questions, considering the CI = -1.97 to -0.59), without differences in neo-
6 Ps mnemonic: natal outcome or maternal morbidity or satisfac-
• Passenger: Is there a malposition or malpresen- tion.20 In this analysis, the number needed to treat
tation or suspected macrosomia? to prevent 1 cesarean by using amniotomy and
• Power: Are contractions adequate in fre- oxytocin together early in labor was 65. However,
quency, duration, and strength? only three studies in this meta-analysis were for
• Pelvis: Is there cephalopelvic disproportion treatment for dystocia, with the combination of
because of a contracted pelvis? amniotomy and oxytocin use showing no reduc-
• Patient: Are there other coexisting clinical issues tion in cesarean delivery rates for women already
such as chorioamnionitis or nonreassuring fetal diagnosed with slow progress in labor.
monitoring that affect the treatment choices? For women with protracted or arrested active-
• Psyche: How are the woman and her support phase labor, the clinician can evaluate the strength
individuals coping with the labor? and frequency of uterine contractions by abdomi-
• Provider: Is there a consistent examiner to nal palpation or an intrauterine pressure catheter
evaluate subtle change? Are there any indica- (IUPC), which allows calculation of Montevideo
tions for consultation or second opinion? units (MVUs) (Figure 2). Two hundred or more
MVUs in 10 minutes is considered evidence of
Treatment of Active-Phase Labor adequate contractions.21 IUPC use may be most
Dystocia helpful if contractions seem to be of sufficient
First Stage Active-Phase Dystocia frequency and duration but are not causing the
Treatment strategies for women with slowly pro- expected cervical change. However, a meta-
gressing active-phase labor include amniotomy, analysis concluded that the use of an IUPC does
pharmacologic augmentation with oxytocin, or both. not appear to change labor duration or cesarean
Amniotomy alone. A meta-analysis showed rates.22 The use of an IUPC may increase the risk
that women of any parity undergoing amniotomy of maternal fever so should not be routine. 23
alone were less likely to have nonprogressing labor If contractions are inadequate, administering
than women who did not (RR 0.75; 95% CI = intravenous (IV) oxytocin increases contraction
0.64 to 0.88); similarly, women who underwent frequency, duration, and strength. There are
amniotomy routinely were less likely to need oxy- numerous approaches to dosage, dosing interval,
tocin augmentation (RR 0.73; 95% CI = 0.57 to and duration of oxytocin treatment. Low-dose

4  Chapter F —
Labor Dystocia

regimens, which attempt to mimic the known to 8% without increasing maternal or fetal mor-
steady-state pharmacodynamics of oxytocin, start bidity.26,27 Expectant management for longer time
at 1 to 2 mU/min and increase by 1 to 2 mU/ frames (eg, 6 hours) is under study to assess the
min every 30 minutes to maximum doses of 36 potential for further decreasing cesarean delivery
mU/min.21 High-dose regimens, which should be rates versus increasing the risk of complications
routinely used only in nulliparous women, have including chorioamnionitis and postpartum hem-
starting doses of 4 to 6 mU/min and incremen- orrhage as labor duration increases.
tal increases of 1 to 6 mU/min up to maximums In clinical practice, it is sometimes challeng-
of 36 mU/min.21 High-dose oxytocin for labor ing to separate out whether a cesarean delivery
augmentation appears to result in shorter labors is being done for lack of labor progress or for
and to reduce the likelihood of cesarean delivery,24 fetal intolerance to oxytocin needed to affect that
but further trials are needed to assess important progress. Clinicians should follow the National
potential adverse outcomes and address women’s Institute of Child Health and Human Develop-
experience of this treatment strategy. ment (NICHD) categories of FHR monitoring
Appendix 1 lists sample oxytocin orders. Any use to classify abnormalities (Chapter E: Intrapartum
of oxytocin is usually combined with the routine Fetal Surveillance).
use of continuous electronic FHR monitoring or
the use of structured intermittent fetal ausculta- Second Stage Labor Dystocia
tion every 15 minutes in stage one and every 5 Labor dystocia can also occur in the second stage
minutes in stage two.25 Oxytocin is a drug with of labor and is characterized by prolonged dura-
the potential for adverse effects so its use should be tion or arrest of descent (Table 2). The median
reserved for clearly defined indications.17 second-stage duration lasts longer for nulliparous
Once oxytocin has been started to augment women than traditionally defined with the 95th
slow labor, recent studies again suggest the need percentile at 2.8 hours without regional anes-
for a more patient approach in monitoring labor thesia and 3.6 hours with regional anesthesia.3
progress.3 Assuming fetal well-being, the clinician For multiparous women, the 95th percentiles for
and mother should wait for at least 4 hours of second-stage duration with and without regional
adequate contractions after oxytocin augmenta- anesthesia remained around 2 hours and 1 hour,
tion before operative intervention for arrested respectively.10 Studies on the neonatal and mater-
dilation. A study showed that not performing a nal effects of a prolonged second stage yield mixed
cesarean delivery for labor dystocia until at least results,28 but the likelihood of vaginal delivery
4 hours of no cervical change during active labor diminishes when the second stage lasts longer than
rather than the traditional 2 hours lowered the the time frames noted in Table 2.29,30 Prolongation
cesarean delivery rate for arrest of labor from 26% of the second stage beyond an arbitrary time limit

Figure 2. Measurement of Uterine Contraction Strength by Montevideo Units

— Chapter F  5

You might also like