Endocrine Disorders During Pregnancy 2006
Endocrine Disorders During Pregnancy 2006
Endocrine Disorders During Pregnancy 2006
CONTENTS
Foreword
Derek LeRoith
Preface
Susan J. Mandel
Adrenal Disorders in Pregnancy
John R. Lindsay and Lynnette K. Nieman
xi
xiii
In normal gestation, hypercortisolism and relative hyperaldosteronism are not usually clinically apparent. In contrast, adrenal disorders
that do occur during pregnancy contribute to significant maternal
and fetal morbidity. This article reviews the natural history, causes,
diagnosis, and treatment of adrenal causes of Cushings syndrome,
adrenocortical hypofunction, primary hyperaldosteronism, and the
management of adrenal pheochromocytoma in pregnancy.
21
VOLUME 35
53
79
This article discusses the risks of pregnancy for mothers with type
1 diabetes and their babies, emphasizing appropriate preconception counseling and management goals; optimizing glycemic monitoring and intensifying insulin therapy during the metabolically
dynamic process of pregnancy; appropriately evaluating women
and their fetuses for complications of both diabetes and intensive
diabetes therapy during pregnancy; and peripartum and postpartum glycemic control.
99
vi
CONTENTS
117
Thyroid physiology and pathophysiology during pregnancy are reviewed focusing on the distinction between thyroid disorders and
normal gravid physiology using the history, clinical examination,
and laboratory testing. Management of hypothyroidism is discussed, including the effects of insufficient maternal levothyroxine
therapy on the fetus and recommendations for optimizing maternal levothyroxine therapy. Therapy of hyperthyroidism is reviewed, with attention to the fetal effects of maternal antithyroid
therapy. In addition, diagnosis and therapy of fetal hyperthyroidism is presented. A practical evaluation of thyroid nodules during
pregnancy is presented.
137
Hypertension in Pregnancy
Caren G. Solomon and Ellen W. Seely
157
CONTENTS
vii
Endocrinology of Parturition
Victoria Snegovskikh, Joong Shin Park, and Errol R. Norwitz
173
193
Index
viii
205
CONTENTS
FORTHCOMING ISSUES
June 2006
Impaired Glucose Tolerance and Cardiovascular
Disease
Willa A. Hsueh, MD, and Preethi Srikanthan, MD
Guest Editors
September 2006
Molecular Basis of Inherited Pancreatic Disorders
Markus M. Lerch, MD, Thomas Griesbacher, MD,
and David C. Whitcomb, MD, Guest Editors
December 2006
Growth Hormones
Ken K.Y. Ho, MD, Guest Editor
RECENT ISSUES
December 2005
Endocrinology of Aging
Johannes D. Veldhuis, MD, Guest Editor
June 2005
Cushings Syndrome
James W. Findling, MD, and Hershell Raff, PhD
Guest Editors
March 2005
Type 2 Diabetes and Cardiovascular Disease
Daniel Einhorn, MD, and Julio Rosenstock, MD
Guest Editors
Foreword
0889-8529/06/$ - see front matter 2005 Elsevier Inc. All rights reserved.
doi:10.1016/j.ecl.2005.11.001
endo.theclinics.com
xii
FOREWORD
Preface
It has been 11 years since the last publication of an issue of the Endocrinology and Metabolism Clinics of North America that focused on endocrine
disorders during pregnancy. In the preface, Lois Jovanovic, guest editor of
and contributing author in that issue, wrote that there were three endocrine
components to mammalian pregnancy: fetal, placental, and maternal endocrinology. And, of these, she observed, the maternal was the most understood and the focus of many of the articles contained in that issue.
Fortunately, in the ensuing 11 years, we have learned much about the other
two components, specically the inuences of maternal endocrine disorders
on the fetoplacental unit.
In this issue, I have selected topics that cover each of the main subspecialties (adrenal, pituitary, diabetes, thyroid, and calcium/bone) within adult
endocrinology. For each area, the authors have delineated the current
understanding of its physiology and pathophysiology during gestation as
well as outlined the maternal therapy to optimize pregnancy outcome. In addition, I have included an article on the impact of polycystic ovarian syndrome on fertility, relevant because of its prevalence and the availability
of therapy. Cardiovascular endocrinology, a newer subspecialty within
our discipline, is well-represented by the article on pregnancy and hypertensive disorders. There are two other nontraditional articles. Recently, we
have gained understanding of the eects of the intrauterine milieu on the future endocrine development of the child and this is presented in the section
on imprinting. Lastly, the conclusion of pregnancy, parturition, represents
a complex hormonal interplay that is described in the article on endocrinology of parturition.
0889-8529/06/$ - see front matter 2005 Elsevier Inc. All rights reserved.
doi:10.1016/j.ecl.2005.10.001
endo.theclinics.com
xiv
PREFACE
I have had the pleasure and honor of working with renowned experts in
their respective elds and in large part because of them, it has been a privilege to serve as the Guest Editor of this issue of the Endocrinology and Metabolism Clinics of North America. I thank my colleagues for their
outstanding contributions to this volume, which is a valuable and timely addition to its eld.
Susan J. Mandel, MD, MPH
Division of Endocrinology, Diabetes, and Metabolism
University of Pennsylvania School of Medicine
415 Curie Boulevard
611 CRB
Philadelphia, PA 19104, USA
E-mail address: smandel@mail.med.upenn.edu
The hypothalamic-pituitary-adrenal (HPA) axis and the renin-angiotensin system (RAS) are up-regulated during normal pregnancy. Pregnancy
represents a state of relative hypercortisolism, resulting from the interaction
of the maternal HPA axis and the fetal-placental unit. Consistent with
a physiologic role, the RAS maintains normal sodium balance and volume
homeostasis. In normal gestation, hypercortisolism and relative hyperaldosteronism are not usually clinically apparent. In contrast, adrenal disorders
that do occur during pregnancy contribute to signicant maternal and fetal
morbidity. This article reviews the natural history, causes, diagnosis, and
treatment of adrenal causes of Cushings syndrome, adrenocortical hypofunction, primary hyperaldosteronism, and the management of adrenal
pheochromocytoma in pregnancy.
endo.theclinics.com
nodular adrenal disease and adrenocorticotropic hormone (ACTH)-independent hyperplasia, caused possibly by aberrant receptor stimulation, accounted for eight cases of the pregnancies [1]. The overall increased
incidence of adrenal Cushings syndrome suggests that anovulation may
be less prevalent in this cause or that unrecognized, illicit luteinizing hormone/human chorionic gonadotropin receptor expression was considered
to be adrenal adenoma [6,7]. In the latter setting, Cushings syndrome would
not be triggered until pregnancy is established.
Cushings syndrome in gestation is associated with maternal morbidity,
including preeclampsia, hypertension, diabetes, wound breakdown, opportunistic infections, and fracture [4,8]. Maternal death was reported in two
cases [5,9]. Hypercortisolism eects on the fetus include increased rates of
spontaneous abortion, perinatal death, premature birth, and intrauterine
growth retardation [2,4]. As a result of this fetal and maternal morbidity,
early diagnosis and treatment of Cushings syndrome in pregnancy are critical. However, the physiologic changes of pregnancy complicate this goal.
Plasma cortisol, cortisol-binding protein (CBG), and urinary free cortisol
(UFC) increase during the second and third trimesters (Fig. 1) [10,11]. There
are no formal studies of how the usual diagnostic criteria should be modied
to allow for pregnancy-induced hypercortisolism.
Screening and dierential diagnosis of Cushings syndrome
The interpretation of screening tests for hypercortisolism is more dicult
in pregnancy, particularly in the second and third trimesters. In the rst trimester, cortisol excretion is similar to that of nonpregnant women. It then
increases up to threefold by term, to overlap values seen in pregnant women
with Cushings syndrome [10]. As a result, it is likely that only the UFC values in the second and third trimester greater than three times the upper limit
of normal can be interpreted as indicating Cushings syndrome. Unfortunately, there are limited data on the upper range of UFC in normal pregnancy, using modern antibody-based assays, and no reports of UFC
measured by structural assays such as mass spectrometry. Plasma cortisol
diurnal variation is preserved in normal pregnancy, albeit with a higher
nighttime nadir. Although the loss of diurnal variation is characteristic of
Cushings syndrome, diagnostic thresholds for evening plasma or salivary
cortisol levels in pregnant patients have not been developed [1,10]. Furthermore, the suppression of serum and urinary cortisol by dexamethasone is
blunted in pregnancy (Fig. 2). Thus, this screening test has an increased potential for false-positive results in pregnancy. Although the evidence is limited, the present authors suggest a combination of greater than threefold
elevation of UFC and elevated midnight salivary cortisol as a diagnostic
strategy for the identication of Cushings syndrome in pregnancy [12,13].
The rst step in the dierential diagnosis is to discriminate between
ACTH-independent and ACTH-dependent hypercortisolism, which may
Fig. 1. Serial increases in serum cortisol (open circles) and ACTH (closed circles) during pregnancy in normal controls throughout pregnancy. This graph was modied from the series of ve
normal pregnant women studied by Carr and colleagues (1981). The bars adjacent to the right
axis are summary data derived from a recent series [1] to denote the range of serum cortisol observed in Cushings syndrome in pregnancy (open diamonds, median and range; n 52); ACTH
values for Cushings disease (closed triangles, median and range, n 18); and adrenal Cushings
syndrome (open triangles, median and range, n 17). (Modied from Carr BR, Parker CR Jr,
Madden JD, et al. Maternal plasma adrenocorticotropin and cortisol relationships throughout
human pregnancy. Am J Obstet Gynecol 1981;139(4):41622.)
be achieved by measuring plasma ACTH levels [14]. A two-site immunometric assay is preferred to radioimmunoassay, unless the latter can reliably discriminate low or suppressed ACTH levels (%10 pg/mL [2.2 pmol/L]) [14,15].
To avoid falsely low results, it is important to collect the sample in prechilled
ethylenediaminetetraacetic acid tubes, transport it to an ice bath, and prepare it promptly by refrigerated centrifugation and plasma separation.
In the nonpregnant population, plasma ACTH suppression (%5 pg/mL
[1.1 pmol/L]) identies ACTH-independent primary adrenal causes of Cushings syndrome. However, in a recent review of Cushings syndrome in pregnancy, mean plasma ACTH levels were nonsuppressed in approximately
half of those patients who had primary adrenal disorders, perhaps because
of continued stimulation of the maternal HPA axis by placental CRH or because of placental secretion of ACTH [1,16]. As a result, the recommended
diagnostic ACTH thresholds in the general population may lead to inaccurate diagnoses in pregnancy [15].
If plasma ACTH is suppressed, no further biochemical testing is needed,
and imaging of the adrenal glands, discussed below, then will localize the
Fig. 2. Change in plasma cortisol before (closed circles) and after (open circles) the administration
of 1 mg of dexamethasone (Dex) in pregnant women. Blood was drawn at 8 AM. A single dose of
dexamethasone was administered orally at 11 PM, and blood was drawn at 8 AM on the following
day. (From Odagiri E, Ishiwatari N, Abe Y, et al. Hypercortisolism and the resistance to dexamethasone suppression during gestation. Endocrinol Jpn 1988;35:68590; with permission.)
UFC elevated
Elevated midnight
salivary cortisol
CS confirmed
Adrenal Imaging
US/MRI
Investigate CD
YES
Fig. 3. Diagnostic algorithm for the dierential diagnosis of Cushings syndrome (CS) in
pregnancy.
1947 [28]. The present authors believe there have been no reports of maternal deaths from AI in pregnancy since the 1950s, with the introduction of
cortisone, earlier diagnosis, and improved antenatal care. However, a reported case of maternal death at 8 months after delivery illustrates the importance of careful postpartum follow-up [26]. Some cases that were not
diagnosed during pregnancy had normal maternal and fetal outcomes, suggesting that women with unrecognized AI benet from transplacental passage of cortisol from the fetus. Therefore, AI may only become apparent
in the immediate postpartum period [29]. In women known to have the disease, careful titration of glucocorticoid and mineralocorticoid replacement
are required to avoid an adrenal crisis and potential side eects, including
hypertension and exacerbation of preeclampsia [26,30].
Gestational AI has been associated with high rates of intrauterine growth
retardation, low birth weight [31,32], and fetal mortality [33]. A recent Italian series reported improved miscarriage rates (16 of 104 pregnancies) compared with intrauterine death rates in the 1950s of 40% to 50% [34]. Most of
the earlier reports consisted of previously unrecognized cases or preceded
the availability of modern glucocorticoid regimens [35]. These dierences
have led to the speculation that, treated properly, women do not have increased complications of gestation. There does not appear to be an increased
risk of preterm abortion or congenital anomalies resulting from AI alone,
when patients are treated adequately [36]. However, when AI is associated
with other autoimmune conditions, including positive circulating anticardiolipin antibodies, lupus anticoagulant, and diabetes, there may be additional risks of preterm abortion [3739].
Diagnosis
Clinical and laboratory features
Most cases of primary AI are diagnosed before pregnancy. During gestation, a new diagnosis should be considered in a case with classic symptoms
of excessive fatigue, malaise, weight loss, vomiting, orthostasis, abdominal
pain, hyperpigmentation, or biochemical disturbance [30,39,40]. Hypoglycemia, salt craving, malaise, seizures, or even coma should prompt testing of
the HPA axis in pregnancy. Persistent vomiting can be mistaken for hyperemesis gravidarum, potentially leading to a fatal outcome if left undiagnosed [40]. Biochemical disturbance with hyponatremia is characteristic;
however, this is usually more severe than the mild hyponatremia (5-mmol/L
decrease) that occurs in normal pregnancy. Severe hyponatremia or metabolic acidosis is associated with poor outcomes, including fetal death [33].
Hyperkalemia was reported to be absent in several cases of newly diagnosed
primary AI during pregnancy. This may reect the increase in the RAS,
rather than the severity of adrenocortical dysfunction [33,40].
Although criteria for diagnosing AI in pregnancy have not been developed, the previously reported 8 AM third-trimester plasma cortisol levels
and responses to the SCT in normal pregnant women and the results
from the low-dose cosyntropin stimulation testing cited above provide useful information. Taken together, the present authors consider basal or
ACTH-stimulated plasma cortisol levels in the third trimester greater than
30 mg/dL (828 nmol/L) sucient to exclude AI [42].
Tests used for the dierential diagnosis
Plasma ACTH levels dierentiate primary (elevated) from secondary adrenal failure (low or normal) and can help to conrm primary AI in nonpregnant patients who have borderline plasma cortisol levels. An ACTH
level above 100 pg/mL (22 pmol/L) is generally consistent with primary
AI, even in late pregnancy [43]. However, ACTH levels uctuate widely
day-to-day, and a single value cannot be relied on for the diagnosis of either
primary or secondary AI.
Approximately 90% of nonpregnant patients who have idiopathic AI are
positive for 21-hydroxylase antibodies, and antibodies to 17-a-hydroxylase
and side-chain cleavage enzymes are positive in approximately 30% of patients [51]. The presence of adrenal antibodies provides conrmatory evidence for an autoimmune cause, but it cannot be relied on for diagnosing
AI, given the 10% prevalence of negative test results in patients proven to
have AI. The presence of mineralocorticoid deciency is highly suggestive
of primary AI arising from adrenocortical atrophy, and in this setting, plasma
aldosterone-to-renin ratios are low in association with elevated plasma renin activity [52].
Patients who have an autoimmune cause do not require imaging. Imaging
of the adrenal glands can detect the large glands associated with other
causes, such as tuberculous or fungal infection, bilateral metastases, hemorrhage, or infarction [39,53,54]. US imaging is safe but may have limited resolution. Non-gadolinium-enhanced MRI is preferred to CT in pregnancy.
Although MRI provides excellent soft tissue enhancement and has improved
resolution compared with ultrasound [55], deferring adrenal imaging until
the postpartum period should be considered if the patient is clinically stable.
Treatment
The optimal antenatal management of AI occurs in a multidisciplinary
clinic that includes an endocrinologist. The primary roles for the endocrinologist are to provide a diagnosis, monitor the adequacy of corticosteroid or
mineralocorticoid replacement regimens during gestation, crisis, and labor,
and to ensure continuity during the postpartum period. Prepregnancy
counseling should be conducted routinely in women of childbearing age.
Women should be educated regarding the principles of self-administration
10
of intramuscular hydrocortisone (50100 mg) for pregnancy-associated emesis before urgent medical assessment. All patients should wear a medic alert
bracelet or necklace for identication during an emergency.
Physiologic glucocorticoid and mineralocorticoid treatment appears to
be safe during pregnancy [56]. The most critical periods in pregnancy
when the adequacy of glucocorticoid replacement regimens may aect outcomes are in undiagnosed cases during the rst trimester when symptoms of
adrenal crisis can be mistaken for pregnancy-associated emesis and during
the stress of labor and delivery [33].
Several glucocorticoid regimens are available for chronic replacement
during pregnancy. Hydrocortisone is the present authors preferred choice,
at a replacement dose of 12 mg/m2 to 15 mg/m2 of body surface area [57].
Two thirds of the daily dose is given when the patient awakens, with the remainder given in the afternoon to mimic normal diurnal variation. Although this dose may lead to over-replacement in a proportion of
nonpregnant cases, in the absence of more detailed studies during pregnancy, this approach seems reasonable [58]. Glucocorticoid replacement
regimens usually remain stable during gestation, even until late in the third
trimester. Prednisone or prednisolone may be used as alternative glucocorticoid regimens. Prednisone and cortisone acetate are not useful for the
emergency management of adrenal crisis because they require the reduction
of a ketone group to a hydroxyl group on carbon-11 to active metabolites.
Furthermore, they are longer acting compared with hydrocortisone and less
suitable for physiologic replacement [59].
Oral udrocortisone, 0.1 mg daily (ranging between 0.05 mg and 0.2 mg),
is standard mineralocorticoid replacement and has largely replaced salt tablet regimens [59]. Fludrocortisone dosages are usually stable through pregnancy but occasionally may need to be reduced during the third trimester
to avoid the side eects of edema or exacerbation of hypertension [60].
The acute treatment of adrenal crisis includes prompt, rapid glucocorticoid replacement with hydrocortisone, 100 mg to 200 mg IV, as a single bolus. Thereafter a bolus of 50 mg to 100 mg is given every 6 to 8 hours during
the acute period, based on maximal cortisol production rates of 200 mg/d to
400 mg/d [30]. Women with hypoglycemia should receive 5% dextrose infusions, and those with hypotension should receive 0.9% saline. Because hydrocortisone at these dose levels contains adequate mineralocorticoid
activity, udrocortisone is not indicated in the acute period and has been associated with the side eects of pulmonary edema caused by salt and water
retention [30]. A transfer to routine oral therapy is warranted when acute
symptoms have settled or when the patient is tolerating oral uids.
During labor and the postpartum period
Routine glucocorticoid and mineralocorticoid replacement therapy can
be continued until the onset of labor, provided that the patient has no
11
Hypertension in pregnancy
Normal gestation is associated with a fall in blood pressure toward the
end of the rst trimester, reaching a nadir between 22 and 24 weeks. Hypertension during pregnancy, dened by an absolute systolic or diastolic blood
pressure exceeding 140 or 90 mmHg, respectively, complicates approximately
15% of pregnancies [61]. Increased vascular distensibility and reduced
peripheral vascular resistance occurring in pregnancy are accompanied
by physiologic changes, including sodium retention, increased extracellular
uid, and up-regulation of the RAS [62].
Hyperaldosteronism
The prevalence of primary hyperaldosteronism (PA) in nonpregnant hypertensive subjects varies between 1% and 12%, depending on the patient
population, diagnostic methods used, and conrmation by surgery [63]. In
contrast, PA is rare in pregnancy, with approximately 31 cases reported
worldwide since the original description by Crane in 1964 [64,65]. The majority of reported cases arise as a result of adrenal adenoma or hyperplasia.
However, there are rare reports of glucocorticoid-remediable hyperaldosteronism in pregnancy [66].
Plasma renin activity increases early in the rst trimester of normal pregnancy, reaching values almost three- to seven-fold greater than the normal
12
13
Pheochromocytoma in pregnancy
Pheochromocytoma accounts for approximately 0.1% cases of hypertension in the general population. There have been at least 200 cases diagnosed
in pregnancy, and the estimated prevalence at term is approximately 1 in
54,000 [83,84]. Pheochromocytoma is associated with sustained or paroxysmal episodes of hypertension, pallor, headaches, and palpitations [84]. Other
presentations include chest pain, dyspnea, abdominal pain, seizure, or even
sudden death [85]. Although antenatal diagnosis is associated with improved
outcomes, pheochromocytoma can be missed because of unexpectedly normal blood pressure during gestation [83,85]. A recent case illustrated the
14
course of a women who was managed emergently after being diagnosed during labor [86]. However, a delayed diagnosis has been associated with maternal death caused by cerebral edema associated with cardiogenic shock [85].
Untreated pheochromocytoma is associated with increased fetal and maternal morbidity and mortality. The maternal mortality rate was 48% before
1969 and 26% in the 1970s, and subsequently it fell to 17% [8789]. Since
1990, approximately 85% of cases have been diagnosed antenatally, perhaps
because of increased awareness [84]. In one series, antenatal diagnosis reduced the maternal mortality and the fetal loss rate to less than 1% and
15%, respectively [88]. A hypertensive crisis may be precipitated by abdominal palpitation [84], drugs, including metoclopramide [87], or labor. Pheochromocytoma should be actively sought for in patients who have aected
family members with von Hippel-Lindau or multiple endocrine neoplasia
2 syndromes [90]. The lack of proteinuria in pheochromocytoma may dierentiate pheochromocytoma from hypertension associated with preeclampsia. Other potential dierential diagnoses include anxiety, cocaine use,
pulmonary embolism, and alcohol withdrawal [84].
Catecholamine production generally remains stable during gestation [91].
Screening for pheochromocytoma was traditionally undertaken by assessing
elevated 24-hour urinary epinephrine and norepinephrine excretions. Because tumor secretion may be episodic, these methods have been largely superseded by more sensitive and specic techniques. Fractionated urinary
metanephrines and plasma metanephrines are more sensitive for the diagnosis of pheochromocytoma in nonpregnant patients, but their value in pregnancy has yet to be evaluated fully [92]. Dynamic testing with clonidine has
not been validated in pregnancy [93]. Adrenal imaging is performed when
the diagnosis is conrmed biochemically using existing nonpregnant reference ranges. MRI (without gadolinium) has better specicity than ultrasonography and is preferred to CT to minimize exposure to ionizing
radiation. Pheochromocytoma is typically bright on T2-weighted MRI,
with a sensitivity of 93% to 100% [94]. Metaiodobenzylguanidine-, uorine-18 ([18F])-dopa, and [18F]-dopaminelabeled positron emission tomography scanning methods are highly specic, functional imaging modalities
that may be considered for localization in the postpartum period and
have additional utility for assessing extra-adrenal disease, which occurs in
approximately 10% of cases [94].
Adrenalectomy is the preferred denitive treatment of pheochromocytoma following adequate a- and b-blockade for at least 2 weeks before
surgery. Primary medical therapy is indicated for cases diagnosed after 24
weeks gestation because of the attendant diculties and risks of surgery performed at that time. a-Blockade with phenoxybenzamine, 10 mg to 20 mg
twice daily, is begun initially, with titration to a dose of approximately 1
mg/kg/d until hypertension is controlled. After several days of a-blockade,
b-blockers are added to minimize reex tachycardia, despite their possible
association with intrauterine growth retardation [88]. When used as
15
monotherapy, unopposed b-blockade leads to vasoconstriction and potential hypertensive crisis and should be avoided. Metyrosine is a specic inhibitor of cathecholamine synthesis through its eect on tyrosine hydroxylase.
Although it is an FDA category C agent, metyrosine was previously used in
one patient with malignant pheochromocytoma without complication [95].
There are insucient data to recommend its routine use in pregnancy, but
it may be considered for short-term emergency use in cases late in the third
trimester with refractory hypertension or arrhythmia [95]. Phentolamine,
1 mg to 5 mg, is the agent of choice for the treatment of a hypertensive crisis
[86].
The optimal timing of adrenalectomy is late in the rst or early in the second trimester. In the third trimester, a combined caesarian section followed
by adrenalectomy for cases managed conservatively with medical therapy
may be considered. Vaginal delivery is not recommended because of the potential for exacerbating a hypertensive crisis. A close liaison with the anesthesiologist is imperative before adrenalectomy, C-section, or labor.
Pregnancy and congenital adrenal hyperplasia
The prenatal diagnosis and treatment of 21-hydroxylase (CYP21 gene)
deciency are options for couples who have a previously aected infant
and may be requested by an index case before attempting pregnancy. Genetic counseling is guided by molecular investigation of the family, ideally
by CYP21 genotyping of the aected individuals and an evaluation of the
parents to exclude a new mutation in the child. Biochemical testing of the
partner of an aected individual can identify a potential heterozygote, in
which case genotyping should be performed [96].
Couples at risk for an aected infant may choose to undergo prenatal
treatment with dexamethasone coupled with prenatal diagnosis. The goal
of the treatment is to inhibit fetal ACTH and so prevent hyperandrogenism
and virilization of an aected female fetus. Thus, dexamethasone, at a divided daily dose of 20 mg/kg of prepregnancy weight, is started when pregnancy is diagnosed. Dexamethasone is the agent of choice because it is not a
ready substrate for 11-b-hydroxysteroid dehydrogenase type 2. Chorionic villous sampling is performed at 9 to 11 weeks for the evaluation of the fetus
sex and CYP21 gene. Dexamethasone is discontinued if the fetus is male or
an unaected female. As a result of this strategy, treatment would continue
in only one of eight pregnancies. The initiation of therapy before 9 weeks
has resulted in normal genitalia in 11 of 25 aected females and only mild
virilization in another 11, but later treatment or the use of a lower dose of
dexamethasone has resulted in virilization in 23 of 24 aected infants [97].
There are relatively few data on the long-term risks of prenatal dexamethasone treatment for aected or unaected fetuses. One recent study showed
that 174 children (including 48 who had CAH) who received dexamethasone
did not dier in cognitive or motor development from 313 unexposed
16
17
[11] Evans JJ, Sin IL, Du GB, et al. Estrogen-induced transcortin increase and progesterone and
cortisol interactions: implications from pregnancy studies. Ann Clin Lab Sci 1987;17(2):
1015.
[12] Putignano P, Toja P, Dubini A, Giraldi FP, et al. Midnight salivary cortisol versus urinary
free and midnight serum cortisol as screening tests for Cushings syndrome. J Clin Endocrinol Metab 2003;88(9):41537.
[13] Gafni RI, Papanicolaou DA, Nieman LK. Nighttime salivary cortisol measurement as a simple, noninvasive, outpatient screening test for Cushings syndrome in children and adolescents. J Pediatr 2000;137(1):305.
[14] Ra H, Findling JW. A new immunoradiometric assay for corticotropin evaluated in normal
subjects and patients with Cushings syndrome. Clin Chem 1989;35(4):596600.
[15] Arnaldi G, Angeli A, Atkinson AB, et al. Diagnosis and complications of Cushings syndrome: a consensus statement. J Clin Endocrinol Metab 2003;88(12):5593602.
[16] Magiakou MA, Mastorakos G, Rabin D, et al. The maternal hypothalamic-pituitaryadrenal axis in the third trimester of human pregnancy. Clin Endocrinol (Oxf) 1996;44(4):
41928.
[17] Bowerman RA, Silver TM, Jae MH, et al. Sonography of adrenal pheochromocytomas.
AJR Am J Roentgenol 1981;137(6):122731.
[18] Berwaerts J, Verhelst J, Mahler C, Abs R. Cushings syndrome in pregnancy treated by
ketoconazole: case report and review of the literature. Gynecol Endocrinol 1999;13(3):
17582.
[19] Hana V, Dokoupilova M, Marek J, et al. Recurrent ACTH-independent Cushings syndrome in multiple pregnancies and its treatment with metyrapone. Clin Endocrinol (Oxf)
2001;54(2):27781.
[20] Connell JM, Cordiner J, Davies DL, et al. Pregnancy complicated by Cushings syndrome:
potential hazard of metyrapone therapy: case report. Br J Obstet Gynaecol 1985;92(11):
11925.
[21] Close CF, Mann MC, Watts JF, et al. ACTH-independent Cushings syndrome in pregnancy
with spontaneous resolution after delivery: control of the hypercortisolism with metyrapone.
Clin Endocrinol (Oxf) 1993;39(3):3759.
[22] Gormley MJ, Hadden DR, Kennedy TL, et al. Cushings syndrome in pregnancytreatment
with metyrapone. Clin Endocrinol (Oxf) 1982;16(3):28393.
[23] Khir AS, How J, Bewsher PD. Successful pregnancy after cyproheptadine treatment for
Cushings disease. Eur J Obstet Gynecol Reprod Biol 1982;13(6):3437.
[24] Hanson TJ, Ballono LB, Northcutt RC. Amino-glutethimide and pregnancy [letter].
JAMA 1974;230(7):9634.
[25] Leiba S, Weinstein R, Shindel B, et al. The protracted eect of o, p-DDD in Cushings disease and its impact on adrenal morphogenesis of young human embryo. Ann Endocrinol
(Paris) 1989;50(1):4953.
[26] Albert E, Dalaker K, Jorde R, Berge LN. Addisons disease and pregnancy. Acta Obstet Gynecol Scand 1989;68(2):1857.
[27] Stechova K, Bartaskova D, Mrstinova M, et al. Pregnancy in a woman suering from type 1
diabetes associated with Addisons disease and Hashimotos thyroiditis (fully developed autoimmune polyglandular syndrome type 2). Exp Clin Endocrinol Diabetes 2004;112(6):
3337.
[28] Cohen M. Addisons disease complicated by toxemia of pregnancy. Arch Intern Med 1948;
81:87987.
[29] Drucker D, Shumak S, Angel A. Schmidts syndrome presenting with intrauterine growth
retardation and postpartum addisonian crisis. Am J Obstet Gynecol 1984;149(2):22930.
[30] Seaward PG, Guidozzi F, Sonnendecker EW. Addisonian crisis in pregnancy: case report.
Br J Obstet Gynaecol 1989;96(11):134850.
[31] Osler M, Pedersen J. Pregnancy in a patient with Addisons disease and diabetes mellitus.
Acta Endocrinol (Copenh) 1962;41:7987.
18
[32] OShaughnessy RW, Hackett KJ. Maternal Addisons disease and fetal growth retardation:
a case report. J Reprod Med 1984;29(10):7526.
[33] Gradden C, Lawrence D, Doyle PM, et al. Uses of error: Addisons disease in pregnancy.
Lancet 2001;357(9263):1197.
[34] Ambrosi B, Barbetta L, Morricone L. Diagnosis and management of Addisons disease during pregnancy. J Endocrinol Invest 2003;26(7):698702.
[35] Allemang WH. Pregnancy in the absence of adrenal cortical function. Can Med Assoc J
1961;85:11822.
[36] Donnelly JC, OConnell MP, Keane DP. Addisons disease, with successful pregnancy outcome. J Obstet Gynaecol 2003;23(2):199.
[37] Grottolo A, Ferrari V, Mariano M, Zambruni A, et al. Primary adrenal insuciency, circulating lupus anticoagulant and anticardiolipin antibodies in a patient with multiple abortions
and recurrent thrombotic episodes. Haematologica 1988;73(6):5179.
[38] Vengrove MA, Amoroso A. Reversible adrenal insuciency after adrenal hemorrhage.
Ann Intern Med 1993;119(5):439.
[39] Guibal F, Rybojad M, Cordoliani F, et al. Melanoderma revealing primary antiphospholipid syndrome. Dermatology 1996;192(1):757.
[40] George LD, Selvaraju R, Reddy K, Stout TV, et al. Vomiting and hyponatraemia in pregnancy. BJOG 2000;107(6):8089.
[41] Schulte HM, Weisner D, Allolio B. The corticotrophin releasing hormone test in late pregnancy: lack of adrenocorticotrophin and cortisol response. Clin Endocrinol (Oxf) 1990;
33(1):99106.
[42] McKenna DS, Wittber GM, Nagaraja HN, et al. The eects of repeat doses of antenatal corticosteroids on maternal adrenal function. Am J Obstet Gynecol 2000;183(3):66973.
[43] Grinspoon SK, Biller BM. Clinical review 62: laboratory assessment of adrenal insuciency.
J Clin Endocrinol Metab 1994;79(4):92331.
[44] Nieman LK. Dynamic evaluation of adrenal hypofunction. J Endocrinol Invest 2003;
26(Suppl 7):S7482.
[45] May ME, Carey RM. Rapid adrenocorticotropic hormone test in practice: retrospective review. Am J Med 1985;79(6):67984.
[46] Speckart PF, Nicolo JT, Bethune JE. Screening for adrenocortical insuciency with cosyntropin (synthetic ACTH). Arch Intern Med 1971;128(5):7613.
[47] Dickstein G. Hypothalamo-pituitary-adrenal axis testing: nothing is sacred and caution in
interpretation is needed. Clin Endocrinol (Oxf) 2001;54(1):156.
[48] Dorin RI, Qualls CR, Crapo LM. Diagnosis of adrenal insuciency. Ann Intern Med 2003;
139(3):194204.
[49] Courtney CH, McAllister AS, McCance DR, et al. Comparison of one week 0900 h serum
cortisol, low and standard dose synacthen tests with a 4 to 6 week insulin hypoglycaemia
test after pituitary surgery in assessing HPA axis. Clin Endocrinol (Oxf) 2000;53(4):4316.
[50] Nolten WE, Lindheimer MD, Oparil S, et al. Desoxycorticosterone in normal pregnancy: I.
sequential studies of the secretory patterns of desoxycorticosterone, aldosterone, and cortisol. Am J Obstet Gynecol 1978;132(4):41420.
[51] Nigam R, Bhatia E, Miao D, et al. Prevalence of adrenal antibodies in Addisons disease
among north Indian Caucasians. Clin Endocrinol (Oxf) 2003;59(5):5938.
[52] Symonds EM, Craven DJ. Plasma renin and aldosterone in pregnancy complicated by adrenal insuciency. Br J Obstet Gynaecol 1977;84(3):1916.
[53] Ilbery M, Jones AR, Sampson J. Lupus anticoagulant and HELLP syndrome complicated
by placental abruption, hepatic, dermal and adrenal infarction. Aust N Z J Obstet Gynaecol
1995;35(2):2157.
[54] Kelestimur F. The endocrinology of adrenal tuberculosis: the eects of tuberculosis on the
hypothalamo-pituitary-adrenal axis and adrenocortical function. J Endocrinol Invest
2004;27(4):3806.
19
[55] Levine D, Edelman RR. Fast MRI and its application in obstetrics. Abdom Imaging 1997;
22(6):58996.
[56] Sidhu RK, Hawkins DF. Prescribing in pregnancy: corticosteroids. Clin Obstet Gynaecol
1981;8(2):383404.
[57] van der Spuy ZM, Jacobs HS. Management of endocrine disorders in pregnancy: part II. pituitary, ovarian and adrenal disease. Postgrad Med J 1984;60(703):31220.
[58] Esteban NV, Loughlin T, Yergey AL, et al. Daily cortisol production rate in man determined
by stable isotope dilution/mass spectrometry. J Clin Endocrinol Metab 1991;72(1):3945.
[59] Malcho CD, Carey RM. Adrenal insuciency. Curr Ther Endocrinol Metab 1997;6:1427.
[60] Normington EA, Davies D. Hypertension and oedema complicating pregnancy in Addisons
disease. BMJ 1972;2(806):1489.
[61] James PR, Nelson-Piercy C. Management of hypertension before, during, and after pregnancy. Heart 2004;90(12):1499504.
[62] Gabbe. Obstetrics: normal and problem pregnancies. 4th edition. New York: Churchhill and
Livingston; 2002.
[63] Mulatero P, Stowasser M, Loh KC, et al. Increased diagnosis of primary aldosteronism, including surgically correctable forms, in centers from ve continents. J Clin Endocrinol
Metab 2004;89(3):104550.
[64] Okawa T, Asano K, Hashimoto T, et al. Diagnosis and management of primary aldosteronism in pregnancy: case report and review of the literature. Am J Perinatol 2002;19(1):316.
[65] Crane MG, Andes JP, Harris JJ, et al. Primary aldosteronism in pregnancy. Obstet Gynecol
1964;23:2008.
[66] Wycko JA, Seely EW, Hurwitz S, et al. Glucocorticoid-remediable aldosteronism and pregnancy. Hypertension 2000;35(2):66872.
[67] Brown MA, Wang J, Whitworth JA. The renin-angiotensin-aldosterone system in preeclampsia. Clin Exp Hypertens 1997;19(56):71326.
[68] Wilson M, Morganti AA, Zervoudakis I, et al. Blood pressure, the renin-aldosterone system
and sex steroids throughout normal pregnancy. Am J Med 1980;68(1):97104.
[69] Nielsen AH, Schauser KH, Poulsen K. Current topic: the uteroplacental renin-angiotensin
system. Placenta 2000;21(56):46877.
[70] Dorr HG, Heller A, Versmold HT, et al. Longitudinal study of progestins, mineralocorticoids, and glucocorticoids throughout human pregnancy. J Clin Endocrinol Metab 1989;
68(5):8638.
[71] Lindheimer MD, Katz AI, Nolten WE, et al. Sodium and mineralocorticoids in normal and
abnormal pregnancy. Adv Nephrol Necker Hosp 1977;7:3359.
[72] Boonshaft B, OConnell JM, Hayes JM, et al. Serum renin activity during normal pregnancy:
eect of alterations of posture and sodium intake. J Clin Endocrinol Metab 1968;28(11):
16414.
[73] Matsumoto J, Miyake H, Isozaki T, et al. Primary aldosteronism in pregnancy. J Nippon
Med Sch 2000;67(4):2759.
[74] Fujiyama S, Mori Y, Matsubara H, et al. Primary aldosteronism with aldosterone-producing adrenal adenoma in a pregnant woman. Intern Med 1999;38(1):369.
[75] Neerhof MG, Shlossman PA, Poll DS, et al. Idiopathic aldosteronism in pregnancy. Obstet
Gynecol 1991;78(3 Pt 2):48991.
[76] Solomon CG, Thiet M, Moore F Jr, et al. Primary hyperaldosteronism in pregnancy: a case
report. J Reprod Med 1996;41(4):2558.
[77] Gordon RD, Fishman LM, Liddle GW. Plasma renin activity and aldosterone secretion in
a pregnant woman with primary aldosteronism. J Clin Endocrinol Metab 1967;27(3):3858.
[78] Aboud E, De Swiet M, Gordon H. Primary aldosteronism in pregnancy: should it be treated
surgically? Ir J Med Sci 1995;164(4):27980.
[79] Baron F, Sprauve ME, Huddleston JF, et al. Diagnosis and surgical treatment of primary
aldosteronism in pregnancy: a case report. Obstet Gynecol 1995;86(4 Pt 2):6445.
20
[80] Webb JC, Bayliss P. Pregnancy complicated by primary aldosteronism. South Med J 1997;
90(2):2435.
[81] Hecker A, Hasan SH, Neumann F. Disturbances in sexual dierentiation of rat foetuses following spironolactone treatment. Acta Endocrinologica (Copenh) 1980;95(4):5405.
[82] Deruelle P, Dufour P, Magnenant E, et al. Maternal Bartters syndrome in pregnancy treated
by amiloride. Eur J Obstet Gynecol Reprod Biol 2004;115(1):1067.
[83] Botchan A, Hauser R, Kupfermine M, et al. Pheochromocytoma in pregnancy: case report
and review of the literature. Obstet Gynecol Surv 1995;50(4):3217.
[84] Lyman DJ. Paroxysmal hypertension, pheochromocytoma, and pregnancy. J Am Board
Fam Pract 2002;15(2):1538.
[85] Cermakova A, Knibb AA, Hoskins C, et al. Post partum phaeochromocytoma. Int J Obstet
Anesth 2003;12(4):3004.
[86] Strachan AN, Claydon P, Caunt JA. Phaeochromocytoma diagnosed during labour. Br J
Anaesth 2000;85(4):6357.
[87] Takai Y, Seki H, Kinoshita K. Pheochromocytoma in pregnancy manifesting hypertensive
crisis induced by metoclopramide. Int J Gynaecol Obstet 1997;59(2):1337.
[88] Harper MA, Murnaghan GA, Kennedy L, et al. Phaeochromocytoma in pregnancy: ve
cases and a review of the literature. Br J Obstet Gynaecol 1989;96(5):594606.
[89] Hadden DR. Adrenal disorders of pregnancy. Endocrinol Metab Clin North Am 1995;24(1):
13951.
[90] Ahn JT, Hibbard JU, Chapa JB. Atypical presentation of pheochromocytoma as part of
multiple endocrine neoplasia IIa in pregnancy. Obstet Gynecol 2003;102(5 Pt 2):12025.
[91] Peleg D, Munsick RA, Diker D, et al. Distribution of catecholamines between fetal and maternal compartments during human pregnancy with emphasis on L-dopa and dopamine.
J Clin Endocrinol Metab 1986;62(5):9114.
[92] Lenders JW, Pacak K, Eisenhofer G. New advances in the biochemical diagnosis of pheochromocytoma: moving beyond catecholamines. Ann N Y Acad Sci 2002;970:2940.
[93] Del Giudice A, Bisceglia M, DErrico M, et al. Extra-adrenal functional paraganglioma
(phaeochromocytoma) associated with renal-artery stenosis in a pregnant woman. Nephrol
Dial Transplant 1998;13(11):29203.
[94] Ilias I, Pacak K. Current approaches and recommended algorithm for the diagnostic localization of pheochromocytoma. J Clin Endocrinol Metab 2004;89(2):47991.
[95] Devoe LD, ODell BE, Castillo RA, et al. Metastatic pheochromocytoma in pregnancy and
fetal biophysical assessment after maternal administration of alpha-adrenergic, beta-adrenergic, and dopamine antagonists. Obstet Gynecol 1986;68(Suppl 3):S158.
[96] Forest MG. Recent advances in the diagnosis and management of congenital adrenal hyperplasia due to 21-hydroxylase deciency. Hum Reprod Update 2004;10(6):46985.
[97] New MI, Carlson A, Obeid J, et al. Prenatal diagnosis for congenital adrenal hyperplasia in
532 pregnancies. J Clin Endocrinol Metab 2001;86(12):56517.
[98] Meyer-Bahlburg HF, Dolezal C, Baker SW, et al. Cognitive and motor development of children with and without congenital adrenal hyperplasia after early-prenatal dexamethasone.
J Clin Endocrinol Metab 2004;89(2):6104.
[99] Clayton PE, Miller WL, Obereld SE, et al. Consensus statement on 21-hydroxylase deciency from the European Society for Paediatric Endocrinology and the Lawson Wilkins Pediatric Endocrine Society. Horm Res 2002;58(4):18895.
* Corresponding authors. Basic Medical Sciences, Health Sciences Centre, 300 Prince
Philip Drive, St. Johns, NL, A1B 3V6 Canada (C.S. Kovacs); Calcium Metabolism and
Osteoporosis Program, American University of BeirutMedical Center, P.O. Box 11-0236,
Riad E1 Solh 4407 2020, Beirut, Lebanon (G.E.-H. Fuleihan).
E-mail addresses: ckovacs@mun.ca (C.S. Kovacs); gf01@aub.edu.lb (G.E.-H. Fuleihan).
0889-8529/06/$ - see front matter 2005 Elsevier Inc. All rights reserved.
doi:10.1016/j.ecl.2005.09.004
endo.theclinics.com
22
Fig. 1. Calcium homeostasis in human pregnancy and lactation compared with normal. The
thickness of arrows indicates a relative increase or decrease with respect to the normal and nonpregnant state. (Adapted from Kovacs CS, Kronenberg HM. Maternal-fetal calcium and bone
metabolism during pregnancy, puerperium and lactation. Endocr Rev 1997;18:83272. 1997
The Endocrine Society; with permission.)
human data are unavailable. The reader also is referred to several other
comprehensive reviews on the subject [13].
23
Fig. 2. Longitudinal changes in calcium, phosphorus, and calcitropic hormone levels that occur
during pregnancy and lactation. Normal adult ranges are indicated by the shaded areas. The
progression in PTHrP levels is depicted by a dashed line to reect that the data are less complete; the implied comparisons of PTHrP levels in late pregnancy and lactation are uncertain
extrapolations because no reports followed patients serially. In both situations, PTHrP levels
are elevated. (Adapted from Kovacs CS, Kronenberg HM. Maternal-fetal calcium and bone metabolism during pregnancy, puerperium and lactation. Endocr Rev 1997;18:83272. 1997 The
Endocrine Society; with permission.)
24
not be mistaken for evidence of physiologic hyperparathyroidism of pregnancy, an erroneous concept that has persisted in some modern texts [4,5].
The decline in total serum calcium is an unimportant artifact of a nonphysiologic measurement; the ionized calcium is the relevant measurement and always should be assayed if there is any doubt about the true value of the
serum calcium during pregnancy (or at any time). Serum phosphorus levels
also are normal during pregnancy.
As observed by longitudinal measurements during pregnancy with modern two-site intact immunoradiometric assays (IRMA), serum parathyroid hormone (PTH) decreases to the low-normal range (ie, 1030% of the
mean nonpregnant value) during the rst trimester, then increases steadily
to the mid-normal range by term [610]. As judged by the intact serum
PTH level, the parathyroids are modestly suppressed beginning early in the
rst trimester and return to apparently normal function by the end of pregnancy. First-generation PTH assays in the 1970s and 1980s were insensitive
and measured multiple, biologically inactive fragments of PTH; a few studies
with these assays had detected higher levels of PTH during pregnancy in humans. Those early studies of PTH in pregnancy, combined with the observation that total serum calcium decreases during pregnancy, reinforced the
erroneous concept that secondary hyperparathyroidism occurs during pregnancy. Modern intact assays have made it clear that in well-nourished
women, ionized calcium is normal throughout pregnancy, and that PTH is
suppressed during early pregnancy. Bio-intact PTH assays have been developed that detect true full-length PTH [11]; the levels are likely similar to
levels obtained with the more widely used intact PTH assays, but no study
has examined this. In contrast to the normal suppression of PTH during
pregnancy, there is evidence that PTH may increase above normal in late
pregnancy in women from Malay, who have very low intakes of calcium [12].
Total 1,25(OH)2D3 levels double early in pregnancy and maintain this
increase until term; free 1,25(OH)2D3 levels are increased from the third trimester and possibly earlier. The increase in 1,25(OH)2D3 may be largely independent of changes in PTH because PTH levels typically are decreasing at
the time of the increase in 1,25(OH)2D3. The maternal kidneys likely account
for most, if not all, of the increase in 1,25(OH)2D3 during pregnancy, although the decidua, placenta, and fetal kidneys may contribute a small
amount. The relative contribution of the maternal kidneys is based on several
lines of evidence [1], including the report of an anephric woman on hemodialysis who had low 1,25(OH)2D3 levels before and during a pregnancy [13].
The renal 1a-hydroxylase may be upregulated in response to factors such
as PTH-related protein (PTHrP), estradiol, prolactin, and placental lactogen
(evidence from animal studies is reviewed by Kovacs and Kronenberg [1]).
Serum calcitonin levels also increase during pregnancy, with the C cells of
the thyroid, breast, and placenta possibly contributing to the circulating level
of calcitonin. It has been postulated that calcitonin protects the maternal skeleton from excessive resorption of calcium, but this hypothesis is unproved.
25
26
27
transfer occurring at this stage of pregnancy compared with the peak rate
of calcium transfer in the third trimester. One might have anticipated that
markers of bone turnover would increase particularly in the third trimester;
however, no further increase was seen at that time.
Changes in skeletal calcium content have been assessed in humans through
the use of sequential bone density studies during pregnancy. Because of concerns about fetal radiation exposure, few such studies have been done. Such
studies are confounded by changes in body composition and weight during
normal pregnancy, which can lead to artifactual changes in bone density. Using single-photon or dual-photon absorptiometry (SPA/DPA), several prospective studies did not nd a signicant change in cortical or trabecular
bone density during pregnancy [1]. Several more recent studies have used
DXA before conception (range 18 months prior, but not always stated)
and after delivery (range 16 weeks postpartum) [2127]. Most studies involved 16 or fewer subjects. One study found no change in lumbar spine
bone density measurements obtained preconception and within 1 to 2 weeks
postdelivery [23], whereas the other studies reported decreases of 4% to 5% in
lumbar spine bone density with the postpartum measurement taken 1 to 6
weeks postdelivery. The puerperium is associated with bone density losses
of 1% to 3% per month in women who lactate (see lactation section), and
it is important that the postpartum measurement be done as soon as possible
after delivery. Other longitudinal studies have found a progressive decrease
during pregnancy in indices thought to correlate with bone mineral density,
as determined by ultrasound measurements at a peripheral site, the os calcis
[28]. Although the longitudinal studies with SPA/DPA suggested no change
in trabecular or cortical bone density during pregnancy, the subsequent evidence from preconception and postdelivery DXA measurements and peripheral ultrasound measurements suggests that there may be a small net loss of
maternal bone mineral content during normal human pregnancy. None of all
the aforementioned studies could address the question as to whether skeletal
calcium content increases early in pregnancy in advance of the third trimester,
as has been observed in normal mice. Further studies, with larger numbers of
patients, are needed to clarify the extent of bone loss during pregnancy.
It seems certain that any acute changes in bone metabolism during pregnancy do not normally cause long-term changes in skeletal calcium content
or strength. Numerous studies of osteoporotic or osteopenic women have
failed to nd a signicant association of parity with bone density or fracture
risk [1,29]; however, a few studies of women with extremely low calcium or
vitamin D intake found that pregnancy may compromise skeletal strength
and density (see later). Although most clinical studies could not separate
out the eects of parity from the eects of lactation, it may be reasonable
to conclude that if parity has any eect on bone density or fracture risk,
it normally must be only a modest eect. A more recent study of twins indicated that there may be a small protective eect of parity and lactation on
maintaining bone mineral content [30].
28
29
Fig. 3. The calcium receptor (represented schematically) is expressed in mammary tissue during
lactation (but not pregnancy), wherein it has several key functions as elucidated from studies in
mice [33]. The calcium receptor monitors the systemic concentration of calcium to control
PTHrP synthesis and the supply of calcium to the breast. An increase in systemic calcium or
the administration of a calcimimetic inhibited PTHrP, whereas a decrease in systemic calcium
stimulated PTHrP. The calcium receptor also directly regulates the calcium and uid composition of milk. The administration of calcium or a calcimimetic stimulated the transport of calcium into the breast, and the administration of a calcimimetic enhanced the entry of water
into the milk, making it less viscous. (From Kovacs CS. Calcium and bone metabolism during
pregnancy and lactation. J Mammary Gland Biol Neoplasia 2005;10(2):10518. 2005 Springer
Science and Business Media BV; with permission.)
tissue and stimulates resorption of calcium from the maternal skeleton, renal
tubular reabsorption of calcium, and (indirectly) suppression of PTH
(Fig. 4). In a sense, the breast becomes an accessory parathyroid gland during lactation, but the hyperparathyroidism of lactation is increased secretion of PTHrP, not PTH. The strongest evidence in support of this model
comes from the study of mice in which the PTHrP gene was ablated at
the onset of lactation, but only within mammary tissue [32]. The lactational
decrease in bone mineral content was signicantly blunted in the absence of
mammary gland production of PTHrP. Other evidence for the central role
of PTHrP in lactation comes from humans, in that PTHrP levels correlate
negatively with PTH levels and positively with the ionized calcium levels
of lactating women [31,34], and that higher PTHrP levels correlate with
greater losses of bone mineral density during lactation in humans [35]. Observations in aparathyroid women provide evidence of the impact of PTHrP
in calcium homeostasis during lactation (see later).
Calcitonin levels are elevated in the rst 6 weeks of lactation. Studies in
mice lacking the gene that encodes calcitonin indicate that calcitonin may
modulate the rate of skeletal resorption during lactation. Calcitonin-null
mice lost more than 50% of skeletal mineral content during 3 weeks of
30
Fig. 4. The breast is a central regulator of skeletal demineralization during lactation. Suckling
induces release of prolactin. Suckling and prolactin inhibit the hypothalamic gonadotropin-releasing hormone (GnRH) pulse center, which suppresses the gonadotropins (luteinizing hormone [LH], follicle-stimulating hormone [FSH]), leading to low levels of the ovarian sex
steroids (estradiol and progesterone). PTHrP production and release from the breast is controlled by several factors, including suckling, prolactin, and the calcium receptor. PTHrP enters
the bloodstream and combines with systemically low estradiol levels to upregulate bone resorption markedly. Increased bone resorption releases calcium and phosphate into the bloodstream,
which reach the breast ducts and are actively pumped into the breast milk. PTHrP also passes
into the milk at high concentrations, but whether PTHrP plays a role in regulating calcium
physiology of the neonate is unknown. Calcitonin (CT) may inhibit skeletal responsiveness to
PTHrP and low estradiol. (From Kovacs CS. Calcium and bone metabolism during pregnancy
and lactation. J Mammary Gland Biol Neoplasia 2005;10(2):10518. 2005 Springer Science and
Business Media BV; with permission.)
31
32
and bone metabolism that occur in reproductive-age women who have estrogen deciency induced by gonadotropin-releasing hormone agonist therapy for endometriosis and other conditions. Six months of acute estrogen
deciency induced by gonadotropin-releasing hormone agonist therapy
leads to 1% to 4% losses in trabecular (but not cortical) bone density, increased urinary calcium excretion, and suppression of 1,25(OH)2D3 and
PTH levels [1]. During lactation, women are not as estrogen decient, but
they lose more bone mineral density (at trabecular and cortical sites),
have normal (as opposed to low) 1,25(OH)2D3 levels, and have reduced
(as opposed to increased) urinary calcium excretion. The dierence between
isolated estrogen deciency and lactation may be due to the eects of other
factors (eg, PTHrP) that add to the eects of estrogen withdrawal in lactation (Fig. 5). The relative inuences of estrogen deciency and PTHrP have
been partially discerned in normal mice, in which it has been shown that
treatment with pharmacologic doses of estrogen blunted, but did not abolish, the normal demineralization that occurs during lactation [43].
The bone density losses of lactation are substantially reversed during weaning at a rate of 0.5% to 2% per month [1,29,40]. The mechanism for this restoration of bone density is uncertain and largely unexplored, but preliminary
evidence from animal models suggests that PTH, calcitriol, calcitonin, and estrogen may not be required to achieve that restoration. In the long-term, the
consequences of lactation-induced depletion of bone mineral seem clinically
unimportant in most women. Most epidemiologic studies of premenopausal
Fig. 5. Acute estrogen deciency (eg, gonadotropin-releasing hormone analogue therapy) increases skeletal resorption and raises the blood calcium; PTH is suppressed, and renal calcium
losses are increased. During lactation, the combined eects of PTHrP (secreted by the breast)
and estrogen deciency increase skeletal resorption, reduce renal calcium losses, and raise the
blood calcium, but calcium is directed into the breast milk. (From Kovacs CS, Kronenberg
HM. Maternal-fetal calcium and bone metabolism during pregnancy, puerperium and lactation.
Endocr Rev 1997;18:83272. 1997 The Endocrine Society; with permission.)
33
34
35
36
receptor, and at the same time it treats premature labor associated with hypercalcemia [68,69]. Experience with any of the aforementioned pharmacologic therapies is limited to individual case reports [49]; consequently, no
medical therapy can be claimed to be better than any other. Medical therapy
should be coupled with maternal surveillance and the monitoring of serum
calcium and electrolytes and the initiation of antenatal testing with serial fetal
ultrasound starting at 28 weeks of gestation. Parathyroidectomy is recommended postpartum in cases that were followed medically during pregnancy.
Lactation is not contraindicated in women with untreated hyperparathyroidism, but worsening of hypercalcemia and accelerated skeletal losses may be
anticipated because of the combined eects of PTHrP and hyperparathyroidism to stimulate bone resorption.
Neonatal hypoparathyroidism secondary to maternal hyperparathyroidism is usually transient (see earlier) and is treated with calcium supplementation and calcitriol. These neonates also should be fed milk formulas high
in calcium and low in phosphate to minimize the risk of hypocalcemia. The
prevalence and severity of complications from hyperparathyroidism in
mothers and neonates have and will continue to decrease over time, owing
to increased surveillance, earlier intervention, and improved surgical and anesthetic technology [52,60,61].
Familial benign hypocalciuric hypercalcemia
Familial benign hypocalciuric hypercalcemia (FBHH) is an autosomal
dominant disorder that is caused by inactivating mutations in the calciumsensing receptor that cause hypercalcemia and hypocalciuria [70,71]. In
contrast to patients with hyperparathyroidism, patients with FBHH do
not experience bone demineralization or nephrolithiasis. FBHH has been
reported in pregnancy with no clinical sequelae in the mother [72]. As anticipated, maternal hypercalcemia has caused suppression of PTH synthesis in the fetus, however, and subsequent hypocalcemia and tetany in the
neonate [72,73]. The treatment of neonates is similar to that of children
born to mothers with hyperparathyroidism (see earlier).
The calcium-sensing receptor is expressed in the epithelial ducts of breast
tissue and has been shown to modulate the production of PTHrP and the
transport of calcium into milk in a mouse model [33]. Activating mutations
of this receptor in women with FBHH theoretically could enhance the degree of skeletal demineralization during lactation and the calcium content
of milk, but this has not been studied.
Hypoparathyroidism
Patients usually are known to have hypoparathyroidism or aparathyroidism before pregnancy, and the therapeutic dilemma revolves around adjustment of the treatment, which may vary widely. In 1966, OLeary et al [74]
reported two cases of hypoparathyroidism treated with high doses of
37
calcium and vitamin D wherein the mothers gave birth to healthy infants
after uncomplicated pregnancies. Despite physiologic increments in endogenous calcitriol levels during pregnancy, several studies since have documented increased requirements for exogenous calcium and calcitriol therapy
as pregnancy progressed in patients with hypoparathyroidism [7578]. Conversely, in numerous other case reports, women with hypoparathyroidism
have been reported to require less calcium and vitamin D supplementation
during pregnancy [1]. Potential explanations for requiring less supplementation during pregnancy include pregnancy-induced increments in calcitriol
from placental sources, the potential eect of PTHrP in the maternal circulation to stimulate the renal 1a-hydroxylase, and other pregnancy-related
factors (eg, prolactin or placental lactogen) that may stimulate the renal
1a-hydroxylase or enhance intestinal calcium absorption independently of
calcitriol. The last-mentioned has been reported exclusively in animal models [1]. In some case reports, it seems that the normal, artifactual decrease in
total serum calcium during pregnancy was the parameter that led to treatment with increased calcium and calcitriol supplementation. Although few
cases report measurements of ionized calcium, several do mention that the
increments in vitamin D were due to maternal symptoms of hypocalcemia
or tetany.
Consequently, there is no established therapeutic regimen for the treatment of hypoparathyroidism during pregnancy, but numerous principles exist that help to guide treatment decisions. Calcitriol levels normally increase
during pregnancy and contribute (at least in part) to the enhanced intestinal
calcium absorption of pregnancy; most women should receive an increase in
the dosage of calcitriol at least initially. The total serum calcium is less informative, and the ionized calcium should be monitored in these patients.
Undertreatment results in maternal hypocalcemia; increases the risk of premature labor and of neonatal secondary hyperparathyroidism; and may lead
to neonatal skeletal demineralization, subperiosteal bone resorption, and osteitis brosa cystica [79]. Conversely, overtreatment may lead to maternal
hypercalcemia and neonatal hypoparathyroidism and raises the potential
concerns of teratogenicity that has been shown using older vitamin D preparations [80,81]. The active forms of vitamin D, such as calcitriol and 1a-calcidiol, have the advantages of a shorter half-life and lower risk of toxicity. A
study reported the outcome of pregnancy in 10 women treated with calcitriol
at doses of 0.25 mg/d to 3.25 mg/d [75]. In 8 of 10 pregnancies, healthy infants
were delivered. In two cases, serious adverse events occurred, including premature closure of the frontal fontanelle and stillbirth, but the causative role
of calcitriol could not be established [75]. Details regarding nine additional
cases of hypoparathyroidism and vitamin Dresistant rickets were provided
in the same publication and conrmed the lack of toxicity or teratogenicity
from vitamin D supplementation during pregnancy [75].
In contrast to the conicting literature on the eects of pregnancy on
hypoparathyroidism, calcium and vitamin D or calcitriol requirements in
38
39
40
41
42
Neonatal complications
Administration of intravenous magnesium to mothers before delivery increased neonatal serum magnesium and decreased PTH levels, whereas the
eect on neonatal total and ionized calcium levels varied [97,99]. Studies
evaluating the impact of neonatal hypermagnesemia on neonatal outcomes
have yielded conicting results [97,104106]. Respiratory depression and hypotonia were reported in 16 cases of neonatal hypermagnesemia [106]. A follow-up study of 35 infants born to toxemic mothers treated with magnesium
sulfate for 2 to 4 days suggested that neonates whose mothers had received
prolonged administration may be more likely to manifest respiratory depression [105]. Cord blood and neonatal serum magnesium levels were of little
diagnostic value to the clinical picture except in cases of severe hypermagnesemia [105,107], conrming the general observation that circulating serum
magnesium levels do not reect intracellular and total body magnesium
stores. Conversely, a study of 118 infants born to mothers who had received
intramuscular magnesium in doses of 10 to 95 g concluded that the neonatal
death rate was lower than that of the total newborn population [104]. Respiratory depression, hypotonia, and need for intubation were not evaluated in
that report, however [104]. Neonatal bone abnormalities have been reported
with long-term use of magnesium sulfate. The rst report by Lamm et al
[108] described a congenital form of rickets manifested by defective ossication of bone and enamel in the teeth of the ospring of mothers who had
received magnesium sulfate during pregnancy. Several cases of abnormal
mineralization of metaphyses since have been reported in neonates born
to mothers who received prolonged intravenous magnesium and had high
serum magnesium levels [109111]. The proposed mechanism for defective
mineralization of bones involves the inhibition of calcication of osteoid
in which calcium-binding sites are occupied by magnesium [109,110].
Some of these conicting ndings regarding neonatal morbidity from maternal administration of magnesium sulfate may be explained by the route
and duration of magnesium sulfate administration, by the variability in
the ranges of cord magnesium levels reached, and by the gestational age
of the neonates. Postdelivery, there was a delay in normalization of the serum magnesium level for a few days resulting from the limitation of magnesium excretion by the newborns immature kidneys [97].
Management issues
There are no guidelines for the monitoring of pregnant women receiving
magnesium sulfate. Neonates born to mothers receiving long-term magnesium sulfate and experiencing severe hypermagnesemia (O 7 mg/dL) are
more likely to have hypotonia, respiratory depression, and bone abnormalities [97,105,107,111]. Subjects receiving such therapy for periods exceeding
1 or 2 days should be monitored carefully, with the measurement of maternal serum calcium and magnesium levels, coupled with monitoring the
fetal movement. Symptomatic neonates can be managed by maintaining
43
ventilation for 24 to 48 hours and providing intravenous uids for electrolyte balance for a few days, after which marked clinical improvement is usually noted [105]. Intravenous calcium to antagonize the central nervous
system depression and peripheral neuromuscular blockade has been used,
with careful monitoring of the heart rate [105].
Low calcium intake
There are limited data that low calcium intake in the mother may adversely
aect fetal mineral accretion and maternal bone mineral metabolism [112].
In women with low dietary calcium intake, there are diering results as to
whether or not calcium supplementation during pregnancy improved maternal or neonatal bone density [3]. There is short-term evidence that maternal
turnover was reduced when 1.2 g of calcium was given for 20 days to 31 Mexican women with a mean calcium intake of 1 g during weeks 25 to 30 of gestation [113]. In a double-blind study conducted in 256 pregnant women, 2 g of
calcium supplementation improved bone mineral content in infants of supplemented mothers who were in the lowest quintile of calcium intake [114].
During lactation, there is no rm evidence that low calcium intake leads
to impaired breast milk quality or accentuates maternal bone loss [115].
Even in women with very low calcium intakes, the same amount of mineral
was lost during lactation from the skeleton compared with women who had
supplemented calcium intakes, and the breast milk calcium content was
unaected by calcium intake or vitamin D status [116118]. Conversely,
because high calcium intakes do not aect the degree of skeletal demineralization that occurs during lactation [3841], it is unlikely that increasing calcium supplementation above normal would aect skeletal demineralization.
In general, the physiologic changes in calcium and bone metabolism that
usually occur during pregnancy and lactation are likely to be sucient for
fetal bone growth and breast milk production in women with reasonably
sucient calcium intake [115]. The inclusion of calcium supplementation
for pregnant women with low calcium intake could be defended, however,
and is strengthened further by the possible link between low calcium intake,
preeclampsia, and increased blood pressure in the ospring [112]. Increased
calcium intake also is recommended in adolescent mothers to meet the need
of reproduction and maternal bone growth [115]. There is evidence that the
skeleton of an undernourished adolescent recovers fully from lactational
losses, but there is some concern that peak bone mass might not be attained
subsequently [119].
Vitamin D deciency
In humans and in animal models, vitamin D deciency or the absence
of the vitamin D receptor can lead to adverse neonatal outcomes, including neonatal rickets, craniotabes, decreased wrist ossication centers, and
44
impaired tooth enamel formation [120]. These features generally are not
present at birth, but appear postnatally as intestinal calcium absorption becomes vitamin D dependent. Although vitamin D supplementation of pregnant mothers at risk for vitamin D deciency improved neonatal serum
calcium concentrations and resulted in a trend for greater height and length
in the ospring [112], a Cochrane review of 232 women in two trials reported conicting results [121]. Although there is no evidence to indicate
a benecial eect of vitamin D supplementation during pregnancy above
the amounts needed to prevent vitamin D deciency, optimal levels for vitamin D supplementation are unclear [122]. An arbitrary daily recommended
intake has been set at 400 IU/d, but likely needs revision upward [123]. Recommendations for vitamin D supplementation either for women of childbearing age or for lactating women were not mentioned in the new US
dietary guidelines issued in 2005 [124].
Scientic data pertaining to vitamin D supplementation during lactation
are even scarcer than data on vitamin D supplementation during pregnancy.
An arbitrary daily recommended intake has been set at 400 IU/d, but
may be insucient [123]. Whether vitamin D deciency impairs the ability
to restore maternal skeleton postweaning is unclear [1]. Lactating mothers
supplemented with 1000 IU to 2000 IU of vitamin D for 15 weeks experienced increments in circulating maternal 25-hydroxyvitamin D3 levels of
16 ng/mL to 23 ng/mL [122]. It has been suggested that vitamin D supplementation of lactating mothers would improve vitamin D nutrition in the
mother and the breastfeeding infant; this has not been shown yet, but is currently under investigation [122]. Breastfed infants of vitamin Ddecient
mothers should receive vitamin D supplementation to avoid nutritional rickets [125]. There is low penetrance of vitamin D into breast milk, and it is
more ecient to give the vitamin D supplement directly to the infant, although supplementing the mother with high doses has been shown to
work [126].
Hypercalcemia of malignancy
Hypercalcemia of malignancy, an extremely rare occurrence, has been reported in two casesdone in metastatic breast cancer and the other in renal
cell carcinoma [127,128]. In both reports, the disease was rapidly progressive
and resulted in premature delivery at 29 and 32 weeks of gestation and maternal demise within 4 months postpartum. On the rst day postdelivery,
both infants were hypercalcemic, and one subsequently developed hypocalcemia from transient hypoparathyroidism. Intravenous pamidronate was
used shortly before delivery in one case with normalization of maternal serum calcium within 5 days of pamidronate administration [128]. Treatment
in such cases includes adequate aggressive hydration with close monitoring,
furosemide, and possibly calcitonin. Because bisphosphonates cross the placenta, their use should be reserved for life-threatening situations.
45
Summary
Studies of pregnant women indicate that the fetal calcium demand is met
largely by intestinal calcium absorption, which from early pregnancy onward more than doubles. The studies of biochemical markers of bone turnover, DXA, and ultrasound are inconclusive, but suggest that the maternal
skeleton also contributes calcium to the developing fetus. In contrast, during
lactation, skeletal calcium resorption is the dominant mechanism by which
calcium is supplied to the breast milk; renal calcium conservation is also apparent. Lactation produces an obligatory skeletal calcium loss regardless of
maternal calcium intake, but the calcium is completely restored to the skeleton after weaning through mechanisms that are not understood. The adaptations during pregnancy and lactation lead to novel presentations and
management issues for known disorders of calcium and bone metabolism,
such as primary hyperparathyroidism, hypoparathyroidism, and vitamin
D deciency. Finally, although some women experience fragility fractures
as a consequence of pregnancy or lactation, in most women the changes
in calcium and bone metabolism during pregnancy and lactation are normal
and without adverse consequences in the long-term.
References
[1] Kovacs CS, Kronenberg HM. Maternal-fetal calcium and bone metabolism during pregnancy, puerperium and lactation. Endocr Rev 1997;18:83272.
[2] Kovacs CS. Calcium and bone metabolism in pregnancy and lactation. J Clin Endocrinol
Metab 2001;86:23448.
[3] Prentice A. Pregnancy and lactation. In: Glorieux FH, Petifor JM, Juppner H, editors.
Pediatric bone: biology and diseases. New York: Academic Press; 2003. p. 24969.
46
[4] Chesney RW, Specker BL, McKay CP. Mineral metabolism during pregnancy and lactation. In: Coe FL, Favus MJ, editors. Disorders of bone and mineral metabolism. 2nd edition. Philadelphia: Lippincott, Williams & Wilkins; 2002. p. 34759.
[5] Taylor RN, Lebovic DI. The endocrinology of pregnancy. In: Greenspan FS, Gardner DG,
editors. Basic and clinical endocrinology. 7th edition. New York: Lange Medical Books/
McGraw-Hill; 2004. p. 63757.
[6] Dahlman T, Sjoberg HE, Bucht E. Calcium homeostasis in normal pregnancy and puerperium: a longitudinal study. Acta Obstet Gynecol Scand 1994;73:3938.
[7] Gallacher SJ, Fraser WD, Owens OJ, et al. Changes in calciotrophic hormones and biochemical markers of bone turnover in normal human pregnancy. Eur J Endocrinol 1994;
131:36974.
[8] Cross NA, Hillman LS, Allen SH, et al. Calcium homeostasis and bone metabolism during
pregnancy, lactation, and postweaning: a longitudinal study. Am J Clin Nutr 1995;61:
51423.
[9] Rasmussen N, Frolich A, Hornnes PJ, Hegedus L. Serum ionized calcium and intact parathyroid hormone levels during pregnancy and postpartum. Br J Obstet Gynaecol 1990;97:
8579.
[10] Seki K, Makimura N, Mitsui C, et al. Calcium-regulating hormones and osteocalcin levels
during pregnancy: a longitudinal study. Am J Obstet Gynecol 1991;164:124852.
[11] Gao P, DAmour P. Evolution of the parathyroid hormone (PTH) assaydimportance
of circulating PTH immunoheterogeneity and of its regulation. Clin Lab 2005;51:
219.
[12] Singh HJ, Mohammad NH, Nila A. Serum calcium and parathormone during normal pregnancy in Malay women. J Matern Fetal Med 1999;8:95100.
[13] Turner M, Barre PE, Benjamin A, et al. Does the maternal kidney contribute to the increased circulating 1,25-dihydroxyvitamin D concentrations during pregnancy? Miner
Electrolyte Metab 1988;14:24652.
[14] Woodrow JP, Noseworthy CS, Fudge NJ, et al. Calcitonin/calcitonin gene-related peptide
protect the maternal skeleton from excessive resorption during lactation [abstract]. J Bone
Miner Res 2003;18(Suppl 2):S37.
[15] Horwitz MJ, Tedesco MB, Sereika SM, et al. Direct comparison of sustained infusion of
human parathyroid hormone-related protein-(136). J Clin Endocrinol Metab 2003;88:
16039.
[16] Kovacs CS, Lanske B, Hunzelman JL, et al. Parathyroid hormone-related peptide (PTHrP)
regulates fetal-placental calcium transport through a receptor distinct from the PTH/
PTHrP receptor. Proc Natl Acad Sci U S A 1996;93:152338.
[17] Cornish J, Callon KE, Nicholson GC, Reid IR. Parathyroid hormone-related protein(107139) inhibits bone resorption in vivo. Endocrinology 1997;138:1299304.
[18] Marie PJ, Cancela L, Le Boulch N, Miravet L. Bone changes due to pregnancy and lactation: inuence of vitamin D status. Am J Physiol 1986;251:E4006.
[19] Sharpe CJ, Fudge NJ, Kovacs CS. A rapid 35% ux in bone mass occurs during pregnancy
and lactation cycles in mice [abstract]. International Bone and Mineral Society Meeting,
Osaka, Japan, June 37, 2003. Bone 2003;32(Suppl):S227.
[20] Purdie DW, Aaron JE, Selby PL. Bone histology and mineral homeostasis in human pregnancy. Br J Obstet Gynaecol 1988;95:84954.
[21] Naylor KE, Iqbal P, Fledelius C, et al. The eect of pregnancy on bone density and bone
turnover. J Bone Miner Res 2000;15:12937.
[22] Black AJ, Topping J, Durham B, et al. A detailed assessment of alterations in bone turnover, calcium homeostasis, and bone density in normal pregnancy. J Bone Miner Res
2000;15:55763.
[23] Ritchie LD, Fung EB, Halloran BP, et al. A longitudinal study of calcium homeostasis during human pregnancy and lactation and after resumption of menses. Am J Clin Nutr 1998;
67:693701.
47
[24] Ulrich U, Miller PB, Eyre DR, et al. Bone remodeling and bone mineral density during
pregnancy. Arch Gynecol Obstet 2003;268:30916.
[25] Kaur M, Pearson D, Godber I, et al. Longitudinal changes in bone mineral density during
normal pregnancy. Bone 2003;32:44954.
[26] Gambacciani M, Spinetti A, Gallo R, et al. Ultrasonographic bone characteristics during
normal pregnancy: longitudinal and cross-sectional evaluation. Am J Obstet Gynecol
1995;173:8903.
[27] Pearson D, Kaur M, San P, et al. Recovery of pregnancy mediated bone loss during lactation. Bone 2004;34:5708.
[28] To WW, Wong MW, Leung TW. Relationship between bone mineral density changes in
pregnancy and maternal and pregnancy characteristics: a longitudinal study. Acta Obstet
Gynecol Scand 2003;82:8207.
[29] Sowers M. Pregnancy and lactation as risk factors for subsequent bone loss and osteoporosis. J Bone Miner Res 1996;11:105260.
[30] Paton LM, Alexander JL, Nowson CA, et al. Pregnancy and lactation have no long-term
deleterious eect on measures of bone mineral in healthy women: a twin study. Am J
Clin Nutr 2003;77:70714.
[31] Dobnig H, Kainer F, Stepan V, et al. Elevated parathyroid hormone-related peptide levels
after human gestation: relationship to changes in bone and mineral metabolism. J Clin Endocrinol Metab 1995;80:3699707.
[32] VanHouten JN, Dann P, Stewart AF, et al. Mammary-specic deletion of parathyroid hormone-related protein preserves bone mass during lactation. J Clin Invest 2003;112:142936.
[33] VanHouten J, Dann P, McGeoch G, et al. The calcium-sensing receptor regulates mammary gland parathyroid hormone-related protein production and calcium transport.
J Clin Invest 2004;113:598608.
[34] Kovacs CS, Chik CL. Hyperprolactinemia caused by lactation and pituitary adenomas is
associated with altered serum calcium, phosphate, parathyroid hormone (PTH), and
PTH-related peptide levels. J Clin Endocrinol Metab 1995;80:303642.
[35] Sowers MF, Hollis BW, Shapiro B, et al. Elevated parathyroid hormone-related peptide associated with lactation and bone density loss. JAMA 1996;276:54954.
[36] Woodrow JP, Ho AO, Gagel RF, Kovacs CS. Calcitonin treatment rescues calcitonin-null
mice from excessive bone resorption during lactation [abstract]. J Bone Miner Res 2004;
19(Suppl):SA518.
[37] Laskey MA, Prentice A. Eect of pregnancy on recovery of lactational bone loss [letter].
Lancet 1997;349:15189.
[38] Cross NA, Hillman LS, Allen SH, Krause GF. Changes in bone mineral density and
markers of bone remodeling during lactation and postweaning in women consuming
high amounts of calcium. J Bone Miner Res 1995;10:131220.
[39] Kalkwarf HJ, Specker BL, Bianchi DC, Ranz J, Ho M. The eect of calcium supplementation on bone density during lactation and after weaning. N Engl J Med 1997;337:5238.
[40] Polatti F, Capuzzo E, Viazzo F, et al. Bone mineral changes during and after lactation.
Obstet Gynecol 1999;94:526.
[41] Koltho N, Eiken P, Kristensen B, Nielsen SP. Bone mineral changes during pregnancy
and lactation: a longitudinal cohort study. Clin.Sci (Colch) 1998;94:40512.
[42] Laskey MA, Prentice A, Hanratty LA, et al. Bone changes after 3 mo of lactation: inuence
of calcium intake, breast-milk output, and vitamin D-receptor genotype. Am J Clin Nutr
1998;67:68592.
[43] VanHouten JN, Wysolmerski JJ. Low estrogen and high parathyroid hormone-related peptide levels contribute to accelerated bone resorption and bone loss in lactating mice. Endocrinology 2003;144:55219.
[44] Wermers RA, Khosla S, Atkinson EJ, et al. The rise and fall of primary hyperparathyroidism: a population-based study in Rochester, Minnesota, 19651992. Ann Intern Med 1997;
126:43340.
48
[45] Heath H, Hodgson SF, Kennedy MA. Primary hyperparathyroidism: incidence, morbidity, and potential economic impact in a community. N Engl J Med 1980;302:
18993.
[46] Kelly TR. Primary hyperparathyroidism during pregnancy. Surgery 1991;110:102834.
[47] Haenel LC, Mayeld RK. Primary hyperparathyroidism in a twin pregnancy and review of
fetal/maternal calcium homeostasis. Am J Med Sci 2000;319:1914.
[48] Kort KC, Schiller HJ, Numann PJ. Hyperparathyroidism and pregnancy. Am J Surg 1999;
177:668.
[49] Schnatz PF, Curry SL. Primary hyperparathyroidism in pregnancy: evidence-based management. Obstet Gynecol Surv 2002;57:36576.
[50] El-Hajj Fuleihan G. Pathogenesis and etiology of primary hyperparathyroidism. In: Rose
BD, editor. UpToDate 13.1. Wellesley (MA): UpToDate; 2005.
[51] Kristoersson A, Dahlgren S, Lithner F, Jarhult J. Primary hyperparathyroidism in pregnancy. Surgery 1985;97:32630.
[52] Shangold MM, Dor N, Welt SI, et al. Hyperparathyroidism and pregnancy: a review. Obstet Gynecol Surv 1982;37:21728.
[53] Clark D, Seeds JW, Cefalo RC. Hyperparathyroid crisis and pregnancy. Am J Obstet Gynecol 1981;140:8402.
[54] Carella MJ, Gossain VV. Hyperparathyroidism and pregnancy: case report and review.
J Gen Intern Med 1992;7:44853.
[55] Croom RD, Thomas CG. Primary hyperparathyroidism during pregnancy. Surgery 1984;
96:110918.
[56] Inabnet WB, Baldwin D, Daniel RO, Staren ED. Hyperparathyroidism and pancreatitis
during pregnancy. Surgery 1996;119:7103.
[57] Matthias GS, Helliwell TR, Williams A. Postpartum hyperparathyroid crisis: case report.
Br J Obstet Gynaecol 1987;94:80710.
[58] Negishi H, Kobayashi M, Nishida R, et al. Primary hyperparathyroidism and simultaneous
bilateral fracture of the femoral neck during pregnancy. J Trauma 2002;52:3679.
[59] Hess HM, Dickson J, Fox HE. Hyperfunctioning parathyroid carcinoma presenting as
acute pancreatitis in pregnancy. J Reprod Med 1980;25:837.
[60] Wagner G, Transhol L, Melchior JC. Hyperparathyroidism and pregnancy. Acta Endocrinol (Copenh) 1964;47:54964.
[61] Ludwig GD. Hyperparathyroidism in relation to pregnancy. N Engl J Med 1962;267:
63742.
[62] Delmonico FL, Neer RM, Cosimi AB, et al. Hyperparathyroidism during pregnancy. Am J
Surg 1976;131:32837.
[63] Kaplan EL, Burrington JD, Klementschitsch P, et al. Primary hyperparathyroidism, pregnancy, and neonatal hypocalcemia. Surgery 1984;96:71722.
[64] Ip P. Neonatal convulsion revealing maternal hyperparathyroidism: an unusual case of late
neonatal hypoparathyroidism. Arch Gynecol Obstet 2003;268:2279.
[65] Bruce J, Strong JA. Maternal hyperparathyroidism and parathyroid deciency in the child,
with account of eect of parathyroidectomy on renal function, and of attempt to transplant
part of tumor. QJM 1955;24:30719.
[66] Petit D, Clark R. Hyperparathyroidism and pregnancy. Am J Surg 1947;74:8604.
[67] Schnatz PF. Surgical treatment of primary hyperparathyroidism during the third trimester.
Obstet Gynecol 2002;99(5 Pt 2):9613.
[68] Rajala B, Abbasi RA, Hutchinson HT, Taylor T. Acute pancreatitis and primary hyperparathyroidism in pregnancy: treatment of hypercalcemia with magnesium sulfate. Obstet
Gynecol 1987;70(3 Pt 2):4602.
[69] Dahan M, Chang RJ. Pancreatitis secondary to hyperparathyroidism during pregnancy.
Obstet Gynecol 2001;98(5 Pt 2):9235.
[70] Brown EM, Pollak M, Seidman CE, et al. Calcium-ion-sensing cell-surface receptors.
N Engl J Med 1995;333:23440.
49
[71] El-Hajj Fuleihan G, Heath HH. Familial benign hypocalciuric hypercalcemia and neonatal
severe hyperparathyroidism. In: Bilezikian JP, Marcus R, Levine MA, editors. The parathyroids: basic and clinical concepts. 2nd edition. New York: Academic Press; 1994.
p. 60723.
[72] Powell BR, Buist NR. Late presenting, prolonged hypocalcemia in an infant of a woman
with hypocalciuric hypercalcemia. Clin Pediatr (Phila) 1990;29:2413.
[73] Thomas BR, Bennett JD. Symptomatic hypocalcemia and hypoparathyroidism in two infants of mothers with hyperparathyroidism and familial benign hypercalcemia. J Perinatol
1995;15:236.
[74] OLeary JA, Klainer LM, Neuwirth RS. The management of hypoparathyroidism in pregnancy. Am J Obstet Gynecol 1966;94:11037.
[75] Callies F, Arlt W, Scholz HJ, et al. Management of hypoparathyroidism during pregnancydreport of twelve cases. Eur J Endocrinol 1998;139:2849.
[76] Caplan RH, Beguin EA. Hypercalcemia in a calcitriol-treated hypoparathyroid woman
during lactation. Obstet Gynecol 1990;76:4859.
[77] Salle BL, Berthezene F, Glorieux FH, et al. Hypoparathyroidism during pregnancy: treatment with calcitriol. J Clin Endocrinol Metab 1981;52:8103.
[78] Sadeghi-Nejad A, Wolfsdorf JI, Senior B. Hypoparathyroidism and pregnancy: treatment
with calcitriol. JAMA 1980;243:2545.
[79] Landing BH, Kamoshita S. Congenital hyperparathyroidism secondary to maternal hypoparathyroidism. J Pediatr 1970;77:8427.
[80] Taussig HB. Possible injury to the cardiovascular system from vitamin D. Ann Intern Med
1966;65:1195200.
[81] Friedman WF, Mills LF. The relationship between vitamin D and the craniofacial and dental anomalies of the supravalvular aortic stenosis syndrome. Pediatrics 1969;43:128.
[82] Mather KJ, Chik CL, Corenblum B. Maintenance of serum calcium by parathyroid hormone-related peptide during lactation in a hypoparathyroid patient. J Clin Endocrinol
Metab 1999;84:4247.
[83] Shomali ME, Ross DS. Hypercalcemia in a woman with hypoparathyroidism associated
with increased parathyroid hormone-related protein during lactation. Endocr Pract 1999;
5:198200.
[84] Breslau NA, Zerwekh JE. Relationship of estrogen and pregnancy to calcium homeostasis
in pseudohypoparathyroidism. J Clin Endocrinol Metab 1986;62:4551.
[85] Nordin BE, Roper A. Postpregnancy osteoporosis: a syndrome? Lancet 1955;1:4314.
[86] Phillips AJ, Ostlere SJ, Smith R. Pregnancy-associated osteoporosis: does the skeleton recover? Osteoporos Int 2000;11:44954.
[87] Khovidhunkit W, Epstein S. Osteoporosis in pregnancy. Osteoporos Int 1996;6:34554.
[88] Smith R, Athanasou NA, Ostlere SJ, Vipond SE. Pregnancy-associated osteoporosis. QJM
1995;88:86578.
[89] Reid IR, Wattie DJ, Evans MC, Budayr AA. Post-pregnancy osteoporosis associated with
hypercalcaemia. Clin Endocrinol (Oxf) 1992;37:298303.
[90] Dunne F, Walters B, Marshall T, Heath DA. Pregnancy associated osteoporosis. Clin Endocrinol (Oxf) 1993;39:48790.
[91] Lakhanpal S, Ginsburg WW, Luthra HS, Hunder GG. Transient regional osteoporosis:
a study of 56 cases and review of the literature. Ann Intern Med 1987;106:44450.
[92] Arayssi TK, Tawbi HA, Usta IM, Hourani MH. Calcitonin in the treatment of transient
osteoporosis of the hip. Semin Arthritis Rheum 2003;32:38897.
[93] Curtiss PH Jr, Kincaid WE. Transitory demineralization of the hip in pregnancy: a report
of three cases. J Bone Joint Surg Am 1959;41:132733.
[94] Khastgir G, Studd JW, King H, et al. Changes in bone density and biochemical markers of
bone turnover in pregnancy-associated osteoporosis. Br J Obstet Gynaecol 1996;103:7168.
[95] Brown EM, El-Hajj Fuleihan G, Chen CJ, Kifor O. A comparison of the eects of
divalent and trivalent cations on parathyroid hormone release, 3#,5#-cyclic-adenosine
50
[96]
[97]
[98]
[99]
[100]
[101]
[102]
[103]
[104]
[105]
[106]
[107]
[108]
[109]
[110]
[111]
[112]
[113]
[114]
[115]
[116]
[117]
51
[118] Prentice A, Yan L, Jarjou LM, et al. Vitamin D status does not inuence the breast-milk
calcium concentration of lactating mothers accustomed to a low calcium intake. Acta Paediatr 1997;86:10068.
[119] Bezerra FF, Mendonca LM, Lobato EC, et al. Bone mass is recovered from lactation to
postweaning in adolescent mothers with low calcium intakes. Am J Clin Nutr 2004;80:
13226.
[120] Specker B. Vitamin D requirements during pregnancy. Am J Clin Nutr 2004;80(Suppl):
1740S7S.
[121] Mahomed K, Gulmezoglu AM. Vitamin D supplementation in pregnancy. Cochrane Database Syst Rev 2000;2:CD000228.
[122] Hollis BW, Wagner CL. Assessment of dietary vitamin D requirements during pregnancy
and lactation. Am J Clin Nutr 2004;79:71726.
[123] Hollis BW. Circulating 25-hydroxyvitamin D levels indicative of vitamin D suciency:
implications for establishing a new eective dietary intake recommendation for vitamin D.
J Nutr 2005;135:31722.
[124] Department of Health and Human Services and the Department of Agriculture. Dietary
guidelines for Americans 2005. Executive summary. January 12, 2005. Available at:
http://www.health.gov/dietaryguidelines/dga2005/document/html/executivesummary.htm.
Accessed April 25, 2005.
[125] Thomson K, Morley R, Grover SR, Zacharin MR. Postnatal evaluation of vitamin D and
bone health in women who were vitamin D-decient in pregnancy, and in their infants. Med
J Aust 2004;181:4868.
[126] Hollis BW, Wagner CL. Vitamin D requirements during lactation: high-dose maternal supplementation as therapy to prevent hypovitaminosis D for both the mother and the nursing
infant. Am J Clin Nutr 2004;80(Suppl):1752S8S.
[127] Usta IM, Chammas M, Khalil AM. Renal cell carcinoma with hypercalcemia complicating
a pregnancy: case report and review of the literature. Eur J Gynaecol Oncol 1998;19:5847.
[128] Illidge TM, Hussey M, Godden CW. Malignant hypercalcaemia in pregnancy and antenatal administration of intravenous pamidronate. Clin Oncol (R Coll Radiol) 1996;8:2578.
[129] Richards GE, Brewer ED, Conley SB, Saldana LR. Combined hypothyroidism and hypoparathyroidism in an infant after maternal 131I administration. J Pediatr 1981;99:1413.
54
LANGER
muscles, white adipose tissue, and liver and suppression of hepatic glucose
production. The risk factors associated with type 2 diabetes and GDM
are comparable (eg, obesity, ethnicity, family history). b-Cell adaptation
to insulin resistance is impaired in women with GDM and may be a universal
response to insulin resistance because it is found in many ethnic groups.
Women with a history of GDM are at an increased risk for subsequent development of type 2 diabetes (50%80%). Type 2 diabetes and GDM arguably may be the same disease with dierent names [46].
55
with diabetes do not target the actual normal levels in pregnancies of nondiabetic women (Table 1) [1,23,24].
Optimizing clinical outcome for various diabetic complications in pregnancy occurs at dierent levels of blood glucose. A decreased rate of congenital anomalies was observed when the postprandial threshold was less
than 140 mg/dL or the preprandial threshold was less than 120 mg/dL
[2528]. In contrast, a mean blood glucose of less than 100 mg/dL to 110
mg/dL was associated with fewer large-for-gestational-age (LGA) or macrosomic newborns [10,18,2931]. This association suggests that there are clinical thresholds for optimizing pregnancy rather than an absolute number for
normoglycemia that in many cases is unobtainable. Because macrosomia
and fetal hyperinsulinemia are the central complications in GDM, the targeted threshold needs to be mean blood glucose of 90 mg/dL to 100 mg/dL
and postprandial blood glucose of 110 mg/dL to 120 mg/dL.
DCCT [17]b ACOG [23] ADA [24] 4th Intl [1] Parretti [21] Yogev [22]
70120
70120
6090
60105
! 105
d
! 95
d
5560 G 5
NA
75 G 12
78 G 11
d
d
! 180
O 65
! 130140
! 120
d
6090
! 155
! 130
d
d
! 140
! 120
d
d
98105 G 5
8595 G 6
d
6065 G 5
97.0 G 11
105 G 13
d
68.3 G 10
NA
100
74.7 G 5.2
84.0 G 18
56
LANGER
57
size, they chose to conclude that . The study suggests that intensive treatment of GDM may have little eect on birth weight, birth trauma, operative
delivery, or neonatal metabolic disorders [36].
Treatment modalities in gestational diabetes
The introduction of new pharmacologic alternatives for treatment (insulin analogues and oral antidiabetic agents) and their use in pregnancy make
it worthwhile to consider proven and potential benets during gestation.
Although treatment modalities for achieving targeted levels of glycemic
control in type 1 diabetes, type 2 diabetes, and GDM dier, diet, exercise,
insulin, and oral antidiabetic drugs are the chief means of reducing blood
glucose concentrations. In pregnant women with type 2 diabetes, oral antidiabetic drugs have not been tested adequately in terms of whether targeted
glycemic levels can be achieved.
Diet and exercise: modalities that enhance glucose control
Diet is the mainstay of treatment in GDM whether or not pharmacologic
therapy is introduced. Dietary control with a reduction in fat intake and the
substitution of complex carbohydrates for rened carbohydrates seeks to
achieve and maintain the maternal blood glucose prole essential during gestation. Two current approaches are recommended: decreasing the proportion
of carbohydrates to 35% to 40% in a daily regimen of three meals and three
to four snacks [1,23,24,37] or lowering the glycemic index so that carbohydrates account for approximately 60% of daily intake. The assignment of
daily caloric intake is similar for women with either GDM or pregestational
diabetes and is calculated based on prepregnancy body mass index (BMI)
[1,23,24]. In general, for normal-weight women (BMI 2025), 30 kcal/kg
should be prescribed; for overweight and obese women (BMI O 2534),
calories should be restricted to 25 kcal/kg; and for morbidly obese women
(BMI O 34), calories should be restricted to 20 kcal/kg or less. Caloric restrictions of 30% in obese patients are associated with the same rate of macrosomia as in the general population. When caloric restrictions are applied,
free fatty acids and ketone bodies may increase (starvation ketosis), requiring
daily assessment of the ketones in the morning urine; if positive, blood assessment needs to be done [38]. A moderate exercise program for pregnant diabetic women who are willing and able may improve postprandial blood
glucose levels and insulin sensitivity [24,39]. Some women are less able to exercise owing to issues of socioeconomic limitations, obesity, and multiparity.
Insulin therapy
Types of insulin to manage diabetes in pregnancy
Insulin production has progressed from animal species to human insulin
preparations produced with recombinant DNA technology to the current
58
LANGER
Table 2
Studies reporting the use of insulin analogs in pregnancy
Study
design
Type of
diabetes
CR
RCT
Retro
Retro
Retro
Retro
1
GDM
1 and 2
1 and 2
1
1 and 2, GDM
2
19
10
16
12
20
d
23
d
21
42
57
RCT
Prosp
CR
Retro
1
1
GDM
1
16
36
1
25
17
33
35
RCT
RCT
Retro
Retro
GDM
Diabetes
1
PGDM
25
9
7
25
24
d
d
46
Retro
RCT
CR
1
GDM
1
50
d
d
26
d
d
Neonatal outcome
Other
DPR
Anomalies
Complications
LGA
2/2
d
d
d
0/12
L: 1/20
H
H: 9/57
d
d
d
L: 2/25
H: 2/35
d
d
0/7
d
d
d
6/76
d
d
d
d
d
d
d
d
d
d
d
d
d
d
d
d
d
d
Hypoglycemia
Lispro: 17%
Human: 23%
Hypoglycemia: 41%
d
d
d
16%
d
d
d
d
d
d
d
d
d
24%
Macrosomia O 4000 g
d
d
57%
15a
1b
d
d
3/10
NP
NP
d
d
d
d
NP
d
d
d
d
NP
d
d
d
d
P (6 cases)
d
d
Lispro: 44%
Human: 30%
35%
d
d
59
Abbreviations: CR, case report; DPR, diabetes proliferative retinopathy; H, human; L, lispro; NP, no progression; P, progression; Prosp, prospective;
RCT, randomized control trial; Retro, retrospective; d, no data.
a
Insulin aspart.
b
Insulin glargine.
Study
(rst author, year [Ref.])
60
LANGER
labor; insulin lispro was not detected in the umbilical cord blood of the
infants [45].
Using an in vitro model in which human placentas were perfused, Challier and coworkers [63] reported evidence of human insulin in the fetal perfusate after infusion into the maternal compartment of the placenta.
Boskovic and coworkers [64] evaluated 11 term human placentas obtained
from uncomplicated pregnancies immediately after delivery. No placental
transfer was detected during perfusion with insulin lispro (100 mU/mL and
200 mU/mL). In contrast, there was a concentration-dependent transfer to
the fetal perfusate at insulin lispro levels of 580 mU/mL and higher. Finally,
the investigators compared actual maternal serum level and administered
doses of insulin lispro. Mothers treated with 50 U of insulin lispro achieved
serum concentrations greater than 200 mU/mL with an apparent linear correlation between dose and levels [64]. The investigators did not evaluate placentas of diabetic mothers, however. Placentas generally are aected by the
disease, which may inuence the perfusion characteristics. One can speculate
that even in the presence of lower insulin doses, placental transfer may occur. Although it is unlikely that insulin lispro in therapeutic doses would
cross the placenta, the high dose of insulin required in pregnancy, especially
in GDM and type 2 diabetes, to achieve established levels of glycemic control needs to be weighed against the potential for placental transfer and adverse outcome for the fetus.
Before using insulin analogues to treat pregnant women, several issues
need to be addressed: Can pregnant women achieve targeted levels of glycemic control with the use of insulin lispro? The response is yes; the quality of
glycemic control parallels the accepted criteria recommended during pregnancy. Is the quality of glycemic control during pregnancy with the use of
insulin lispro comparable to that with the use of regular insulin? Is the incidence of hypoglycemic incidents similar? Six published studies compared the
use of insulin lispro and regular insulin in pregnancy. Most of the studies
found no signicant dierence with respect to glycemic control and the incidence of hypoglycemia. Two of the studies were performed on women with
GDM with a total of 60 women treated with insulin lispro. In the study by
Persson and coworkers [50], there was a lower postprandial glucose concentration after breakfast and a slightly higher rate of hypoglycemia (! 55 mg/dL)
in the insulin lispro group. If there is no advantage in using insulin lispro
compared with regular insulin, is insulin lispro as safe in pregnancy as regular insulin? Insulin lispro may be more user-friendly with negligible dierences in glycemic control and hypoglycemia. These assets cannot currently
justify extensive use of the drug, however, before establishing its safety in
well-controlled clinical trials.
Which patients should receive pharmacologic therapy?
When diet fails to achieve targeted levels of glycemic control, insulin and
antidiabetic agents are validated treatment options. Available guidelines
61
dier regarding the threshold of fasting plasma glucose at which pharmacologic therapy (glyburide or insulin) should be initiated [23,24,65]. Some authors recommend a threshold of fasting plasma glucose 95 mg/dL or greater
[1,65] whereas others recommend a threshold of 105 mg/dL or greater
[23,24]. Using a fasting plasma glucose threshold of 95 mg/dL or greater decreases the rate of macrosomia and LGA infants [66,67].
Most authorities agree on initiation of drug therapy with elevated postprandial values (R 120 mg/dL for 2 hours or R 140 mg/dL for 1 hour).
Using these standards, approximately 30% to 50% of women with GDM
require pharmacologic therapy when diet therapy alone fails to reduce glycemic levels. When patients who qualied for diet therapy were evaluated,
only patients who achieved established levels of glycemic control improved
insulin secretion and sensitivity. Patients who failed to achieve glycemic
control, although exhibiting slightly improved insulin sensitivity, did not
achieve the same level of insulin response and sensitivity as nondiabetic
women [68]. Studies using continuous blood glucose monitoring have shed
new light on the existing controversy whether to test blood glucose at 1 or
2 hours postprandial in pregnant women. The author found that the time
from start of meal to the postprandial peak is approximately 80 to 90 minutes depending on the type of diabetes and regardless of level of glycemia. In
nondiabetic pregnant women, the peak postprandial value was 110 mg/dL.
The association between this physiologic characteristic and pregnancy outcome needs to be evaluated before changing current clinical thresholds
[22,69].
Can the fetus provide a marker for pharmacologic initiation?
Three randomized controlled studies addressed the use of fetal abdominal
circumference to guide insulin therapy. This approach combined maternal
glucose and fetal growth parameters. The studies suggest that some women,
despite glucose levels above established targets (R 105 mg/dL) may not derive a fetal benet from intensied therapy [7072]. In a randomized study
with a large sample size, the author found similar results. In the authors
study, subjects who did not achieve targeted levels of glycemic control
had higher rates of macrosomia and LGA infants, however, regardless of
abdominal circumference [73]. The limitation of the use of abdominal circumference at 28 weeks gestation as a measure for insulin initiation is
that it is snapshot information, whereas fetal growth is longitudinal. Most
environmental eects (eg, glucose) occur during the third trimester, which
is also the time of most fetal growth. A fetus at 28 weeks gestation in the
40th percentile of growth can double its weight and reach the 85th percentile
under the inuence of elevated blood glucose. Using the fetus as an additional marker for the decision-making process in initiating pharmacologic
therapy is an attractive approach. It should not be used as a single predictor,
but in conjunction with GDM severity parameters and level of glycemic
control throughout pregnancy.
62
LANGER
63
85 1 (unit) 85. The total insulin dose is divided so that two thirds is administered in the morning, which is further split in a ratio of 2:1 (intermediate and rapid-acting), and one third is administered with supper and
bedtime in a ratio of 1:1 (rapid-acting and intermediate). The rapid-acting
dose is administered with supper, and the intermediate dose is taken before
bedtime. If after 3 to 7 days the GDM patient has not achieved the desired
level of glycemic control, the total insulin dose should increase by 10% to
20% and thereafter adjusted when needed. The actual total insulin dose in
GDM women is 40% higher than the calculated (starting) dose [76]. The decreased insulin sensitivity characterizes pregnancy and in particular GDM
patients. As a rule of thumb, before every insulin administration, self-monitoring blood glucose assessment needs to occur.
Oral antidiabetic agents as alternatives to insulin therapy
A variety of oral agents may be alternatives to insulin therapy for women
with GDM. Sulfonylureas are insulin secretagogues (eg, glyburide and
glipizide). The primary action of glyburide is to increase insulin secretion, decreasing hepatic glucose production with resultant reversal of hyperglycemia and indirect improvement of insulin sensitivity [78] Antidiabetic
drug groups include meglitinides (insulin secretagogues, such as the rapidacting repaglinide, which limit postprandial hyperglycemia), biguanides
(eg, metformin, which decreases insulin resistance), a-glucosidase inhibitors
(eg, acarbose, which reduces intestinal absorption of starch and glucose),
and thiazolidinediones (eg, rosiglitazone and pioglitazone). All have been
used successfully in the treatment of nonpregnant patients with type 2 diabetes.
Each may be used alone or in combination with other oral agents or insulin.
Most of these drugs have not been studied in pregnancy or only minimally so. The most data regarding safety in pregnancy for oral antidiabetic
drugs are with the use of glyburide. To date, 1261 women treated with glyburide have been reported in the literature. For insulin analogues and oral
antidiabetic drugs, none of the studies were blinded (Table 3). Many experts
and authoritative bodies have recommended using glyburide as an alternative to insulin [65,7983]. Others have not rmly advocated the use of oral
agents in pregnancy and recommend further evaluation [24,84,85]. The
use of oral agents is a pragmatic alternative to insulin therapy in pregnancy
because of ease of administration and patient satisfaction with a noninvasive
treatment. However valid these reasons, the introduction of a new drug is
unjustied if improvements in pregnancy outcome and cost-eectiveness
are not evaluated denitively.
Is there increased risk for fetal anomalies with the use of oral
antidiabetic drugs?
For a drug to be potentially eective and safe in pregnancy, it should not
cross the placenta or should not be detrimental to the fetus at concentrations
64
Table 3
Studies reporting use of oral antidiabetic agents in pregnancy
No. of patients
Type of
Regular
diabetes Glyburide insulin Metformin Other
Achievement
of good control
Langer,
2000 [78]
RCT
GDM
201
203
Lim,
1997 [93]
Prosp, observ
GDM
33
21
No signicant
dierence
Conway,
2004 [94]
Kremer,
2004 [95]
Chmait,
2004 [96]
Prosp, observ
GDM
75
84%
Prosp, observ
GDM
73
81%
Prosp, observ
GDM
69
82%
Neonatal outcome
Complications
LGA
No dierence in
metabolic
complications,
congenital
anomalies, and
PNM
No signicant
dierence
in metabolic
complications
and PNM
NA
Caesarean
section: 36%
No signicant
dierence
NA
Macrosomia:
19%
Macrosomia: 7%
LANGER
Study
(rst author, Study
year [Ref.]
design
Prosp, observ
GDM
22
22
82%
Fines,
2003 [98]
Retro
case-control
GDM
40
44
NA
Velazques,
2003 [99]
Case series
GDM
31
Improved level
of glycemic
control in the
glyburide
group: 82%
Improved level
of glycemic
control
Pendsey,
RCT
2002 [100]
23
Repaglinide: 23
Glueck,
2004 [90]
Prosp, observ
PCOS
42
Glueck,
2002 [91]
Prosp and
PCOS
retro, observ
33
w/o metformin: 39
No signicant
No signicant
dierence
dierence in
between insulin
the rate
between
and glyburide
groups
insulin- and
glyburidetreated groups
No dierence in
Less macrosomia
ponderal index
in the glyburide
group (5/40 vs
between the
groups
11/44)
No hypoglycemic LGA in the
events in the
glyburide and
glyburide group
in the insulin
group: 16%
and 29%
GDM developed
in 7.1% of
patients
GDM developed
in 3% of
patients treated
with metformin
vs 27% w/o
treatment
Gilson,
2002 [97]
65
66
Table 3 (continued )
Study
(rst author, Study
year [Ref.]
design
Type of
Regular
diabetes Glyburide insulin Metformin Other
2
Achievement
of good control
78
GDM
and 2
GDM
and 2
Neonatal outcome
Complications
Drug deemed safe
in rst trimester
Reduced PNM
126
60
GDM: 81.4%
Type 2 diabetes:
46.2%
Hellmuth,
Prosp, observ
2000 [104]
GDM
42
50
Sulfonylurea: 68
No signicant
dierence in
neonatal
morbidity.
Higher rate of
preeclampsia
(32% vs 10%)
and PNM
(11.6% vs
1.3%) in
metformin
group
LGA
LANGER
Coetzee,
Retro
1984 [101]
Coetzee,
Retro
1986 [102]
Coetzee,
Prosp, observ
1979 [103]
No. of patients
Notelovitz,
RCT
1971 [105]
GDM
and 2
GDM
Moore,
RCT
2005 [107]
Ramos,
Retro
2005 [108]
GDM
GDM
25
30
31
236
316
32
Tolbutamide
chlorpropamin:
2 52
Using oral
hypoglycemic:
80%
Diet treated: 27
Using insulin:
36%
Mean blood
glucose similar
in all groups
Blood glucose
similar
Blood glucose
similar
No signicant
dierence in
PNM,
metabolic
complications,
and congenital
anomalies
Signicantly
lower rate
of maternal
hypoglycemia
in glyburide
group
Perinatal outcome
similar
Perinatal outcome
similar
Abbreviations: NA, not available; Observ, observational; PNM, perinatal mortality; Prosp, prospective; RCT, randomized controltrial; Retro, retrospective; w/o, without.
Yogev,
Prosp
2004 [106]
52
67
68
LANGER
that are clinically indicated for the mother. Case reports and small retrospective studies in women receiving rst-generation sulfonylureas raised concern
about congenital anomalies [86,87]. Potential eects on the fetus, mainly hypoglycemia and growth stimulation [87], were reported. A report of increased
rates of congenital malformations involved 20 type 2 diabetes patients who
had hyperglycemia before conception (hemoglobin A1c O 8). It is impossible
to determine if the reported rate of anomalies was due to the use of the drug
or the preexisting hyperglycemia [86]. In contrast, several studies showed that
anomalies in the infants of women who received oral antidiabetic agents were
associated with altered maternal glucose metabolism and not the drug [88]. A
meta-analysis failed to show an increased risk for fetal anomalies with sulfonylureas [89]. Metformin seems to be unassociated with congenital malformations in patients with polycystic ovary syndrome and reduces the
occurrence of GDM and spontaneous abortion (see Table 3) [9092].
Does glyburide cross the placenta?
The placenta of the diabetic mother is characterized by capillary dilation,
relatively immature villous structure, and chronic disturbances in intervillous circulation. The author examined the placentas of nondiabetic and diabetic mothers in vitro and showed that glyburide (glibenclamide) does not
cross the human placenta from the maternal to fetal circulation in signicant
amounts. There was virtually no drug transport even when concentrations
three to four times higher than peak therapeutic levels were employed
[109111]. The author also showed that rst-generation sulfonylureas diffused across the placenta most freely [109111]. There is evidence that the
qualitative aspects of transfer are comparable between placentas obtained
during the rst and third trimesters [64,112]. In mothers treated with therapeutic plasma concentrations of glyburide, the drug was undetectable in the
cord blood of their neonates [78]. No data exist on the long-term eects on
the infant when oral antidiabetic drugs and insulin analogues are used in the
mother. Glyburide exhibits less transfer across the placenta compared with
other agents, underscoring its potential therapeutic usefulness.
Because glyburide does not cross the placenta, it cannot aect neonatal
hypoglycemia or fetal anomalies. In addition, most GDM patients are identied between 24 and 28 weeks gestation. The fetus is not exposed to the
drug during organogenesis. In rare cases of early diagnosis in the rst trimester (perhaps type 2) and recognized type 2 diabetes, current data suggest
that the use of glyburide or metformin would not increase the rate of anomalies inuenced by the level of glycemic control. Although a randomized
study would be the ideal model to address this issue, it is highly unlikely
that it would be performed for ethical considerations (see Table 3).
Glyburide pharmacology and administration
The pharmacologic mechanism of action of glyburide is to increase insulin secretion, and its secondary eect is to decrease insulin resistance by
69
70
LANGER
Table 4
Comparison of selected pregnancy outcomes in conventional versus intensied insulin therapy,
glyburide versus insulin, and nondiabetic subjects
Therapy
comparison [18]a
Criteria
LGA (%)b
Macrosomia (%)
Ponderal index O 2.85
Overall Caesarean section (%)
Induction of labor (%)
PET (%)
CHTN (%)
5-Min Apgar score ! 7 (%)
Neonatal ICU admission (%)
Hypoglycemia (%) ! 40 mg/dL
Polycythemia (%) O 60%
Hyperbilirubinemia (%)
O 11 mg/dL
Hypocalcemia (%) ! 8 mg/dL
Respiratory complications (%)
Shoulder dystocia
Perinatal mortality
Stillbirth
Neonatal death
Comparison [78]
Nondiabetic
Conventional Intensied Glyburide Insulin [8,18]
20.1
13.6
21.7
21.5
27
5.6
6.1
3.2
25.6
20.0
12.0
17.5
13.1
7.1
13.8
15.0
22
5.9
5.8
2.1
6.3
3.8
0.7
7.9
12.0
7.2
9.0
23.0
33
6.0
6.0
3.1
6.0
9.0
2.0
6.0
13.0
4.7
12.0
24.0
34
6.0
9.1
4.2
7.0
6.0
3.0
4.0
11.9
8.1
22.0
13.7
13.0
7.6
6.2
2.5
4.7
2.5
1.4
6.4
4.0
6.2
1.4
0.3
2.3
0.4
1.0
2.0
1.5
1.0
3.0
1.6
NA
2.1
0.5
4/1000
2/1000
1/1000
3/1000
5/1000
5/1000
5/1000 4/1000
5/1000 4.7/1000
Abbreviations: CHTN, chronic hypertension; ICU, intensive care unit; NA, not available;
PET, preeclampsia.
a
Conventional therapy: fasting plasma glucose and 2-h postprandial once weekly, visualized
self-monitoring blood glucose 4 times daily; intensied therapy: memory-based self-monitoring
blood glucose 7 times daily.
b
LGA O 90th percentile; macrosomia R 4000 g.
severity when fasting plasma glucose results were between 95 mg/dL and 139
mg/dL. Of patients, 71% required a 10-mg daily dose of glyburide to
achieve glycemic control. In all disease severity levels, glyburide and insulin-treated subjects had similar success rates in achieving targeted glucose
levels and pregnancy outcomes [113]. Achieving the established level of glycemic control, not the mode of therapy, seems to be the key to improving
pregnancy outcome in GDM.
Is glyburide therapy less costly than insulin therapy?
The costs of alternative therapies should be addressed when dierent
medications exhibit similar eectiveness and safety. Goetzel and Wilkins
[114] compared the costs of insulin and glyburide and observed that glyburide is considerably less costly (average savings per patient of $166$200
based on rates in 2000). Glyburide is a cost-eective, patient-friendly, potentially adherence-enhancing therapy that produces perinatal outcome comparable to insulin therapy.
Mechanism of action
Sulfonylureas
Glimepiride (Amaryl)
Glipizide (Glucotrol)
Glipizide-GITS (Glucotrol XL)
Glyburide (DiaBeta, Glynase,
Micronase)
Meglitinides
Nateglinide (Starlix)
Repaglinide (Prandin)
Biguanide
Metformin (Glucophage)
Glitazones
Pioglitazone (Actos)
Rosiglitazone (Avandia)
Pregnancy
category
Decrease in
FPG (mg/dL)
Decrease in
hemoglobin A1C (%)
6070
1.52
C
C
d
B
921
C
C
Unknown
Minimal
d
No
Unknown
Unknown
d
No
Unknown
Unknown
Unknown
Unknown
Yes
No
Unknown
Unknown
Animals
Animals
0.92
B
5980
Excreted in
breast milk
0.50.8
C
C
5978
Cross
placenta
Table 5
Oral antidiabetic drug classication
1.42.6
71
72
Table 5 (continued )
Drug (trade name)
Mechanism of action
Alpha-glucosidase inhibitors
Acarbose (Precose)
Miglitol (Glyset)
Decrease in
FPG (mg/dL)
Decrease in
hemoglobin A1C (%)
2030
0.51
B
B
Dose-dependent
B
B
B
B
B
C
B
Cross
placenta
Excreted in
breast milk
Unknown
Unknown
Animals
Animals
Unknown
Minimal
No
No
No
Unknown
No
No
No
No
No
No
No
No
Dose-dependent
LANGER
Insulin
Aspart (Novolog)
Lispro (Humalog)
Regular
NPH
Lente
Glargine (Lantus)
Ultralente
Pregnancy
category
73
74
LANGER
75
[27] Kitzmiller JL, Gavin LA, Gin GD, et al. Preconception care of diabetes: glycemic control
prevents congenital anomalies. JAMA 1991;265:7316.
[28] Pregnancy outcomes in the Diabetes Control and Complications Trial. Am J Obstet Gynecol 1996;174:134353.
[29] Langer O. Is normoglycemia the correct threshold to prevent complications in the pregnant
diabetic patient? Diabetes Rev 1996;4:210.
[30] Drexel H, Bicher A, Sailer S, et al. Prevention of perinatal morbidity by tight metabolic control in gestational diabetes mellitus. Diabetes Care 1988;11:7618.
[31] Roversi GD, Garguilo M, Nicolini U, et al. Maximal tolerated insulin therapy in gestational diabetes. Diabetes Care 1980;3:47984.
[32] Prospective Diabetes Study Group UK. Intensive blood glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients
with type 2 diabetes. Lancet 1998;352:83753.
[33] Langer N, Langer O. Gestational diabetes vs. pre-existing diabetes: comparison of pregnancy mood proles. Diabetic Educator 2000;26:66772.
[34] Langer O, Langer N. Is cultural diversity a factor in self-monitoring blood glucose in gestational diabetes? J Assoc Minority Physicians 1995;6:737.
[35] Langer N, Langer O. Emotional adjustment to diagnosis and intensied treatment of gestational diabetes. Obstet Gynecol 1994;84:32934.
[36] Garner P, Okun N, Keely E, et al. A randomized controlled trial of strict glycemic control
and tertiary level obstetric care in the management of gestational diabetes: a pilot study. Am
J Obstet Gynecol 1997;177:1905.
[37] Major CA, Henry MJ, De Veciana M, Morgan MA. The eects of carbohydrate restriction in patients with diet-controlled gestational diabetes. Obstet Gynecol 1998;91:
6004.
[38] Dornhorst A, Nicholls JSD, Probst F, et al. Calorie restriction for treatment of gestational
diabetes. Diabetes 1991;404(Suppl 2):1614.
[39] Artal R. Exercise: the alternative therapeutic intervention for gestational diabetes. Clin Obstet Gynecol 2003;46:47987.
[40] Owens DR, Zinman B, Bolli GB. Insulins today and beyond. Lancet 2001;358:73946.
[41] Hirsch I. Insulin analogues. N Engl J Med 2005;352:17483.
[42] Kurtzhals P, Schaer L, Sorensen A, et al. Correlations of reception binding and metabolic
and mitogen potentials of insulin analogues designed for clinical use. Diabetes 2000;49:
9991005.
[43] Holleman F, Hoekstra JBL. Insulin lispro. N Engl J Med 1997;337:17683.
[44] Kitzmiller JL, Main E, Ward B, et al. Insulin lispro and the development of proliferative
diabetic retinopathy during pregnancy [letter]. Diabetes Care 1999;22:8746.
[45] Jovanovic L, Ilic S, Pettitt D, et al. Metabolic and immunologic eects of insulin lispro in
gestational diabetes. Diabetes Care 1999;22:14227.
[46] Diamond T, Kormas N. Possible adverse fetal eect of insulin lispro [letter]. N Engl J Med
1997;337:100910.
[47] Bhattacharyya A, Vice P. Insulin lispro, pregnancy, and retinopathy. Diabetes Care 1999;
22:21012.
[48] Buchbinder A, Miodovnik M, McElvy S, et al. Is insulin lispro associated with the development or progression of diabetic retinopathy during pregnancy? Am J Obstet Gynecol
2000;183:11625.
[49] Bhattacharyya A, Brown S, Hughes S, Vice P. Insulin lispro and regular insulin in pregnancy.
QJM 2001;94:25560.
[50] Persson B, Swahn ML, Hjertberg R. Insulin lispro therapy in pregnancies complicated by
type 1 diabetes mellitus. Diabetes Res Clin Pract 2002;58:11521.
[51] Loukovaara S, Immonen I, Teramo KA, Kaaja R. Progression of retinopathy during
pregnancy in type 1 diabetic women treated with insulin lispro. Diabetes Care 2003;26:
11938.
76
LANGER
[52] Durand-Gonzalez KN, Guillausseau N, Anciaux ML, et al. Allergy to insulin in a woman
with gestational diabetes mellitus: transient eciency of continuous subcutaneous insulin
lispro infusion. Diabetes Metab 2003;29:4324.
[53] Garg S, Frias JP, Anil S, et al. Insulin lispro therapy in pregnancies complicated by type 1
diabetes: glycemic control and maternal and fetal outcomes. Endocr Pract 2003;9:18793.
[54] Mecacci F, Carignani L, Cioni R, et al. Maternal metabolic control and perinatal outcome
in women with gestational diabetes treated with regular or lispro insulin: comparison with
non-diabetic pregnant women. Eur J Obstet Gynaecol Reprod Biol 2003;111:1924.
[55] Carr K, Idama TO, Masson EA. A randomized controlled trial of insulin lispro given before
or after meals in pregnant women with type 1 diabetesdthe eect on glycaemic excursion.
J Obstet Gynaecol 2004;24:3826.
[56] Idama TO, Lindow SW, French M. Preliminary experience with the use of insulin lispro in
pregnant diabetic women. J Obstet Gynaecol 2001;21:3501.
[57] Cypryk K, Sobczak M, Pertynska-Marczewska M, et al. Pregnancy complications and perinatal outcome in diabetic women treated with Humalog (insulin lispro) or regular human
insulin during pregnancy. Med Sci Monit 2004;10:12932.
[58] Masson EA, Patmore JE, Brash PD, et al. Pregnancy outcome in Type 1 diabetes mellitus
treated with insulin lispro (Humalog). Diabet Med 2003;20:4650.
[59] Pettitt DJ, Opsina P, Kolaczynski JW, Jovanovic L. Comparison of an insulin analogue,
insulin aspart, and regular human insulin with no insulin gestational diabetes mellitus. Diabetes Care 2003;26:1836.
[60] Devlin JT, Hothersall L, Wilkins JL. Use of insulin glargine during pregnancy in a type 1
diabetic woman. Diabetes Care 2002;25:10956.
[61] Bauman WA, Yalow RS. Transplacental passage of insulin complexed to antibody. Proc
Natl Acad Sci U S A 1981;78:458890.
[62] Menon RK, Cohen RM, Sperling MA, et al. Transplacental passage of insulin in pregnant
women with insulin-dependent diabetes mellitus: its role in fetal macrosomia. N Engl J Med
1990;323:30915.
[63] Challier JC, Haugel S, Desmaizieres V. Eects of insulin on glucose uptake and metabolism
in the human placenta. J Clin Endocrinol Metab 1986;62:8037.
[64] Boskovic R, Feig DS, Derewlany L, et al. Transfer of insulin lispro across the human placenta. Diabetes Care 2003;26:13904.
[65] Reece EA, Homko C, Miodovnik M, Langer O. A consensus report of the diabetes in pregnancy study group of North America Conference. J Matern Fetal Neonat Med 2002;12:
3624.
[66] Langer O. Maternal glycemic criteria for insulin therapy in gestational diabetes mellitus.
Diabetes Care 1998;21(Suppl 2):B918.
[67] Langer O, Berkus MD, Brustman L, et al. Rationale for insulin management in gestational
diabetes mellitus. Diabetes 1991;40(Suppl2):18690.
[68] Langer O. Management of gestational diabetes. Clin Obstet Gynecol 1999;93:97882.
[69] Haroush AB, Yogev Y, Langer O, et al. Postprandial glucose prole characteristics in diabetic pregnancies. Am J Obstet Gynecol 2004;2:57681.
[70] Buchanan TA, Kjos SL, Montoro MN, et al. Use of fetal ultrasound to select metabolic
therapy for pregnancies complicated by mild gestational diabetes. Diabetes Care 1994;
17:27583.
[71] Schaefer-Graf U, Kjos S, Fauzan O, et al. A randomized trial evaluating a predominantly
fetal growth-based strategy to guide management of gestational diabetes in Caucasian
women. Diabetes Care 2004;27:297302.
[72] Kjos S, Schaefer-Graf U, Sardesi S, et al. A randomized controlled trial using glycemic plus
fetal ultrasound parameters versus glycemic parameters to determine insulin therapy in gestational diabetes with fasting hyperglycemia. Diabetes Care 2001;11:190410.
[73] Rosenn B, Langer O, Brustman L, et al. Its not just the abdominal circumference: glycemic
control still matters [abstract]. American Journal of Obstetrics and Gynecology 2004;189:165.
77
[74] McFarland MB, Langer O, Conway DL, Berkus M. Dietary therapy for gestational diabetes: how long is long enough? Obstet Gynecol 1999;93:97882.
[75] Langer O, Mazze RS. Diabetes in pregnancy: evaluating self-monitoring performance and
glycemic control with memory-based technology. Am J Obstet Gynecol 1986;155:6358.
[76] Langer O, Anyaegbunam A, Brustman L, et al. Gestational diabetes: Insulin requirements
in pregnancy. Am J Obstet Gynecol 1987;157:66975.
[77] DeWitt DE, Hirsch IB. Outpatient insulin therapy in type 1 and type 2 diabetes mellitus.
JAMA 2003;289:225464.
[78] Langer O, Conway DL, Berkus MD, et al. A comparison of glyburide and insulin in women
with gestational diabetes mellitus. N Engl J Med 2000;343:11348.
[79] Saade G. Gestational diabetes mellitus: a pill or a shot? [editorial] Obstet Gynecol 2005;105:
4567.
[80] Cefalo RC. A comparison of glyburide and insulin in women with gestational diabetes mellitus. Obstet Gynecol Surv 2001;56:1267.
[81] Gabbe SG, Graves CR. Management of diabetes mellitus complicating pregnancy. Obstet
Gynecol 2003;102:85768.
[82] Koren G. The use of glyburide in gestational diabetes: An ideal example of bench to bedside. Pediatr Res 2001;49:734.
[83] Ryan EA. Glyburide was as safe and eective as insulin in gestational diabetes. Evidence
Based Medicine 2001;6:79.
[84] Coustan DR. Oral hypoglycemic agents for the Ob/Gyn. Contemp Obstet Gynecol 2001;4563.
[85] Jovanovic L. The use of oral agents during pregnancy to treat gestational diabetes. Curr
Diab Rep 2001;1:6970.
[86] Piacquadio K, Hollingsworth DR, Murphy H. Eects of in-utero exposure to oral hypoglycemic drugs. Lancet 1991;338:8669.
[87] Kemball ML, McIver C, Milner RD, et al. Neonatal hypoglycemia in infants of diabetic
mothers given sulfonylurea drugs in pregnancy. Arch Dis Child 1970;45:696701.
[88] Towner D, Kjos SL, Leung B, et al. Congenital malformations in pregnancies complicated
by NIDDM. Diabetes Care 1995;18:144651.
[89] Gutzin S, Kozer E, Magee L, et al. The safety of oral hypoglycemic agents in the rst trimester of pregnancy: a meta-analysis. Can J Clin Pharmacol 2003;10:17983.
[90] Glueck CJ, Goldenberg N, Wang P, et al. Metformin during pregnancy reduces insulin, insulin resistance, insulin secretion, weight, testosterone and development of gestational diabetes: prospective longitudinal assessment of women with polycystic ovary syndrome
from preconception throughout pregnancy. Hum Reprod 2004;19:51021.
[91] Glueck CJ, Wang P, Kobayashi S, et al. Metformin therapy throughout pregnancy reduces
the development of gestational diabetes in women with polycystic ovary syndrome. Fertil
Steril 2002;77:5205.
[92] Jakubowicz DJ, Juorno MJ, Jakubowica S. Eects of metformin on early pregnancy loss in
the polycystic ovary syndrome. J Clin Endocrinol Metab 2002;87:5249.
[93] Lim JM, Tayob Y, OBrien PM, Shaw RW. A comparison between the pregnancy outcome
of women with gestation diabetes treated with glibenclamide and those treated with insulin.
Med J Malay 1997;52:37781.
[94] Conway DL, Gonzales O, Skiver D. Use of glyburide for the treatment of gestational diabetes: the San Antonio experience. J Matern Fetal Neonatal Med 2004;15:515.
[95] Kremer CJ, Du P. Glyburide for the treatment of gestational diabetes. Am J Obstet Gynecol 2004;190:14389.
[96] Chmait R, Dinise T, Moore T. Prospective observational study to establish predictors of
glyburide success in women with gestational diabetes mellitus. J Perinatol 2004;24(10):
61722.
[97] Gilson G, Murphy N. Comparison of oral glyburide with insulin for the management of
gestational diabetes mellitus in Alaskan native women. Am J Obstet Gynecol 2002;187:
S152.
78
LANGER
[98] Fines V, Moore T, Castle S. A comparison of glyburide and insulin treatment in gestational
diabetes mellitus on infant birth weight and adiposity. Am J Obstet Gynecol 2003;189:S108.
[99] Velazquez MD, Bolnick J, Cloakey D. The use of glyburide in the management of gestational diabetes. Obstet Gynecol 2003;101(Suppl):88S.
[100] Pendsey SP, Sharma RR, Chalkhore SS. Repaglinde: a feasible alternative to insulin in
management of gestational diabetes mellitus. Diabetes Res Clin Pract 2002;56(Suppl):S46.
[101] Coetzee EJ, Jackson WP. Oral hypoglycaemics in the rst trimester and fetal outcome.
S Afr Med J 1984;65:6357.
[102] Coetzee EJ, Jackson WP. The management of non-insulin-dependent diabetes during pregnancy. Diabetes Res Clin Pract 198586;1:2817.
[103] Coetzee EJ, Jackson WP. Metformin in management of pregnant insulin-independent diabetics. Diabetologia 1979;16:2415.
[104] Hellmuth E, Damm P, Molsted-Pedersen L. Oral hypoglycaemic agents in 118 diabetic
pregnancies. Diabet Med 2001;17:50711.
[105] Notelovitz M. Sulphonylurea therapy in the treatment of the pregnant diabetic. S Afr Med J
1971;45:2269.
[106] Yogev Y, Ben-Haroush A, Chen R, et al. Undiagnosed asymptomatic hypoglycemia: diet,
insulin, and glyburide for gestational diabetic pregnancy. Obstet Gynecol 2004;104:8893.
[107] Moore L, Briery C, Martin R, et al. Metformin vs. insulin in A2 diabetics: a randomized
clinical trial. Am J Obstet Gynecol 2004;191(Suppl):S8.
[108] Jacobson GF, Ramos GA, Ching JY, et al. Comparison of glyburide and insulin for the
management of gestational diabetes in a large managed care organization. Am J Obstet
Gynecol 2005;193(1):11824.
[109] Elliot B, Langer O, Schenker S, Johnson R. Insignicant transfer of glyburide occurs across
the human placenta. Am J Obstet Gynecol 1991;165:80712.
[110] Elliot B, Schenker S, Langer O, et al. Comparative placental transport of oral hypoglycemic
agents: a model of human placental drug transfer. Am J Obstet Gynecol 1994;171:65360.
[111] Elliot B, Langer O, Schussling F. A model of human placental drug transfer. Am J Obstet
Gynecol 1997;176:52730.
[112] Ng WW, Miller RK. Transport of nutrients in the early human placenta: amino acid, creatinine, vitamin B12. Trophoblast Res 1983;1:12133.
[113] Langer O, Yogev Y, Xenakis EMJ. Insulin and glyburide therapy: dosage, severity level of
gestational diabetes and pregnancy outcome. Am J Obstet Gynecol 2004;192:1349.
[114] Goetzel L, Wilkins I. Glyburide compared to insulin for the treatment of gestational diabetes mellitus: a cost analysis. J Perinatol 2002;22:4036.
[115] Feig DS, Briggs GG, Kraemer JM, et al. Transfer of glyburide and glipzide into breast milk.
Diabetes Care 2005;28:18515.
Using 2002 birth data [1], it is estimated that diabetes aects an estimated
8% of the more than 4 million pregnancies that come to term annually in the
United States. Identifying women who require aggressive monitoring and
treatment of their diabetes to minimize both maternal and fetal complications during and after pregnancy is a signicant challenge for physicians
and the health system because almost 75% of pregnancy-related diabetes occurs in women with gestational diabetes or undiagnosed type 2 diabetes.
Conversely, although type 1 diabetes is estimated to account for only 1%
to 2% of the pregnancies complicated by diabetes (w6000 births in the United
States annually), screening is not an issue because the diagnosis of type 1
diabetes is generally well established in a womans preconception years.
As recently as the 1980s, type 1 diabetes and pregnancy were a deadly combination for mothers and especially for fetuses, with rates of fetal perinatal
mortality as high as 25% to 30% [2]. Advances over the past 2 decades
in home glucose monitoring and insulin administration have provided
the technology needed to not only allow women to successfully survive pregnancy but also to decrease the risks of diabetic fetopathy to those of the
nondiabetic population [3,4].
Pregnancy in diabetic women is associated with an increase in risk to
both the fetus and the mother. Early in the pregnancy there is an emergency
* Corresponding author. Sansum Diabetes Research Institute, 2219 Bath Street, Santa
Barbara, CA 93105.
E-mail address: ljovanovic@sansum.org (L. Jovanovic).
0889-8529/06/$ - see front matter 2005 Elsevier Inc. All rights reserved.
doi:10.1016/j.ecl.2005.09.008
endo.theclinics.com
80
to normalize the blood glucose to prevent congenital anomalies and spontaneous abortions. As the pregnancy progresses, the mother is at an increased
risk for syncope, hypoglycemia, or diabetic ketoacidosis, all of which need
emergency attention. Later in the pregnancy, she is at risk for accelerated
retinopathy, with the risk of blindness, pregnancy-induced hypertension
and preeclampsia-eclampsia, urinary tract infections, including pyelonephritis, and polyhydramnios. The greatest concern is the increased risk of sudden death in utero for the fetus. All of these dreaded complications can
be obviated or at least minimized with careful planning of the pregnancy
and attention to glucose control.
The additional risks of pregnancy for mothers with type 1 diabetes and
their babies place the emphasis of optimizing maternal and fetal health of
these pregnancies and the focus of this article on (1) appropriate preconception counseling and management goals; (2) optimizing glycemic monitoring
and intensifying insulin therapy during the metabolically dynamic process of
pregnancy; (3) appropriately evaluating women and their fetuses for complications of both diabetes and intensive diabetes therapy during pregnancy;
and (4) peripartum and postpartum glycemic control.
Congenital anomalies
There is an increased prevalence of congenital anomalies and spontaneous abortions in diabetic women who are in poor glycemic control during
the period of fetal organogenesis, which is nearly complete at 7 weeks postconception [5]. Thus, a woman may not even know she is pregnant at this
time. For this reason, prepregnancy counseling and planning are essential
in women of childbearing age who have diabetes. Because organogenesis
is complete so early in the fetus development, if a woman presents to her
health care team and announces that she has missed her period by only
a few days, if the blood glucose levels are normalized immediately [610],
there is still is a chance to prevent cardiac anomalies by swiftly normalizing
the glucose levels (although the neural tube defects are probably already set
by the time the period is missed).
Women with type 2 diabetes are less likely to have preconception care
and counseling, often because the diabetes has not yet been diagnosed,
and thus, they are at even greater risk of bearing a birth-defective child.
In one study [11], 40% of women with type 1 diabetes but only 14% with
type 2 diabetes received preconception care.
Another problem for some women with type 2 diabetes is gaining access
to adequate medical care before and after conception. Nowhere is this more
evident than in Hispanic women, in whom the higher prevalence of obesity,
type 2 and gestational diabetes, and increased fertility rates place them at
higher risk for maternal and neonatal complications [1214]. Pima Indian
women are also at increased risk; they have a 19-fold higher incidence of
81
type 2 diabetes compared with the general United States population, with an
age-adjusted prevalence rate approximately eight times higher [15,16].
A measurement of glycosylated hemoglobin A1c (HbA1c) can, in early
pregnancy, estimate the level of glycemic control during the period of fetal
organogenesis. There are two important observations in this regard: rst,
HbA1c values early in pregnancy are correlated with the rates of spontaneous abortion and major congenital malformations [1719]. Although most
studies have been performed in women with type 1 diabetes, the same risk
of hyperglycemia applies to those with type 2 diabetes [20,21]; and second,
normalizing blood glucose concentrations before and early in the pregnancy
can reduce the risks of spontaneous abortion and congenital malformations
nearly to that of the general population [22,23].
One report compared 110 women who were 6 to 30 weeks pregnant at
the time of referral with 84 women who were recruited before conception
and then put on a daily glucose-monitoring regimen [19]. The mean blood
glucose concentration was between 60 and 140 mg/dL (3.3 mmol/L and 7.8
mmol/L) in 50% of the latter group of women. The incidence of anomalies
was 1.2% in the women recruited before conception versus 10.9% in those
rst seen during pregnancy. Very similar ndings were noted in another
study: 1.4% versus 10.4% incidence of congenital abnormalities [23]. Major congenital malformations, which either require surgical correction or
signicantly aect the health of the child, are more common in infants of diabetic mothers (13% versus 2% in infants of nondiabetic mothers) [24].
The increased rate of spontaneous abortion in poorly controlled diabetic
women is believed to be secondary to hyperglycemia, maternal vascular disease, including uteroplacental insuciency, and possibly immunologic factors [25]. In addition, animal studies suggest that hyperglycemia regulates
the expression of an apoptosis (programmed cell death) regulatory gene as
early as the preimplantation blastocyst stage, resulting in increased DNA
fragmentation [26]. These ndings emphasize the importance of glycemic
control at the earliest stages of conception.
Ideally, if a diabetic woman plans her pregnancy there is time to create
algorithms of care that are individualized, and a woman can be given
choices. When a diabetic woman presents in her rst few weeks of pregnancy, there is no time for individualization; rather, rigid protocols must
substituted urgently to provide optimal control within 24 to 48 hours.
Deleterious eects of strict glycemic control
Despite the clear benets to the fetus of strict glycemic control, there
is a hazard of hypoglycemia. Major complications of hypoglycemia can usually be prevented with careful monitoring and education of the mother
[7,23,27]. Very strict glycemic control (mean blood glucose % 56 mg/dL)
may be deleterious to the fetus and should be avoided. In one study, the
most common cause of maternal mortality was related to hypoglycemia [28].
82
Retinopathy
There are three reported situations in which the rapid normalization of
blood glucose level increases the risk for the deterioration of diabetic retinopathy: puberty [29], pregnancy [30], and insulin-like growth factor
(IGF)-1 treatment [31]. If two of these events occur in the same patient,
the risk for retinopathy progression is potentiated [30,32]. All three situations are associated with increased serum concentrations of growth-promoting factors. It is hypothesized that when the blood glucose level is rapidly
decreased, there is increased retinal extravasation of serum proteins. If there
is a concomitant increase in the concentration of serum growth-promoting
factors, a predisposed retina may deteriorate [3335].
Pregnancy per se is the condition most frequently reported in which the
rapid normalization of blood glucose is associated with retinal deterioration
[7,32, 3436]. Normal pregnancy is associated with a high concentration of
many growth-promoting factors [3638]. Hill and colleagues [33] have reported that a potent mitogenic and angiogenic factor normally absent
from the adult circulation becomes detectable by 14 weeks of gestation
and is maximal at 22 to 32 weeks of gestation. A placental growth hormone
variant had been found to increase throughout pregnancy, along with the
human somatomammotropin prolactin [36]. Maternal IGF-1 production
has also been shown to increase signicantly above nonpregnant levels
[37]. It is well known that diabetes mellitus is associated with perturbations
of growth hormone IGF-1 in cases of poor metabolic control [38].
If treatment with lispro insulin, a short-acting insulin analog, allows for
increasing the rate whereby the hyperglycemic state is normalized, then lispro insulin may play a role in causing the rapid deterioration of retinopathy.
It is unlikely, in the few case reports of rapid deterioration of retinopathy in
patients taking lispro insulin, that deterioration was caused by the IGF-1 activity of this insulin, because pregnancy alone has such elevated IGF-1
levels.
Human insulin binds to the IGF-1 receptor with an anity of 0.1% to
0.2% of the anity of IGF-1. A comparison of lispro and human insulin
was made to determine the relative IGF-1 receptor binding anity in human
placental membranes, skeletal muscle, smooth muscle cells, and mammary
epithelial cells. Lispro had a slightly higher anity for the human placental
membranes compared with human insulin. No other dierences were observed in any other cell lines. Despite the suggested increased anity, it
should be noted that the absolute anity for the IGF-1 receptor is extremely
low for both lispro and human insulin. Concentrations more that 1000 times
above normal physiologic range are needed to reach a receptor binding afnity of 50%. IGF-1 is a much larger protein chain than insulin, and there is
a 49% homology between human insulin and IGF-1. The reversal of the
Asx28 and Asx29 amino aids in lispro increases this homology to 51% because of the analogous position in the IGF-1 molecule. It has been shown
83
that insulin lispro has the same anity for the IGF-1 receptor as human insulin and that the dissociation kinetics of insulin lispro on the insulin receptor are identical to those of insulin, indicating that insulin lispro should have
no excess mitogenic eect through the IGF-1 or insulin receptor [39,40].
Phelps and colleagues [32] have clearly shown that the deterioration of
retinopathy correlated signicantly with the levels of plasma glucose at entry
and with the magnitude of improvement in glycemia during the rst 6 to 14
weeks after entry, although the 13 patients with no retinopathy at baseline
did not progress to proliferative retinopathy. However, one of the women
developed moderate hemorrhages, exudates, and intraretinal microaneurysms. Of their 20 patients with an initial background of retinopathy, two patients progressed to proliferative retinopathy. Laatikainen and colleagues
[41] conrmed that the decrease in hemoglobin HbA1c levels was the most
rapid in the two patients with the worst progression. They concluded that
a rapid near-normalization of glycemic control during pregnancy can accelerate the progression of retinopathy in poorly controlled diabetic patients.
The Diabetes in Early Pregnancy (DIEP) study [42] reported the results of
155 type 1 diabetic women who underwent retinal angiography in the rst
few weeks of gestation and then at term (within 1 week before delivery).
In the 140 patients who did not have proliferative retinopathy at baseline,
the progression of retinopathy was seen in 10.3%, 21.1%, 18.8%, and
54.8% of patients with no retinopathy, microaneurysms only, mild nonproliferative retinopathy, and moderate to severe nonproliferative retinopathy
at baseline, respectively. Proliferative retinopathy developed in 6.3% of patients with mild and 29% of patients with moderate to severe baseline retinopathy. The elevated glycosylated hemoglobin at baseline and the
magnitude of improvement of glucose control through week 14 were associated with a higher risk of progression of retinopathy (the adjusted odds ratio
[OR] for progression in those with a glycohemoglobin level R 6 standard
deviations [SD] above the control mean versus those ! 2 SD was 2.7;
95% condence interval [CI], 1.1%7.2%; P 0.039). Independent of retinal status, the DIEP study also reported that the duration of diabetes increased the risk of progression such that after 6 years duration of
diabetes the OR was 3.0 (95% CI, 0.5%17.4%); after 11 to 15 years, the
OR was 9.7 (95% CI, 1.9%49.0%); and after more than 16 years, the
OR was 15.0 (95% CI, 3.0%74.5%); however, hyperglycemia was a stronger risk factor. Additional evidence has been reported by the Diabetes Control and Complications Trial [30]. For women in the conventional care
group who became pregnant and thus had immediate intensication of glucose control (n 135), the retinal status worsened in 47% of the patients,
and the OR for progression by the second trimester was 2.6, compared
with diabetic women in the conventional group who did not become
pregnant.
There is one case report in the literature, which clearly shows that the
combination of pregnancy and rapid normalization of severe hyperglycemia
84
85
suggestion that screening pregnant women for hypothyroidism by measuring thyrotropin may be worthwhile and that treating women with serum
thyrotropin concentrations at or above the 98th percentile could lead to
an increase of approximately 4 points in IQ scores in their children. Because there is an increased risk of gestational hypothyroidism in diabetic
women, all diabetic pregnant women should be screened for hypothyroidism
early in their pregnancy and treated immediately if hypothyroidism is documented [51]. Notably, Graves disease is not more common in type 1 diabetic
women than in the general population [46].
Diabetic ketoacidosis
Diabetic ketoacidosis, a complication associated with a high mortality
rate in the fetus, may occur. In addition, ketonemia during pregnancy has
been associated with decreased intelligence in ospring [5254], but in these
reports there is no mention of an association with fetal malformations. In
early pregnancy, ketonuria sometimes occurs in women who are limiting
their caloric intake because of nutritional recommendations [55]. Ketonuria
resulting from caloric restriction has been shown subsequently not to be associated with decreased intelligence in the ospring, unless it is also associated with severe hyperglycemia (blood glucose levels O 180 mg/dL). Thus,
women with moderate to large ketonuria associated with hyperglycemia
should immediately alert their physician. Also, if a type 1 diabetic woman
is using an insulin infusion pump [53], even short periods of interruption
in the infusion may result in ketoacidosis caused by the increased metabolic
rate in pregnancy. Thus, many centers are recommending the use of an additional injection of neutral protamine Hagedorn (NPH) insulin at bedtime,
in a volume that is 0.1 times the patients weight in kg, for an appropriate
adjustment in the overnight basal metabolism [27,55].
Pregnancy-induced hypertension
Normal rst trimester blood pressure is less than 120/80 mmHg [56]. Fetal complications from maternal hypertension include intrauterine growth
retardation and fetal demise. The only antihypertensive mediations that
have been proven to be safe over the past two generations are methyldopa
and hydralazine. When additional medication is needed to maintain normotension, the use of labetolol has been recommended, along with the judicious
use of calcium channel blockers and even diuretics. Nifedipine has also been
reported to be safe when added after the rst trimester. The use of angiotensin converting enzyme (ACE) inhibitors in pregnancy have been associated
with congenital anomalies, specically renal agenesis or renal failure, and
thus, they are absolutely contraindicated in pregnancy [57]. However,
Hod and colleagues [57] have shown that when ACE inhibitors are used
86
before conception, they decrease the degree of proteinuria, and when they
are discontinued at the time of conception, the decreased proteinuria is sustained throughout pregnancy, with an improved maternal and fetal outcome. Although the management of elevated blood pressure in a diabetic
pregnant woman is an urgent priority, the means to achieve these goals
are limited in pregnancy.
Diabetic nephropathy
Diabetic nephropathy, when not associated with hypertension, does not
have an impact on fetal outcome unless the kidney function is more than
50% impaired. Normal creatinine clearance is increased in pregnancy because of the increased metabolic rate and the increased cardiac output by
the 10th to 12th week of gestation. Thus, a depression of the creatinine
clearance below 50 mL/min is associated with increased fetal loss. Proteinuria greater than 250 mg/dL in the rst trimester has been associated with
nephrotic syndrome by the third trimester, requiring bed rest and in some
cases replacement of protein losses with parenteral supplementation of albumin [25]. Maternal anasarca is not associated with fetal hypdrops, as long as
the maternal nutrition includes enough protein for fetal growth [55].
87
must be monitored with treatment goals that are lower than those that are
used outside of pregnancy, to goals of 60 to 90 mg/dL. Understanding these
nondiabetic pregnancy, normal glucose ranges provides additional support to
the previous observation that the macrosomia risk increases with increasing
maximal postprandial hyperglycemia R 120 mg/dL [4]. While striving to
achieve normoglycemia in a type 1 diabetic womans pregnancy, fasting, preprandial, and 1-hour postprandial SMBG testing [4,63] must be carried out to
dose insulin safely yet meet the fasting-preprandial glucose goals of 60 mg/dL
to 90 mg/dL and 1-hour postprandial glucose goals of 100 mg/dL to 120 mg/
dL [64].
Because of the high frequency of SMBG testing required in pregnancy,
the use of alternative site SMBG testing is appealing, but the dynamically
changing blood glucose concentrations after eating may be identied at nger sites before they are detected at forearm or thigh sites [65]. Because no
studies have evaluated the use of blood glucose values from alternative sites
in pregnancy, alternative site SMBG testing must be discouraged. The eventual availability of real-time continuous glucose monitoring sensors holds
the promise of not only signicantly reducing the number of nger-stick
blood glucose assessments but also raising the awareness of glycemic excursions [66,67], which can occur commonly, particularly in women with type 1
diabetes, between SMBG measurements. At the time of this writing, no such
system is available currently for commercial use.
Glycosylated hemoglobin testing
Until real-time continuous glucose monitoring systems are in routine
clinical use to accurately assess 24-hour glycemia, frequent HbA1c monitoring will remain valuable as an assessment of blood glucose excursions occurring at non-SMBG time points. Whereas the HbA1c testing frequency in a
nonpregnant population is commonly timed to assess a new steady state of
glycemic control, during the compressed time frame of pregnancy (in which
2 weeks represents 5% of a normal gestation), the need to recognize and
treat worsening glycemic control as promptly as possible can be facilitated
by examining HbA1c changes because they reect trends in glycemic control.
Because clinically and statistically signicant dierences in HbA1c can be detected at 2-week intervals [68,69], HbA1c assessment every 2 weeks during
gestation is recommended.
Nondiabetic pregnant women evaluated by continuous glucose monitoring systems maintain mean blood glucose levels of or less than 100 mg/dL
[60], and treating diabetic pregnant women to this mean glucose goal signicantly decreases perinatal mortality [70]. Knowing that a mean blood
glucose goal of 100 mg/dL correlates with an HbA1c goal of 5% [71], discordant SMBG and HbA1c results should prompt investigation because poor
control, undetected by the patients current SMBG routine, is assumed. Additionally, when using HbA1c measurement as an indicator of glycemic
88
Treatment
There are several components to the treatment of diabetes in pregnant
women, the administration of insulin, exercise, and diet.
Insulin doses
The starting insulin dose is calculated to be 0.7 U/kg/d, divided into three
to four injections of short- and intermediate-acting insulin. Thecapillary
glucose needs to be measured before and 1 hour after each meal, at bedtime, and at 3 AM to assure that there is around-the-clock glucose control.
Each day the insulin is adjusted based on the blood glucose measurements
such that the optimal dose of insulin is derived on a daily basis. Only human
89
insulin should be used in pregnant women. Most women with type 1 diabetes require at least three injections per day [41,45,48]. A two-injection regimen can cause nocturnal hypoglycemia if the evening meal dose of
intermediate-acting insulin peaks during the middle of the night [27].
Total daily insulin requirements typically rise during gestation. This is
caused primarily by the eects of human placental lactogen, which has somatotropic properties. The average insulin requirement in pregnant women
with type 1 diabetes rises from 0.7 U/kg/d in the rst trimester, often increasing to 0.8 U/kg/d for weeks 18 to 26, 0.9 U/kg/d for weeks 26 to 36,
and 1.0 U/kg/d for weeks 36 to term. Massively obese women may need
initial doses of 1.5 U/kg/d to 2.0 U/kg/d to overcome the combined insulin
resistance of pregnancy and obesity [54]. Declining insulin requirements
may occur at 9 to 12 weeks of gestation and again at 38 to 40 weeks of
gestation [27]. These changes in insulin requirement are thought to be
caused by the luteal-placental shift in progesterone seen in the late rst trimester and to the onset of labor, with increased glucose use that occurs
when uterine contractions commence.
The total daily dose is divided such that 50% is given as a basal dose, using an insulin infusion pump or NPH insulin, divided into two to three daily
injections spaced 8 to 12 hours apart. The meal-related dose then is the other
50% of the insulin requirement. Recent reports have shown that insulin lispro is safe in pregnancies complicated by type 1 diabetes [77]. However, the
studies using insulin aspart and the long-acting insulin analogs detemir and
glargine have not yet been published.
Some clinicians have recommended using insulin pumps to achieve optimal
glycemic control during pregnancy [78,79]. Most pregnant women require at
least three infusion rates in a 24-hour period, in particular a low-dose basal
from 12 midnight to 4 AM, an increased rate in the early morning hours,
from 4 AM to 10 AM, to counteract the increased release of the anti-insulin hormones cortisol and growth hormone, and an intermediate basal during the
rest of the day, from 10 AM to 12 midnight. A protocol for calculating infusion
rates has been described elsewhere [80].
Role of exercise
Gestational diabetes diers from a pregnancy in type 1 diabetes because
the former is a disorder primarily of impaired glucose clearance. As a result,
therapies that overcome peripheral resistance to insulin, such as exercise, are
preferable to insulin. In comparison, in women with type 1 diabetes who are
already taking insulin, the benets of exercise are not so clear. Exercise can
contribute to the brittleness of diabetes, with the risk of exercise-induced
hypoglycemia. Women who exercised before pregnancy can usually continue under the supervision of their obstetrician. However, exercise is not recommended in women who are deconditioned and did not exercise before
pregnancy [80].
90
Diet
The optimal diet takes into account caloric intake, carbohydrate content,
and the distribution of meals throughout the day. The appropriate caloric
intake depends on the pregravid weight, with the following general
recommendations:
Fetal surveillance
The high perinatal mortality once associated with a diabetic pregnancy
has decreased signicantly, largely caused by improved glycemic control
[81]. In the past, unexplained fetal death occurred in 10% to 30% of type
1 diabetic pregnancies, typically after the 36th week of gestation in women
with poor glycemic control, associated with macrosomia, hydramnios, preeclampsia, and vascular disease. Fetal surveillance, therefore, is of utmost
importance in optimizing a good outcome for both mother and fetus, especially in the perilous third trimester. Ultrasonography is the most useful tool
for the assessment of the fetus. It can be used to (1) estimate gestational age;
(2) screen for structural anomalies; (3) evaluate growth; (4) assess amniotic
uid volume; and (5) determine fetal status dynamically through Doppler
and biophysical studies.
Ultrasonographic estimates of gestational age are most accurate if they
are performed in early pregnancy. The gestational age, determined by
a crownrump length measured in the rst trimester, will be accurate within 5
days. Ultrasonographic estimates of gestational age are not reliable after the
28th week and cannot be used to determine the estimated date of delivery
[82].
Macrosomia is more apparent in some fetal structures, such as the liver
and abdomen. As a result, ultrasonographic estimates that use the fetal
91
92
fetal testing should be performed twice per week. Doppler umbilical artery
velocimetry has shown increased placental resistance in women with vasculopathy and poor glycemic control, which increase the risk of intrauterine
growth retardation and preeclampsia [82,88].
Postpartum
Insulin requirements drop sharply after delivery, and the new mother may
not require insulin for 24 to 72 hours. Insulin requirements should be recalculated at this time, at approximately 0.6 U/kg/d, based on postpartum weight.
Postpartum caloric requirements are approximately 25 kcal/kg/d and somewhat higher (27 kcal/kg/d) in lactating women.
Glycemic control is somewhat more erratic in lactating diabetic women,
with more frequent episodes of hypoglycemia. Because the risk of lifethreatening hypoglycemia is increased in the immediate postpartum period,
93
94
[9] Miller E, Hare JW, Cloherty JP, et al. Elevated maternal hemoglobin A1C in early pregnancy
and major congenital anomalies in infants of diabetic mothers. N Engl J Med 1981;304(22):
13314.
[10] Fuhrmann K, Ruher H, Semmler K, et al. Prevention of congenital malformations in infants
of insulin dependent diabetic mothers. Diabetes Care 1983;6:219.
[11] Janz NK, Herman WH, Becker MP, et al. Diabetes and pregnancy: factors associated with
seeking pre-conception care. Diabetes Care 1995;18(2):15765.
[12] Forsbach G, Contreras-Soto JJ, Fong G. Prevalence of gestational diabetes and macrosomic
infants in Mexican population. Diabetes Care 1988;11:235.
[13] Hollingsworth DR, Vaucher Y, Yamamoto TR. Diabetes in pregnancy in Mexican Americans. Diabetes Care 1991;14:695.
[14] Mestman JH. Outcome of diabetes screening in pregnancy and perinatal morbidity in infants
of mothers with mild impairment in glucose tolerance. Diabetes Care 1980;3:447.
[15] Knowler WC, Bennett PH, Hamman RF. Diabetes incidence and prevalence in Pima Indians: a 19-fold greater incidence than in Rochester, Minnesota. Am J Epidemiol 1978;108:
497.
[16] Knowler WC, Pettit DJ, Saad MF. Diabetes mellitus in Pima Indians: incidence, risk factors,
and pathogenesis. Diabet Metab Rev 1990;6:1.
[17] Damm P, Molsted-Pederson L. Signicant decrease in congenital malformations in newborn
infants of an unselected population of diabetic women. Am J Obstet Gynecol 1989;161:1163.
[18] Greene MF, Hare JW, Cloherty JP, et al. First trimester hemoglobin A1C and risk for major
malformation and spontaneous abortion in diabetic pregnancy. Teratology 1989;39:225.
[19] Ylinen K, Aula P, Stenman UH, et al. Risk of minor and major fetal malformations in diabetics with high haemoglobin A1C values in early pregnancy. BMJ 1984;289(6441):3456.
[20] Becerra JE, Khoury MJ, Cordero JF. Diabetes mellitus during pregnancy and risks for specic birth defects: a population-based case-control study. Pediatrics 1990;85:1.
[21] Towner D, Kjos SL, Leung B. Congenital malformations in pregnancies complicated by
NIDDM. Diabetes Care 1995;18:1446.
[22] Fuhrmann K, Reiher H, Semmler K, et al. The eect of intensied conventional insulin therapy before and during pregnancy on the malformation rate in ospring of diabetic mothers.
Exp Clin Endocrinol 1984;83:173.
[23] Steel JM, Johnstone FD, Hepburn DA, et al. Can prepregnancy care of diabetic women reduce the risk of abnormal babies? BMJ 1990;301:1070.
[24] Cousins L. Etiology and prevention of congenital anomalies among infants of overt diabetic
women. Clin Obstet Gynecol 1991;34:481.
[25] Kitzmiller JL, Watt N, Driscoll SG. Decidual arteriopathy in hypertension and diabetes in
pregnancy and immunouorescent studies. Am J Obstet Gynecol 1981;141:773.
[26] Moley KH, Chi MM, Knudson CM, et al. Hyperglycemia induces apoptosis in preimplantation embryos through cell death eector pathways. Nat Med 1998;4:1421.
[27] Jovanovic L, Mills Jl, Knopp RH, et al, for the National Institute of Child Health and Human Development-Diabetes in Early Pregnancy Study Group. Declining insulin requirement in the late rst trimester of diabetic pregnancy. Diabetes Care 2001;24:11306.
[28] Leinonen PJ, Hiilesmaa VK, Kaaja RJ, et al. Maternal mortality in type 1 diabetes. Diabetes
Care 2001;24:15012.
[29] Daneman D, Drash AL, Lobes LA. Progressive retinopathy with improved control in diabetic dwarsm (Mauriacs syndrome). Diabetes Care 1981;4:3604.
[30] Lachin J, Clearly P, Molitch M, et al, for the DCCT Research Group. Pregnancy increases
the risk of complication in the DCCT. Diabetes 1998;47(Suppl 1):S1091.
[31] Van Ballegooie E, Hooymans JMM, Timerman Z. Rapid deterioration of diabetic retinopathy during treatment with continuous subcutaneous insulin infusion. Diabetes Care 1984;7:
23641.
[32] Phelps RL, Sakol L, Metzger BE, et al. Changes in diabetic retinopathy during pregnancy:
correlations with regulation of hyperglycemia. Arch Ophthalmol 1986;104:180610.
95
[33] Hill DJ, Clemmons DR, Riley SC, et al. Immunohistochemical localization of insulin like
growth factors and IGF binding proteins-1,-2, and -3 in human placenta and fetal membranes. Placenta 1993;14:112.
[34] Larinkari J, Laatikainen L, Ranta T. Metabolic control and serum hormone levels in relationship to retinopathy in diabetic pregnancy. Diabetologia 1982;22:32731.
[35] Merimee TJ, Zapf J, Froesch ER. Insulin-like growth factors: studies in diabetics with and
without retinopathy. N Engl J Med 1983;309:52731.
[36] MacLeod JN, Worsley I, Ray Y, et al. Human growth hormone variant is a biologically active somatogen and lactogen. Endocrinology 1991;128:1298302.
[37] Gluckman PD. The endocrine regulation of fetal growth in late gestation: the role of insulinlike growth factors. T J Clin Endocinol Metab 1995;80:104750.
[38] Holly JMP, Amiel SA, Sandhu RR, et al. The role of growth hormone in diabetes mellitus.
J Endocrinol 1988;118:35364.
[39] DiMarchi RD, Chance RE, Long HB, et al. Preparation of an insulin with improved pharmacokinetics relative to human insulin through consideration of structural homology with
insulin-like growth factor-1. Horm Res 1994;41(Suppl 2):S936.
[40] Llewelyn J, Slieker LJ, Zimmermann JL. Pre-clinical studies on insulin lispro. Drugs Today
(Barc) 1998;34(Suppl C):C1121.
[41] Laatikainen L, Teramo K, Hieta-Heikurainen H, et al. A controlled study of he inuence of
continuous subcutaneous insulin infusion treatment on diabetic retinopathy during pregnancy. Acta Medica Scandinavica 1987;221:36776.
[42] Chew EY, Mills JL, Metzger BE, et al, for the National Institute of Child Health and Human
Development-Diabetes in Early Pregnancy Study. Metabolic control and progression of retinopathy: The Diabetes In Early Pregnancy Study. Diabetes Care 1995;18:6317.
[43] Hagay ZJ, Schachter M, Pollack A, et al. Case report: development of proliferative retinopathy in a gestational diabetes patient following rapid metabolic control. Eur J Obstet Gynecol Reprod Biol 1994;57:2113.
[44] Anderson JH, Brunelle RL, Koivisto VA. Reduction of postprandial hyperglycemia and frequency of hypoglycemia in IDDM patients on insulin-analog treatment. Diabetes 1997;46:
26570.
[45] Jovanovic L, Ilic S, Pettitt DJ, et al. The metabolic and immunologic eects of insulin lispro
in gestational diabetes. Diabetes Care 1999;22:14227.
[46] Betterle C, Zanette F, Pedini B, et al. Clinical and subclinical organ-specic autoimmune
manifestations in Type 1 (insulin-dependent) diabetic patients and their rst-degree relatives.
Diabetologia 1989;26:4316.
[47] Bech K, Hoier-Madsen M, Feldt-Rasmussen U, et al. Thyroid function and autoimmune
manifestations in insulin-dependent diabetes mellitus during and after pregnancy. Acta Endocrinol (Copenh) 1991;124:5349.
[48] Jovanovic L, Peterson CM. De novo clinical hypothyroidism in pregnancies complicated by
type 1 diabetes, subclinical hypothyroidism, and proteinuria: a new syndrome. Am J Obstet
Gynecol 1988;159:4426.
[49] Alvarez-Marfany M, Roman SH, Drexler Aj, et al. Long-term prospective study of postpartum thyroid dysfunction in women with insulin dependent diabetes mellitus. J Clin Endocrinol Metab 1994;79:106.
[50] Gerstein HC. Incidence of postpartum thyroid dysfunction in patients with type I diabetes
mellitus. Ann Intern Med 1993;118:41923.
[51] Haddow JE, Palomaki GE, Allan WC, et al. Maternal thyroid deciency during pregnancy
and subsequent neuropsychological development of the child. N Engl J Med 1999;341:
54955.
[52] Rizzo T, Metzger BE, Nurns WJ. Correlations between antepartum maternal metabolism
and intelligence of ospring. N Engl J Med 1991;325:911.
[53] Jovanovic L, Metzger BE, Knopp RH, et al. The Diabetes in Early Pregnancy Study: betahydroxybutyrate levels in type 1 diabetic pregnancy compared with normal pregnancy.
96
[54]
[55]
[56]
[57]
[58]
[59]
[60]
[61]
[62]
[63]
[64]
[65]
[66]
[67]
[68]
[69]
[70]
[71]
[72]
[73]
[74]
[75]
97
[76] Chez RA, Curcio FD III. Ketonuria in normal pregnancy. Obstet Gynecol 1987;69:272.
[77] Wyatt JW, Frias JL, Hoyme HE, et al, for the IONS study group. Congenital anomaly rate in
ospring of pre-gestational diabetic women treated with insulin lispro during pregnancy.
Diabet Med 2004;22(6):8037.
[78] Coustan DR, Reece EA, Sherwin RS, et al. A randomized clinical trial of insulin pump vs.
intensive conventional therapy in diabetic pregnancies. JAMA 1986;255:631.
[79] Rudolf MC, Coustan DR, Sherwin RS, et al. Ecacy of the insulin pump in the home treatment of pregnant patients. Diabetes 1981;30:891.
[80] Bornstein K, Jovanovic L. Type 1 diabetic woman and safe insulin protocols. In: Ruderman
N, editor. ADA handbook on exercise in diabetes. Alexandria (VA): American Diabetes Association. Inc; 2002. pp. 51132.
[81] Kitzmiller JL, Cloherty JP, Younger MD, et al. Diabetic pregnancy and perinatal morbidity.
Am J Obstet Gynecol 1978;131(5):56080.
[82] Landon MB, Gabbe SG, Brunner JP, et al. Doppler umbilical artery velocimetry in pregnancy
complicated by insulin-dependent diabetes mellitus. Obstet Gynecol 1989;73:961.
[83] Benedetti TJ, Gabbe SG. Shoulder dystocia: a complication of fetal macrosomia and prolonged second stage of labor with mid-pelvic delivery. Obstet Gynecol 1978;52:526.
[84] Golditch IM, Kirkman K. The large fetus: management and outcome. Obstet Gynecol 1978;
52:26.
[85] Shepard MJ, Richards VA. An evaluation of two equations for predicting fetal weight by ultrasound. Am J Obstet Gynecol 1982;142:47.
[86] Milunsky A. Prenatal diagnosis of neural tube defects: VIII. the importance of serum alphafetoprotein screening in diabetic pregnant women. Am J Obstet Gynecol 1982;142:1030.
[87] Reece EA, Hobbins JC. Ultrasonography and diabetes mellitus in pregnancy. In: Sanders
RG, editor. The principles and practice of ultrasonography in obstetrics and gynecology.
3rd edition. Norwalk (CT): Appleton-Century-Crofts; 1985. p. 297.
[88] Bracero L, Schulman H, Fleischer A. Umbilical artery velocimetry in diabetes and pregnancy.
Obstet Gynecol 1986;68:654.
[89] Jovanovic L, Peterson CM. Insulin and glucose requirements during the rst stage of labor in
insulin-dependent diabetic women. Am J Med 1983;75:607.
[90] Jovanovic L. Glucose and insulin requirements during labor and delivery: the case for normoglycemia in pregnancies complicated by diabetes. Endocr Pract 2004;10:405.
[91] Jovanovic L, Peterson CM. Maternal milk and plasma glucose and insulin levels: studies in
normal and diabetic subjects. J Am Coll Nutr 1989;8:12531.
100
MOLITCH
Prolactinoma
Hyperprolactinemia is responsible for about one third of all cases of female
infertility [17]. Hyperprolactinemia impairs the hypothalamic-pituitaryovarian axis at several levels, the primary site of inhibition being at the
hypothalamus, where it inhibits the pulsatile secretion of GnRH [18]. The
dierential diagnosis of hyperprolactinemia is extensive [18].
For patients with prolactinomas, the choice of therapy may have important consequences for decisions regarding pregnancy. Transsphenoidal surgery is curative in 50% to 60% of cases and rarely causes hypopituitarism
when it is performed on women with microadenomas. For patients with
101
102
MOLITCH
Prior therapy
No. of patients
Symptomatic
enlargementa
Microadenomas
Macroadenomas
Macroadenomas
None
None
Yes
376
86
71
6 (1.3%)
20 (23.2%)
2 (2.8%)
103
Table 2
Eect of bromocriptine on pregnancies
Bromocriptine
Criteria
No.
Pregnancies
Spontaneous abortion
Termination
Ectopic
Hydatidiform moles
6239
620
75
31
11
100.0
9.9
1.2
0.5
0.2
100.0
10.015.0
d
0.51.0
0.050.7
4139
3620
519
100.0
87.5
12.5
100.0
85.0
15.0
5120
5031
89
100.0
9.3
1.7
100.0
8.7
1.7
5213
5030
93
90
100.0
96.5
1.8
1.7
100.0
95.0
3.04.0
O2.0
Data from Krupp P, Monka C, Richter K. The safety aspects of infertility treatments. In:
Program of the Second World Congress of Gynecology and Obstetrics. Rio de Janeiro, Brazil,
August 1988. p. 9.
throughout gestation is limited to only slightly more than 100 women, but
no abnormalities were noted in the infants except one with an undescended
testicle and one with a talipes deformity [32]. Because bromocriptine crosses
the placenta [33], however, it should not be used any longer than necessary
during pregnancy.
Pergolide has been shown to cross the placenta in mice, but no teratogenicity was seen in doses of 60 mg/kg/d [34]. Detailed data are available on
the safety during early gestation for only one patient treated with pergolide
for Parkinsons disease [35]. In this pregnancy, no teratogenicity or developmental abnormalities were found in the child, but the authors stated in this
report that, in premarketing studies of pergolide for endocrine disorders,
two major and three minor congenital abnormalities were described among
38 pregnancies, but a causal relationship has not been established [35].
Other information from the manufacturer (Eli Lilly & Co) stated that
they had only limited data on pregnancies in which the fetus was exposed
to pergolide, nding that 7.2% of pregnancy outcomes resulted in spontaneous abortions, 7.2% in minor malformations, 14.3% in intentional abortions, and 28.6% in healthy infants; for 43.4%, no information was
available [36]. This limited information is sucient to recommend against
using pergolide when a woman wishes to get pregnant.
Some early publications reported no detrimental eects on pregnancy or
fetal development in women who became pregnant during treatment with
104
MOLITCH
quinagolide [37]. A more recent review of 176 pregnancies, in which quinagolide was maintained for a median duration of 37 days, reported 24 spontaneous abortions, 1 ectopic pregnancy, and 1 stillbirth at 31 weeks
gestation [38]. Nine fetal malformations were reported in this group: spina
bida, trisomy 13, Down syndrome, talipes, cleft lip, arrhinencephaly, and
Zellweger syndrome [38]. Quinagolide also should not be used if pregnancy
is desired.
Cabergoline has been shown to cross the placenta in animal studies [16],
but such data are lacking in humans. Data on exposure of the fetus or embryo during the rst several weeks of pregnancy have been reported in more
than 350 cases, and such use has not shown an increased percentage of spontaneous abortion, premature delivery, multiple gestation, or congenital
abnormalities [3943]. No alterations in newborn weights were observed
[3943]. Available data from 107 infants followed for 1 to 72 months showed
normal physical and mental development [39].
With respect to using a dopamine agonist to facilitate ovulation and fertility, bromocriptine has the largest safety database and has a proven safety
record for pregnancy. Although the database for cabergoline use in pregnancy is much smaller, it does not seem to exert any deleterious eects on
pregnant women, and the incidence of malformation in their ospring is
not greater than in the general population. For a woman who is intolerant
to bromocriptine and who is doing well with cabergoline, continuation of cabergoline for facilitating pregnancy seems reasonable. The safety databases
for pergolide and quinagolide are limited, but they seem to raise considerable
concerns, so these drugs should not be used when fertility is desired. The effects of transsphenoidal surgery during gestation are not known specically,
but would not be expected to be signicantly dierent from the eects of
other types of surgery (unless hypopituitarism should ensue) [44].
Management of prolactinoma in pregnancy
The risks of surgery versus medical therapy for prolactinoma should be
explained in detail to each patient. For patients with microadenomas or intrasellar macroadenomas, bromocriptine or cabergoline therapy generally is
preferred to surgery because it is safe for the fetus when discontinued early
in gestation, and it poses only a small risk of tumor enlargement for the
mother. Such patients should be seen each trimester and assessed for symptoms such as headaches or visual problems; visual eld testing needs to be
done only when clinically indicated. When the tumor is large or extends
to the optic chiasm or into the cavernous sinus, the following approaches
should be considered: (1) preoperative surgical debulking, (2) intensive monitoring without bromocriptine therapy, or (3) continuous bromocriptine
therapy. The safety of the last approach has not been established, but based
on the few cases cited earlier, it probably is not harmful. Patients with macroadenomas should be seen monthly for such assessments, and visual elds
105
106
MOLITCH
may cause an exacerbation of acromegaly in a few cases [47], but this does
not seem to be a sucient enough risk to advise against pregnancy.
Eects of acromegaly on pregnancy
Certain complications of acromegaly are potentially harmful to the mother
and the fetus. Carbohydrate intolerance is present in 50% of patients with
acromegaly, and overt diabetes is seen in 10% to 20% [55]. Insulin resistance secondary to the increased levels of GH may increase the risk of gestational diabetes. There is increased salt retention, and hypertension occurs
in 25% to 35% of patients. In addition, cardiac disease is present in about
one third of patients. There may be a specic cardiomyopathy associated
with acromegaly, and coronary artery disease may be increased [55]. The
risks for gestational diabetes, hypertension, and heart disease likely are increased in women with acromegaly during pregnancy.
Management of acromegaly and pregnancy
The considerations regarding the use of bromocriptine and cabergoline in
women with prolactinomas also apply to women with acromegaly. For most
patients, these drugs should not be continued during pregnancy. Data on the
use of octreotide during pregnancy are limited. Only 14 pregnant patients
treated with octreotide, octreotide long-acting-release, and lanreotide have
been reported; no malformations were found in their children [54]. Octreotide crosses the placenta [59] and can aect developing fetal tissues. It does
not bind with high anity to the placenta and has no eect on the placental
GH variant [60]. Because octreotide crosses the placenta, and because data
documenting safety are limited, it is recommended that octreotide and other
somatostatin analogues be discontinued if pregnancy is considered, and that
contraception be used when these drugs are administered. Considering the
prolonged nature of the course of most patients with acromegaly, interruption of medical therapy for 9 to 12 months should not have a particularly
adverse eect on the long-term outcome. These drugs can control tumor
growth, and for enlarging tumors, their reintroduction during pregnancy
may be warranted versus operating.
Cushings syndrome
Slightly more than 100 cases of Cushings syndrome in pregnancy have
been reported [6176]. The distribution of causes of Cushings syndrome in
pregnancy diers markedly from that in the nonpregnant population. Less
than 50% of pregnant patients described had pituitary adenomas, a similar
number had adrenal adenomas, and more than 10% had adrenal carcinomas
[6173]. Only three reports have described pregnancies associated with the
ectopic ACTH syndrome [63,67]. In many cases, the hypercortisolism rst
became apparent during pregnancy, with improvement after parturition,
107
108
MOLITCH
[76], but catheterization was performed via the direct jugular vein approach
rather than the femoral vein approach to minimize fetal irradiation; in these
cases, clearly increased central-to-peripheral ACTH gradients were found.
Eects of Cushings syndrome on pregnancy
Cushings syndrome is associated with a fetal mortality of 25% from
spontaneous abortion, stillbirth, and early neonatal death because of extreme prematurity [6173,76]. Premature labor occurs in more than 50%
of cases, regardless of cause [6173,76]. The passage of cortisol across the
placenta occasionally results in suppression of the fetal adrenals [69]. This
passage seems to be uncommon, but the neonate should be tested for this
potential problem and given exogenous corticosteroids until the results of
the evaluation are known.
Maternal complications also may occur. Hypertension develops in most
patients. Diabetes and myopathy are frequent. Postoperative wound infection and dehiscence are common after cesarean section. The pregnancy
seems to induce an amelioration of Cushings syndrome in some patients,
but an exacerbation in others [6167,73].
Management of Cushings syndrome during pregnancy
In data from the literature summarized from two reviews [61,64], fetal
loss rates of 9% and 24% and premature labor rates of 20% and 47%
were found in 11 and 17 women who were treated during pregnancy compared with fetal loss rates of 30% and 38% and premature labor rates of
48% and 72% in 26 and 43 women in whom treatment was delayed. Treatment during pregnancy has been advocated [61,64,76].
Medical therapy for Cushings syndrome during pregnancy is not very
eective [63,64,73]. A few case reports have documented the ecacy of metyrapone [76]. Ketoconazole has been given to two patients with complications
of intrauterine growth retardation, but no malformations or other perinatal
disorders [70,71]. Use of other drugs, such as aminoglutethimide, mitotane,
bromocriptine, and cyproheptadine, has been limited [73]; because of potential toxicity to the fetus, aminoglutethimide and mitotane should be
avoided. Transsphenoidal resection of a pituitary ACTH-secreting adenoma
has been performed successfully in several patients during the second trimester [62,65,68,69,72,73,76]. Although any surgery poses risks for the mother
and fetus [44], with Cushings syndrome, the risks of not operating seem to
be considerably higher than the risks of proceeding with surgery.
109
had been stopped, had to be reinstituted to control tumor size [77], and in
a second, octreotide was continued during pregnancy for tumor size control.
The most pressing issue with such tumors is the need to control hyperthyroidism during pregnancy, and that can be done with standard antithyroid
drugs [78]. With growing macroadenomas, octreotide may be necessary for
tumor size control [77,79].
Clinically nonfunctioning adenomas
Pregnancy would not be expected to inuence tumor size in patients with
clinically nonfunctioning adenomas, and only two cases have been reported
in which tumor enlargement during pregnancy resulted in a visual eld defect [25,80]. The lactotroph hyperplasia that occurs during pregnancy can be
quite signicant, however [2,3]. MRI obtained during pregnancy and the immediate postpartum period has shown that this hyperplasia may cause the
normal pituitary to increase to 12 mm in height [57]. It would be expected
that if this lactotroph hyperplasia were to occur in a patient with a preexisting clinically nonfunctioning adenoma, this hyperplasia could push up
the clinically nonfunctioning adenoma to cause chiasmal compression or
headaches. In the second case reported, the patient responded rapidly to
bromocriptine treatment, probably owing to shrinkage of the lactotroph hyperplasia with decompression of the chiasm and probably with little or no
direct eect on the tumor itself [80]. Most clinically nonfunctioning adenomas are gonadotroph adenomas [81]. Two patients have been reported who
had gonadotroph adenomas secreting intact follicle-stimulating hormone
with a resultant ovarian hyperstimulation syndrome [82,83]; both became
pregnant, one after having the follicle-stimulating hormone hypersecretion
controlled by bromocriptine [82] and the second after surgical removal of
the tumor [83].
Hypopituitarism
Hypopituitarism may occur because of tumor compression of the hypothalamus or pituitary stalk or from prior neurosurgery. Hormone decits
can be partial or complete, and loss of gonadotropin secretion is common.
Induction of ovulation may be dicult, and a variety of techniques have
been used, including administration of human chorionic gonadotropin
and follicle-stimulating hormone (in the past as human menopausal gonadotropin) [8487], pulsatile GnRH [8890], and in vitro fertilization [91].
In women, the only hormone replacements to be considered during pregnancy are thyroid and adrenal hormones. Because of the increased thyroxine
turnover that occurs during pregnancy, thyroxine levels decrease, and TSH
levels increase with a xed thyroxine dose over the course of gestation
[15,92]. The average increase in thyroxine needed in these patients is about
0.05 mg/d. Because patients with hypothalamic-pituitary dysfunction may
110
MOLITCH
not elevate their TSH levels normally in the face of increased need for thyroxine, it may be appropriate to increase the thyroxine supplementation by
0.025 mg after the rst trimester and by an additional 0.025 mg after the second trimester. There are no data to support this approach, however.
Because the cortisol production rate normally is increased in pregnancy
[11,12], the dose of long-term glucocorticoid replacement theoretically ought
to be increased during pregnancy. This dose increase does not seem to be
necessary in practice, however, and patients usually can be kept on their
standard replacement doses of glucocorticoids. Additional glucocorticoids
are needed for the stress of labor and delivery, such as 75 mg of hydrocortisone intravenously every 8 hours with rapid tapering postpartum. If there
is signicant stress during the pregnancy, such as infection, that would require prolonged high doses of glucocorticosteroids, the steroid of choice is
prednisolone, which does not cross the placenta [93]. Even high doses of
prednisone are generally quite safe [94], however, and suppression of neonatal adrenal function in ospring of women taking prednisone during pregnancy is rare [95]. Glucocorticoids also may pass to the neonate in breast
milk, but the amounts (0.14% of maternal blood levels) are not sucient
to alter neonatal adrenal function, even with large maternal doses of prednisone [96].
Lymphocytic hypophysitis
Lymphocytic hypophysitis usually presents in the peripartum period as
a mass lesion indistinguishable from a pituitary adenoma. It is characterized
by massive inltration of the pituitary by lymphocytes and plasma cells with
destruction of the normal parenchyma. The disorder is thought to have an
autoimmune basis. Most cases occur in association with pregnancy, and
women present during pregnancy or postpartum with symptoms of varying
degrees of hypopituitarism or symptoms related to the mass lesion, such as
headaches or visual eld defects. Mild hyperprolactinemia and diabetes insipidus also may be found. On CT or MRI, a sellar mass is found, which
may extend in an extrasellar fashion and may cause visual eld defects.
The condition usually is confused with that of a pituitary tumor and cannot
be distinguished from a tumor except by biopsy. By virtue of the hypopituitarism it produces, lymphocytic hypophysitis also can be confused clinically
with Sheehans syndrome except that there is no history of obstetric hemorrhage [97,98].
The diagnosis of lymphocytic hypophysitis should be considered in women
with symptoms of hypopituitarism or mass lesions of the sella during pregnancy or postpartum, especially in the absence of a history of obstetric hemorrhage. An evaluation of pituitary function and CT or MRI are
warranted. If prolactin levels are only modestly elevated (! 150 ng/mL)
in the presence of a large mass, the diagnosis is unlikely to be an enlarging
prolactinoma and more likely to be hypophysitis or a nonsecreting tumor.
111
Hormone replacement therapy should be instituted promptly when hypopituitarism is determined to be present. For unclear reasons, there seems
to be a particular predilection for impaired ACTH secretion, and this
axis must be evaluated carefully and treated to avoid adrenal insuciency
[99]. Unless there are visual eld defects, uncontrollable headaches, or radiologic evidence of progressive enlargement of the sellar mass, rapid surgical intervention is not warranted because some women may undergo
a spontaneous regression of the mass and return of pituitary function
[99101]. Surgery generally does not result in improvement in endocrine
function, however [102,103]. Although high doses of glucocorticoids
have been advocated to reduce the inammation [103], there have been
no controlled studies documenting the benet of this approach, and the
author does not recommend this.
Sheehans syndrome
Sheehans syndrome consists of pituitary necrosis secondary to ischemia
occurring within hours of delivery [104,105]. It is usually secondary to hypotension and shock from an obstetric hemorrhage. Pituitary enlargement during pregnancy apparently predisposes to the risk for ischemia with occlusive
spasm of the arteries to the anterior pituitary and stalk [104,105]. The degree
of ischemia and necrosis dictates the subsequent patient course. Modern obstetric techniques have resulted in Sheehans syndrome being found rarely in
current practice [106].
Acute necrosis is suspected in the setting of an obstetric hemorrhage in
which hypotension and tachycardia persist after adequate replacement of
blood products. In addition, the woman fails to lactate and may have hypoglycemia [104,105,107]. Investigation should include levels of ACTH, cortisol, prolactin, and free thyroxine. The ACTH stimulation test would be
normal because the adrenal cortex would not be atrophied. Thyroxine levels
may prove normal initially because the hormone has a half-life of 7 days.
Prolactin levels are usually low, although they are generally 5- to 10-fold elevated in the puerperium. Treatment with saline and stress doses of corticosteroids should be instituted immediately after drawing the blood tests.
Additional pituitary testing with subsequent therapy should be delayed until
recovery. Diabetes insipidus also may occur secondary to vascular occlusion
with atrophy and scarring of the neurohypophysis [108].
When milder forms of infarction occur, the diagnosis of Sheehans syndrome may be delayed for months or years [107,108]. These women generally have a history of amenorrhea, decreased libido, failure to lactate, breast
atrophy, loss of pubic and axillary hair, fatigue, and symptoms of secondary
adrenal insuciency with nausea, vomiting, diarrhea, and abdominal pain
[107,108]. Some women experience only partial hypopituitarism and may
have normal menses and fertility [109]. Although women may have episodes
of transient polydipsia and polyuria, many have impaired urinary
112
MOLITCH
References
[1] Central Brain Tumor Registry of the United States (CTBRUS). Statistical report: primary
brain tumors in the US 19972001.Available at: http://www.cbtrus.org/. Accessed April 8,
2005.
[2] Golubo LG, Ezrin C. Eect of pregnancy on the somatotroph and the prolactin cell of the
human adenohypophysis. J Clin Endocrinol Metab 1969;29:15338.
[3] Scheithauer BW, Sano T, Kovacs KT, et al. The pituitary gland in pregnancy: a clinicopathologic and immunohistochemical study of 69 cases. Mayo Clin Proc 1990;65:46174.
[4] Rigg LA, Lein A, Yen SSC. Pattern of increase in circulating prolactin levels during human
gestation. Am J Obstet Gynecol 1977;129:4546.
[5] Gonzalez JG, Elizondo G, Saldivar D, et al. Pituitary gland growth during normal pregnancy: an in vivo study using magnetic resonance imaging. Am J Med 1988;85:21720.
[6] Elster AD, Sanders TG, Vines FS, Chen MYM. Size and shape of the pituitary gland during pregnancy and post partum: measurement with MR imaging. Radiology 1991;181:
5315.
[7] Dinc H, Esen F, Demirci A, et al. Pituitary dimensions and volume measurements in pregnancy and post partum: MR assessment. Acta Radiol 1998;39:649.
[8] Frankenne F, Closset J, Gomez F, et al. The physiology of growth hormones (GHs) in pregnant women and partial characterization of the placental GH variant. J Clin Endocrinol
Metab 1988;66:117180.
[9] Eriksson L, Frankenne F, Eden S, et al. Growth hormone 24-h serum proles during pregnancy lack of pulsatility for the secretion of the placental variant. Br J Obstet Gynaecol
1989;106:94953.
[10] Beckers A, Stevenaert A, Foidart J-M, et al. Placental and pituitary growth hormone secretion during pregnancy in acromegalic women. J Clin Endocrinol Metab 1990;71:72531.
[11] Carr BR, Parker CR Jr, Madden JD, et al. Maternal plasma adrenocorticotropin and cortisol relationships throughout human pregnancy. Am J Obstet Gynecol 1981;139:41622.
[12] Nolten WE, Lindheimer MD, Rueckert PA, et al. Diurnal patterns and regulation of cortisol secretion in pregnancy. J Clin Endocrinol Metab 1980;51:46672.
[13] Rees LH, Burke CW, Chard T, et al. Possible placental origin of ACTH in normal human
pregnancy. Nature 1975;254:6202.
[14] Sasaki A, Shinkawa O, Yoshinaga K. Placental corticotropin-releasing hormone may be
a stimulator of maternal pituitary adrenocorticotropic hormone secretion in humans.
J Clin Invest 1989;84:19972001.
[15] Glinoer D. The regulation of thyroid function in pregnancy: pathways of endocrine adaptation from physiology to pathology. Endocr Rev 1997;18:40433.
[16] Jeppsson S, Rannevik G, Liedholm P, Thorell JI. Basal and LHRH stimulated secretion of
FSH during pregnancy. Am J Obstet Gynecol 1977;127:326.
[17] Kredentser JV, Hoskins CF, Scott JZ. Hyperprolactinemia: a signicant factor in female
infertility. Am J Obstet Gynecol 1981;139:2647.
[18] Molitch ME. Disorders of prolactin secretion. Endocrinol Metab Clin North Am 2001;30:
585610.
[19] Webster J, Piscitelli G, Polli A, et al. A comparison of cabergoline and bromocriptine in the
treatment of hyperprolactinemic amenorrhea. N Engl J Med 1994;331:9049.
[20] Molitch ME. Dopamine resistance of prolactinomas. Pituitary 2003;6:1927.
[21] Bevan JS, Webster J, Burke CW, Scanlon MF. Dopamine agonist and pituitary tumor
shrinkage. Endocr Rev 1992;13:22040.
113
[22] Biller BMK, Molitch ME, Vance ML, et al. Treatment of prolactin-secreting macroadenomas with the once-weekly dopamine agonist cabergoline. J Clin Endocrinol Metab 1996;81:
233843.
[23] Colao A, DiSarno A, Landi ML, et al. Macroprolactinoma shrinkage during cabergoline
treatment is greater in na ve patients than in patients pretreated with other dopamine agonists: a prospective study of 110 patients. J Clin Endocrinol Metab 2000;85:224752.
[24] Molitch ME. Pregnancy and the hyperprolactinemic woman. N Engl J Med 1985;312:
136470.
[25] Kupersmith MJ, Rosenberg C, Kleinberg D. Visual loss in pregnant women with pituitary
adenomas. Ann Intern Med 1994;121:4737.
[26] Rossi AM, Vilska S, Heinonen PK. Outcome of pregnancies in women with treated or untreated hyperprolactinemia. Eur J Obstet Gynaecol Reprod Biol 1995;63:1436.
[27] Musolino NRC, Bronstein MD. Prolactinomas and pregnancy. In: Bronstein MD, editor.
Pituitary tumors and pregnancy. Norwell (MA): Kluwer Academic Publishers; 2001.
p. 91108.
[28] Liu C, Tyrrell JB. Successful treatment of a large macroprolactinoma with cabergoline during pregnancy. Pituitary 2001;4:17985.
[29] Krupp P, Monka C, Richter K. The safety aspects of infertility treatments. In: Program of
the Second World Congress of Gynecology and Obstetrics, Rio de Janeiro, Brazil, 1988.
p. 9.
[30] Krupp P, Monka C. Bromocriptine in pregnancy: safety aspects. Klin Wochenschr 1987;65:
8237.
[31] Raymond JP, Goldstein E, Konopka P, et al. Follow-up of children born of bromocriptinetreated mothers. Horm Res 1985;22:23946.
[32] Konopka P, Raymond JP, Merceron RE, Seneze J. Continuous administration of bromocriptine in the prevention of neurological complications in pregnant women with prolactinomas. Am J Obstet Gynecol 1983;146:9358.
[33] Bigazzi M, Ronga R, Lancranjan I, et al. A pregnancy in an acromegalic woman during
bromocriptine treatment: eects on growth hormone and prolactin in the maternal, fetal,
and amniotic compartments. J Clin Endocrinol Metab 1979;48:912.
[34] Buelke-Sam J, Byrd RA, Johnson JA, et al. Developmental toxicity of the dopamine agonist
pergolide mesylate in CD-1 mice: I. gestational exposure. Neurotoxicol Teratol 1991;13:
28395.
[35] De Mari M, Zenzola A, Lamberti P. Antiparkinsonian treatment in pregnancy. Mov Disord 2002;17:4289.
[36] Acharya V. Review of pregnancy reports in patients on pergolide treatment. Data on le.
Indianapolis (IN): Eli Lilly & Co; 2004.
[37] Morange I, Barlier A, Pellegrini I, et al. Prolactinomas resistant to bromocriptine: longterm ecacy of quinagolide and outcome of pregnancy. Eur J Endocrinol 1996;135:
41320.
[38] Webster J. A comparative review of the tolerability proles of dopamine agonists in the
treatment of hyperprolactinaemia and inhibition of lactation. Drug Saf 1996;14:22838.
[39] Robert E, Musatti L, Piscitelli G, Ferrari CI. Pregnancy outcome after treatment with the
ergot derivative, cabergoline. Reprod Toxicol 1996;10:3337.
[40] Garceau R. Pregnancies reported to Pharmacia Upjohn in patients receiving cabergoline.
Data on le. Kalamazoo (MI): Pharmacia & Upjohn; 1997.
[41] Verhelst J, Abs R, Maiter D, et al. Cabergoline in the treatment of hyperprolactinemia:
a study in 455 patients. J Clin Endocrinol Metab 1999;84:251822.
[42] Ricci E, Parazzini F, Motta T, et al. Pregnancy outcome after cabergoline treatment in early
weeks of gestation. Reprod Toxicol 2002;16:7913.
[43] Musolino NRC, Bronstein MD. Prolactinomas and pregnancy. In: Bronstein MD, editor.
Pituitary tumors and pregnancy. Norwell (MA): Kluwer Academic Publishers; 2001.
p. 91108.
114
MOLITCH
[44] Brodsky JB, Cohen EN, Brown BW Jr, et al. Surgery during pregnancy and fetal outcome.
Am J Obstet Gynecol 1980;138:11657.
[45] Divers WA, Yen SSC. Prolactin-producing microadenomas in pregnancy. Obstet Gynecol
1983;62:4259.
[46] Colao A, Merola B, Ferone D, Lombardi G. Acromegaly. J Clin Endocrinol Metab 1997;
82:277781.
[47] Herman-Bonert V, Seliverstow M, Melmed S. Pregnancy in acromegaly: successful therapeutic outcome. J Clin Endocrinol Metab 1998;83:72731.
[48] Mozas J, Ocon E, Lopez de la Torre M, et al. Successful pregnancy in a woman with acromegaly treated with somatostatin analog (octreotide) prior to surgical resection. Int J Gynecol Obstet 1999;65:713.
[49] DeMenis E, Billeci D, Marton E. Uneventful pregnancy in an acromegalic patient treated
with slow-release lanreotide: a case report. J Clin Endocrinol Metab 1999;84:1489.
[50] Hierl T, Ziegler R, Kasperk C. Pregnancy in persistent acromegaly. Clin Endocrinol (Oxf)
2000;53:2623.
[51] Neal JM. Successful pregnancy in a woman with acromegaly treated with octreotide.
Endocr Pract 2000;6:14850.
[52] Fassnacht M, Capeller B, Arlt W, et al. Octreotide LAR treatment throughout pregnancy
in an acromegalic woman. Clin Endocrinol (Oxf) 2001;55:4115.
[53] Bronstein MD, Salgado LR, Musolino NR. Medical management of pituitary adenomas:
the special case of management of the pregnant woman. Pituitary 2002;5:99107.
[54] Serri O, Lanoie G. Successful pregnancy in a woman with acromegaly treated with octreotide long-acting release. Endocrinologist 2003;13:179.
[55] Molitch ME. Clinical manifestations of acromegaly. Endocrinol Metab Clin North Am
1992;21:597614.
[56] Barkan AL, Stred SE, Reno K, et al. Increased growth hormone pulse frequency in acromegaly. J Clin Endocrinol Metab 1989;69:122533.
[57] Chang-DeMoranville BM, Jackson IMD. Diagnosis and endocrine testing in acromegaly.
Endocrinol Metab Clin North Am 1992;21:64968.
[58] Okada Y, Morimoto I, Ejima K, et al. A case of active acromegalic woman with a marked
increase in serum insulin-like growth factor-1 levels after delivery. Endocr J 1997;44:11720.
[59] Caron P, Gerbeau C, Pradayrol L. Maternal-fetal transfer of octreotide. N Engl J Med
1995;333:6012.
[60] Caron P, Buscail L, Beckers A, et al. Expression of somatostatin receptor SST4 in human
placenta and absence of octreotide eect on human placental growth hormone concentration during pregnancy. J Clin Endocrinol Metab 1997;82:37716.
[61] Bevan JS, Gough MH, Gillmer MD, Burke CW. Cushings syndrome in pregnancy: the
timing of denitive treatment. Clin Endocrinol (Oxf) 1987;27:22533.
[62] Casson IF, Davis JC, Jereys RV, et al. Successful management of Cushings disease during
pregnancy by transsphenoidal adenectomy. Clin Endocrinol (Oxf) 1987;27:4238.
[63] Aron DC, Schnall AM, Sheeler LR. Cushings syndrome and pregnancy. Am J Obstet Gynecol 1990;162:24452.
[64] Buescher MA, McClamrock HD, Adashi EY. Cushings syndrome in pregnancy. Obstet
Gynecol 1992;79:1307.
[65] Ross RJ, Chew SL, Perry L, et al. Diagnosis and selective cure of Cushings disease during
pregnancy by transsphenoidal surgery. Eur J Endocrinol 1995;132:7226.
[66] Chico A, Manzanares JM, Halperin I, et al. Cushings disease and pregnancy. Eur J Obstet
Gynaecol Reprod Biol 1996;64:1436.
[67] Guilhaume B, Sanson ML, Billaud L, et al. Cushings syndrome and pregnancy: aetiologies
and prognosis in twenty-two patients. Eur J Med 1992;1:839.
[68] Pinette MG, Pan YQ, Oppenheim D, et al. Bilateral inferior petrosal sinus corticotropin
sampling with corticotropin-releasing hormone stimulation in a pregnant patient with
Cushings syndrome. Am J Obstet Gynecol 1994;171:5634.
115
[69] Kreines K, DeVaux WD. Neonatal adrenal insuciency associated with maternal Cushings syndrome. Pediatrics 1971;47:5169.
[70] Amado JA, Pesquera C, Gonzalez EM, et al. Successful treatment with ketoconazole of
Cushings syndrome in pregnancy. Postgrad Med J 1990;66:2213.
[71] Berwaerts J, Verhelst J, Mahler C. Abs R. Cushings syndrome in pregnancy treated by
ketoconazole: case report and review of the literature. Gynecol Endocrinol 1999;13:
17582.
[72] Mellor A, Harvey RD, Pobereskin LH, Sneyd JR. Cushings disease treated by trans-sphenoidal selective adenomectomy in mid-pregnancy. Br J Anaesth 1998;80:8502.
[73] Madhun ZT, Aron DC. Cushings disease in pregnancy. In: Bronstein MD, editor. Pituitary
tumors and pregnancy. Norwell (MA): Kluwer Academic Publishers; 2001. p. 14972.
[74] Wallace C, Toth EL, Lewanczuk RZ, Siminoski K. Pregnancy-induced Cushings syndrome in multiple pregnancies. J Clin Endocrinol Metab 1996;81:1521.
[75] Hana V, Dokoupilova M, Marek J, Plavka R. Recurrent ACTH-independent Cushings
syndrome in multiple pregnancies and its treatment with metyrapone. Clin Endocrinol
(Oxf) 2001;54:27781.
[76] Lindsay JR, Jonklaas J, Oldeld EH, Nieman LK. Cushings syndrome during pregnancy:
personal experience and review of the literature. J Clin Endocrinol Metab 2005;90:
307783.
[77] Caron P, Gerbeau C, Pradayrol L, et al. Successful pregnancy in an infertile woman with
a thyrotropin-secreting macroadenoma treated with the somatostatin analog (octreotide).
J Clin Endocrinol Metab 1996;81:11648.
[78] Blackhurst G, Strachan MW, Collie D, et al. The treatment of a thyrotropin-secreting pituitary macroadenoma with octreotide in twin pregnancy. Clin Endocrinol (Oxf) 2002;56:
4014.
[79] Chaiamnuay S, Moster M, Katz MR, Kim YN. Successful management of a pregnant
woman with a TSH secreting pituitary adenoma with surgical and medical therapy. Pituitary 2003;6:10913.
[80] Masding MG, Lees PD, Gawne-Cain ML, Sandeman DD. Visual eld compression by
a non-secreting pituitary tumour during pregnancy. J R Soc Med 2003;96:278.
[81] Molitch ME. Clinically non-functioning adenomas. In: Bronstein MD, editor. Pituitary
tumors and pregnancy. Norwell (MA): Kluwer Academic Publishers; 2001. p. 1239.
[82] Murata Y, Ando H, Nagasaka T, et al. Successful pregnancy after bromocriptine therapy
in an anovulatory woman complicated with ovarian hyperstimulation caused by folliclestimulating hormone-producing plurihormonal pituitary microadenoma. J Clin Endocrinol Metab 2003;88:198893.
[83] Sugita T, Seki K, Nagai Y, et al. Successful pregnancy and delivery after removal of gonadotrope adenoma secreting follicle-stimulating hormone in a 29-year-old amenorrheic
woman. Gynecol Obstet Invest 2005;59:13843.
[84] Golan A, Abramov L, Yedwab G, David MP. Pregnancy in panhypopituitarism. Gynecol
Obstet Invest 1990;29:2324.
[85] Verdu LI, Martin-Caballero C, Garcia-Lopez G, Cueto MJ. Ovulation induction and normal pregnancy after panhypopituitarism due to lymphocytic hypophysitis. Obstet Gynecol
1998;91:8502.
[86] Volz J, Heinrich U, Volz-Koster S. Conception and spontaneous delivery after total
hypophysectomy. Fertil Steril 2002;77:6245.
[87] Kitajima Y, Endo T, Yamazaki K, et al. Successful twin pregnancy in panhypopituitarism
caused by suprasellar germinoma. Obstet Gynecol 2003;102:12057.
[88] Gompel A, Mauvais-Jarvis P. Induction of ovulation with pulsatile GnRH in hypothalamic
amenorrhea. Hum Reprod 1988;3:4737.
[89] Martin KA, Hall JE, Adams JM, Crowley WF Jr. Comparison of exogenous gonadotropins and pulsatile gonadotropin-releasing hormone for induction of ovulation in hypogonadotropic amenorrhea. J Clin Endocrinol Metab 1993;77:1259.
116
MOLITCH
[90] Hall JE, Martin KA, Whitney HA, et al. Potential for fertility with replacement of hypothalamic gonadotropin-releasing hormone in long term female survivors of cranial tumors.
J Clin Endocrinol Metab 1994;79:116672.
[91] Suganuma N, Furuhashi M, Ando T, et al. Successful pregnancy and delivery after in vitro
fertilization and embryo transfer in a patient with primary hypopituitarism. Fertil Steril
2000;73:10578.
[92] Mandel SJ, Larsen PR, Seely EW, Brent GA. Increased need for thyroxine during pregnancy in women with primary hypothyroidism. N Engl J Med 1990;323:916.
[93] Beitins IZ, Bayard F, Ances IG, et al. The transplacental passage of prednisone and prednisolone in pregnancy near term. J Pediatr 1972;81:93645.
[94] Turner ES, Greenberger PA, Patterson R. Management of the pregnant asthmatic patient.
Ann Intern Med 1980;93:90518.
[95] Kenny FM, Preeyasombat C, Spaulding JS, Migeon CJ. Cortisol production rate: IV. infants born of steroid-treated mothers and of diabetic mothers: infants with trisomy syndrome and with anencephaly. Pediatrics 1966;37:9606.
[96] McKenzie SA, Selley JA, Agnew JE. Secretion of prednisolone into breast milk. Arch Dis
Child 1975;50:8946.
[97] Thodou E, Asa SL, Kontogeorgos G, et al. Lymphocytic hypophysitis: clinicopathological
ndings. J Clin Endocrinol Metab 1995;80:230211.
[98] Pressman EK, Zeidman SM, Reddy UM, et al. Dierentiating lymphocytic adenohypophysitis from pituitary adenoma in the peripartum patient. J Reprod Med 1995;40:2519.
[99] Gillam M, Molitch ME. Lymphocytic hypophysitis. In: Bronstein MD, editor. Pituitary
tumors and pregnancy. Norwell (MA): Kluwer Academic Publishers; 2001. p. 13148.
[100] Leiba S, Schindel B, Weinstein R, et al. Spontaneous postpartum regression of pituitary
mass with return of function. JAMA 1986;255:2302.
[101] McGrail KM, Beyerl BD, Black PM, et al. Lymphocytic adenohypophysitis of pregnancy
with complete recovery. Neurosurgery 1987;20:7913.
[102] Leung GK, Lopes MB, Thorner MO, et al. Primary hypophysitis: a single-center experience
in 16 cases. J Neurosurg 2004;101:26271.
[103] Caturegli P, Newschaer C, Olivi A, et al. Autoimmune hypophysitis. Endocr Rev 2005;
26(5):599614.
[104] Sheehan HL, Davis JC. Pituitary necrosis. Br Med Bull 1968;24:5970.
[105] Kelesxtimur F. Sheehans syndrome. Pituitary 2003;6:1818.
[106] Feinberg E, Molitch M, Peaceman A. Frequency of Sheehans syndrome. Fertil Steril
2005;84:9759.
[107] Ozbey N, Inanc S, Aral F, et al. Clinical and laboratory evaluation of 40 patients with Sheehans syndrome. Isr J Med Sci 1994;30:8269.
[108] Sheehan HL. The neurohypophysis in post-partum hypopituitarism. J Pathol Bacteriol
1963;85:14569.
[109] Grimes HG, Brooks MH. Pregnancy in Sheehans syndrome: report of a case and review.
Obstet Gynecol Surv 1980;35:4818.
[110] Iwasaki Y, Oiso Y, Yamauchi K, et al. Neurohypophyseal function in post-partum hypopituitarism: impaired plasma vasopressin response to osmotic stimuli. J Clin Endocrinol
Metab 1989;68:5605.
[111] Bakiri F, Bendib S-E, Maoui R, et al. The sella turcica in Sheehans syndrome: computerized tomographic study in 54 patients. J Endocrinol Invest 1991;14:1936.
118
The most recognized alteration in maternal thyroid physiology is the increase in thyroxine-binding globulin (TBG). This increase begins early in the
rst trimester, plateaus during midgestation, and persists until shortly after
delivery [5]. Elevated TBG results from a decrease in its hepatic clearance,
owing to estrogen-induced sialylation [6]. The increased concentration of
TBG expands the extrathyroidal pool, triggering a concomitant increase
in maternal thyroid hormone synthesis and elevation of total thyroxine
(T4) and triiodothyronine (T3) levels [5]. In addition to TBG, maternal glomerular ltration rate is increased during early gestation and maintained
throughout pregnancy; this results in an increased renal clearance of iodide,
which indirectly stimulates the maternal thyroid machinery [7]. Lastly,
transplacental passage of T4 and iodide and placental metabolism of iodothyronines stimulate the maternal thyroid by depleting the maternal circulation of thyroid hormone and its precursors [8].
In addition to indirect stimuli, there is strong evidence that human chorionic gonadotropin (hCG) has intrinsic thyrotropic activity. A signicant
degree of homology exists between the hormone-specic b-subunits and
the extracellular receptor binding domains of hCG and TSH [9,10]. hCG secretion begins shortly after conception, peaks around gestational week 10,
and gradually declines thereafter to a nadir by about week 20 [5]. It is believed that the high serum concentrations of hCG during early pregnancy
directly activate the TSH receptor. This belief is substantiated by the nding
that serum hCG concentrations during early pregnancy are negatively correlated with serum TSH concentrations and positively correlated with free
T4 concentrations [5,11]. Serum TSH concentrations vary throughout pregnancy, with the rst-trimester values being lower than the values before conception and during the second and third trimesters [12]. Using an assay with
a detection limit of 0.05 mU/L, researchers found that during the rst trimester, 9% of pregnant women without thyrotoxic symptoms had subnormal serum TSH concentrations (O 0.05 mU/L, but ! 0.4 mU/L), whereas
an additional 9% had suppressed concentrations (! 0.05 mU/L) [13]. Ultimately, the nadir of maternal serum TSH levels mirrors the peak in
hCG concentrations [5]. A more recent study has reported the upper 95%
condence interval (CI) for serum TSH levels in the rst trimester to be
2.5 mU/L [14].
Although consistent patterns of total T4 and TSH during pregnancy
are well documented, the same is not true for free T4 concentrations, particularly during the rst trimester. Free T4 concentrations during this time
have been reported to be higher, lower, and the same as concentrations before conception [5]. These discrepant ndings are likely due to a combination
of shortcomings in methodology and dietary iodine intake among study participants [12]. Based on the available data, it is likely that free T4 and T3 concentrations increase slightly during the rst trimester in response to elevated
hCG. Subsequently, free T4 values decline as pregnancy progresses and nadir during the third trimester with measured values in commercial assays
119
that may be lower than the assays published reference ranges [12]. Currently, none of the manufacturers of the automated free T4 assays has provided trimester-specic reference ranges. Until this information is available,
the serum total T4 concentration may be a better reection of T4 production
during pregnancy. To account for the increased thyroid hormone production
during gestation, the normal reference range for total T4 should be adjusted
by a factor of 1.5 for pregnant patients [12,15].
Hypothyroidism
Screening studies have shown that an elevated serum TSH concentration
is found in 2.5% of pregnancies [16,17]. In an iodine-sucient environment,
hypothyroidism during pregnancy is caused primarily by Hashimotos thyroiditis and prior radioactive iodine treatment or surgical ablation of
Graves disease [18]. Nevertheless, the clinician should keep in mind the
less common causes (eg, overtreatment of hyperthyroidism with thionamides, transient hypothyroidism owing to postpartum thyroiditis, medications that alter the absorption or metabolism of levothyroxine, and
pituitary/hypothalamic disease) because the diagnosis and management of
these patients may be dierent.
Diagnosis
The diagnosis of hypothyroidism during pregnancy is crucial because of
its potential adverse eects on the mother and the child. Only 20% to 30%
of patients with overt hypothyroidism develop symptoms consistent with
disease, however, whereas most patients with subclinical disease are entirely
asymptomatic [19,20]. When symptoms do exist, the diagnosis often is overlooked, and a womans complaints may be attributed to pregnancy itself.
The diagnosis of primary hypothyroidism is made by documenting an elevated serum TSH concentration; however, most commercial laboratories
have not established trimester-specic reference ranges. Patients with central
hypothyroidism resulting from pituitary or hypothalamic disease do not
manifest an elevated serum TSH level during pregnancy.
Pregnancy outcome
Despite the association between overt hypothyroidism and infertility, hypothyroid women may become pregnant [21]. The likelihood that either maternal or fetal complications will arise depends on the severity of disease and
adequacy of treatment. Gestational hypertension occurs more often in
overtly hypothyroid women (36%) than in women with subclinical disease
(25%) or the general population (8%) [22]. A separate study reported
a markedly increased use of cesarean section because of fetal distress among
women who were severely hypothyroid (56%) at their initial antenatal visit
120
121
compared development between children born to women with hypothyroxinemia (free T4 % 10th percentile) and normal serum TSH concentrations
(! 2.2 mU/L) at gestational week 12 with children of controls (free T4
50th90th percentiles and normal TSH). They found a signicant delay in
mental and motor development for 1-year-olds and 2-year-olds born to
the women with hypothyroxinemia. Infants of mothers with hypothyroxinemia at 12 weeks that normalized during pregnancy did not dier signicantly
from controls. In addition, infants in the control group whose mothers became hypothyroxinemic after 12 weeks of gestation did not show impaired
development [31]. The potential inuences of iodine nutrition and other maternal comorbidities were not addressed in this study. Although the relative
contribution of maternal versus fetal thyroid hormone to neurologic development is not fully understood, these ndings suggest that maternal hormone is important during the rst trimester, before the fetal thyroid gland
begins to function, and later in gestation.
Treatment
The treatment of hypothyroidism during pregnancy depends on the timing of diagnosis, the severity of disease, and possibly the cause of thyroid
dysfunction (Box 1). The starting levothyroxine dose for women with overt
hypothyroidism should be 2 mg/kg/d ) [12]. This dosage is higher than that
required for full replacement in nonpregnant patients and accounts for the
increased thyroidal demand of normal pregnancy. If the initial serum TSH
level is only slightly elevated (ie, TSH ! 10 mU/L), an initial dose of 0.1
mg/d of levothyroxine may be sucient [32].
The likelihood that hypothyroid women who receive levothyroxine replacement will need a dosage increase may depend on the cause of their thyroid dysfunction. In one study, 76% of hypothyroid women versus 47% of
women with Hashimotos thyroiditis required an increase in levothyroxine
dosage during pregnancy [2]. In addition, because women with Hashimotos
thyroiditis may have residual thyroid function that can respond partially to
the stimulatory eects of pregnancy, they require a smaller mean dosage increase (25%) than hypothyroid women (45%) [2].
As previously discussed, the stimulatory eects in pregnancy begin early
in the rst trimester and persist until delivery. Maternal requirements must
be monitored closely throughout gestation. A serum TSH level should be
checked as soon as pregnancy is conrmed. In a prospective study, the median time for dosage increase was 8 weeks gestation [1]. In addition, 25% of
women with a normal TSH concentration during the rst trimester and 37%
with a normal TSH in the second trimester eventually require an increase
in levothyroxine dosage [2]. Women with a serum TSH level less than
2.5 mU/L at initial screening should have their thyroid function re-evaluated
every 3 to 4 weeks during the rst half of pregnancy and every 6 weeks during the latter half.
122
123
immediately after delivery and have their serum TSH level re-evaluated in 6
weeks.
Hyperthyroidism
The prevalence of hyperthyroidism during pregnancy ranges from
0.1% to 0.4%, with Graves disease accounting for 85% of cases [32,39].
Single toxic adenoma, multinodular toxic goiter, and subacute thyroiditis
124
constitute most of the remaining cases during pregnancy, whereas exogenous thyroid hormone and hydatidiform molar disease are extremely rare
[40].
Graves disease
Graves disease is the most common cause of hyperthyroidism during
pregnancy [32,39]. Similar to other autoimmune diseases, the activity level
of Graves disease may uctuate during gestation, with exacerbation during
the rst trimester and gradual improvement during the latter half. Patients
with Graves disease also may experience an exacerbation shortly after delivery [44]. With this possibility in mind, there are several dierent clinical
scenarios by which a woman may present with Graves disease during pregnancy. First, a woman with stable Graves disease receiving thionamide
therapy may experience an exacerbation during early pregnancy. Second,
a woman in remission may experience a relapse of disease. Lastly, a woman
without prior history may be diagnosed with Graves disease de novo during
pregnancy.
125
Diagnosis
The diagnosis of Graves disease may be dicult to make clinically because of the presence of hypermetabolic symptoms in normal pregnancy,
such as palpitations, irritability, and heat intolerance. The thyroid examination is often dierent from that of normal pregnancy, hyperemesis gravidarum, and gestational thyrotoxicosis. Women with Graves disease usually
have a goiter (with or without bruit), but the accompanying autoimmune
syndromes of ophthalmopathy and possibly dermopathy are still quite rare.
Laboratory studies also are helpful, revealing a suppressed serum TSH
level and usually elevated free and total T4 serum concentrations. As described previously, however, 60% of women with hyperemesis gravidarum
have a subnormal serum TSH, and nearly 50% have an elevated serum
free T4 concentration [45]. In situations in which doubt exists, measurement
of serum total T3 concentration and T3 resin uptake may be helpful because
only 12% of women with hyperemesis gravidarum have an elevated free T3
index [45]. Finally, TSH receptor antibodies are usually present with
Graves disease and may aid in conrming the diagnosis.
Pregnancy outcome
The risk of complications for the mother and the child is related to the
duration and control of maternal hyperthyroidism. The highest incidence
occurs in cases with the poorest control, and the lowest incidence occurs
in cases with adequate treatment [40,4648]. The frequency of preterm labor
is highest among untreated mothers (88%) compared with partially treated
mothers (25%) and adequately treated mothers (8%) [46]. In addition, untreated women are twice as likely to develop preeclampsia during pregnancy
than women receiving antithyroid medication [40]. As for the fetus, stillbirth
is much more common among untreated women (50%) than either partially
treated women (16%) or adequately treated women (0%) [46]. Finally, studies have indicated that children born to mothers with uncontrolled hyperthyroidism are more likely to be small for gestational age and to have
congenital malformations unrelated to thionamide therapy [47,48].
Treatment
The most important thing to remember when treating Graves disease
during pregnancy is that two patients are involved, the mother and fetus.
The goal of therapy is to control maternal disease, while minimizing to potential for fetal hypothyroidism and hyperthyroidism (Box 2).
Thionamides
Thionamide antithyroid drugs, propylthiouracil (PTU) and methimazole
(MMI), are the mainstay of treatment of Graves disease during pregnancy.
These medications decrease thyroid hormone production by inhibiting
126
127
128
discontinuation, of thionamide treatment [60]. If improvement is not apparent, consideration should be given to more aggressive diagnostic testing,
such as periumbilical blood sampling, with treatment tailored to the specic
ndings (eg, intra-amniotic levothyroxine therapy for persistent hypothyroidism) [61,62].
Aside from the hypothyroidism, the other potential side eects of thionamide therapy must be considered. The maternal side eects are the same for
any individual taking these medications, the most common being a rash. As
for the fetus/neonate, several cross-sectional studies have found no eect on
cognitive or somatic development among infants exposed to PTU or MMI
[6366]. Several rare birth defects have been observed in neonates born to
mothers taking MMI during pregnancy (eg, aplasia cutis, choanal atresia,
esophageal atresia, and minor dysmorphic features) [67,68]. None of these
ndings has been reported with PTU. The authors prefer PTU as rst-line
therapy. If a woman is unable to tolerate PTU, the authors prescribe
MMI, especially after rst-trimester organogenesis.
Treatment guidelines
The initial dosage of antithyroid medication depends on the severity of
disease. PTU should be used preferentially given the multiple case reports
of congenital defects and dysmorphic features occurring in infants exposed
to MMI [67,68]. If one is allergic or intolerant of PTU, however, MMI
should be substituted (especially after rst-trimester organogenesis) before
recommending thyroidectomy. Antithyroid medication should be titrated
to maintain maternal T4 concentrations within the upper third, or slightly
above, of the normal range for pregnancy. Because pregnancy reference
ranges have not been reported for most free T4 assays, the authors recommend using the serum total T4 concentration for thionamide dose titration.
In addition, the authors adopt a pregnancy reference range for total T4 to be
1.5 times the nonpregnant reference range owing to thyroidal stimulation
during pregnancy (see section on normal gravid thyroid physiology) [15].
Clinicians should re-evaluate maternal thyroid function every 2 to 4 weeks
and adjust thionamide therapy accordingly. Serum TSH levels are not helpful in the management of Graves disease during early pregnancy because of
the lag time that exists between the normalization of thyroid hormones and
TSH, but may be useful later in gestation when the disease is controlled. The
authors do not exceed 600 mg/d of PTU or 40 mg/d of MMI for extended
periods. One should continue to titrate PTU to the lowest possible dose that
maintains control. Graves disease may ameliorate as pregnancy progresses.
As a result, thionamide therapy may be discontinued in 30% of women during the nal weeks of pregnancy [69]. Cord blood testing for TSH and T4
is recommended in all infants born to mothers with active Graves disease.
In addition, to assess the risk for fetal or neonatal hyperthyroidism, measurement of TSH receptor antibodies at 26 to 28 weeks gestation is recommended (see later).
129
b-Adrenergic blockers
b-Adrenergic blockers are used often in the treatment of hyperthyroidism
because of the presence of adrenergic signs and symptoms (eg, tachycardia,
palpitations, and diaphoresis). A retrospective analysis reported, however,
an increased frequency of rst-trimester miscarriages in mothers treated
for 6 to 12 weeks with a thionamide and propranolol (24%) compared
with a thionamide alone (5.5%) [70]. This dierence existed despite similar
thyroid hormone concentrations. Although this was a small study, the authors recommend avoiding b-adrenergic blocking agents if possible in the
rst trimester.
Iodides
Because of past reports of neonatal hypothyroidism after exposure to iodine, its use during pregnancy has been severely limited. Most published
work consists of case reports, however, in which euthyroid mothers were exposed to iodine-rich food, medicine, contrast media, or disinfectant agents
or hyperthyroid mothers were simultaneously treated with iodine and thionamides [71]. A study investigating the use of low-dose iodine (640 mg/d of
potassium iodide) for the treatment of Graves disease during pregnancy revealed improved maternal thyroid function and normal neonatal outcome
[71]. Specically, none of the neonates exposed to potassium iodide exhibited a goiter, and only 6% had an elevated TSH by way of the cord
blood. Although this evidence does not make iodine a rst-line therapy
for pregnant women with Graves disease, it does suggest that low-dose potassium iodide may be considered in special circumstances, such as in preparation for thyroidectomy or in thionamide-intolerant patients refusing
surgery.
Surgery
Subtotal thyroidectomy for the treatment of Graves disease during pregnancy is reserved for specic situations: when persistently high dosages of
thionamides (PTU O 600 mg/d, MMI O 40 mg/d) are required to control
maternal disease, if a patient is allergic or intolerant of both thionamides, if
a patient is noncompliant with medical therapy, or if compressive symptoms
occur in the mother because of goiter size [32]. Historically, it has been
recommended that surgery occur during the second trimester, before gestational week 24, in an attempt to minimize the risk of miscarriage [59]. Women who are persistently hyperthyroid before surgery should be prepared with
a b-adrenergic blocking agent and a 10- to 14-day course of potassium iodide with the intent of treating the hyperthyroid state and minimizing the
potential for perioperative disease exacerbation.
Radioactive iodine therapy
Radioactive iodine therapy is contraindicated for the treatment of
Graves disease during pregnancy. Fetal tissue is more radiosensitive than
130
adult tissue and may be susceptible to congenital defects during early development [72]. Additionally, the fetal thyroid gland begins to concentrate iodine after gestational week 10, predisposing the fetus/neonate to congenital
hypothyroidism [73,74].
131
132
state [88]. Nodules with benign cytology should be observed and followed by
an endocrinologist after delivery. Nodules with cytologic ndings consistent
with follicular neoplasm may require further evaluation after delivery with
radioactive iodine imaging, which is contraindicated during pregnancy,
and possibly surgery. If the ne-needle aspiration cytology is consistent
with thyroid cancer, surgery is recommended. Currently, there is no consensus on the appropriate timing of surgery. Some experts recommend operating during the second trimester, before 24 weeks gestation, to minimize the
risk of miscarriage [21]. Thyroid cancer discovered during pregnancy is not
more aggressive, however, than that diagnosed in a similar aged group of
nonpregnant women, leading other experts to advocate postponing denitive surgery until after delivery in most patients [89,90]. A retrospective
study of pregnant women with dierentiated thyroid cancer found there
to be no dierence in either recurrence or survival rates between women operated on during or after their pregnancy [90].
The authors have adopted a clinical practice inuenced by both schools
of thought. If a malignant nodule is discovered early in pregnancy, it should
be monitored with repeat ultrasound at midtrimester. If the nodule has
grown signicantly, surgery should be considered with the assistance of an
experienced anesthesiologist. Postoperative radioactive iodine therapy, if indicated, must be delayed until after the delivery. Conversely, if the nodule
size is stable, or the malignancy is not discovered until the second half of
pregnancy, surgery can occur after the patient has delivered. In all women
with cytology indicative of papillary thyroid cancer, regardless of surgical
timing, levothyroxine suppression therapy should be initiated to maintain
the serum TSH in the subnormal, but detectable, range (ie, 0.10.3 mU/L).
In addition, women with previously diagnosed or treated dierentiated
thyroid cancer require an increased levothyroxine dosage during pregnancy.
The recommended schedule for serum TSH testing is the same as that outlined for hypothyroid women (see Box 1). The authors adopt a therapeutic
target for serum TSH of 0.1 to 0.8 mU/L for patients with papillary or follicular thyroid cancer, depending on their risk of recurrence, and less than
2.5 mU/L for patients with medullary thyroid cancer.
Summary
Thyroid disorders are common among pregnant women. Diagnosis of
thyroid dysfunction is complicated, however, by nonspecic symptoms,
the hypermetabolic state of pregnancy, and normal gravid thyroid physiology, which results in alterations of maternal serum TSH and thyroxine concentrations. If untreated, hypothyroidism and hyperthyroidism may
adversely aect the mother and fetus. In cases of primary hypothyroidism,
maternal serum TSH concentrations should be monitored throughout pregnancy, and the levothyroxine dosage should be adjusted to maintain
133
maternal serum TSH levels at 2.5 mU/L or less. Conversely, in cases of true
hyperthyroidism (e.g., Graves disease), maternal thyroid function should be
evaluated every 2 to 4 weeks, and thionamide therapy (preferably PTU)
should be given, titrated to achieve maternal serum total T4 concentration within the upper one third of normal to slightly elevated range for pregnancy. Fetal ultrasonography and measurement of maternal serum TSH
receptor antibody concentrations should be employed to prevent fetal/neonatal complications. Finally, the evaluation of nodular thyroid disease and
cancer is similar in pregnant and nonpregnant patients. Use of radioactive
iodine is contraindicated during pregnancy, however, and should be postponed until after delivery.
References
[1] Alexander EK, Marqusee E, Lawrence J, et al. Timing and magnitude of increases in levothyroxine requirements during pregnancy in women with hypothyroidism. N Engl J Med
2004;351:241.
[2] Kaplan M. Monitoring thyroxine treatment during pregnancy. Thyroid 1992;2:147.
[3] Mandel SJ, Larsen PR, Seely EW, et al. Increased need for thyroxine during pregnancy in
women with primary hypothyroidism. N Engl J Med 1990;323:91.
[4] Glinoer D, Delange F, Laboureur I, et al. Maternal and neonatal thyroid function at birth in
an area of marginally low iodine intake. J Clin Endocrinol Metab 1992;75:800.
[5] Glinoer D, DeNayer P, Bourdoux P, et al. Regulation of maternal thyroid during pregnancy.
J Clin Endocrinol Metab 1990;71:276.
[6] Ain KB, Mori Y, Refeto S. Reduced clearance rate of thyroxine-binding globulin (TGB)
with increased sialylation: a mechanism for estrogen-induced elevation of serum TBG concentration. J Clin Endocrinol Metab 1987;65:689.
[7] Dworkin H, Jacquez J, Beierwalters W. Relationship of iodine ingestion to iodine excretion
in pregnancy. J Clin Endocrinol Metab 1966;26:1329.
[8] Burrow GN, Fisher DA, Larsen PR. Maternal and fetal thyroid function. N Engl J Med
1994;331:1072.
[9] Hershman JM. Physiological and pathological aspects of the eect of human chorionic gonadotropin on the thyroid. Best Pract Res Clin Endocrinol Metab 2004;18:249.
[10] Smits G, Govaerts C, Nubourgh I, et al. Lysine 183 and glutamic acid 157 of the TSH receptor: two interacting residues with a key role in determining specicity toward TSH and human CG. Mol Endocrinol 2002;16:722.
[11] Kimura M, Amino N, Tamaki H, et al. Physiologic thyroid activation in normal early pregnancy is induced by circulating hCG. Obstet Gynecol 1990;75:775.
[12] Mandel SJ, Spencer CA, Hollowell JG. Are detection and treatment of thyroid insuciency
in pregnancy feasible? Thyroid 2005;15:44.
[13] Glinoer D, DeNayer P, Robyn C, et al. Serum levels of intact human chorionic gonadotropin
(HCG) and its free a and b subunits, in relation to maternal thyroid stimulation during normal pregnancy. J Endocrinol Invest 1993;16:881.
[14] Haddow JE, Palomaki GE, Allan WC, et al. Maternal thyroid deciency during pregnancy
and subsequent neuropsychological development of the child. N Engl J Med 1999;341:549.
[15] Demers L, Spencer CA. Laboratory medicine practice guidelines: laboratory support for the
diagnosis and monitoring of thyroid disease. Thyroid 2003;13:6.
[16] Glinoer D. The regulation of thyroid function in pregnancy: pathways of endocrine adaptation from physiology to pathology. Endocr Rev 1997;18:404.
[17] Klein RZ, Haddow JE, Faix JD, et al. Prevalence of thyroid deciency in pregnant women.
Clin Endocrinol (Oxf) 1991;35:41.
134
[18] Neale D, Burrow G. Thyroid disease and pregnancy. Obstet Gynecol Clin North Am 2004;
31:893.
[19] Davis LE, Leveno KJ, Cunningham FG. Hypothyroidism complicating pregnancy. Obstet
Gynecol 1988;72:108.
[20] Montoro M, Collea JV, Frasier SD, et al. Successful outcome of pregnancy in women with
hypothyroidism. Ann Intern Med 1981;94:31.
[21] Mestman JH, Goodman TM, Montoro MM. Thyroid disorders of pregnancy. Endocrinol
Metab Clin North Am 1995;24:41.
[22] Leung AS, Millar LK, Koonings PP, et al. Perinatal outcome in hypothyroid pregnancies.
Obstet Gynecol 1993;81:349.
[23] Wasserstrum N, Anania CA. Perinantal consequences of maternal hypothyroidism in early
pregnancy and inadequate replacement. Clin Endocrinol (Oxf) 1995;42:353.
[24] Casey B, Dashe JS, Wells CE, et al. Subclinical hypothyroidism and pregnancy outcomes.
Obstet Gynecol 2005;105:239.
[25] Stagnaro-Green A, Chen X, Bogden JD, et al. The thyroid and pregnancy: a novel risk factor
for very preterm delivery. Thyroid 2005;15:351.
[26] Rotondi M, Caccavale C, DiSerio C, et al. Successful outcome of pregnancy in a thyroidectomized parathyroidectomized young woman aected by severe hypothyroidism. Thyroid
1999;9:1037.
[27] Fisher D. Endocrinology of fetal development. In: Wilson JD, Foster DW, editors. Textbook of endocrinology. Philadelphia: Saunders; 1999. p. 2073102.
[28] Fisher DA. Fetal thyroid function: diagnosis and managment of fetal thyroid disorders. Clin
Obstet Gynecol 1997;40:16.
[29] Morreale de Escobar G, Obregon MJ, Escobar del Rey F. Maternal thyroid hormones early
in pregnancy and fetal brain development. Best Pract Res Clin Endocrinol Metab 2004;18:
225.
[30] Man EB, Jones WS. Thyroid function in human pregnancy: V. incidence of maternal serum
low butanol-extractable iodines and of normal gestational TBG and TPBA capacities: retardation of 8-month-old infants. Am J Obstet Gynecol 1969;104(6):898908.
[31] Pop VJ, Brouwers EP, Vader HL, et al. Maternal hypothyroxinaemia during early pregnancy and subsequent child development: a 3-year follow-up study. Clin Endocrinol (Oxf)
2003;59:282.
[32] Mandel SJ. Thyroid disease and pregnancy. In: Copper DS, editor. Medical management of
thyroid disease. New York: Marcel Dekker; 2001. p. 387418.
[33] Smallridge RC, Ladenson PW. Hypothyroidism in pregnancy: consequences to neonatal
health. J Clin Endocrinol Metab 2001;86:2349.
[34] Hollowell JG, Staehling NW, Flanders WD, et al. Serum TSH, T4, and thyroid antibodies in
the United States population (1988 to 1994): National Health and Nutrition Examination
Survey (NHANES III). J Clin Endocrinol Metab 2002;82:489.
[35] Stagnaro-Green A, Glinoer D. Thyroid autoimmunity and the risk of miscarriage. Best Pract
Res Clin Endocrinol Metab 2004;18:167.
[36] Prummel MF, Wiersinga WM. Thyroid autoimmunity and miscarriage. Eur J Endocrinol
2004;150:751.
[37] Stagnaro-Green A, Roman SH, Cobin RH, et al. Detection of at-risk pregnancy by means of
highly sensitive assays for thyroid autoantibodies. JAMA 1990;264:1422.
[38] Glinoer D, Riahi M, Grun J, et al. Risk of subclinical hypothyroidism in pregnant
women with asymptomatic autoimmune thyroid disorders. J Clin Endocrinol Metab
1994;79:197.
[39] Glinoer D. Thyroid hyperfunction during pregnancy. Thyroid 1998;8:859.
[40] Millar LK, Wing DA, Leung AS, et al. Low birth weight and preeclampsia in pregnancies
complicated by hyperthyroidism. Obstet Gynecol 1994;84:946.
[41] Yoshikawa N, Nishikawa M, Horimoto M, et al. Thyroid-stimulating activity in sera of normal pregnant women. J Clin Endocrinol Metab 1989;69:891.
135
[42] Yoshimura M, Hershman JM. Thyrotropic action of human chorionic gonadotropin. Thyroid 1995;5:425.
[43] Goodwin TM, Montoro M, Mestman JH, et al. The role of chorionic gonadotropin in transient hyperthyroidism of hyperemesis gravidarum. J Clin Endocrinol Metab 1992;75:1333.
[44] Amino N, Tanizawa O, Mori H, et al. Aggravation of thyrotoxicosis in early pregnancy and
after delivery in Graves disease. J Clin Endocrinol Metab 1982;55:108.
[45] Goodwin GM, Montoro M, Mestman JH. Transient hyperthyroidism and hyperemesis
gravidarum: clinical aspects. Am J Obstet Gynecol 1992;167:648.
[46] Davis LE, Lucas MJ, Hankins GDV, et al. Thyrotoxicosis complicating pregnancy. Am J
Obstet Gynecol 1989;160:63.
[47] Mitsuda N, Tamaki H, Amino N, et al. Risk factors for developmental disorders in infants
born to women with Graves disease. Obstet Gynecol 1992;80:359.
[48] Momotani N, Ito K, Hamada N, et al. Maternal hyperthyroidism and congenital malformation in the ospring. Clin Endocrinol (Oxf) 1984;20:695.
[49] Wing DA, Millar LK, Koonings PP, et al. A comparison of propylthiouracil versus methimazole in the treatment of hyperthyroidism in pregnancy. Am J Obstet Gynecol 1994;170:90.
[50] Marchant B, Brownlie BEW, McKay Hart D, et al. The placental transfer of propylthiouracil, methimazole and carbimazole. J Clin Endocrinol Metab 1977;45:1187.
[51] Mortimer R, Cannell G, Addison R, et al. Methimazole and propylthiouracil equally cross
the perfused human term placental lobule. J Clin Endocrinol Metab 1997;82:3099.
[52] Lamberg BA, Ikonen E, Teramo K, et al. Treatment of maternal hyperthyroidism with antithyroid agents and changes in thyrotrophin and thyroxine in the newborn. Acta Endocrinol
(Copenh) 1981;97:186.
[53] Mortimer RH, Tyack SA, Galligan JP, et al. Graves disease in pregnancy: TSH receptor
binding inhibiting immunoglobulins and maternal and neonatal thyroid function. Clin Endocrinol (Oxf) 1990;32:141.
[54] Cheron RG, Kaplan MM, Larsen PR, et al. Neonatal thyroid function after propylthiouracil
therapy for maternal Graves disease. N Engl J Med 1981;304:525.
[55] Gardner DF, Cruikshank DP, Hays PM, et al. Pharmacology of propylthiouracil (PTU) in
pregnant hyperthyroid women: correlation of maternal PTU concentrations with cord serum
thyroid function test. J Clin Endocrinol Metab 1986;62(21):21720.
[56] Momotani N, Noh J, Oyanagi H, et al. Antithyroid drug therapy for Graves disease during
pregnancy. N Engl J Med 1986;315:24.
[57] Momotani N, Noh JY, Ishikawa N, et al. Eects of propylthiouracil and methimazole on
fetal thyroid status in mothers with graves hyperthyroidism. J Clin Endocrinol Metab
1997;82:3633.
[58] Peleg D, Cada S, Peleg A, et al. The relationship between maternal serum thyroid-stimulating immunoglobulin and fetal and neonatal thyrotoxicosis. Obstet Gynecol 2002;99:1040.
[59] Mestman JH. Hyperthyroidism in pregnancy. Best Pract Res Clin Endocrinol Metab 2004;
18:267.
[60] Ochoa-Maya M, Frates NC, Lee-Parritz A, et al. Resolution of fetal goiter after discontinuation of propylthiouracil in a pregnant woman with Graves hyperthyroidism. Thyroid
1999;9:1111.
[61] Davidson KM, Richards DS, Schatz DA, et al. Successful in utero treatment of fetal goiter
and hypothyroidism. N Engl J Med 1991;324:543.
[62] Van Loon AJ, Derksen J, Bos AF, et al. In utero diagnosis and treatment of fetal goitrous
hypothyroidism, caused by maternal use of propylthiouracil. Prenat Diagn 1995;15:599.
[63] Burrow GN, Klatskin EH, Genel M. Intellectual development in children whose mothers received propylthiouracil during pregnancy. Yale J Biol Med 1978;51:151.
[64] Eisenstein Z, Weiss M, Katz Y, et al. Intellectual capacity of subjects exposed to methimazole or propylthiouracil in utero. Eur J Pediatr 1992;1:558.
[65] McCarroll AM, Hutchinson M, McAuley R, et al. Long-term assessment of children exposed in utero to carbimazole. Arch Dis Child 1976;51:532.
136
[66] Messer PM, Haua BP, Olbricht T, et al. Antithyroid drug treatment of Graves disease in
pregnancy: long-term eects on somatic growth, intellectual development and thyroid function of the ospring. Acta Endocrinol (Copenh) 1990;123:311.
[67] Clementi M, Gianantonio ED, Pelo E, et al. Methimazole embryopathy: delineation of the
phenotype. Am J Med Genet 1999;83:43.
[68] Mandel SJ, Brent GA, Larsen PR. Review of antithyroid drug use during pregnancy and report of a case of aplasia cutis. Thyroid 1994;4:129.
[69] Mestman JH. Hyperthyroidism in pregnancy. Clin Obstet Gynecol 1997;40:45.
[70] Sherif IH, Oyan WT, Bosairi S, et al. Treatment of hyperthyroidism in pregnancy. Acta Obstet Gynecol Scand 1991;70:461.
[71] Momotani N, Hisaoka T, Noh J, et al. Eects of iodine on thyroid status of fetus versus
mother in treatment of Graves disease complicated by pregnancy. J Clin Endocrinol Metab
1992;75:738.
[72] Masiukiewicz US, Burrow GN. Hyperthyroidism in pregnancy: diagnosis and treatment.
Thyroid 1999;9:647.
[73] Evans P, Webster J, Evans W, et al. Radioiodine treatments in unsuspected pregnancy. Clin
Endocrin 1998;48:281.
[74] Stoer SS, Hamburger JI. Inadvertent 131I therapy for hyperthyroidism in the rst trimester
of pregnancy. J Nucl Med 1976;17:146.
[75] Burrow GN. Thyroid function and hyperfunction during gestation. Endocr Rev 1993;14:
194.
[76] Polak M, Le Gac I, Vuillard E, et al. Fetal and neonatal thyroid function in relation to maternal Graves disease. Best Pract Res Clin Endocrinol Metab 2004;18:289.
[77] Matsuura N, Fujiedu K, Iida Y, et al. TSH-receptor antibodies in mothers with Graves disease and outcome in their ospring. Lancet 1988;331:14.
[78] Kilpatrick S. Umbilical blood sampling in women with thyroid disease in pregnancy: is it
necessary? Obstet Gynecol 2003;189:1.
[79] Skuza KA, Sills IN, Stene M, et al. Prediction of neonatal hyperthyroidism in infants born to
mothers with Graves disease. J Pediatr 1996;128:264.
[80] Stoer R, Welsh J, Hellwig C, et al. Nodular goiter: incidence morphology before and after
iodine prophylaxis, and clinical diagnosis. Arch Intern Med 1960;106:10.
[81] Vander JB, Gaston EA, Dawber TR. The signicance of nontoxic thyroid nodules. Ann Intern Med 1968;69:537.
[82] Hintze G, Windeler J, Baumert J, et al. Thyroid volume and goiter prevalence in the elderly
as determined by ultrasound and their relationships to laboratory indices. Acta Endocrinol
(Copenh) 1991;124:12.
[83] Knudsen N, Perrild H, Christiansen E, et al. Thyroid structure and size and two year followup of solitary cold thyroid nodules in an unselected population with borderline iodine deciency. Eur J Endocrinol 2000;142:224.
[84] Mazzaferri EL. Management of a solitary thyroid nodule. N Engl J Med 1993;328:553.
[85] Struve CW, Haupt S, Ohlen S. Inuence of frequency of previous pregnancies on the prevalence of thyroid nodules in women without clinical evidence of thyroid disease. Thyroid
1993;3:7.
[86] Kung A, Chau M, Lao T, et al. The eect of pregnancy on thyroid nodule formation. J Clin
Endocrinol Metab 2002;87:1010.
[87] Mandel SJ. A 64-year-old woman with a thyroid nodule. JAMA 2004;292:2632.
[88] Tan GH, Gharib H, Goellner JR, et al. Management of thyroid nodules in pregnancy. Arch
Intern Med 1996;156:2317.
[89] Herzon FS, Morris DM, Segal MN, et al. Coexistent thyroid cancer and pregnancy. Arch
Otolaryngol Head Neck Surg 1994;120:1191.
[90] Moosa M, Mazzaferri EL. Outcome of dierentiated thyroid cancer diagnosed in pregnant
women. J Clin Endocrinol Metab 1997;82:2862.
Polycystic ovary syndrome (PCOS) is the most common cause of anovulatory infertility in the United States, aecting 5% to 10% of women of reproductive age [1,2]. It is typically characterized by hyperandrogenism and
chronic anovulation. Hyperandrogenism may present as hirsutism, acne, or
male-pattern alopecia. Anovulation manifests as irregular menstrual cycles,
usually amenorrhea or oligomenorrhea, and infertility [3]. In the past, abnormally regulated luteinizing hormone (LH) levels alone were believed to
account for these symptoms. However, recent evidence suggests that hyperinsulinemic insulin resistance plays a pathogenic role in PCOS by increasing
circulating ovarian androgen concentrations, perhaps via P450c17a stimulation [4,5], thus impeding ovulation [68]. Obese as well as lean women with
PCOS manifest insulin resistance independent of fat mass [6,9]. Presumably
as a result of their insulin resistance, women with PCOS have also been
found to be at increased risk for the development of type 2 diabetes mellitus
[10], dyslipidemia [11], and possibly cardiovascular disease.
The presence of hyperinsulinemic insulin resistance in PCOS creates an
interesting paradox because peripheral tissues, such as skeletal muscle, are
resistant to insulin in terms of glucose metabolism, whereas the ovary remains sensitive to insulin with regard to stimulation of testosterone biosynthesis, suggesting tissue-specic dierences in insulin sensitivity. The exact
underlying mechanism of insulin resistance in PCOS remains poorly characterized. Although recent studies have implicated a post-insulin receptor
138
defect as the likely mechanism [1215], further work must be done to elucidate a specic target. Of note, attention is concurrently being focused on the
possibility of PCOS as a genetic disorder, expressing itself only after interaction with other environmental factors [1618].
139
Aside from weight loss, other therapies for anovulatory infertility that
target insulin resistance have become widely popular in recent years because
of the growing recognition that insulin resistance likely hampers fertility and
decreases the ecacy of other pharmacologic therapies in PCOS. Of the
insulin-sensitizing drugs, metformin, an oral biguanide used to treat type
2 diabetes mellitus, is the most comprehensively evaluated drug, with proven
eectiveness in lean as well as obese women with PCOS [4,5,23]. An
improvement in insulin sensitivity and reduction in circulating insulin by
metformin may enhance ovulation in PCOS by decreasing intraovarian concentrations of androgens, normalizing gonadotropin secretory dynamics, or
a combination of these and other processes.
The ecacy of metformin monotherapy in promoting ovulation and
fertility in PCOS was recently conrmed in a meta-analysis by Lord and
colleagues [24]. The meta-analysis consisted of 13 randomized controlled
clinical trials that included 428 women with PCOS. Ovulation was achieved
in 46% of women with PCOS who received metformin compared with 24%
who received placebo (odds ratio [OR] 3.88, 95% condence interval [CI]:
2.256.69) [24]. Subsequently, a pilot study comparing metformin and lifestyle modication showed no dierence in ovulation rates between the
two groups; however, the study had a 39% dropout rate, potentially biasing
the results [25]. A further study showed that the addition of metformin to
a hypocaloric diet in obese women with PCOS resulted in improvement in
menstrual irregularity when compared with the placebo and hypocaloric
diet arm [26]. Similarly, in clomiphene-resistant patients, the combination
of clomiphene and metformin has been shown to improve ovulation [27]
and pregnancy rates [24,28] when compared with clomiphene alone.
Newer insulin sensitizers known as the thiazolidinediones (TZDs) have
also received attention with regard to their potential role in the treatment
of PCOS. Troglitazone, the rst drug of this class, has been shown to improve ovulation rates, hyperandrogenemia, and insulin resistance compared
with placebo [29]. Its mechanism of action, although partially systemic, is
also thought to involve direct ovarian action because thecal cells possess
the peroxisome proliferatoractivated receptor-g (PPARg) nuclear receptors that these drugs target. In thecal cell cultures, troglitazone impedes
stimulation of androgen biosynthesis by the combination of LH and insulin [30]. However, troglitazone was withdrawn from the worldwide market
because of hepatocellular toxicity.
Rosiglitazone is another TZD that is commercially available, and it has
also been shown to normalize menses and improve insulin resistance in
women with PCOS [31]. Additionally, when rosiglitazone was administered
to nonobese women who had PCOS with normal insulin sensitivity, ovulation frequency and androgen levels improved compared with placebo even
though insulin parameters did not change [32]. This supports the theory
that TZDs as a class may have direct inhibitory eects on ovarian androgen
biosynthesis. Pioglitazone, the most recently developed TZD, also improved
140
141
well, including plasma concentrations of triglycerides, small dense low-density lipoprotein (LDL) particles, free fatty acids, plasminogen activator
inhibitor-1 (PAI-1), tissue plasminogen activator antigen (TPA Ag), vascular
cell adhesion molecules (VCAMs), leptin, and tumor necrosis factora
(TNF-a) [39]. Thus, normal pregnancy itself is a state of heightened insulin resistance. Further, in women with PCOS, pregnancy may augment the
preexisting insulin resistance and hyperinsulinemia, resulting in fatal and
nonfatal complications.
142
and obese women who conceived after IVF or intracytoplasmic sperm injection (ICSI) to determine their miscarriage rates. Obese women were again
found to have an increased risk of EPL (22% versus 12%; P .03) and a decreased live birth rate compared with lean women. Obesity, characterized by
hyperinsulinemic insulin resistance, was shown to be an independent risk factor for EPL [51].
Investigators have examined the eects of insulin-sensitizing drugs on the
rate of miscarriage in PCOS. Glueck and colleagues [52] performed a prospective cohort pilot study of women with PCOS, comparing those treated
with metformin before conception who continued the drug throughout pregnancy with historical controls who did not receive metformin. Results
showed that the rate of rst trimester pregnancy loss was 39% in the historical controls but only 11% in the metformin group. Ten women in the metformin group had had previous pregnancies while not receiving metformin.
In this subcohort, the miscarriage rate decreased from 73% to 10% with the
addition of metformin (P ! .002). Overall, no evidence of teratogenicity
was noted in the metformin group. Before metformin therapy, fasting serum
insulin levels correlated positively with PAI activity (r 0.60, P .004).
Over a median treatment period of 6 months (before conception), a reduction in fasting serum insulin was also positively correlated with a reduction
in PAI activity (r 0.65, P .04), suggesting that hyperinsulinemia and
PAI activity may be closely related; perhaps both can help to explain the etiology of EPL in PCOS.
Glueck and coworkers [53] went on to complete a follow-up prospective
study conrming that EPL decreased from 62% to 26% (P ! .0001) when
women with PCOS continued metformin throughout pregnancy. Again, no
teratogenic adverse events occurred. In this study, PAI activity was again
lower on metformin compared with premetformin entry levels. However,
logistic regression revealed that entry fasting serum insulin was the only signicant variable associated with total (previous and current) rst trimester
miscarriage (OR 1.32 for each 5-mIU/mL increment in insulin, 95% CI:
1.091.60, P .005).
A larger retrospective study of women with PCOS performed by an independent group also showed a decrease in EPL in those who continued metformin throughout pregnancy compared with controls (8.8% versus 42%;
P ! .001) [40]. This trend remained signicant in the subgroup analysis
of women with a prior history of miscarriage. No adverse fetal outcomes
were noted in the metformin group, except for 1 of 62 live births that suffered from achondroplasia, a hereditary condition thought to be unrelated
to metformin. Additionally, those women who remained on metformin
had signicantly decreased serum androgen concentrations and improved
insulin sensitivity during pregnancy compared with controls; both are plausible explanations for the decrease in miscarriage rate.
Given the consistency among these studies, insulin resistance seems to
play an important role in EPL in women with PCOS. It is plausible that
143
144
145
146
between those with and without pregestational insulin resistance, one might
consider other determinants of PCOS as possible explanations.
The hypothesis that PCOS is associated with GDM has not been supported by all studies. For example, a retrospective cohort study by Haakova
and coworkers [67] compared pregnant women with PCOS with age- and
weight-matched pregnant controls and found no statistical dierence in
the development of GDM. Nevertheless, consistency among many studies
combined with their large magnitudes of eect is supportive evidence in favor of insulin resistance causing women with PCOS to be at increased risk
for developing GDM. This eect has been noted independent of age and
BMI. Several reports prospectively document insulin resistance preceding
GDM, implying a cause-eect relation. Further, the association is biologically feasible because insulin resistance is the main oender implicated in
type 2 diabetes mellitus.
Consequences of GDM are signicant and include the long-term risk of
developing diabetes later in life as well as signicant immediate perinatal
risks, including macrosomia, hypoglycemia, stillbirth, jaundice, and respiratory distress syndrome. Rates of birth trauma and cesarean section are also
increased. Another potential complication is the development of pregnancyinduced hypertensive disorders (PIHDs), such as preeclampsia and gestational hypertension. A recent case-control study examined this relation
and found that women with GDM have a 1.5-fold increased risk of developing PIHDs [68]. Ethnicity was found to modify the association, with the
greatest risk existing among black women [68].
Given the signicant attendant complications, some investigators have
examined potential drug therapy to prevent the onset of GDM in women
with PCOS. One study compared women with PCOS who conceived while
taking metformin and continued taking it throughout pregnancy with
women with PCOS who conceived without metformin therapy [58]. GDM developed in 3% of the metformin pregnancies compared with 31% of the
nonmetformin pregnancies (OR 0.093, 95% CI: 0.0110.795; P .03).
After adjustment for age at delivery, metformin was associated with a 10fold reduction in GDM in women with PCOS. Further, metformin was
shown to decrease BMI, insulin levels, insulin resistance, and insulin secretion; these eects were maintained throughout pregnancy. Metformin did
not result in any major fetal malformations or fetal hypoglycemia, again
supporting its safety in pregnancy. Nevertheless, the utility of metformin administration to women with PCOS during pregnancy to prevent development of GDM has not been rigorously tested in a randomized clinical trial.
Pregnancy-induced hypertensive disorders
PIHDs complicate 3% to 5% of pregnancies in previously normotensive
women [39] and usually develop during the third trimester. Although hypertension in these women typically resolves by 6 weeks postpartum, they are at
147
148
149
with controls (11.5% versus 0.3%; P ! .05), and preeclampsia was found
more frequently among insulin-resistant patients with PCOS than in noninsulin-resistant patients with PCOS (22% versus 7%; P ! .05) and controls. BMI was similar among the insulin-resistant and noninsulin-resistant
women with PCOS. This was the rst prospective study in women with
PCOS to support the association between hyperinsulinemia and PIHDs
proposed earlier by other investigators.
Nonetheless, some of the available data do not support the proposed hypothesis. A study by Mikola and coworkers [64] found no signicant dierence in the incidence of preeclampsia between women with PCOS and
a normal control population. After multiple logistic regression, nulliparity
was the only signicant risk factor for preeclampsia; BMI greater than
25 kg/m2, multiple pregnancy, and PCOS had no predictive value. When
de Vries and colleagues [75] matched cases and controls for parity, however,
women with PCOS maintained a signicant association with preeclampsia.
A more recent study by Haakova and coworkers [67] also retrospectively analyzed the relationship between women with PIHDs and women with
PCOS. Although a trend toward increased PIHDs occurred in the women
with PCOS, no statistically signicant dierence was found when compared
with age- and weight-matched controls.
Clearly, the limited data on the association between PCOS and PIHDs
are conicting. The eects of age, BMI, ovulation induction, and parity
have been studied, however, and the magnitude of association among positive studies remains quite large.
Taken one step further, the impact of insulin resistance in women with
PCOS who develop preeclampsia has also been examined, suggesting a possible mechanistic link. Several reports document hyperinsulinemia or hyperglycemia developing before preeclampsia, providing support for insulin
resistance in disease pathogenesis [69,77,78]. Additionally, insulin resistance
or its associated features have been shown to occur many years after a preeclamptic rst pregnancy [7982]. Persistence of insulin resistance postpartum
further suggests a role for its causality in the development of preeclampsia
[82]. Laivuori and colleagues [83] found an association between preeclamptic
pregnancy and the existence of hyperandrogenemia 17 years after delivery,
suggesting that these women could have had hyperinsulinemic PCOS during
their reproductive years (although all reportedly had normal menstruation)
and may be at risk for its metabolic consequences after menopause.
Several theoretic mechanisms exist whereby hyperinsulinemia could predispose to PIHDs in women with PCOS. For example, hyperinsulinemia
may increase renal sodium reabsorption [84] and stimulate the sympathetic
nervous system [85]. Insulin resistance or associated hyperglycemia may impair endothelial function [86]. Endothelial dysfunction may decrease prostacyclin production [87]. Studies in animal models have shown that androgens
increase vasoconstriction in response to pressors [88] and also decrease the
synthesis of prostacyclin [89], potentially playing a further role in the
150
development of PIHDs. Interestingly, the plasma total renin level was found
to be higher in normotensive women with PCOS compared with healthy
controls, independent of insulin resistance [90]. Although many possible theories exist, more studies need to be done to conrm a mechanistic link between hyperinsulinemia and preeclampsia.
Preeclampsia may have acute and chronic consequences. Acute problems
primarily revolve around delivery complications and increased perinatal
morbidity. Chronic eects, however, are more dicult to study because
women who develop these problems often have a remote diagnosis of
PIHD that is dicult to ascertain [39]. One study was able to show that
the risk of hypertension is increased almost threefold after 2 to 24 years
of follow-up in women with a history of preeclampsia or eclampsia compared with controls [91]. Other long-term consequences that have been reported include hypertriglyceridemia, hyperuricemia, hyperandrogenemia,
and abnormal brachial artery ow-mediated (endothelium-dependent) dilatation [39], consistent with the eects of insulin resistance.
Given this increase in cardiovascular risk factors, some investigators have
attempted to determine if mortality attributable to cardiovascular causes is
increased among women with a prior history of preeclampsia. One study
found that mortality attributable to cardiovascular causes was increased
among Norwegian women who had preeclampsia and preterm delivery
(ie, severe preeclampsia) compared with preeclamptic women with term delivery or women with preterm delivery alone [92]. Another study found that
women with a discharge diagnosis of preeclampsia were twice as likely to be
admitted to the hospital for or to die from ischemic heart disease compared
with women with uncomplicated pregnancies [93]. These ndings suggest
that preeclampsia may serve as a marker for future cardiovascular disease.
Whether this association is independent of obesity and other predictors of
cardiovascular risk still remains to be seen.
Given the available evidence, it is possible to imagine that interventions
targeted at improving insulin sensitivity may decrease the likelihood of preeclampsia and later life complications, including risk of cardiovascular
events. For instance, weight reduction before pregnancy and avoidance of
excessive weight gain during pregnancy may contribute to improved insulin
sensitivity and decreased risk for developing preeclampsia. When lifestyle
modication cannot be maintained, however, incentive to use insulin-sensitizing drug therapy, such as metformin, may exist. Studies that examine the
eect of metformin on the incidence of preeclampsia and its complications
in women with PCOS are currently needed.
Future directions
In summary, infertility in PCOS has multiple facets (eg, anovulation, decreased implantation), and several treatment options exist including those
that target insulin resistance. Once pregnancy is achieved, however, women
151
are faced with several potential complications, including EPL, GDM, and
PIHDs. All these disorders are potentially linked by their association with
hyperinsulinemia and insulin resistance. Hence, it is notable that preliminary studies suggest that metformin, which is an insulin-sensitizing agent
that is a pregnancy class B drug, may decrease the risk of EPL and GDM
in women with PCOS. Furthermore, given the long-term metabolic consequences of PCOS, including type 2 diabetes, hyperlipidemia, and cardiovascular disease, some experts speculate as to whether administering metformin
during pregnancy may not only prevent pregnancy complications but also
retard or prevent some of these long-term health risks. In fact, the occurrence of EPL, GDM, and PIHDs may be identied as predictors of cardiovascular risk in women with PCOS.
Although it is reassuring that metformin administration during pregnancy
appears to be safe, rigorous randomized controlled trials must be conducted
to conrm metformins ecacy and safety under these conditions before
its use during pregnancy can be recommended.
References
[1] Knochenhauer ES, Key TJ, Kahsar-Miller M, et al. Prevalence of the polycystic ovary syndrome in unselected black and white women of the southeastern United States: a prospective
study. J Clin Endocrinol Metab 1998;83(9):307882.
[2] Asuncion M, Calvo RM, San Millan JL, et al. A prospective study of the prevalence of the
polycystic ovary syndrome in unselected Caucasian women from Spain. J Clin Endocrinol
Metab 2000;85(7):24348.
[3] Franks S. Polycystic ovary syndrome. N Engl J Med 1995;333(13):85361.
[4] Nestler JE, Jakubowicz DJ. Decreases in ovarian cytochrome P450c17alpha activity and
serum free testosterone after reduction of insulin secretion in polycystic ovary syndrome.
N Engl J Med 1996;335(9):61723.
[5] Nestler JE, Jakubowicz DJ. Lean women with polycystic ovary syndrome respond to insulin
reduction with decreases in ovarian P450c17alpha activity and serum androgens. J Clin
Endocrinol Metab 1997;82(12):40759.
[6] Chang RJ, Nakamura RM, Judd HL, et al. Insulin resistance in nonobese patients with polycystic ovarian disease. J Clin Endocrinol Metab 1983;57(2):3569.
[7] Franks S, Gilling-Smith C, Watson H, et al. Insulin action in the normal and polycystic ovary.
Endocrinol Metab Clin North Am 1999;28(2):36178.
[8] Dunaif A. Insulin resistance and the polycystic ovary syndrome: mechanism and implications for pathogenesis. Endocr Rev 1997;18(6):774800.
[9] Dunaif A, Segal KR, Futterweit W, et al. Profound peripheral insulin resistance, independent of obesity, in polycystic ovary syndrome. Diabetes 1989;38(9):116574.
[10] Legro RS, Kunselman AR, Dodson WC, et al. Prevalence and predictors of risk for type 2
diabetes mellitus and impaired glucose tolerance in polycystic ovary syndrome: a prospective,
controlled study in 254 aected women. J Clin Endocrinol Metab 1999;84(1):1659.
[11] Talbott E, Clerici A, Berga SL, et al. Adverse lipid and coronary heart disease risk proles in
young women with polycystic ovary syndrome: results of a case-control study. J Clin Epidemiol 1998;51(5):41522.
[12] Rosenbaum D, Haber RS, Dunaif A. Insulin resistance in polycystic ovary syndrome: decreased expression of GLUT-4 glucose transporters in adipocytes. Am J Physiol 1993;264:
E197202.
152
[13] Dunaif A, Wu X, Lee A, et al. Defects in insulin receptor signaling in vivo in the
polycystic ovary syndrome (PCOS). Am J Physiol Endocrinol Metab 2001;281(2):
E3929.
[14] Book CB, Dunaif A. Selective insulin resistance in the polycystic ovary syndrome. J Clin
Endocrinol Metab 1999;84(9):31106.
[15] Nestler JE, Jakubowicz DJ, Reamer P, et al. Ovulatory and metabolic eects of D-chiroinositol in the polycystic ovary syndrome. N Engl J Med 1999;340(17):131420.
[16] Legro RS, Driscoll D, Strauss JF, et al. Evidence for a genetic basis for hyperandrogenemia
in polycystic ovary syndrome. Proc Natl Acad Sci USA 1998;95(25):1495660.
[17] Franks S, McCarthy M. Genetics of ovarian disorders: polycystic ovary syndrome. Rev Endocr Metab Disord 2004;5(1):6976.
[18] Franks S, Gharani N, Waterworth D, et al. Genetics of polycystic ovary syndrome. Mol Cell
Endocrinol 1998;145(12):1238.
[19] Homburg R. The management of infertility associated with polycystic ovary syndrome. Reprod Biol Endocrinol 2003;(1):109.
[20] Jakubowicz DJ, Essah PA, Seppala M, et al. Reduced serum glycodelin and insulin-like
growth factor-binding protein-1 in women with polycystic ovary syndrome during rst trimester of pregnancy. J Clin Endocrinol Metab 2004;89(2):8339.
[21] Norman RJ. Metformindcomparison with other therapies in ovulation induction in polycystic ovary syndrome [editorial]. J Clin Endocrinol Metab 2004;89(10):4797800.
[22] Parsanezhad ME, Alborzi S, Motazedian S, et al. Use of dexamethasone and clomiphene citrate in the treatment of clomiphene citrate-resistant patients with polycystic ovary syndrome
and normal dehydroepiandrostenedione sulfate levels: a prospective, double-blind, placebocontrolled trial. Fertil Steril 2002;78(5):10014.
[23] Fleming R, Hopkinson ZE, Wallace M, et al. Ovarian function and metabolic factors in
women with oligomenorrhea treated with metformin in a randomized double-blind placebocontrolled trial. J Clin Endocrinol Metab 2002;87(2):56974.
[24] Lord JM, Flight I, Norman RJ. Metformin in polycystic ovary syndrome: systematic review
and meta-analysis. BMJ 2003;327(7421):9515.
[25] Hoeger KM, Kochman L, Wixom N, et al. A randomized, 48-week, placebo-controlled trial
of intensive lifestyle modication and/or metformin therapy in overweight women with polycystic ovary syndrome: a pilot study. Fertil Steril 2004;82(2):4219.
[26] Pasquali R, Gambineri A, Biscotti D, et al. Eect of long-term treatment with metformin
added to hypocaloric diet on body composition, fat distribution, and androgen and insulin
levels in abdominally obese women with and without the polycystic ovary syndrome. J Clin
Endocrinol Metab 2000;85(8):276774.
[27] Nestler JE, Jakubowicz DJ, Evans WS, et al. Eects of metformin on spontaneous and clomiphene-induced ovulation in the polycystic ovary syndrome. N Engl J Med 1998;338(26):
187680.
[28] Kashyap S, Wells GA, Rosenwaks Z. Insulin-sensitizing agents as primary therapy for patients with polycystic ovary syndrome. Hum Reprod 2004;19(11):247483.
[29] Azziz R, Ehrmann D, Legro RS, et al. Troglitazone improves ovulation and hirsutism in the
polycystic ovary syndrome: a multicenter, double-blind, placebo-controlled trial. J Clin Endocrinol Metab 2001;86(4):162632.
[30] Veldhuis J, Zhang G, Garmey J. Troglitazone, an insulin-sensitizing thiazolidinedione, represses combined stimulation by LH and insulin of de novo androgen biosynthesis by thecal
cells in vitro. J Clin Endocrinol Metab 2002;87(3):112933.
[31] Belli SH, Gragna MN, Oneto A, et al. Eect of rosiglitazone on insulin resistance, growth
factors, and reproductive disturbances in women with polycystic ovary syndrome. Fertil
Steril 2004;81(3):6249.
[32] Baillargeon JP, Jakubowicz DJ, Iuorno MJ, et al. Eects of metformin and rosiglitazone,
alone and in combination, in nonobese women with polycystic ovary syndrome and normal
indices of insulin sensitivity. Fertil Steril 2004;82(4):893902.
153
[33] Brettenthaler N, De Geyter C, Huber PR, et al. Eect of the insulin sensitizer pioglitazone on
insulin resistance, hyperandrogenism, and ovulatory dysfunction in women with polycystic
ovary syndrome. J Clin Endocrinol Metab 2004;89(8):383540.
[34] Glueck CJ, Moreira A, Goldenberg N, et al. Pioglitazone and metformin in obese women
with the polycystic ovary syndrome not optimally responsive to metformin. Hum Reprod
2003;18(8):161825.
[35] Saleh AM, Khalil HS. Review of nonsurgical and surgical treatment and the role of insulinsensitizing agents in the management of infertile women with polycystic ovary syndrome.
Acta Obstet Gynecol Scand 2004;83(7):61421.
[36] Farquhar C, Lilford R, Marjoribanks J, et al. Laparoscopic drilling by diathermy or laser
for ovulation induction in anovulatory polycystic ovary syndrome. Cochrane Database Syst
Rev 2005;(3):CD001122.
[37] Palomba S, Orio F Jr, Nardo LG, et al. Metformin administration versus laparoscopic ovarian diathermy in clomiphene citrate-resistant women with polycystic ovary syndrome: a prospective parallel randomized double-blind placebo-controlled trial. J Clin Endocrinol Metab
2004;89(10):48019.
[38] Urman B, Tiras B, Yakin K. Assisted reproduction in the treatment of polycystic ovary syndrome. Reprod Biomed Online 2004;8(4):41930.
[39] Seely EW, Solomon CG. Insulin resistance and its potential role in pregnancy-induced hypertension. J Clin Endocrinol Metab 2003;88(6):23938.
[40] Jakubowicz DJ, Iuorno MJ, Jakubowicz S, et al. Eects of metformin on early pregnancy
loss in the polycystic ovary syndrome. J Clin Endocrinol Metab 2002;87(2):5249.
[41] Regan L, Owen EJ, Jacobs HS. Hypersecretion of luteinising hormone, infertility, and miscarriage. Lancet 1990;336(8724):11414.
[42] Watson H, Kiddy DS, Hamilton-Fairley D, et al. Hypersecretion of luteinizing hormone
and ovarian steroids in women with recurrent early miscarriage. Hum Reprod 1993;8(6):
82933.
[43] Homburg R, Armar NA, Eshel A, et al. Inuence of serum luteinising hormone concentrations on ovulation, conception, and early pregnancy loss in polycystic ovary syndrome. BMJ
1988;297(6655):10246.
[44] Balen AH, Tan S, MacDougall J, et al. Miscarriage rates following in-vitro fertilization are
increased in women with polycystic ovaries and reduced by pituitary desensitization with buserelin. Hum Reprod 1993;8(6):95964.
[45] Homburg R, Levy T, Berkovitz D, et al. Gonadotropin-releasing hormone agonist reduces
the miscarriage rate for pregnancies achieved in women with polycystic ovarian syndrome.
Fertil Steril 1993;59(3):52731.
[46] Cliord K, Rai R, Watson H, et al. Does suppressing luteinising hormone secretion reduce
the miscarriage rate? Results of a randomised controlled trial. BMJ 1996;312(7045):
150811.
[47] Okon MA, Laird SM, Tuckerman EM, et al. Serum androgen levels in women who have recurrent miscarriages and their correlation with markers of endometrial function. Fertil Steril
1998;69(4):68290.
[48] Tulppala M, Stenman UH, Cacciatore B, et al. Polycystic ovaries and levels of gonadotrophins and androgens in recurrent miscarriage: prospective study in 50 women. Br J Obstet
Gynaecol 1993;100(4):34852.
[49] Glueck CJ, Wang P, Fontaine RN, et al. Plasminogen activator inhibitor activity: an independent risk factor for the high miscarriage rate during pregnancy in women with polycystic
ovary syndrome. Metabolism 1999;48(12):158995.
[50] Hamilton-Fairley D, Kiddy D, Watson H, et al. Association of moderate obesity with a poor
pregnancy outcome in women with polycystic ovary syndrome treated with low dose gonadotrophin. Br J Obstet Gynaecol 1992;99(2):12831.
[51] Fedorcsak P, Storeng R, Dale PO, et al. Obesity is a risk factor for early pregnancy loss after
IVF or ICSI. Acta Obstet Gynecol Scand 2000;79(1):438.
154
[52] Glueck CJ, Phillips H, Cameron D, et al. Continuing metformin throughout pregnancy in
women with polycystic ovary syndrome appears to safely reduce rst-trimester spontaneous
abortion: a pilot study. Fertil Steril 2001;75(1):4652.
[53] Glueck CJ, Wang P, Goldenberg N, et al. Pregnancy outcomes among women with polycystic ovary syndrome treated with metformin. Hum Reprod 2002;17(11):285864.
[54] Hellmuth E, Damm P, Molsted-Pedersen L. Oral hypoglycaemic agents in 118 diabetic pregnancies. Diabet Med 2000;17(7):50711.
[55] Glueck CJ, Goldenberg N, Praniko J, et al. Height, weight, and motor-social development
during the rst 18 months of life in 126 infants born to 109 mothers with polycystic ovary
syndrome who conceived on and continued metformin through pregnancy. Hum Reprod
2004;19(6):132330.
[56] Coetzee EJ, Jackson WP. The management of non-insulin-dependent diabetes during pregnancy. Diabetes Res Clin Pract 1985;1(5):2817.
[57] Ferrara A, Hedderson MM, Quesenberry CP, et al. Prevalence of gestational diabetes mellitus detected by the National Diabetes Data Group or the Carpenter and Coustan plasma
glucose thresholds. Diabetes Care 2002;25(9):162530.
[58] Glueck CJ, Wang P, Kobayashi S, et al. Metformin therapy throughout pregnancy reduces
the development of gestational diabetes in women with polycystic ovary syndrome. Fertil
Steril 2002;77(3):5205.
[59] Ferrara A, Kahn HS, Quesenberry CP, et al. An increase in the incidence of gestational diabetes mellitus: Northern California, 19912000. Obstet Gynecol 2004;103(3):52633.
[60] Lanzone A, Caruso A, Di Simone N, et al. Polycystic ovary disease: a risk factor for gestational diabetes? J Reprod Med 1995;40(4):3126.
[61] Holte J, Gennarelli G, Wide L, et al. High prevalence of polycystic ovaries and associated
clinical, endocrine, and metabolic features in women with previous gestational diabetes mellitus. J Clin Endocrinol Metab 1998;83(4):114350.
[62] Anttila L, Karjala K, Penttila TA, et al. Polycystic ovaries in women with gestational diabetes. Obstet Gynecol 1998;92(1):136.
[63] Radon PA, McMahon MJ, Meyer WR. Impaired glucose tolerance in pregnant women with
polycystic ovary syndrome. Obstet Gynecol 1999;94(2):1947.
[64] Mikola M, Hiilesmaa V, Halttunen M, et al. Obstetric outcome in women with polycystic
ovary syndrome. Hum Reprod 2001;16(2):2269.
[65] Urman B, Sarac E, Dogan L, et al. Pregnancy in infertile PCOD patients. J Reprod Med
1997;42(8):5015.
[66] Bjercke S, Dale PO, Tanbo T, et al. Impact of insulin resistance on pregnancy complications
and outcome in women with polycystic ovary syndrome. Gynecol Obstet Invest 2002;54(2):
948.
[67] Haakova L, Cibula D, Rezabek K, et al. Pregnancy outcome in women with PCOS and in
controls matched by age and weight. Hum Reprod 2003;18(7):143841.
[68] Bryson CL, Ioannou GN, Rulyak SJ, et al. Association between gestational diabetes and
pregnancy-induced hypertension. Am J Epidemiol 2003;158(12):114853.
[69] Joe GM, Esterlitz JR, Levine RJ, et al. The relationship between abnormal glucose tolerance and hypertensive disorders of pregnancy in healthy nulliparous women. Am J Obstet
Gynecol 1998;179(4):10327.
[70] Suhonen L, Teramo K. Hypertension and pre-eclampsia in women with gestational glucose
intolerance. Acta Obstet Gynecol Scand 1993;72(4):26972.
[71] Hamasaki T, Yasuhi I, Hirai M, et al. Hyperinsulinemia increases the risk of gestational hypertension. Int J Gynaecol Obstet 1996;55(2):1415.
[72] Bauman WA, Maimen M, Langer O. An association between hyperinsulinemia and hypertension during the third trimester of pregnancy. Am J Obstet Gynecol 1988;159(2):
44650.
[73] Diamant YZ, Rimon E, Evron S. High incidence of preeclamptic toxemia in patients with
polycystic ovarian disease. Eur J Obstet Gynecol Reprod Biol 1982;14(3):199204.
155
[74] Gjonnaess H. The course and outcome of pregnancy after ovarian electrocautery in women
with polycystic ovarian syndrome: the inuence of body-weight. Br J Obstet Gynaecol 1989;
96(6):7149.
[75] de Vries MJ, Dekker GA, Schoemaker J. Higher risk of preeclampsia in the polycystic ovary
syndrome: a case control study. Eur J Obstet Gynecol Reprod Biol 1998;76(1):915.
[76] Kashyap S, Claman P. Polycystic ovary disease and the risk of pregnancy-induced hypertension. J Reprod Med 2000;45(12):9914.
[77] Solomon CG, Carroll JS, Okumura K, et al. Higher cholesterol and insulin levels are associated with increased risk for pregnancy-induced hypertension. Am J Hypertens 1999;12(3):
27682.
[78] Solomon CG, Graves SW, Greene MF, et al. Glucose intolerance as a predictor of hypertension in pregnancy. Hypertension 1994;23(6 Pt 1):71721.
[79] Fuh MMT, Yin C-S, Pei D, et al. Resistance to insulin-mediated glucose uptake and hyperinsulinemia in women who had preeclampsia during pregnancy. Am J Hypertens 1995;8(7):
76871.
[80] Nisell H, Erikssen C, Persson B, et al. Is carbohydrate metabolism altered among women
who have undergone a preeclamptic pregnancy? Gynecol Obstet Invest 1999;48(4):2416.
[81] He S, Silveira A, Hamsten A, et al. Haemostatic, endothelial, and lipoprotein parameters
and blood pressure levels in women with a history of preeclampsia. Thromb Haemost
1999;81(4):53842.
[82] Laivuori H, Tikkanen MJ, Ylikorkala O. Hyperinsulinemia 17 years after preeclamptic rst
pregnancy. J Clin Endocrinol Metab 1996;81(8):290811.
[83] Laivuori H, Kaaja R, Rutanen E-M, et al. Evidence of high circulating testosterone in women
with prior preeclampsia. J Clin Endocrinol Metab 1998;83(2):3447.
[84] Defronzo RA. The eect of insulin on renal sodium metabolism. A review with clinical implications. Diabetologia 1981;21(3):16571.
[85] Rowe JW, Young JB, Minaker KL, et al. Eect of insulin and glucose infusions on sympathetic nervous system activity in normal man. Diabetes 1981;30(3):21925.
[86] Calles-Escandon J, Cipolla M. Diabetes and endothelial dysfunction: a clinical perspective.
Endocr Rev 2001;22(1):3652.
[87] Report of the National High Blood Pressure Education Program Working Group on High
Blood Pressure in Pregnancy. Am J Obstet Gynecol 2000;183(1 Suppl):S122.
[88] Baker PJ, Ramey ER, Ramwell PW. Androgen-mediated sex dierences of cardiovascular
responses in rats. Am J Physiol 1978;235(2):H2426.
[89] Wakasugi M, Noguchi T, Kazama YI, et al. The eects of sex hormones on the synthesis of
prostacyclin (PGI2) by vascular tissues. Prostaglandins 1989;37(4):40110.
[90] Hacihaneoglu B, Seyisoglu H, Karsidag K, et al. Inuence of insulin resistance on total renin level in normotensive women with polycystic ovary syndrome. Fertil Steril 2000;73(2):
2615.
[91] Sibai BM, el-Nazer A, Gonzalez-Ruiz A. Severe preeclampsia-eclampsia in young primigravid women: subsequent pregnancy outcome and remote prognosis. Am J Obstet Gynecol
1986;155(5):10116.
[92] Irgens HU, Reisaeter L, Irgens LM, et al. Long term mortality of mothers and fathers after
pre-eclampsia: population based cohort study. BMJ 2001;323(7323):12137.
[93] Smith GC, Pell JP, Walsh D. Pregnancy complications and maternal risk of ischaemic heart
disease: a retrospective cohort study of 129,290 births. Lancet 2001;357(9273):20026.
Hypertension in Pregnancy
Caren G. Solomon, MD, MPHa, Ellen W. Seely, MDb,*
a
158
HYPERTENSION IN PREGNANCY
159
160
Primiparity
Prior personal or family history of preeclampsia
Obesity
Hypertension
Diabetes mellitus
Renal disease
Collagen vascular disease
Thrombophilia
Multiple gestation
Data from Roberts JN, Pearson GD, Cutler JA, Lindheimer MD. Summary
of the NHLBI Working Group on Research on Hypertension in Pregnancy. Hypertension Preg 2003;22:10927; and Duckitt K, Harrington D. Risk factors for preeclampsia at antenatal booking: systematic review of controlled studies. BMJ
2005;330:565.
HYPERTENSION IN PREGNANCY
161
Even in the absence of frank obesity, increasing body mass index (BMI) is
associated with increased risk for preeclampsia [13]; similarly, increased
blood pressure even within the normal range is a recognized risk factor
[14]. Black race also may predispose, although the association between black
race and preeclampsia may be explained by the high prevalence of obesity
and chronic hypertension in this population.
Other recognized risk factors for preeclampsia include a prior personal or
family history of preeclampsia [12]. A maternal history of preeclampsia increases the risk of preeclampsia in a womans daughter and her sons female
partner [15]. There may be several susceptibility genes for preeclampsia. Associations have been described between risk for preeclampsia and polymorphisms the genes for factor V Leiden, angiotensinogen, and endothelial
nitric oxide synthase, although these have not been conrmed consistently
in other populations [16]. Conditions linked to preeclampsia, in addition
to preexisting hypertension, include diabetes mellitus, renal disease, collagen
vascular disease [1], and possibly polycystic ovary syndrome [17].
An increased risk of preeclampsia also has been reported in multiparous
women when there is a change in paternity [18], with shorter length of sexual
relationship, and with prior use of barrier contraceptives; these observations
have been suggested to support a possible immunologic cause of the disorder. More recent data have suggested, however, that relationships between
these factors and preeclampsia may be explained by other confounding factors [19]. The risk associated with a change in paternity may be explained by
the length of the interval between pregnancies; longer intervals seem to predispose to preeclampsia regardless of paternity [20].
Women who are current cigarette smokers during pregnancy seem to have
a decreased risk of developing preeclampsia, even after adjustment for BMI
[21]; in contrast, women who stop smoking before pregnancy do not have decreased risk. Possible mediators of the risk reduction associated with cigarette
smoking include increase in nitric oxide production, decrease in thromboxane production, or alteration in cell immunity; better understanding of these
factors might lead to preventive strategies without the hazards of smoking.
Decreased placental perfusion is considered a central feature of preeclampsia (see section on pathophysiology later). Conditions associated
with increased placental size, such as multiple gestation [22], increase the
risk for preeclampsia. The risk for severe preeclampsia was reported to be signicantly greater with triplet pregnancy compared with twin pregnancy [23].
Assessment of risk factors for preeclampsia should be a part of routine
prenatal care. Women who have identied risk factors may benet from
closer surveillance during pregnancy [24].
Pathophysiology of preeclampsia
Despite decades of research, the cause of preeclampsia remains uncertain.
Many theories have been proposed.
162
Maternal factors
Endothelial dysfunction is a hallmark of preeclampsia and involves the
placental and systemic circulations [25]. It is unclear whether endothelial
dysfunction is primary (causal) or a secondary manifestation of disease.
Signs of endothelial dysfunction include hypertension, proteinuria, microangiopathic hemolytic anemia, and organ hypoperfusion. Several laboratory
abnormalities consistent with abnormal endothelial function have been observed in women with preeclampsia, including decreased levels of prostacyclin (with an increased ratio of thromboxane to prostacyclin) [26] and
increased levels of endothelin-1 [27] (a growth factor derived from endothelial cells that causes vasoconstriction). Increased pressor responsiveness to
angiotensin II (in contrast to the refractoriness to this substance observed
in normal pregnancy) also is characteristic of preeclampsia.
Insulin resistance is a well-recognized correlate of endothelial dysfunction
and may play a role in the pathogenesis of preeclampsia [28]. In normal
pregnancy, insulin resistance increases with gestation, and insulin resistance
is greater in women with preeclampsia. Studies of women with established
preeclampsia revealed higher levels of insulin and glucose, increased levels
of triglycerides and free fatty acids [29], and elevated levels of cytokines
(eg, tumor necrosis factor-a [30]) associated with the insulin-resistant state.
Although abnormalities observed in cross-sectional studies of preeclampsia could be a result rather than a cause of disease, several of these abnormalities have been shown to precede the development of preeclampsia.
Prospective studies of pregnant women who subsequently developed preeclampsia have shown higher levels of glucose [31], insulin [32], triglycerides,
free fatty acids [33], and tumor necrosis factor-a [34] and lower levels of highdensity lipoprotein [35] and sex hormonebinding globulin [36] than in women
who remain normotensive through pregnancy [28]. More recently, elevated
levels of the cytokine soluble fms-like tyrosine kinase-1(st-1) and lower
levels of placental growth factor (PlGF) were reported to antedate the development of preeclampsia [37]; high levels of st-1 are proposed to antagonize
vasodilatory eects of PlGF-1 and vascular endothelium-derived growth factor, resulting in placental vascular insuciency and other systemic eects.
Follow-up studies of women with a history of preeclampsia have revealed
several metabolic abnormalities, including increased insulin resistance [38]
and abnormal endothelial relaxation [39]. These observations, coupled
with the abnormalities observed in women before the development of preeclampsia, have suggested the importance of underlying maternal factors
and factors specic to the pregnancy in the pathogenesis of disease.
Fetoplacental factors
During normal pregnancy, fetal trophoblasts transform maternal spiral
arteries from resistance muscular vessels to low-resistance vessels by
HYPERTENSION IN PREGNANCY
163
replacing endothelial cells and smooth muscle cells of the media [40]. Several
factors (eg, nitric oxide and vascular adhesion molecules) seem to play a role
in successful trophoblastic transformation. In preeclampsia, the trophoblastic invasion is incomplete such that spiral arteries maintain increased vascular tone, leading to decreased placental perfusion. It is currently uncertain
which mediators of trophoblastic invasion are defective in preeclampsia.
Predictors of preeclampsia
Given the increased maternal and fetal morbidity and mortality associated
with preeclampsia, attempts have been made to identify markers predictive of the development of preeclampsia. Several purported markers (eg,
hyperinsulinemia and hypocalciuria) have been associated with a higher
risk of preeclampsia, but the overlap in levels between women who have
and have not developed the disease is too great for them to serve as useful
discriminators in practice. Assessments of vascular function also have been
proposed as discriminatory markers; however, assessment of angiotensin II
pressor response [41] is too invasive and impractical and Doppler assessment of uterine artery blood ow is too nonspecic [42] for clinical use. It
is currently uncertain whether serum levels of st-1 or serum or urinary levels of PlGF [43] will prove to be useful in distinguishing between pregnant
women who will and will not develop preeclampsia.
Prevention of preeclampsia
The lack of predictive markers for the development of preeclampsia has
complicated eorts to prevent the disease. Nonetheless, several interventions
have been studied in populations considered to be at increased risk based on
historical factors, such as primiparity.
Aspirin
Based on the observation of relative increases in thromboxane relative to
prostacylin levels in women with preeclampsia, aspirin or other antiplatelet
agents have been proposed as preventive agents. Although several small trials suggested a benet to aspirin, a large multicenter, placebo-controlled,
randomized trial [44] including 6927 women at increased risk for preeclampsia found no signicant reduction overall in the risk of preeclampsia with
aspirin therapy (60 mg/d). Subgroup analyses indicated a possible benet
in reducing risk in women who delivered before 32 weeks gestation, but
such women could not be reliably identied prospectively. Another large
randomized trial of low-dose aspirin (60 mg/d) in high-risk women also
showed no signicant benet [45].
164
Calcium
Similar to aspirin, calcium supplementation seemed to be a promising
preventive strategy in small studies. In a multicenter, placebo-controlled,
randomized trial of 4589 healthy nulliparous women [46], supplementation
with 2 g of elemental calcium daily did not reduce signicantly the risks of
preeclampsia or pregnancy-associated hypertension among the group overall or among women with low calcium intake or low urinary calcium levels
at baseline.
Antioxidants
Because preeclampsia has been considered to reect a state of increased
oxidant stress, it has been hypothesized that supplementation with antioxidants might reduce risk for this disorder. A randomized trial of supplementation with vitamins E and C was terminated early, however, after the rates
of preeclampsia did not dier materially between women receiving and not
receiving supplementation [47]. Low levels of u-3 fatty acids have been associated with increased rates of preeclampsia [48], but it is not known
whether supplementation reduces this risk.
Lifestyle modications
Women who are lean before pregnancy have a lower risk of preeclampsia
than women with higher pregravid BMI [12]. It is not known, however, if
weight loss before pregnancy in obese women reduces risk. Some retrospective data have indicated that higher levels of leisure-time physical activity in
the rst 20 weeks of pregnancy are associated with a signicantly reduced
risk for preeclampsia [49]. However, another retrospective report found
no signicant reduction in the risk for preeclampsia with greater leisuretime or nonleisure-time physical activity [50].
Management of preeclampsia
Women with established or suspected preeclampsia require close clinical
surveillance. The goal is to avoid the development of eclampsia or other complications in the mother, but prolong the pregnancy as long as possible, as
long as the mother and fetus are stable. Indications for immediate delivery
have been outlined by the Working Group on High Blood Pressure in Pregnancy and include maternal and fetal factors [1]. Maternal indications include gestational age 38 weeks or greater; platelet count less than 100,000
cells/mm3; progressive decline in either liver or renal function; signs of placental abruption; or symptoms of persistent severe headaches, visual
changes, severe epigastric pain, nausea, or vomiting. Fetal indications for delivery include abnormal nonstress testing or ultrasound scans indicating severe growth restriction, unfavorable biophysical prole, or oligohydramnios.
HYPERTENSION IN PREGNANCY
165
166
Table 1
Pharmacologic therapy for hypertensive pregnancy
Agent
Route of
administration
By mouth
Calcium channel
blockers
By mouth
Contraindications
d
May exacerbate asthma and heart
failure
May exacerbate asthma and heart
failure
Caution when used with magnesium
sulfate
FDA
classicationa
B
C
C
C
C
C
C
a
Categories: A (not listed), adequate and well-controlled studies have failed to show a risk
to the fetus in the rst trimester of pregnancy (and there is no evidence of risk in later trimesters); B, animal reproduction studies have failed to show a risk to the fetus and there are no
adequate and well-controlled studies in pregnant women; C, animal reproduction studies
have shown an adverse eect on the fetus and there are no adequate and well-controlled studies
in humans, but potential benets may warrant use of the drug in pregnant women despite potential risks; D (not listed), positive evidence of human fetal risk based on adverse reaction data
from investigational or marketing experience or studies in humans, but potential benets may
warrant use of the drug in pregnant women despite potential risks; and X (not listed), studies in
animals or humans have showed fetal abnormalities and/or there is positive evidence of human
fetal risk based on adverse reaction data from investigational or marketing experience, and the
risks involved in use of the drug in pregnant women clearly outweigh potential benets.
b
ACEIs and ARBs are contraindicated in the second and third trimesters of pregnancy because of the risk of fetal or neonatal renal failure and other complications (category D).
c
Short-acting calcium channel blockers have been used in this setting, but are not approved
for treating hypertensive crisis even in nonpregnant patients, and may increase risk for cardiovascular events.
HYPERTENSION IN PREGNANCY
167
168
mortality compared with women who were normotensive and gave birth at
term [70]. Studies of long-term risks may be biased, however, by possible
misdiagnosis of chronic hypertension as preeclampsia, and further research
is needed to better understand the long-term risks associated with a history
of preeclampsia and to identify eective interventions. Currently, lifestyle
interventions to control BMI and reduce insulin resistancedincluding dietary changes and increased physical activitydare reasonable and should
be encouraged.
Summary
New-onset hypertension complicates 5% to 10% of pregnancies. Preeclampsia is associated with substantial risk to the mother and the fetus.
Recognized risk factors include primiparity and preexisting chronic hypertension. Many biochemical abnormalities (eg, hyperinsulinemia, hyperlipidemia, elevated levels of tumor necrosis factor-a, and, more recently, elevated
levels of st-1 and reduced levels of PlGF) also have been recognized to predispose to disease. Currently, there is no marker for disease that has proved
suciently discriminatory to be used in practice, and no interventions are
known to be eective in preventing the disease. The cure for preeclampsia
is delivery of the fetus, but delivery earlier than necessary poses major risks
to the fetus; management involves close follow-up of the mother and the
fetus to prolong the pregnancy as long as is safely possible. Although
no antihypertensive medications are approved for use in pregnancy, clinical experience has supported certain choices (including methyldopa,
b-blockers, and calcium channel blockers for long-term management and
hydralazine, labetalol, and nitroprusside for acute management). Angiotensin-converting enzyme inhibitors and angiotensin receptor blockers are contraindicated in the second and third trimesters of pregnancy. Women with
a history of preeclampsia are at increased risk of preeclampsia in subsequent
pregnancies and may be at increased risk for subsequent hypertension and
cardiovascular disease. Further research is needed to understand causes of
preeclampsia better and to identify interventions that can prevent its occurrence and reduce subsequent risks in women who have had preeclampsia.
References
[1] Giord RW, August PA, Cunningham G. Report of the National High Blood Pressure Education Program Working Group on High Blood Pressure in Pregnancy. Am J Obstet Gynecol 2000;183:S122.
[2] Sibai B, Kupferminc M. Pre-eclampsia. Lancet 2005;365:78599.
[3] Waugh JJ, Clark TJ, Divakaran TG, et al. Accuracy of urinalysis dipstick technique in predicting signicant proteinuria in pregnancy. Obstet Gynecol 2004;103:76977.
[4] Sibai BM, Lindheimer M, Hauth J, et al. Risk factors for preeclampsia, abruptio placentae,
and adverse neonatal outcomes among women with chronic hypertension. National Institute
HYPERTENSION IN PREGNANCY
[5]
[6]
[7]
[8]
[9]
[10]
[11]
[12]
[13]
[14]
[15]
[16]
[17]
[18]
[19]
[20]
[21]
[22]
[23]
[24]
[25]
[26]
[27]
[28]
169
170
[29] Lorentzen B, Birkeland KI, Endresen MJ, et al. Glucose intolerance in women with preeclampsia. Acta Obstet Gynecol Scand 1998;77:227.
[30] Vince GC, Startkey PM, Austgulen R, et al. Interkleukin-6, tumor necrosis factor and soluble tumor factor receptors in women with pre-eclampsia. Br J Obstet Gynaecol 1995;102:20.
[31] Solomon CG, Graves SW, Greene MF, et al. Glucose intolerance as a predictor of hypertension in pregnancy. Hypertension 1994;23:71721.
[32] Solomon CG, Carroll JS, Okumura K, et al. Higher cholesterol and insulin levels are associated with increased risk for pregnancy-induced hypertension. Am J Hypertens 1999;12:
27682.
[33] Lorentzen B, Endresen MJ, Clausen T, et al. Fasting serum free fatty acids and triglycerides
are increased before 20 weeks of gestation in women who later develop preeclampsia. Hypertens Pregnancy 1994;13:1039.
[34] Serin YS, Ozcelik B, Bapbou M, et al. Predictive value of tumor necrosis factor-[alpha]
(TNF-[alpha]) in preeclampsia. Eur J Obstet Gynecol Reprod Biol 2002;100:1435.
[35] Kaaje R, Laivuori H, Laasko M, et al. Evidence of a state of increased insulin resistance in
preeclampsia. Metabolism 1999;48:8926.
[36] Wolf M, Sandler L, Muniz K, et al. First trimester insulin resistance and subsequent preeclampsia: prospective study. J Clin Endocrinol Metab 2002;87:15638.
[37] Levine RJ, Maynard SE, Qian C, et al. Circulating angiogenic factors and risk of preeclampsia. N Engl J Med 2004;350:67283.
[38] Fuh MMT, Yin C-S, Pei D, et al. Resistance to insulin-mediated glucose uptake and hyperinsulinemia in women who had preeclampsia during pregnancy. Am J Hypertens 1995;8:
76871.
[39] Chambers JC, Fusi L, Malik I, et al. Association of maternal endothelial dysfunction with
preeclampsia. JAMA 2001;285:160712.
[40] Kaufmann P, Black S, Huppertz B. Endovascular trophoblast invasion: implications for the
pathogenesis of intrauterine growth retardation and preeclampsia. Biol Reprod 2003;69:17.
[41] Gant NF, Daley GL, Chand S, et al. A study of angiotensin II pressor response throughout
primigravid pregnancy. J Clin Invest 1973;52:26829.
[42] Irion O, Masse J, Forest J-C, et al. Prediction of pre-eclampsia, low birthweight for gestation, and prematurity by uterine blood ow velocuite waveform analysis in low risk nulliparous women. Br J Obstet Gynaecol 1998;105:4229.
[43] Levine RJ, Thadani R, Qian C, et al. Urinary placental growth factor and risk of preeclampsia. JAMA 2005;293:7785.
[44] CLASP (Collaborative Low-dose Aspirin Study in Pregnancy) Collaborative group.
CLASP: a randomized trial of low-dose aspirin for the prevention and treatment of preeclampsia among 9364 pregnant women. Lancet 1994;343:61929.
[45] Caritis S, Sibai B, Hauth J, et al. Low dose aspirin to prevent preeclampsia in women at high
risk. N Engl J Med 1998;338:7015.
[46] Levine RJ, Hauth JC, Curet LB, et al. Trial of calcium to prevent preeclampsia. N Engl J
Med 1997;337:6976.
[47] Beazley D, Ahokas R, Livingston J, et al. Vitamin C and E supplementation in women at
high risk for preeclampsia: a double-blind, placebo-controlled trial. Am J Obstet Gynecol
2005;192:5201.
[48] Williams MA, Zigheim RW, King IB, et al. Omega three fatty acids in maternal erythrocytes
and risk of preeclampsia. Epidemiology 1995;6:2327.
[49] Marcoux S, Brisson J, Fabia J. Eect of leisure time physical activity on the risk of preeclampsia and gestational hypertension. J Epidemiol Community Health 1989;43:14752.
[50] Saftlas AF, Logsden-Sackett N, Wang W, et al. Work, leisure-time physical activity, and risk
of preeclampsia and gestational hypertension. Am J Epidemiol 2004;160:75865.
[51] Sibai BM, Mercer BM, Schi E, et al. Aggressive versus expectant management of severe preeclampsia at 28 to 32 weeks gestation: a randomized controlled trial. Am J Obstet Gynecol
1994;171:81822.
HYPERTENSION IN PREGNANCY
171
[52] Visser W, Wallenburg HC. Maternal and perinatal outcome of temporizing management in
254 consecutive patients with severe pre-eclampsia remote from term. Eur J Obstet Gynaecol
Reprod Biol 1995;63:14754.
[53] Oettle C, Hall D, Roux A, et al. Early onset severe pre-eclampsia: expectant management at
a secondary hospital in close association with a tertiary institution. Br J Obstet Gynaecol
2005;112:848.
[54] Sibai BM, Gonzalez AR, Mabie WC, et al. A comparison of labetalol plus hospitalization
versus hospitalization alone in the management of preeclampsia remote from term. Obstet
Gynecol 1987;70:3237.
[55] Sibai BM, Barton JR, Akl S, et al. A randomized prospective comparison of nifedipine and
bed rest versus bed rest alone in the management of preeclampsia remote from term. Am J
Obstet Gynecol 1992;167:87984.
[56] Ablalos E, Duley L, Steyn W. Antihypertensive drug therapy for mild to moderate hypertension in pregnancy. Cochrane Database System Review 2001:CD002252.
[57] Ounsted M, Cockburn J. Maternal hypertension with superimposed preeclampsia: eect in
child development at 7.5 years. Br J Obstet Gynaecol 1983;90:6449.
[58] Butters L, Kennedy S, Rubin PC. Atenolol in essential hypertension during pregnancy. BMJ
1990;301:5879.
[59] Hanssens M, Keirse MJ, Vankelecom F, et al. Fetal and neonatal eects of treatment with
angiotensin-converting enzyme inhibitors in pregnancy. Obstet Gynecol 1991;78:12835.
[60] Alwan S, Polifka JE, Friedman JM. Angiotensin II receptor antagonist treatment during
pregnancy. Birth Defects Res A Clin Mol Teratol 2005;73(2):12330.
[61] Mabie WC, Gonzalez AR, Sibai BM, et al. A comparative trial of labetalol and hydralazine
in the acute management of severe hypertension complicating pregnancy. Obstet Gynaecol
1987;70:32833.
[62] Paterson-Brown S, Robson SC, Redfern N, et al. Hydralazine boluses for the treatment of
severe hypertension in pre-eclampsia. Br J Obstet Gynaecol 1994;101:40913.
[63] Grossman E, Messerli FH, Grodzicki T, et al. Should a moratorium be placed on sublingual
nifedipine capsules given for hypertensive emergencies and pseudoemergencies? JAMA
1996;276:132831.
[64] Davis WB, Wells SR, Kuller JA, et al. Analysis of the risks associated with calcium channel
blockade: implications for the obstetrician-gynecologist. Obstet Gynecol Surv 1999;
54(Suppl):17982.
[65] Do women with pre-eclampsia and their babies benet from magnesium sulfate? The Magpie
Trial: arandomised placebo-controlled trial. Lancet 2002;359:187790.
[66] Lucas MJ, Leveno KJ, Cunningham FG. A comparison of magnesium sulfate with phenytoin for the prevention of eclampsia. N Engl J Med 1995;333:2015.
[67] Belford MA, Anthony J, Saade GR, et al. A comparison of magnesium sulfate and nimodipine for the prevention of eclampsia. N Engl J Med 2003;348:30411.
[68] Seely EW. Hypertension in pregnancy: a potential window into long-term cardiovascular
risk in women. J Clin Endocrinol Metab 1999;84:185861.
[69] Sibai BM, el-Nazer A, Gonzalez-Ruiz A. Severe preeclampsia-eclampsia in young primigravid women: subsequent pregnancy outcome and remote prognosis. Am J Obstet Gynecol
1986;155:10116.
[70] Irgens HU, Reisaeter L, Irgens LM, et al. Long term mortality of mothers and fathers after
pre-eclampsia: population based cohort study. BMJ 2001;323:12137.
Endocrinology of Parturition
Victoria Snegovskikh, MDa,*,
Joong Shin Park, MD, PhDb,
Errol R. Norwitz, MD, PhDa
a
Reproductive success is critical for survival of the species. The timely onset of labor and delivery is an important determinant of perinatal outcome.
Preterm birth (dened as delivery before 37 weeks gestation) and post-term
pregnancy (dened as pregnancy continuing beyond 42 weeks) are both associated with a signicant increase in perinatal morbidity and mortality. The
factors responsible for the timing of labor in the human are complex and, as
yet, are not completely understood. This article reviews the current understanding of the parturition cascade responsible for the spontaneous onset
of labor at term and discusses preterm labor and post-term pregnancy.
Historical context
Considerable evidence suggests that the fetus is in control of the timing of
labor. Horsedonkey crossbreeding experiments in the 1950s resulted in
a gestational length intermediate between that of horses (340 days) and
that of donkeys (365 days) [13], suggesting a role for the fetal genotype in
the initiation of labor. The mechanism by which the fetus triggers labor at
term has been demonstrated elegantly in domestic ruminants such as sheep
and cows and involves the activation at term of the fetal hypothalamicpituitary-adrenal (HPA) axis, leading to a surge in adrenal cortisol production. Fetal cortisol then acts to up-regulate directly the activity of placental
17a-hydroxylase/17,20-lyase (CYP17) enzyme, which catalyzes the conversion of pregnenolone to 17b-estradiol. The switch in progesterone:estrogen
* Corresponding author.
E-mail address: vica@inbox.ru (V. Snegovskikh).
0889-8529/06/$ - see front matter 2005 Elsevier Inc. All rights reserved.
doi:10.1016/j.ecl.2005.09.012
endo.theclinics.com
174
SNEGOVSKIKH
et al
ratio at term provides the impetus for uterine prostaglandin production and
labor [49]. However, human placentae lack the CYP17 enzyme, which is
critical to this pathway [2], and, as such, this mechanism does not apply in
humans. During the Hippocratic period, it was believed that the fetus presented head down so that it could kick its legs up against the fundus of
the uterus and propel itself through the birth canal. Although we have
moved away from this simple and mechanical view of labor, the factors responsible for the initiation and maintenance of labor at term are not well dened. The slow progress in our understanding of labor in humans is the
result, in large part, of the absence of an adequate animal model. Parturition
in most animals results from changes in circulating hormone levels in the maternal and fetal circulations at the end of pregnancy (endocrine events),
whereas labor in humans results from a complex dynamic biochemical dialog
that exists between the fetoplacental unit and the mother (paracrine and autocrine events).
Diagnosis of labor
Labor is the physiologic process by which a fetus is expelled from the uterus
and is common to all viviparous species. Labor remains a clinical diagnosis. It
requires the presence of regular painful uterine contractions, which increase in
frequency and intensity, leading to progressive cervical eacement and dilatation. In normal labor, there appears to be a time-dependent relationship
between these elements: the biochemical connective tissue changes in the
cervix usually precede uterine contractions that, in turn, precede cervical
dilatation. All of these events occur usually before spontaneous rupture of
the fetal membranes [10]. The mean duration of human singleton pregnancy
is 280 days (40 weeks) from the rst day of the last normal menstrual period.
Term is dened as the period from 37.0 to 42.0 weeks of gestation.
ENDOCRINOLOGY OF PARTURITION
175
Fig. 1. Proposed parturition cascade for labor induction at term. The spontaneous induction
of labor at term in the human is regulated by a series of paracrine-autocrine hormones acting
in an integrated parturition cascade responsible for promoting uterine contractions. COX-2,
cyclooxygenase 2; OT, oxytocin; PGDH, prostaglandin dehydrogenase; PGEM, 13,14dihydro-15-keto-PGE2; PGFM, 13,14-dihydro-15-keto-PGF2a; PLA2, phospholipase A;
SROM, spontaneous rupture of the fetal membranes; 11b-HSD, 11b-hydroxysteroid dehydrogenase; 16-OH DHEAS, 16-OH-dehydroepiandrostendione sulfate.
176
SNEGOVSKIKH
et al
Regardless of whether the trigger for labor begins within or outside the
fetus, the nal common pathway for labor ends in the maternal tissues of
the uterus and is characterized by the development of regular phasic uterine
contractions. As in other smooth muscles, myometrial contractions are mediated through the ATP-dependent binding of myosin to actin. In contrast
to vascular smooth muscle, however, myometrial cells have a sparse innervation, which is further reduced during pregnancy [12]. The regulation of
the contractile mechanism of the uterus is therefore largely humoral or dependent on intrinsic factors within the myometrial cells.
ENDOCRINOLOGY OF PARTURITION
177
(see Fig. 1) [16]. DHEAS is converted in the fetal liver to 16-hydroxy DHEAS
and then travels to the placenta where it is metabolized into estradiol (E2),
estrone (E1), and estriol (E3). In the rhesus monkey, an infusion of C19 precursor (androstenedione) leads to preterm delivery [17]. This eect is blocked
by the concurrent infusion of an aromatase inhibitor [18], demonstrating that
conversion to estrogen is important. However, a systemic infusion of estrogen failed to induce delivery, suggesting that the action of estrogen is likely
paracrine-autocrine [17,19,20]. In addition to DHEAS, the fetal adrenal
glands also produce copious amounts of cortisol. Cortisol acts to prepares
fetal organ systems for extrauterine life and to promote expression of a number of placental genes, including corticotropin releasing hormone (CRH),
oxytocin, and prostaglandins (especially prostaglandin E2 [PGE2]).
CRH is a peptide hormone released by the hypothalamus but is also expressed by placental and chorionic trophoblasts and amnionic and decidual
cells [2123]. CRH stimulates pituitary ACTH secretion and adrenal cortisol
production. In the mother, cortisol inhibits hypothalamic CRH and pituitary
ACTH release, creating a negative feedback loop. In contrast, cortisol
stimulates CRH release by the decidual, trophoblastic, and fetal membranes
[2326]. CRH, in turn, further drives maternal and fetal HPA activation,
thereby establishing a potent positive feed-forward loop. In normal pregnancy,
the increased production of CRH from decidual, trophoblastic, and fetal
membranes leads to an increase in circulating cortisol beginning in midgestation [27]. The eects of CRH are enhanced by a fall in maternal plasma
CRH-binding protein near term [28]. CRH also enhances prostaglandin production by amnionic, chorionic, and decidual cells [23]. Prostaglandins, in
turn, stimulate CRH release from the decidual and fetal membranes [24].
The rise in prostaglandins ultimately results in parturition [29]. CRH also
can directly aect myometrial contractility [30]. Taken together, these factors
suggest that placental CRH serves as a placental clock that controls the timing of labor [31,32]. A longitudinal measurement of CRH throughout pregnancy suggests that the placental clock may be set to run fast or slow as early
as the rst or second trimester of pregnancy [31,3335]. Once the speed of the
placental clock is set, the timing of delivery may be predetermined.
Role of estrogens in the onset of labor
Human pregnancy is characterized by a hyperestrogenic state of unparallel magnitude in the entire mammalian kingdom. The placenta is the primary source of estrogens, and concentrations of estrogens increase in the
maternal circulation with increasing gestational age. Placental estrone and
17b-estradiol are derived primarily from maternal C19 androgens (testosterone and androstenedione), whereas estriol is derived almost exclusively from
the fetal C19 estrogen precursor (DHEAS). Estrogens do not themselves
cause uterine contractions but do promote a series of myometrial changes,
including increasing the number of prostaglandin receptors, oxytocin
178
SNEGOVSKIKH
et al
receptors, and gap junctions, and up-regulating the enzymes responsible for
muscle contractions (myosin light chain kinase, calmodulin) [3639] that enhance the capacity of the myometrium to generate contractions.
Role of progesterone in the onset of labor
The administration of a progesterone receptor antagonist such as RU486 readily induces abortion if given before 7 weeks (49 days) of gestation
[40]. Similarly, the surgical removal of the corpus luteum, the source of progesterone, before 7 weeks results in pregnancy loss [41]. Taken together,
these data suggest that adequate production of progesterone by the corpus
luteum is critical to the maintenance of early pregnancy until the placenta
takes over this function at approximately 7 to 9 weeks of gestation (hence,
its name: pro-gestational steroid hormone). The role of progesterone in later
pregnancy, however, is less clear.
In contrast to most animal species, the circulating levels of progesterone
during human labor are similar to levels measured 1 week prior [2,42], suggesting that the systemic withdrawal of progesterone is not a prerequisite for
labor in humans. This is in contrast to most laboratory animals (with the
noted exceptions of the guinea pig and armadillo) in which progesterone
withdrawal is an essential component of parturition. However, circulating
hormone levels do not necessarily reect tissue levels, and there is increasing
evidence from both in vitro [4345] and in vivo studies [4648] that the spontaneous onset of labor at term may be preceded by a physiologic (functional)
withdrawal of progesterone activity at the level of the uterus. In one clinical
trial, Meis and colleagues [47] randomly assigned 459 patients at high risk
for preterm delivery by virtue of a previous preterm birth to receive a weekly
intramuscular injection of 17a-hydroxyprogesterone caproate (250 mg) or
a matching placebo, beginning at 16 to 20 weeks of gestation and continuing
until 36 weeks. Prophylaxis with 17a-hydroxyprogesterone signicantly reduced the risk of delivery at less than 37 weeks (36% versus 55% in the placebo
group [relative risk [RR], 0.66; 95% CI, 0.54%0.81%]), less than 35 weeks
(21% versus 31% [RR, 0.67; 95% CI, 0.48%0.93%]), and less than 32 weeks
(11% versus 20% [RR, 0.58; 95% CI, 0.37%0.91%]). Progesterone likely
maintains uterine quiescence during the latter half of pregnancy by limiting
the production of stimulatory prostaglandins and inhibiting the expression
of contraction-associated protein genes (ion channels, oxytocin and prostaglandin receptors, and gap junctions) within the myometrium [9,49]. The molecular mechanisms by which progesterone maintains uterine quiescence are
not known, but the progesterone receptor is likely critical to its action. In support of this hypothesis, the administration of the progesterone receptor antagonist RU-486 at term leads to increased uterine activity and the induction of
labor [50].
Cortisol and progesterone appear to have antagonistic actions within the
fetoplacental unit. For example, cortisol increases prostaglandin production
ENDOCRINOLOGY OF PARTURITION
179
180
SNEGOVSKIKH
et al
16
12
11.0 11.0
11.9
11.4 11.6 11.8 11.6
12.1
0
1990 1991 1992 1993 1994 1995 1996 1997 1998 1999 2000 2001 2002
Fig. 2. Preterm births in the United States, 19902002. (Data from the National Center for
Health Statistics, nal natality data, and the March of Dimes Perinatal Data Center, 2003.
Available at http://marchofdimes.com/peristats.)
Iatrogenic
preterm births
Preterm births
for maternal or
fetal indication
Preterm PROM
30%
Intra-amniotic
infection
20%
25%
25%
Spontaneous
(idiopathic) preterm
labor
- Diabetes
- IUGR
- Preeclampsia
- Placenta previa
- Placental abruption
Fig. 3. Causes of preterm births. (Data from Tucker JM, Goldenberg RL, Davis RO, et al. Etiologies of preterm birth in an indigent population: is prevention a logical expectation? Obstet
Gynecol 1991;77:3437.)
ENDOCRINOLOGY OF PARTURITION
181
182
SNEGOVSKIKH
et al
Fetal stress
Maternal stress
(uteroplacental insufficiency)
Activation of maternal HPA axis
Adrenal
Cortisol
cox-2 in amnion
PGDH in chorion
Decidua
Placenta
Membranes
DHEAS
Placenta
Membranes
Estrogen
CRH
+
Prostaglandins
Contractions
Cervical
change
Rupture of
membranes
Fig. 4. Maternal and fetal HPA axis and preterm birth. COX-2, cyclooxygenase 2; MLCK,
myosin light chain kinase; OTR, oxytocin receptors; PGDH, prostaglandin dehydrogenase.
183
ENDOCRINOLOGY OF PARTURITION
IL-6
CRH
Uterotonins
(PGs, endothelin)
FasL
IL-8
Proteases
(MMPs)
Apoptosis
Contractions
Cervical
change
Rupture of
membranes
cytokine network and the presence of cytokines in the maternal or fetal compartment several weeks before delivery.
Decidual hemorrhage
Decidual hemorrhage (abruption) presenting as vaginal bleeding in more
than one trimester of pregnancy is associated with a three- and sevenfold increased risk, respectively, of preterm birth [84] and pPROM [85]. The risk of
preterm birth is even higher (100-fold) in such women if they also had
a previous pregnancy complicated by pPROM [86]. The causes of decidual
hemorrhage-associated preterm birth include older, parous, married, and
college-educated women [87], a prole quite distinct from that of both infection- and stress-mediated preterm labor.
Thrombin, a plasma protease that converts brinogen into brin, is
formed from prothrombin by the action of prothrombinase (factor Xa). Recent studies have shown that thrombin stimulates myometrial contractions
by activating phosphatidylinositol-signaling pathways in a dose-dependent
fashion [88]. Thrombin also increases expression of plasminogen activators
and MMPs (Fig. 6) [89,90]. The release of thrombin associated with placental abruption may therefore directly initiate the nal common pathway leading to preterm labor.
Pathologic uterine distention
Excessive uterine stretching caused by multiple-birth pregnancy or polyhydramnios is associated with preterm labor. Preterm birth rates exceed
50% for twin pregnancies, 80% for triplet pregnancies, and 90% for quadruplet pregnancies. The mechanism is not clear but appears to involve a signal initiated by the mechanical stretching of uterine myometrial, cervical,
and fetal membrane cells that is transmitted through the cellular
184
SNEGOVSKIKH
et al
Hemorrhage
Decidual activation with release of clotting
factors including factor Vlla / tissue factor
Xa
X
Va
thrombin (IIa)
prothrombin (II)
fibrin
fibrinogen
plasminogen
uPA / tPA
PAI-1
plasmin
clot
+
active MMPs
ECM degradation
Contractions
Cervical
change
Rupture of
membranes
Fig. 6. Hemorrhage and preterm labor. ECM, extracellular matrix; PAI-1, plasminogen activator inhibitor 1; tPA, tissue-type plasminogen activator; uPA, urokinase plasminogen activator.
Post-term pregnancy
Post-term (prolonged) pregnancy is dened as a pregnancy that has extended to or beyond 42 weeks (294 days) from the rst day of the last normal
menstrual period or 14 days beyond the best obstetric estimate of the date of
delivery [93]. Because of the heterogeneity of populations, denitions, the use
of ultrasonography, and local practice patterns (such as the routine induction of labor at term and the management of parturients who previously
have undergone cesarean delivery), the reported incidence of pregnancies
continuing beyond the estimated date of delivery varies widely. In the United
States, approximately 18% of all births occur after 41 weeks, 3% to 14%
(mean 10%) occur after 42 weeks and are therefore post-term, and 4% of
pregnancies will continue to or beyond 43 weeks in the absence of obstetric
intervention [94,95]. The routine early use of ultrasonography to accurately
date pregnancies can reduce the rate of false-positive diagnoses and thereby
ENDOCRINOLOGY OF PARTURITION
185
Uterine distention
Uterine expansile capacity
Myometrial
activation
_
+
Fetal membrane
activation
Cytokine production
Contractions
Cervical
change
Rupture of
membranes
186
SNEGOVSKIKH
et al
the risks of the routine induction of labor (primarily failed induction leading to
cesarean delivery) are lower than reported previously [114,115], recent consensus opinions recommend the routine induction of labor at an earlier gestation
age, specically 41 weeks gestation [93,107].
Summary
Labor is a complex physiologic process involving fetal, placental, and
maternal signals. The timely onset of labor and birth is an important determinant of perinatal outcome. Both preterm labor and delivery and post-term
pregnancy are associated with increased perinatal morbidity and mortality.
Considerable evidence suggests that the fetus is in control of the timing of
labor and, thus, its birth, but exactly how this is achieved in the human is still
unknown. A better understanding of the mechanisms responsible for the
process of labor will further our knowledge about disorders of parturition,
such as preterm labor, and improve the ability of obstetric care providers
to secure a successful pregnancy outcome.
References
[1] Liggins GC. The onset of labour: an overview. In: McNellis D, Challis JRG, MacDonald
PC, et al, editors. The onset of labor: cellular and integrative mechanisms: a National Institute of Child Health and Human Development research planning workshop (November
29December 1, 1987). Ithaca (NY): Perinatology Press; 1988. p. 13.
[2] Liggins GC. Initiation of labor. Biol Neonate 1989;55:36694.
[3] Turnbull AC, Anderson AB. Evidence of a foetal role in determining the length of gestation. Postgrad Med J 1969;45:657.
[4] Liggins BJ, Fairclough RJ, Grieves SA, et al. The mechanism of initiation of parturition in
the ewe. Recent Prog Horm Res 1973;29:11159.
[5] Flint APF, Anderson ABM, Steele PA, et al. The mechanism by which fetal cortisol
controls the onset of parturition in the sheep. Biochem Soc Trans 1975;3:118994.
[6] Thorburn GD, Hollingworth SA, Hooper SB. The trigger for parturition in sheep: fetal
hypothalamus or placenta? J Dev Physiol 1991;15:719.
[7] Matthews SG, Challis JRG. Regulation of the hypothalamo-pituitary-adreno-cortical axis
in fetal sheep. Trends Endocrinol Metab 1996;4:23946.
[8] Norwitz ER, Robinson JN, Challis JR. The control of labor. N Engl J Med 1999;341:6606.
[9] Challis JRG, Matthews SG, Gibb W, et al. Endocrine and paracrine regulation of birth at
term and preterm. Endocr Rev 2000;21:51450.
[10] Du P, Hu RW, Gibbs RS. Management of premature rupture of membranes and unfavorable cervix in term pregnancy. Obstet Gynecol 1984;63:697702.
[11] Myers DA, Nathanielsz PW. Biologic basis of term and preterm labor. Clin Perinatol 1993;
20:928.
[12] Pauerstein CJ, Zauder HL. Autonomic innervation, sex steroids and uterine contractility.
Obstet Gynecol Surv 1970;25:61730.
[13] Lopez Bernal A, Rivera J, Europe-Finner GN, et al. Parturition: activation of stimulatory
pathways or loss of uterine quiescence? Adv Exp Med Biol 1995;395:43551.
[14] Garrioch DB. The eect of indomethacin on spontaneous activity in the isolated human
myometrium and on the response to oxytocin and prostaglandin. Br J Obstet Gynaecol
1978;85:4752.
ENDOCRINOLOGY OF PARTURITION
187
[15] Honnebier MB, Nathanielsz PW. Primate parturition and the role of the maternal circadian
system. Eur J Obstet Gynecol Reprod Biol 1994;55:193203.
[16] Nathanielsz PW. Comparative studies on the initiation of labor. Eur J Obstet Gynecol
Reprod Biol 1998;78:12732.
[17] Mecenas CA, Giussani DA, Owiny JR, et al. Production of premature delivery in pregnant
rhesus monkeys by androstenedione infusion. Nat Med 1996;2:4428.
[18] Giussani DA, Jenkins SL, Mecenas CA, et al. Daily and hourly temporal association
between delta4-androstenedione-induced preterm myometrial contractions and maternal
plasma estradiol and oxytocin concentrations in the 0.8 gestation rhesus monkey. Am J
Obstet Gynecol 1996;174:10505.
[19] Figueroa JP, Honnebier MBOM, Binienda Z, et al. Eect of 48 hour intravenous
D4-androstenedione infusion on pregnant rhesus monkeys in the last third of gestation:
Changes in maternal plasma estradiol concentrations and myometrial contractility. Am J
Obstet Gynecol 1989;161:4816.
[20] Nathanielsz PW, Jenkins SL, Tame JD, et al. Local paracrine eects of estradiol are central
to parturition in the rhesus monkey. Nat Med 1998;4:4569.
[21] Zoumakis E, Makrigiannakis A, Margioris AN, et al. Endometrial corticotropin-releasing
hormone: its potential autocrine and paracrine actions. Ann N Y Acad Sci 1997;828:8494.
[22] Petraglia F, Potter E, Cameron VA, et al. Corticotropin-releasing factor-binding protein is
produced by human placenta and intrauterine tissues. J Clin Endocrinol Metab 1993;77:
91924.
[23] Jones SA, Brooks AN, Challis JR. Steroids modulate corticotropin-releasing hormone production in human fetal membranes and placenta. J Clin Endocrinol Metab 1989;68:82530.
[24] Petraglai F, Coukos G, Volpe A, et al. Involvement of placental neurohormones in human
parturition. Ann N Y Acad Sci 1991;622:33140.
[25] Smith R, Mesiano S, Chan EC, Brown S, et al. Corticotropin-releasing hormone directly
and preferentially stimulates dehydroepiandrosterone sulfate secretion by human fetal adrenal cortical cells. J Clin Endocrinol Metab 1998;83:291620.
[26] Chakravorty A, Mesiano S, Jae RB. Corticotropin-releasing hormone stimulates P450
17alpha- hydroxylase/17,20-lyase in human fetal adrenal cells via protein kinase C.
J Clin Endocrinol Metab 1999;84:37328.
[27] Lockwood CJ, Radunovic N, Nastic D, et al. Corticotropin-releasing hormone and related
pituitary-adrenal axis hormones in fetal and maternal blood during the second half of pregnancy. J Perinat Med 1996;24:24351.
[28] Perkins AV, Eben F, Wolfe CD, et al. Plasma measurements of corticotrophin-releasing
hormone-binding protein in normal and abnormal human pregnancy. J Endocrinol 1993;
138:14957.
[29] Gibb W. The role of prostaglandins in human parturition. Ann Med 1998;30:23541.
[30] Grammatopoulos DK, Hillhouse EW. Role of corticotropin-releasing hormone in onset of
labor. Lancet 1999;354:15469.
[31] McLean M, Bisits A, Davies J, et al. A placental clock controlling the length of human pregnancy. Nat Med 1995;1:4603.
[32] Korebrits C, Ramirez MM, Watson L, et al. Maternal corticotropin-releasing hormone is
increased with impending preterm birth. J Clin Endocrinol Metab 1998;83:158591.
[33] Hobel CJ, Dunkel-Schetter C, Roesch SC, et al. Maternal plasma corticotropin-releasing
hormone associated with stress at 20 weeks gestation in pregnancies ending in preterm
delivery. Am J Obstet Gynecol 1999;180(1 Pt 3):S25763.
[34] Wadhwa PD, Porto M, Garite TJ, et al. Maternal corticotropin-releasing hormone levels in
the early third trimester predicts length of gestation in human pregnancy. Am J Obstet
Gynecol 1998;179:107985.
[35] Leung TN, Chung TK, Madsen G, et al. Elevated mid-trimester maternal corticotrophinreleasing hormone levels in pregnancies that delivered before 34 weeks. Br J Obstet Gynaecol 1999;106:10416.
188
SNEGOVSKIKH
et al
[36] Lye SJ, Nicholson BJ, Mascarenhas M, et al. Increased expression of connexin-43 in the rat
myometrium during labor is associated with an increase in the plasma estrogen:progesterone ratio. Endocrinology 1993;132:23806.
[37] Bale TL, Dorsa DM. Cloning, novel promoter sequence, and estrogen regulation of a rat
oxytocin receptor gene. Endocrinology 1997;138:11518.
[38] Windmoller R, Lye SJ, Challis JR. Estradiol modulation of ovine uterine activity. Can J
Physiol Pharmacol 1983;61:7228.
[39] Matsui K, Higashi K, Fukunaga K, et al. Hormone treatments and pregnancy alter myosin
light chain kinase and calmodulin levels in rabbit myometrium. J Endocrinol 1983;97:119.
[40] Peyron R, Aubeny E, Targosz V, et al. Early termination of pregnancy with mifepristone
(RU 486) and the orally active prostaglandin misoprostol. N Engl J Med 1993;328:150913.
[41] Csapo AI, Pulkkinen M. Indispensability of the human corpus luteum in the maintenance
of early pregnancy: luteectomy evidence. Obstet Gynecol Surv 1978;33:6981.
[42] Hanssens MC, Selby C, Symonds EM. Sex steroid hormone concentrations in preterm
labour and the outcome of treatment with ritodrine. Br J Obstet Gynaecol 1985;92:698702.
[43] Madsen G, Zakar T, Ku CY, et al. Prostaglandins dierentially modulate progesterone receptor-A and -B expression in human myometrial cells: evidence for prostaglandin-induced
functional progesterone withdrawal. J Clin Endocrinol Metab 2004;89:10103.
[44] Grazzini E, Guillon G, Mouillac B, et al. Inhibition of oxytocin receptor function by direct
binding of progesterone. Nature 1998;392:50912.
[45] Condon JC, Jeyasuria P, Faust JM, et al. A decline in the levels of progesterone receptor
coactivators in the pregnant uterus at term may antagonize progesterone receptor function
and contribute to the initiation of parturition. Proc Natl Acad Sci U S A 2003;100:951823.
[46] Keirse MJNC. Progestogen administration in pregnancy may prevent preterm delivery.
Br J Obstet Gynaecol 1990;97:14954.
[47] Meis PJ, Klebano M, Thom E, et al. National Institute of Child Health and Human
Development Maternal-Fetal Medicine Units Network. Prevention of recurrent preterm
delivery by 17 alpha-hydroxyprogesterone caproate. N Engl J Med 2003;348:237985.
[48] Da Fonseca EB, Bittar RE, Carvalho MH, et al. Prophylactic administration of progesterone by vaginal suppository to reduce the incidence of spontaneous preterm birth in women
at increased risk: a randomized placebo-controlled double-blind study. Am J Obstet Gynecol 2003;188:41924.
[49] Norwitz ER, Schust DJ, Fisher SJ. Implantation and the survival of early pregnancy.
N Engl J Med 2001;345:14008.
[50] Neilson JP. Mifepristone for induction of labour. Cochrane Database Syst Rev 2000:(4);
CD002865.
[51] Challis JR, Sloboda DM, Alfaidy N, et al. Prostaglandins and mechanisms of preterm
birth. Reproduction 2002;124:117.
[52] Karalis K, Goodwin G, Majzoub JA. Cortisol blockade of progesterone: a possible molecular mechanism involved in the initiation of human labor. Nat Med 1996;2:55660.
[53] Wilson T, Liggins GC, Whittaker DJ. Oxytocin stimulates the release of arachidonic acid
and prostaglandin F2 alpha from human decidual cells. Prostaglandins 1988;35:77180.
[54] Rush RW, Keirse MJNC, Howat P, et al. Contribution of preterm delivery to perinatal
mortality. BMJ 1976;2:9658.
[55] Villar J, Ezcurra EJ, de la Fuente VG, et al. Preterm delivery syndrome: the unmet need. Res
Clin Forums 1994;16:933.
[56] Martin JA, Hamilton BE, Sutton PD, et al. Births: nal data for 2002. Natl Vital Stat Rep
2003;52:1.
[57] Robertson PA, Sniderman SH, Laros RK Jr, et al. Neonatal morbidity according to gestational age and birth weight from ve tertiary care centers in the United States, 1983 through
1986. Am J Obstet Gynecol 1992;166:162941.
[58] Romero R, Avila C, Brekus CA, et al. The role of systemic and intrauterine infection in
preterm parturition. Ann N Y Acad Sci 1991;662:35575.
ENDOCRINOLOGY OF PARTURITION
189
190
SNEGOVSKIKH
et al
[81] Lockwood CJ, Ghidini A, Wein R, et al. Increased interleukin-6 concentrations in cervical
secretions are associated with preterm delivery. Am J Obstet Gynecol 1994;171:1097102.
[82] Goldenberg RL, Andrews WW, Mercer BM, et al, for the National Institute of Child
Health and Human Development Maternal-Fetal Medicine Units Network. The preterm
prediction study: granulocyte colony-stimulating factor and spontaneous preterm birth.
Am J Obstet Gynecol 2000;182:62530.
[83] Yoon BH, Romero R, Jun JK, et al. Amniotic uid cytokines (interleukin-6, tumor necrosis
factor-alpha, interleukin-1 beta, and interleukin-8) and the risk for the development of
bronchopulmonary dysplasia. Am J Obstet Gynecol 1997;177:82530.
[84] Williams MA, Mittendorf R, Lieberman E, et al. Adverse infant outcomes associated with
rst-trimester vaginal bleeding. Obstet Gynecol 1991;78:148.
[85] Harger JH, Hsing AW, Tuomala RE, et al. Risk factors for preterm premature rupture
of fetal membranes: a multicenter case-control study. Am J Obstet Gynecol 1990;163:
1307.
[86] Ekwo EE, Gosselink CA, Moawad A. Unfavorable outcome in penultimate pregnancy and
premature rupture of membranes in successive pregnancy. Obstet Gynecol 1992;80:16672.
[87] Strobino B, Pantel-Silverman J. Gestational vaginal bleeding and pregnancy outcome. Am
J Epidemiol 1989;129:80615.
[88] Elovitz MA, Ascher-Landsberg J, Saunders T, et al. The mechanisms underlying the stimulatory eects of thrombin on myometrial smooth muscle. Am J Obstet Gynecol 2000;183:
67481.
[89] Rosen T, Schatz F, Kuczynshi E, et al. Thrombin-enhanced matrix metalloproteinase-1 expression: a mechanism linking placental abruption with premature rupture of the membranes. J Matern Fetal Neonatal Med 2002;11:117.
[90] Lockwood CJ, Krikun G, Aigner S, et al. Eects of thrombin on steroid-modulated cultured endometrial stromal cell brinolytic potential. J Clin Endocrinol Metab 1996;81:
10712.
[91] Ou CW, Orsino A, Lye SJ. Expression of connexin-43 and connexin-26 in the rat myometrium during pregnancy and labor is dierentially regulated by mechanical and hormonal
signals. Endocrinology 1997;138:5398407.
[92] Nemeth E, Millar LK, Bryant-Greenwood G. Fetal membrane distention: II. dierentially
expressed genes regulated by acute distention in vitro. Am J Obstet Gynecol 2000;182:607.
[93] American College of Obstetricians and Gynecologists. Management of postterm pregnancy. ACOG Practice Bulletin No. 55. 2004.
[94] Ventura SJ, Martin JA, Curtin SC, et al. Births: nal data for 1998. Natl Vital Stat Rep
2000;48:1100.
[95] Bakketeig LS, Bergsjo P. Post-term pregnancy: magnitude of the problem. In: Chalmers I,
Enkin M, Keirse M, editors. Eective care in pregnancy and childbirth. Oxford: Oxford
University Press; 1991.
[96] Boyd ME, Usher RH, McLean FH, et al. Obstetric consequences of postmaturity. Am J
Obstet Gynecol 1988;158:3348.
[97] Gardosi J, Vanner T, Francis A. Gestational age and induction of labor for prolonged pregnancy. Br J Obstet Gynaecol 1997;104:7927.
[98] Taipale P, Hiilermaa V. Predicting delivery date by ultrasound and last menstrual period on
early gestation. Obstet Gynecol 2001;97:18994.
[99] Neilson JP. Ultrasound for fetal assessment in early pregnancy. Cochrane Database Syst
Rev 2000; CD000182.
[100] Alrevic Z, Walkinshaw SA. Management of post-term pregnancy: to induce or not? Br J
Hosp Med 1994;52:21821.
[101] Mogren I, Stenlund H, Hogberg U. Recurrence of prolonged pregnancy. Int J Epidemiol
1999;28:2537.
[102] Divon MY, Ferber A, Nisell H, et al. Male gender predisposes to prolongation of pregnancy.
Am J Obstet Gynecol 2002;187:10813.
ENDOCRINOLOGY OF PARTURITION
191
[103] Olesen AW, Basso O, Olsen J. Risk of recurrence of prolonged pregnancy. BMJ 2003;326:
476.
[104] Herabutya Y, Prasertsawat PO, Tongyai T, et al. Prolonged pregnancy: the management
dilemma. Int J Gynecol Obstet 1992;37:2538.
[105] Hilder L, Costeloe K, Thilaganathan B. Prolonged pregnancy: evaluating gestation-specic
risks of fetal and infant mortality. Br J Obstet Gynaecol 1998;105:16973.
[106] Cotzias CS, Paterson-Brown S, Fisk NM. Prospective risk of unexplained stillbirth in singleton pregnancies at term: population based analysis. BMJ 1999;319:2878.
[107] Rand L, Robinson JN, Economy KE, et al. Post-term induction of labor revisited. Obstet
Gynecol 2000;96:77983.
[108] Smith GC. Life-table analysis of the risk of perinatal death at term and post term in singleton pregnancies. Am J Obstet Gynecol 2001;184:48996.
[109] Alexander JM, McIntire DD, Leveno KJ. Forty weeks and beyond: pregnancy outcomes by
week of gestation. Obstet Gynecol 2000;96:2914.
[110] Treger M, Hallak M, Silberstein T, et al. Post-term pregnancy: should induction of labor be
considered before 42 weeks? J Matern Fetal Neonatal Med 2002;11:503.
[111] Bakketeig LS, Bergsjo P. Post-term pregnancy: magnitude of the problem. In: Enkin M,
Keirse MJ, Chalmers I, editors. Eective care in pregnancy and childbirth. Oxford: Oxford
University Press; 1989.
[112] Feldman GB. Prospective risk of stillbirth. Obstet Gynecol 1992;79:54753.
[113] Badawi N, Kurinczuk JJ, Keogh JM, et al. Antepartum risk factors for newborn encephalopathy: the Western Australian case-control study. BMJ 1998;317:154953.
[114] Hannah ME, Hannah WJ, Hellmann J, et alfor the Canadian Multicenter Post-Term
Pregnancy Trial Group. Induction of labor as compared with serial antenatal monitoring
in post-term pregnancy: a randomized controlled trial. N Engl J Med 1992;326:158792.
[115] Hannah ME, Hannah WJ, Willan A. Comment on the eectiveness of induction of labor
for postterm pregnancy. Am J Obstet Gynecol 1994;170:71623.
The combined epidemiologic, clinical, and animal studies clearly demonstrate that the intrauterine environment inuences growth and development
of the fetus and the subsequent development of adult diseases. There are
critical specic windows during development, often coincident with periods
of rapid cell division, during which a stimulus or insult may have long-lasting consequences on tissue or organ function after birth. Birth weight is only
one marker of an adverse fetal environment, and conning studies to this
population only may lead to erroneous conclusions regarding etiology.
Studies using animal models of uteroplacental insuciency suggest that mitochondrial dysfunction and oxidative stress play an important role in the
pathogenesis of the fetal origins of adult disease.
* University of Pennsylvania, 421 Curie Boulevard, BRB II/III, Room 1308, Philadelphia,
PA 19104.
E-mail address: rsimmons@mail.med.upenn.edu
0889-8529/06/$ - see front matter 2005 Elsevier Inc. All rights reserved.
doi:10.1016/j.ecl.2005.09.006
endo.theclinics.com
194
SIMMONS
a signicant correlation between low birth weight and the later development
of adult diseases. The associations with low birth weight and increased risk
of coronary heart disease, stroke, and type 2 diabetes remain strong even after adjusting for lifestyle factors, such as smoking, physical activity, occupation, income, dietary habits, and childhood socioeconomic status, and occur
independent of the current level of obesity or exercise [17].
195
that accelerated growth confers an additional risk to the growth-retarded fetus, these conicting results demonstrate the need for additional carefully
designed studies to determine just how childhood growth rates inuence
the later development of cardiovascular disease and type 2 diabetes.
196
SIMMONS
Epidemiologic challenges
These data suggest that low birth weight is associated with glucose intolerance, type 2 diabetes, and cardiovascular disease. The question remains as
to whether these associations reect fetal nutrition or other factors that contribute to birth weight and the observed glucose intolerance. Because of the
retrospective nature of the cohort identication, many confounding variables were not always recorded, such as lifestyle, socioeconomic status, education, maternal age, parental build, birth order, obstetric complications,
smoking, and maternal health. Maternal nutritional status, directly in the
form of diet history or indirectly in the form of BMI, height, and pregnancy
weight gain, were usually not recorded. Instead, birth anthropometric measures were used as proxies for presumed undernutrition in pregnancy.
Genetics versus environment
Several epidemiologic and metabolic studies of twins and rst-degree relatives of patients with type 2 diabetes have demonstrated an important genetic component of diabetes [3235]. The association between low birth
weight and risk of type 2 diabetes in some studies could theoretically be explained by a genetically determined reduced fetal growth rate. In other
words, the genotype responsible for type 2 diabetes may itself cause retarded
fetal growth in utero. This forms the basis for the fetal insulin hypothesis,
which suggests that genetically determined insulin resistance could result
in low insulin-mediated fetal growth in utero as well as insulin resistance in
childhood and adulthood [36]. Insulin is one of the major growth factors in
fetal life, and monogenic disorders that aect fetal insulin secretion or fetal
insulin resistance also aect fetal growth. Mutations in the gene encoding
glucokinase that result in low birth weight and maturity onset diabetes of
the young have been identied [37,38]. Such mutations are rare, however,
and no analogous common allelic variation has yet been discovered.
Recent genetic studies suggest that the increased susceptibility to type 2 diabetes of subjects who are born SGA results from the combination of genetic
factors and an unfavorable fetal environment. Polymorphisms of PPARg2,
a gene involved in the development and metabolic function of adipose tissue,
have been shown to modulate the susceptibility of subjects who are born SGA
to develop insulin resistance later in life [39,40]. The polymorphism is only associated with a higher risk of type 2 diabetes if birth weight is reduced [39,40].
There is obviously a close relation between genes and the environment.
Not only can maternal gene expression alter the fetal environment; the maternal intrauterine environment also aects fetal gene expression.
What animal models can tell us
Animal models have a normal genetic background on which environmental eects during gestation or early postnatal life can be tested for their role
197
in inducing diabetes. The most commonly used animal models are caloric or
protein restriction, glucocorticoid administration, or induction of uteroplacental insuciency in the pregnant rodent. In the rat, maternal dietary protein restriction (approximately 40%50% of normal intake) throughout
gestation and lactation has been reported to alter glucose homeostasis and
hypertension in the adult ospring [4146]. Ospring are signicantly growth
retarded, remain growth retarded throughout life, and develop mild b-cell secretory abnormalities in some cases [4145] and insulin resistance in others
[43,4650]. Aged rats develop hyperglycemia, which is characterized by defects in insulin signaling in muscle, adipocytes, and liver [4750].
Fetal overexposure to glucocorticoids via maternal administration or by
inhibition of placental 11bhydroxysteroid dehydrogenase1 in the rat induces hypertension, glucose intolerance, and abnormalities in hypothalamo-pituitary-adrenocortical (HPA) function after birth [5154].
To extend these experimental studies of growth retardation, we developed
a model of uteroplacental insuciency (IUGR) in the rat that restricts fetal
growth [55,56]. Growth-retarded fetal rats have critical features of a metabolic prole characteristic of growth-retarded human fetuses: decreased levels of glucose, insulin, insulin-like growth factor-I (IGF-I), amino acids, and
oxygen [5759]. By 6 months of age, IUGR rats develop diabetes with a phenotype remarkably similar to that observed in the human being with type 2
diabetes: progressive dysfunction in insulin secretion and insulin action.
Thus, the studies in various animal models support the hypothesis that an
abnormal intrauterine milieu can induce permanent changes in glucose homeostasis after birth and lead to type 2 diabetes in adulthood.
198
SIMMONS
199
200
SIMMONS
[4] Barker DJP, Winter PD, Osmond C, et al. Weight in infancy and death from ischaemic heart
disease. Lancet 1989;ii:57780.
[5] Hales CN, Barker DJP, Clark PMS, et al. Fetal and infant growth and impaired glucose tolerance at age 64. BMJ 1991;303:101922.
[6] Phipps K, Barker DJ, Hales CN, et al. Fetal growth and impaired glucose tolerance in men
and women. Diabetologia 1993;36:2258.
[7] Fall CHD, Osmond C, Barker DJP, et al. Fetal and infant growth and cardiovascular risk
factors in women. BMJ 1995;310:42832.
[8] Barker DJP, Hales CN, Fall CHD, et al. Type 2 diabetes mellitus, hypertension, and hyperlipidemia (syndrome X): relation to reduced fetal growth. Diabetologia 1993;36:627.
[9] Hales CN, Barker DJ. The thrifty phenotype hypothesis. Br Med Bull 2001;60:520.
[10] Valdez R, Athens MA, Thompson GH, et al. Birthweight and adult health outcomes in
a biethnic population in the USA. Diabetologia 1994;37:62431.
[11] Curhan GC, Willett WC, Rimm EB, et al. Birthweight and adult hypertension, diabetes mellitus and obesity in US men. Circulation 1996;94:324650.
[12] Lithell HO, McKeigue PM, Berglund L, et al. Relation of size at birth to non-insulin dependent diabetes and insulin concentrations in men aged 5060 years. BMJ 1996;312:
40610.
[13] Leger J, Levy-Marchal C, Bloch J, et al. Reduced nal height and indications for insulin resistance in 20 year olds born small for gestational age: regional cohort study. BMJ 1997;315:
3417.
[14] Jaquet D, Gaboriau A, Czernichow P, et al. Insulin resistance early in adulthood in subjects
born with intrauterine growth retardation. J Clin Endocrinol Metab 2000;85:14016.
[15] Egeland GM, Skjaerven R, Irgrens LM. Birth characteristics of women who develop gestational diabetes: population based study. BMJ 2000;321:5467.
[16] Forsen T, Eriksson J, Tuomilehto J, et al. The fetal and childhood growth of persons who
develop type 2 diabetes. Ann Intern Med 2000;133:17682.
[17] Rich-Edwards JW, Colditz GA, Stampfer MJ, et al. Birthweight and the risk for type 2
diabetes mellitus in adult women. Ann Intern Med 1999;130:27884.
[18] St. Jeor ST, Hayman LL, Daniels SR, et al. American Heart Association. Prevention Conference VII: obesity, a worldwide epidemic related to heart disease and stroke: group II: agedependent risk factors for obesity and comorbidities. Circulation 2004;110:4715.
[19] Eriksson J, Forsen T, Tuomilehto J, et al. Fetal and childhood growth and hypertension in
adult life. Hypertension 2000;36:7904.
[20] Bavdekar A, Sachdev HS, Fall CHD, et al. Relation of serial changes in childhood bodymass index to impaired glucose tolerance in young adulthood. N Engl J Med 2004;350:
86575.
[21] Bavdekar A, Yajnik CS, Fall CH, et al. Insulin resistance syndrome in 8-year-old Indian
children: small at birth, big at 8 years, or both? Diabetes 1999;48:24229.
[22] Homan PL, Cuteld WS, Robinson EM, et al. Insulin resistance in short children with
intrauterine growth retardation. J Clin Endocrinol Metab 1997;82:4026.
[23] Li C, Johnson MS, Goran MI. Eects of low birth weight on insulin resistance syndrome in
Caucasian and African-American children. Diabetes Care 2001;24:203542.
[24] Yajnik CS, Fall CH, Vaidya U, et al. Fetal growth and glucose and insulin metabolism in
four-year-old Indian children. Diabet Med 1995;12:3306.
[25] Clausen JO, Borch-Johnsen K, Pedersen O. Relation between birth weight and the insulin
sensitivity index in a population sample of 331 young, healthy Caucasians. Am J Epidemiol
1997;146:2331.
[26] Flanagan DE, Moore VM, Godsland IF, et al. Fetal growth and the physiological control of
glucose tolerance in adults: a minimal model analysis. Am J Physiol Endocrinol Metab 2000;
278:E7006.
[27] Phillips DI, Barker DJ, Hales CN, et al. Thinness at birth and insulin resistance in adult life.
Diabetologia 1994;37:1504.
201
[28] Widdowson EM, Southgate DAT, Hey EN. Nutrition and metabolism of the fetus and
infant. In: Visser HKA, editor. Body composition of the fetus and infant in nutrition and
metabolism of the fetus and infant. The Hague: Martinus Nijho; 1979. p. 16977.
[29] Lapillonne A, Braillon P, Chatelain PG, et al. Body composition in appropriate and small for
gestational age infants. Acta Paediatr 1997;86:196200.
[30] Van Assche FA, De Prins F, Aerts L, et al. The endocrine pancreas in small for dates infants.
Br J Obstet Gynaecol 1977;84:7513.
[31] Jensen CB, Storgaard H, Dela F, et al. Early dierential defects of insulin secretion and action in 19-year-old Caucasian men who had low birth weight. Diabetes 2002;51:127180.
[32] Barnett AH, E C, Leslie RDG, et al. Diabetes in identical twins. Diabetologia 1981;20:
8793.
[33] Newman B, Selby JV, King MC, et al. Concordance for type 2 diabetes mellitus in male
twins. Diabetologia 1987;30:7638.
[34] Warram JH, Martin BC, Krolewski AS, et al. Slow glucose removal rate and hyperinsulinemia precede the development of type II diabetes in the ospring of diabetic parents. Ann Intern Med 1990;113:90915.
[35] Vaag A, Henricksen JE, Madsbad S, et al. Insulin secretion, insulin action, and hepatic glucose production in identical twins discordant for NIDDM. J Clin Invest 1995;95:6908.
[36] Hattersley AT, Tooke JE. The fetal insulin hypothesis: an alternative explanation of the
association of low birthweight with diabetes and vascular disease. Lancet 1999;353:
178992.
[37] Froguel P, Zouali H, Vionnet N. Familial hyperglycemia due to mutations in glucokinase:
denition of a subtype of diabetes mellitus. N Engl J Med 1993;328:697702.
[38] Hattersley AT, Beards F, Ballantyne E, et al. Mutations in the glucokinase gene of the fetus
result in reduced birth weight. Nat Genet 1998;19:26870.
[39] Kubaszek A, Markkanen A, Eriksson JG, et al. The association of the K121Q polymorphism
of the plasma cell glycoprotein-1 gene with type 2 diabetes and hypertension depends on size
at birth. J Clin Endocrinol Metab 2004;89:20447.
[40] Eriksson JG, Lindi V, Uusitupa M, et al. The eects of the Pro12Ala polymorphism of the
peroxisome proliferator-activated receptor-gamma2 gene on insulin sensitivity and insulin
metabolism interact with size at birth. Diabetes 2002;517:23214.
[41] Dahri S, Snoeck A, Reusens-Billen B, et al. Islet function in o-spring of mothers on lowprotein diet during gestation. Diabetes 1991;40:11520.
[42] Snoeck A, Remacle C, Reusens B, et al. Eect of a low protein diet during pregnancy on the
fetal rat endocrine pancreas. Biol Neonate 1990;57:10718.
[43] Ozanne SE, Wang CL, Coleman N, et al. Altered muscle insulin sensitivity in the male ospring of protein-malnourished rats. Am J Physiol 1996;271:E112834.
[44] Berney DM, Desai M, Palmer DJ, et al. The eects of maternal protein deprivation on the
fetal rat pancreas: major structural changes and their recuperation. J Pathol 1997;183:
10915.
[45] Wilson MR, Hughes SJ. The eect of maternal protein deciency during pregnancy and lactation on glucose tolerance and pancreatic islet function in adult rat ospring. J Endocrinol
1997;154:17785.
[46] Burns SP, Desai M, Cohen RD, et al. Gluconeogenesis, glucose handling, and structural
changes in livers of the adult ospring of rats partially deprived of protein during pregnancy
and lactation. J Clin Invest 1997;100:176874.
[47] Ozanne SE, Jensen CB, Tingey KJ, et al. Low birthweight is associated with specic changes
in muscle insulin-signaling protein expression. Diabetologia 2005;48:54752.
[48] Ozanne SE, Olsen GS, Hansen LL, et al. Early growth restriction leads to down regulation
of protein kinase C zeta and insulin resistance in skeletal muscle. J Endocrinol 2003;177:
23541.
[49] Petry CJ, Dorling MW, Pawlak DB, et al. Diabetes in old male ospring of rat dams fed a reduced protein diet. Int J Exp Diabetes Res 2001;2:13943.
202
SIMMONS
[50] Fernandez-Twinn DS, Wayman A, Ekizoglou S, et al. Maternal protein restriction leads to
hyperinsulinemia and reduced insulin-signaling protein expression in 21-mo-old female rat
ospring. Am J Physiol Regul Integr Comp Physiol 2005;288:R36873.
[51] Benediktsson R, Lindsay R, Noble J, et al. Glucocorticoid exposure in utero; a new model for
adult hypertension. Lancet 1993;341:33941.
[52] Lindsay RS, Lindsay RM, Edwards CRW, et al. Inhibition of 11b-hydroxysteroid dehydrogenase in pregnant rats and the programming of blood pressure in the ospring. Hypertension 1996;27:12004.
[53] Lindsay RS, Lindsay RM, Waddell B, et al. Programming of glucose tolerance in the rat: role
of placental 11b-hydroxysteroid dehydrogenase. Diabetologia 1996;39:1299305.
[54] Niyirenda MJ, Seckl JR. Intrauterine events and the programming of adulthood disease: the
role of fetal glucocorticoid exposure. Int J Mol Med 1998;2:60714.
[55] Simmons RA, Templeton L, Gertz S, et al. Intrauterine growth retardation leads to type II
diabetes in adulthood in the rat. Diabetes 2001;50:227986.
[56] Boloker J, Gertz S, Simmons RA. Ospring of diabetic rats develop obesity and type II diabetes in adulthood. Diabetes 2002;51:1499506.
[57] Ogata ES, Bussey M, Finley S. Altered gas exchange, limited glucose, branched chain amino
acids, and hypoinsulinism retard fetal growth in the rat. Metabolism 1986;35:95077.
[58] Simmons RA, Gounis AS, Bangalore SA, et al. Intrauterine growth retardation: fetal glucose
transport is diminished in lung but spared in brain. Pediatr Res 1991;31:5963.
[59] Unterman T, Lascon R, Gotway M, et al. Circulating levels of insulin-like growth factor
binding protein-1 (IGFBP-1) and hepatic mRNA are increased in the small for gestational
age fetal rat. Endocrinology 1990;127:20357.
[60] Myatt L, Eis ALW, Brockman DE, et al. Dierential localization of superoxide dismutase
isoforms in placental villous tissue of normotensive, pre-eclamptic, and intrauterine
growth-restricted pregnancies. J Histochem Cytochem 1997;45:14338.
[61] Karowicz-Billinska A, Suzin J, Sieroszewski P. Evaluation of oxidative stress indices during
treatment in pregnant women with intrauterine growth retardation. Med Sci Monit 2002;
8(3): CR211CR6.
[62] Ejima K, Nanri H, Toki N, et al. Localization of thioredoxin reductase and thioredoxin in
normal human placenta and their protective eect against oxidative stress. Placenta 1999;20:
95101.
[63] Kato H, Yoneyama Y, Araki T. Fetal plasma lipid peroxide levels in pregnancies complicated by preeclampsia. Gynecol Obstet Invest 1997;43:15861.
[64] Bowen RS, Moodley J, Dutton MF, et al. Oxidative stress in pre-eclampsia. Acta Obstet Gynecol Scand 2001;80:71925.
[65] Wang Y, Walsh SW. Increased superoxide generation is associated with decreased superoxide dismutase activity and mRNA expression in placental trophoblast cells in pre-eclampsia.
Placenta 2001;22:20612.
[66] Wang Y, Walsh SW. Placental mitochondria as a source of oxidative stress in pre-eclampsia.
Placenta 1998;19:5816.
[67] Peterside IE, Selak MA, Simmons RA. Impaired oxidative phosphorylation in hepatic mitochondria of growth retarded rats alters glucose metabolism. Am J Physiol 2003;285:
E125864.
[68] Selak MA, Storey BT, Peterside IE, et al. Impaired oxidative phosphorylation in skeletal
muscle contributes to insulin resistance and hyperglycemia. Am J Physiol 2003;285:
E1307.
[69] Panten U, Zielman S, Langer J, et al. Regulation of insulin secretion by energy metabolism in
pancreatic b-cell mitochondria. Biochem J 1984;219:18996.
[70] Newgard CB, McGarry JD. Metabolic coupling factors in pancreatic b-cell signal transduction. Annu Rev Biochem 1995;64:689719.
[71] Schuit F. Metabolic fate of glucose in puried islet cells. Glucose regulated anaplerosis in
b-cells. J Biol Chem 1997;272:185729.
203
[72] Mertz RJ, Worley JF, Spencer B, et al. Activation of stimulus-secretion coupling in pancreatic b-cells by specic products of glucose metabolism. J Biol Chem 1996;271(9):483845.
[73] Ortsater H, Liss P, Akerman KEO. Contribution of glycolytic and mitochondrial pathways
in glucose-induced changes in islet respiration and insulin secretion. Pugers Arch Eur J
Physiol 2002;444:50612.
[74] Antinozzi PA, Ishihara H, Newgard CB, et al. Mitochondrial metabolism sets the maximal
limit of fuel-stimulated insulin secretion in a model pancreatic beta cell. A survey of four fuel
secretagogues. J Biol Chem 2002;277:1174655.
[75] Malaisse WJ, Hutton JC, Carpinelli AR, et al. The stimulus-secretion coupling of amino
acid-induced insulin release. Metabolism and cationic eects of leucine. Diabetes 1980;29:
4317.
[76] Lenzen S, Schmidt W, Rustenbeck I, et al. 2-Ketoglutarate generation in pancreatic b-cell
mitochondria regulates insulin secretory action of amino acids and 2-keto acids. Biosci
Rep 1986;6:1639.
[77] Noda M, Yamashita S, Takahashi N, et al. Switch to anaerobic glucose metabolism with
NADH accumulation in the beta-cell model of mitochondrial diabetes. Characteristics of
beta HC9 cells decient in mitochondrial DNA transcription. J Biol Chem 2002;277:
4181726.
[78] Lenzen S, Drinkgern J, Tiedge M. Low antioxidant enzyme gene expression in pancreatic
islets compared with various other mouse tissues. Free Radic Biol Med 1996;20:4636.
[79] Tiedge M, Lortz S, Drinkgern J, et al. Relationship between antioxidant enzyme gene expression and antioxidant defense status of insulin-producing cells. Diabetes 1997;46:173342.
[80] Maechler P, Jornot L, Wollheim CB. Hydrogen peroxide alters mitochondrial activation and
insulin secretion in pancreatic beta cells. J Biol Chem 1999;274:2790513.
[81] Sakai K, Matsumoto K, Nishikawa T, et al. Mitochondrial reactive oxygen species reduce
insulin secretion by pancreatic b-cells. Biochem Biophys Res Commun 2003;300:21622.
[82] Kaneto H, Xu G, Fujii N, et al. Involvement of c-Jun N-terminal kinase in oxidative stressmediated suppression of insulin gene expression. J Biol Chem 2002;277:300108.
[83] Kaneto HH, Xu G, Fujii N, et al. Involvement of protein kinase C beta 2 in c-myc induction
by high glucose in pancreatic beta-cells. J Biol Chem 2002;277:36805.
[84] Kaneto H, Xu G, Song KH, et al. Activation of the hexosamine pathway leads to deterioration of pancreatic beta-cell function through the induction of oxidative stress. J Biol Chem
2001;276:31099104.
[85] Kaneto H, Kajimoto Y, Fujitani Y, et al. Oxidative stress induces p21 expression in pancreatic islet cells: possible implication in beta-cell dysfunction. Diabetologia 1999;42:10937.
[86] Jonas JC, Laybutt DR, Steil GM, et al. High glucose stimulates early response gene c-Myc
expression in rat pancreatic beta cells. J Biol Chem 2001;276:3537581.
[87] Jonas JC, Sharma A, Hasenkamp W, et al. Chronic hyperglycemia triggers loss of pancreatic
beta cell dierentiation in an animal model of diabetes. J Biol Chem 1999;274:1411221.
[88] Efanova IB, Zaitsev SV, Zhivotovsky B, et al. Glucose and tolbutamide induce apoptosis in
pancreatic b-cells. J Biol Chem 1998;273:225017.
[89] Moran A, Zhang HJ, Olson LK, et al. Dierentiation of glucose toxicity from b-cell exhaustion during the evolution of defective insulin gene expression in the pancreatic islet cell line,
HIT-T15. J Clin Invest 2000;99(3):5349.
[90] Donath MY, Gross DJ, Cerasi E, et al. Hyperglycemia-induced b-cell apoptosis in pancreatic islets of Psammomas obesus during development of diabetes. Diabetes 1999;48:
73844.
[91] Silva JP, Kohler M, Gra C, et al. Impaired insulin secretion and b-cell loss in tissue specic
knockout mice with mitochondrial diabetes. Nat Genet 2000;26:33640.
[92] Park J, Suponitsky-Kroyer I, Simmons RA. Epigenetic silencing of Pdx-1 in growth retarded
[IUGR] rats. Endocrinology, in press.
[93] Vuguin P, Raab E, Liu B, et al. Hepatic insulin resistance precedes the development of diabetes in a model of intrauterine growth retardation. Diabetes 2004;53:261722.
204
SIMMONS
[94] Peterside IE, Selak MA, Simmons RA. Impaired oxidative phosphorylation in hepatic mitochondria of growth retarded rats alters glucose metabolism. Am J Physiol 2003;285(6):
E125864.
[95] Selak MA, Storey BT, Peterside IE, et al. Impaired oxidative phosphorylation in skeletal
muscle contributes to insulin resistance and hyperglycemia. Am J Physiol 2003;285:E1307.
[96] Petersen KF, Dufour S, Befroy D, et al. Impaired mitochondrial activity in the insulinresistant ospring of patients with type 2 diabetes. N Engl J Med 2004;350:66471.
Index
Note: Page numbers of article titles are in boldface type.
A
Abortion, spontaneous, gestational Type I
diabetes associated with, 54, 81
Acromegaly, during pregnancy, 105106
Addisons disease, in pregnancy, 611. See
also Primary adrenal insuciency.
Adenomas, pituitary, during pregnancy,
clinically nonfunctioning, 109
Cushings syndrome with,
12, 56
endocrine changes with,
99100
prolactinoma, 100105
thyroid-stimulating
hormone-secreting,
100, 108109
0889-8529/06/$ - see front matter 2005 Elsevier Inc. All rights reserved.
doi:10.1016/S0889-8529(05)00132-5
endo.theclinics.com
206
b-Adrenergic blockade (continued )
for pheochromocytoma, during
pregnancy, 1415
for preeclampsia, during pregnancy,
166167
B
Biophosphonates, for primary
hyperparathyroidism, during
pregnancy, 3536
Birth size, in adult metabolic disease, insulin
inuence, 195
Birth weight, adult metabolic disease and,
193194
insulin factors, 195
Blood pressure, elevated, in pregnancy,
157171. See also Hypertension.
Body mass index (BMI), adult metabolic
disease and, 193194
insulin factors, 195
as preeclampsia risk factor, 160161,
164
infertility and, 147149
Bone biopsy, for skeletal calcium
metabolism, during pregnancy, 2627
Bone metabolism, in pregnancy and
lactation, 2151
adaptations during lactation,
2833
adaptations during pregnancy,
2227
disorders of, 3345
Breast(s), skeletal demineralization role of,
30
Bromocriptine, for acromegaly, during
pregnancy, 106
for Cushings syndrome, during
pregnancy, 108
for hyperprolactinemia, during
pregnancy, 101
eects on pregnancy,
102103
indications for, 104105
for lactotroph hyperplasia, during
pregnancy, 109
C
Cabergoline, for acromegaly, during
pregnancy, 106
for hyperprolactinemia, during
pregnancy, 101, 104105
Calcitonin, for primary
hyperparathyroidism, during
pregnancy, 35
INDEX
207
INDEX
Dehydroepiandrostenedione (DHEAS),
fetal, labor onset role of, 176177
E
Early pregnancy loss (EPL), with polycystic
ovary syndrome, 141144
Emergency management, of preeclampsia,
167
Endocrinology consult, for primary adrenal
insuciency, during pregnancy, 9
Endothelial dysfunction, in preeclampsia,
fetoplacental, 162163
maternal, 162
Environmental factors, of adult metabolic
disease, 196
Estrogen(s), labor onset role of, 177178
Euthyroidism, with autoimmune disease,
during pregnancy, 123
Exercise, for gestational Type I diabetes, 57,
89
208
INDEX
F
Familial benign hypocalciuric
hypercalcemia (FBHH), in pregnancy,
36
Gonadotrophin-releasing hormone
(GnRH), in skeletal calcium
metabolism, during lactation, 30, 32
209
INDEX
210
Hypothalamic-pituitary-adrenal (HPA)
axis, fetal, labor onset role of, 176177
premature, 181182
Hypothyroidism, during pregnancy,
119123
causes of, 119
diagnosis of, 119
pregnancy outcomes with,
119121
treatment of, 121123
fetal, 120121
avoiding, 127128
neonatal, 120121
I
Immunoradiometric assays (IRMA), for
calcium and bone adaptations, during
pregnancy and lactation, 2324, 28
Impaired glucose tolerance (IGT),
developmental adult metabolic disease
and, 193199
in gestational diabetes mellitus,
144145
In vitro fertilization (IVF), for infertility,
140
Infertility, body mass index and, in
preeclampsia risks, 147149
INDEX
L
Labetalol, for preeclampsia, 166167
211
INDEX
M
Macrosomia, with gestational diabetes,
9092
Magnesium sulfate, administration during
pregnancy, 4143
bone and mineral impact of, 41
management issues with, 4243
maternal complications of, 41
neonatal complications of, 42
Magnetic resonance imaging (MRI), in
pregnancy screening, for Cushings
syndrome, 45
for hyperaldosteronism, 12
for pheochromocytoma, 14
for primary adrenal insuciency,
9
Maternal factors, of preeclampsia, 160162
Maternal monitoring, for preeclampsia, 165
Metabolic disease/disorders, in
preeclampsia, 150, 162
origins of adult, 193204
animal models of, 196197
birth size and insulin, 195
catch-up growth, 194195
cellular mechanisms, 197199
epidemiologic challenges of, 196
genetic versus environment, 196
high birth weight, 194
low birth weight, 193194
mitochondrial dysfunction and,
197199
oxidative stress and, 197199
with insulin resistance, 140141, 150
212
Nifedipine, for preeclampsia, 167
Nitroprusside, for preeclampsia, 166167
Nodules, thyroid, during pregnancy,
131132
O
Obesity, as preeclampsia risk factor,
160161, 164
infertility and, 147149
Octreotide, for acromegaly, during
pregnancy, 106
for thyroid-stimulating hormonesecreting tumors, during
pregnancy, 108109
Oral antidiabetic agents, during pregnancy,
administration of, 69
anomalies risks with, 63, 68
classication of, 7172
cost of, 70
insulin therapy versus, 6367,
6970
lactation and, 73
pharmacology of, 6869
placental transfer of, 68
Osteoporosis, with pregnancy and lactation,
3840
clinical features of, 3940
diagnostic studies of, 40
hormonal factors of, 39
laboratory ndings, 40
pathogenesis of, 40
prevalence of, 3839
therapeutic interventions for, 40
Oxidative stress, adult metabolic disease
and, 197199
Oxytocin, labor onset role of, 179
P
Paracrine mediators, of parturition,
176179
Parathyroid hormone (PTH), magnesium
sulfate administration during
pregnancy impact on, 41
maternal, 41
neonatal, 42
mineral ions and, adaptations during
lactation, 2830, 32
adaptations during pregnancy,
2325
Parathyroidectomy, for primary
hyperparathyroidism, during
pregnancy, 3536
INDEX
213
INDEX
superimposed on chronic
hypertension, 157158
Pregnancy, acromegaly during, 105106
adrenal disorders in, 120
congenital adrenal hyperplasia
and, 1516
Cushings syndrome, 16
hyperaldosteronism, 1113
hypertension, 11, 1314
overview of, 1, 16
pheochromocytoma, 1316
primary adrenal insuciency,
611
calcium and bone metabolism during,
2151
adaptations during lactation,
2833
adaptations of, 2227
disorders of, 3345
Cushings syndrome in, 16
causes of, 12
clinical presentation of, 1
diagnosis of, 107108
dierential diagnosis of, 25
eects on pregnancy, 108
incidence of, 2, 106
maternal morbidity associated
with, 2
pathogenesis of, 106107
pituitary dysfunction with,
106108
screening tests for, 2
treatment of, 56, 108
euthyroidism with autoimmune
disease during, 123
familial benign hypocalciuric
hypercalcemia in, 36
Graves disease in, 124130
diagnosis of, 125
epidemiology of, 124
pregnancy outcomes with, 125
treatment of, 125130
b-adrenergic blockers, 129
guidelines for, 126, 128
iodides for, 129
radioactive iodine therapy,
129130
surgery indications, 129
thionamides for, 125128
hyperaldosteronism in, 1113
clinical presentations of, 12
diagnosis of, 1213
prevalence of, 11
RAS activity with, 1112
treatment of, 13
hypercalcemia of malignancy in, 44
hyperemesis gravidarum with, 124
hypertension in, 11, 157171
214
Pregnancy (continued )
chronic antenatal disorders of,
157158
classication of, 157158
clinical manifestations of,
159160
complications of, 158
incidence of, 157, 168
later life risks associated with,
167168
management of, 164167
pathophysiology of, 161163
predictors of, 163
prevention of, 163164
risk factors for, 160161
with hypothyroidism, 119120
with pheochromocytoma, 1314
with polycystic ovary syndrome,
146150
with Type I diabetes, 8586
hyperthyroidism during, 123130
Graves disease, 124130
hyperemesis gravidarum, 124
prevalence of, 123124
hypoparathyroidism during, 3638
hypopituitarism during, 109112
causes of, 109
hormone replacement for,
109110
syndromes of, 110112
hypothyroidism during, 119123
causes of, 119
diagnosis of, 119
pregnancy outcomes with,
119121
treatment of, 121123
insulin resistance during, 140141, 162
labor and delivery, endocrinology of,
173191. See also Parturition.
gestational Type I diabetes
management during, 92
primary adrenal insuciency
management during, 1011
low calcium intake during, 43
lymphocytic hypophysitis during,
110111
magnesium sulfate administration
during, bone and mineral impact
of, 4143
osteoporosis associated with, 3840
pheochromocytoma in, 1316
clinical presentations of, 13
diagnosis of, 14
fetal morbidity/mortality with, 14
maternal morbidity/mortality
with, 14
treatment of, 1415
pituitary disorders in, 99116
acromegaly, 105106
INDEX
adenomas, clinically
nonfunctioning, 109
Cushings syndrome with,
12, 56
endocrine changes with,
99100
thyroid-stimulating
hormone-secreting,
100, 108109
Cushings syndrome, 106108
tumor-related, 12, 56
hypopituitarism, 109112
normal anterior gland
physiology, 99100
prolactinoma, 100105
polycystic ovary syndrome
complications of, 141150
post-term, causes of, 185
complications of, 185186
endocrinology of, 184185
primary adrenal insuciency in, 611
causes of, 6
clinical presentations of, 67
diagnosis of, 79
clinical features, 7
dierential, 89
tests used for, 9
laboratory features, 7
screening tests, 8
fetal morbidity/mortality with, 7
maternal morbidity/mortality
with, 67
treatment of, 910
during labor, 1011
postpartum, 11
primary hyperparathyroidism during,
3336
fetal complications of, 3435
incidence of, 33
management of, 3536
maternal manifestations/
complications of, 34
morbidity with, 33
prolactinoma in, 100105
dopamine agonists for, 101
eects on pregnancy, 102104
gestational tumor growth,
101102
incidence of, 100
management of, 104105
surgical treatment of, 100101
pseudohypoparathyroidism during, 38
Sheehans syndrome during, 111112
thyroid disorders during, 117136
cancer, 132
common incidence of, 117,
132133
euthyroidism with autoimmune
disease, 123
215
INDEX
fetal/neonatal hyperthyroidism,
130131
hyperthyroidism, 123130
hypothyroidism, 119123
nodules, 131132
normal gravid thyroid physiology
versus, 117119
thyroid function during, normal
gravid, 117119
thyrotoxicosis during, 124
Type I diabetes in, 7997
congenital anomalies associated
with, 54, 63, 68, 8081
diabetic ketoacidosis with, 85
diabetic nephropathy with, 86
diet for, 57, 62, 90
exercise for, 57, 89
fetal risks with, 54, 7980, 90
fetal surveillance with, 9092
glucose monitoring for, 81
by self, 8687
glycemic control for, 5378, 81
goals for, 55, 8687
monitoring methods for,
8788
normal prole in, 5455
glycosylated hemoglobin A1c
monitoring for, 81, 8788
hypertension with, 8586
hypoglycemia hazards with, 81
insulin therapy for, diet therapy
versus, 62
doses for, 8889
fetal markers for initiation
of, 61
oral antidiabetic agents
versus, 6367
patient indications for,
6061
placental transfer and safety
concerns, 58, 60
requirements for, 6263
risks of rapid-acting, 8184
types of, 5759
intensied therapy for, 5557
labor and delivery
considerations, 92
management of, 5378
maternal risks with, 7980
oral antidiabetic agents for,
administration of, 69
anomalies risks with, 63, 68
classication of, 7172
cost of, 70
insulin therapy versus,
6367, 6970
lactation and, 73
pharmacology of, 6869
placental transfer of, 68
216
INDEX
S
Seizure prophylaxis, for preeclampsia, 167
T
Tetany, neonatal, with maternal primary
hyperparathyroidism, 34
Thiazolidinediones (TZDs), for infertility,
139140, 143
Thionamides, for Graves disease, during
pregnancy, 125128
Thyroid cancer, during pregnancy, 132
Thyroid disorders, during pregnancy,
117136
cancer, 132
common incidence of, 117,
132133
euthyroidism with autoimmune
disease, 123
fetal/neonatal hyperthyroidism,
130131
hyperthyroidism, 123130
hypothyroidism, 119123
nodules, 131132
normal gravid thyroid physiology
versus, 117119
Thyroid function, during pregnancy,
dysfunctional, 119132. See also
Thyroid disorders.
gestational Type I diabetes and,
8485
normal gravid, 117119
Thyroid nodules, during pregnancy,
131132
Thyroid-stimulating hormone (TSH),
pregnancy physiology of, 117118
INDEX
217
placental transfer and safety
concerns, 58, 60
requirements for, 6263
risks of rapid-acting,
8184
types of, 5759
intensied therapy for, 5557
labor and delivery
considerations, 92
management of, 5378
maternal risks with, 7980
oral antidiabetic agents for,
administration of, 69
anomalies risks with,
63, 68
classication of, 7172
cost of, 70
insulin therapy versus,
6970
lactation and, 70, 73
pharmacology of, 6869
placental transfer of, 68
polycystic ovary syndrome and,
144146
postpartum management of,
9293
prevalence of, 53, 79
retinopathy progression risks
with, 8184
spontaneous abortion rate with,
54, 81
thyroid disease associated with,
8485
treatment of, 8890, 93
evidence-based criteria
for, 73
intensive, 5557
Type 2 diabetes versus, 5354
urinary ketone testing for, 88
U
Ultrasonography, for fetal surveillance,
gestational diabetes indications for,
9092
in pregnancy screening, for Cushings
syndrome, 4
for pheochromocytoma,
14
for primary adrenal
insuciency, 9
for thyroid nodules, 131132
Urinary free cortisol (UFC), in pregnancy
screening, for Cushings syndrome, 2
Urinary ketone testing, for gestational Type
I diabetes, 88
218
Uterine distention, in premature birth,
183184
V
Vitamin D deciency, during pregnancy,
4344
osteoporosis associated with, 39
INDEX
W
Weight loss, for infertility, 138139, 150