Download as pdf or txt
Download as pdf or txt
You are on page 1of 22

Contraception 87 (2013) 706 – 727

Review article

Ethinyl estradiol and 17β-estradiol in combined oral contraceptives:


pharmacokinetics, pharmacodynamics and risk assessment☆
Frank Z. Stanczyk a,⁎, David F. Archer b , Bhagu R. Bhavnani c
a
Departments of Obstetrics and Gynecology and Preventive Medicine, University of Southern California Keck School of Medicine, Los Angeles,
CA 90033, USA
b
Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, VA 23507, USA
c
Department of Obstetrics and Gynecology, University of Toronto, St. Michael's Hospital, Toronto, ON M5B 1W8, Canada
Received 30 October 2012; revised 25 December 2012; accepted 25 December 2012

Abstract

The need to seek improved combined oral contraceptive (COC) efficacy, with fewer health risks and better acceptability, has been
ongoing since the introduction of COCs more than 50 years ago. New progestin formulations combined with lower doses of ethinyl estradiol
(EE), the predominant estrogenic component of COCs, have reduced the incidence of venous thromboembolism and other negative outcomes
of COC treatment. Previous attempts to use endogenous 17β-estradiol (E2) instead of EE were limited primarily by poor cycle control. The
recent introduction of E2-based formulations has renewed interest to determine if there are potential benefits of using E2 in COCs. These
formulations have been shown to have similar efficacy and cycle control as EE-based COCs. This review provides a brief summary of the
pharmacology of EE and E2, including metabolism, pharmacokinetics and pharmacodynamics, as well as adverse effects of these estrogens.
© 2013 Elsevier Inc. All rights reserved.

Keywords: 17β-Estradiol; E2; Combined oral contraceptive; COC; Ethinyl estradiol; EE

1. Introduction ception, including COCs, intrauterine devices/intrauterine


systems, long-acting hormone-releasing implants, weekly
Combination oral contraceptives (COCs) have enabled transdermal patches and monthly vaginal rings. Despite the
millions of women to avoid unintended pregnancies since number of contraceptive options available to women, COCs
their introduction in 1960. Today, women have unprece- remain the most popular form of reversible hormonal birth
dented choices with regard to reversible hormonal contra- control, especially in North America and Europe.
Several progestins are used currently in COC formula-
tions in the United States. Each is combined with one of four
☆ estrogens: mestranol, ethinyl estradiol (EE), estradiol
Notes: Frank Z. Stanczyk is a consultant for Agile Therapeutics and
Merck & Co., Inc. (formerly Schering Plough, N.V. Organon), and has valerate (E2V) and 17β-estradiol (E2). The majority of the
consulted for Bayer Healthcare and Shionogi. David F. Archer is a past and current COCs contain EE.The initial presence of an
consultant for Agile Therapeutics; Bayer Healthcare; CHEMO; Depomed; estrogen in COCs was accidental owing to the presence of
HRA Pharma; Merck & Co., Inc.; Shionogi; Warner Chilcott; Watson the contaminant, mestranol, produced during the synthesis of
Pharmaceuticals and Wyeth Laboratories (now Pfizer Inc.). He has received the progestin norethynodrel [1,2]. Mestranol was later shown
research support from Bayer Healthcare; Duramed; Merck & Co., Inc.;
Warner Chilcott; Watson Pharmaceuticals and Wyeth Laboratories (now to be a prodrug for EE, and it is rarely used in COCs today.
Pfizer Inc) and has received direct lecture fees from Bayer Healthcare; The estrogenic component provides cyclic stability when
Merck & Co., Inc.; and Wyeth Laboratories (now Pfizer Inc.). Bhagu combined with progestins in COCs and enhances the
Bhavnani has nothing to disclose. suppressive effect of the progestin on the hypothalamic–
⁎ Corresponding author. USC Keck School of Medicine, Reproductive
pituitary–ovarian (HPO) axis.
Research Laboratory, Livingston Research Building, 1321 North Mission
Road, Los Angeles, CA 90033, USA. Tel.: +1 323 226 3220; fax: +1 323 Although EE and COCs are safe, there are concerns
226 2850. regarding adverse events (AEs) and side effects, especially in
E-mail address: fstanczyk@socal.rr.com (F.Z. Stanczyk). smokers. Concerns associated with COCs include cancer
0010-7824/$ – see front matter © 2013 Elsevier Inc. All rights reserved.
http://dx.doi.org/10.1016/j.contraception.2012.12.011
F.Z. Stanczyk et al. / Contraception 87 (2013) 706–727 707

(particularly breast cancer), blood clots, heart attacks, stroke, and compare whenever possible the advantages and
weight gain and loss of libido. Side effects include disadvantages of using these estrogens.
breakthrough bleeding, breast tenderness, nausea, headache
and mood changes.
Initial reductions in EE dose (from N 50 mcg to 30 mcg) 2. Materials and methods
were associated with decreased cardiovascular risk [3,4].
Although further reductions in the total dose of EE (from 30 An extensive literature review was performed using the
mcg to b 20 mcg) in COCs may theoretically be more MEDLINE/PubMed database. Additional resources were
tolerable, data from existing randomized controlled trials do identified using reference lists of the obtained articles as well
not have enough statistical power to enable the detection of as pertinent book chapters on the subjects of E2 and EE.
differences between COCs for these rare AEs [5]. Nonethe- Although we focused on the more recent literature, no limit
less, reductions in the dose of EE to ≤ 20 mcg have led to on year of publication was used in this review.
fewer AEs and side effects [5], yet room for improvement
remains. Low-dose COCs result in higher rates of bleeding
irregularities, and women receiving COCs with lower doses 3. Biochemistry of endogenous (physiologic) E2 in
of EE are more likely to discontinue treatment, possibly premenopausal women
leading to higher rates of unintended pregnancy. 3.1. Formation and secretion of hormones during the
Since the late 1970s, researchers and pharmaceutical menstrual cycle
manufacturers have investigated replacing EE with E2 or
E2V in COCs to improve their safety and tolerability profiles, The normal reproductive years of healthy women are
especially with regard to cardiovascular events, hemostatic characterized by several complex physiological changes that
parameters and lipid metabolism. Until recently, efforts to occur on a monthly basis. Some of these changes involve the
incorporate physiologic (endogenous) sex steroids into secretion of the hypothalamic hormone gonadotropin-
COCs have largely failed owing to poor cycle control, releasing hormone (GnRH); the anterior pituitary hormones
especially when E2 was administered as part of a monophasic follicle-stimulating hormone (FSH) and luteinizing hormone
or biphasic regimen [6–12]. (LH); and the ovarian hormones E2, E1 and progesterone.
The first COC with E2V, Femilar®, was a biphasic 21-day This monthly rhythmic pattern of the menstrual cycle
regimen containing a combination of E2V (1 mg) and consists of four distinct phases that include menstruation
cyproterone acetate (1 mg) taken for 10 days, plus E2V (2 and the follicular (proliferative), ovulatory and luteal
mg) and cyproterone acetate (2 mg) taken for 11 days [13]. (secretory) phases. Since this review deals with the use of
Femilar, first marketed in Finland in 1993, showed good E2-based COCs, the biosynthesis and metabolism of
efficacy, but bleeding profiles were more irregular than those endogenous E2 and its potential contribution to the E2
of COCs containing EE [14]. There are two possible derived from the COCs are discussed. To better understand
explanations for these bleeding problems: (a) the proges- this, the formation of E2 and its regulation during the normal
tin-stimulated endometrial 17β-hydroxysteroid dehydroge- menstrual cycle are briefly outlined.
nase (17β-HSD) rapidly converts E2 to estrone (E1), which The menstrual cycle is highly regulated by a delicately
cannot maintain stable endometrial proliferation, and (b) balanced interplay of the HPO axis, as shown schematically
progestins reduce nuclear E2 receptor concentrations and in Fig. 1 and discussed briefly below. As the circulating
thereby decrease nuclear E2 bioavailability and estrogenic levels of progesterone and estrogen from the previous cycle
endometrial proliferation. Thus, it appears that the decrease fall, menstrual bleeding occurs. The mean length of the cycle
in endometrial proliferation depends on the antiestrogenic is 28 (range, 21–35) days. The first 13 to 14 days comprise
effect of the progestin component. Due to this finding, new the follicular phase with ovulation occurring on approxi-
formulations combining E2 with progestins with lower mately days 14 to 16, and the last 14 days comprise the luteal
antiestrogenic potential have been developed to reduce phase. The variability in most menstrual cycles is due to the
endometrial breakdown [15,16]. One formulation consists of length of the follicular phase. Falling or low levels of E2 and
a combination of E2V (1, 2 or 3 mg) and the progestin progesterone during menstruation result in the secretion of
dienogest (2 or 3 mg) used in a multiphasic regimen [17–20]. GnRH, which in turn signals the pituitary to secrete
The other formulation consists of E2 (1.5 mg) combined with increasing levels of FSH, followed by increasing levels of
nomegestrol acetate (NOMAC; 2.5 mg) in a monophasic LH. These changes result in the initiation of growth of new
daily regimen [21–25]. The long-term potential benefits and ovarian follicles and ovulation; the sloughing off of the
adverse effects of these E2-based COCs remain to be endometrium formed during the previous cycle (menstrua-
established due to their recent introduction. tion) usually lasts for 3 to 7 days. The developing ovarian
The aims of this review are (a) to provide a comprehen- follicles start to produce increasing amounts of E2 that, in the
sive background on the biochemistry and physiology of E2, beginning of the new ovarian cycle, do not have a high
and the pharmacology of both E2 and EE, including their enough level in the blood to inhibit the secretion of FSH and
pharmacokinetics and pharmacodynamics, and (b) to discuss LH. Although both FSH and LH are required for ovarian
708 F.Z. Stanczyk et al. / Contraception 87 (2013) 706–727

fertilization of the ovum released during ovulation occurs


and pregnancy has been initiated, in which case it continues
Higher
Highe to produce large amounts of progesterone until the placenta
Brain
ain Cente
Center
takes over the production of progesterone approximately
± ve
Hypothalamic 7–8 weeks after conception.
GnRH
Concurrent with the ovarian secretion of E2, progesterone
and a number of other steroid hormones, there are peptides
E 2 /P Positive/negative feedback

± ve such as inhibin, follistatin and activin that have been shown


Pituitary Gonadotrope to play a role in both the negative and positive feedback
mechanisms discussed above. The endocrinology of the
FSH LH
menstrual cycle has been recently reviewed [35]. The
steroidogenic pathway in the ovary is regulated by LH and
Ovary FSH and will be discussed in a subsequent section.
Follicle Corpus
Ovulation
Luteum 3.2. Biosynthesis of steroid hormones by the human ovary

E2 E2 + P
During the premenopausal reproductive years, the human
ovarian follicle and the corpus luteum produce the sex
steroid hormones E2, progesterone and testosterone, by the
Normal COCs well-established classical pathway of steroidogenesis start-
Targets ing from cholesterol or from acetate, which is converted to
cholesterol by multiple reactions. Since this pathway has
Reproductive Tract Withdrawal
Menstrual Fallopian Tubes Bleeding been extensively reviewed [36–38], only the biosynthetic
Bleeding
Endometrium Prevention of pathway pertaining to the human ovary, from cholesterol to
Cervical Mucus Pregnancy
the sex steroids, is discussed briefly below (Fig. 2).

Fig. 1. Simplified depiction of HPO axis and feedback mechanisms during 3.3. Conversion of cholesterol to pregnenolone
normal menstrual cycle and potential target tissues in normal menstrual
cycle and during administration of COCs. P, progesterone. Cholesterol in the ovary can arise by de novo synthesis
from acetate, plasma low-density lipoprotein cholesterol
(LDL-C) or intracellular stores of cholesterol [34]. The rate-
steroidogenesis, it appears that early development of the
limiting step is the transfer of cholesterol from the cytosol to
follicles is independent of FSH [26–29]. As the dominant
the mitochondria, a process that is regulated by the
follicle continues to grow, increasing amounts of E2 are
steroidogenic acute regulatory (STAR) protein [39–41].
secreted. The rising levels of E2 secreted into the circulation
Cholesterol in the mitochondria is converted to pregneno-
have a negative feedback effect on the HPO axis resulting in
lone, a C21 steroid that is the major precursor of all steroid
a decrease in FSH levels. However, between days 12 and 13,
hormones [36]. This step is catalyzed by the inner
the amount of E2 reaching the pituitary gland attains a critical
mitochondrial membrane cytochrome P450 side-chain
peak level. This dramatic increase in E2 results in a positive
cleavage enzyme (CYP11A1) and is regulated by LH and
feedback signal to the pituitary, triggering an increased
FSH in the theca and granulosa cells. It is mediated further
secretion of LH and, to a lesser extent, FSH — the midcycle
by cyclic adenosine monophosphate, STAR and steroido-
surge. The LH surge induces ovulation which occurs 24 to
genic factor 1 [40–42].
36 h after the LH peak [30–33]. E2 reaches peak values a
day before the LH surge in most normal menstrual cycles 3.4. Conversion of pregnenolone to progesterone
[30–33]. Coincidental with the surge of gonadotropins, there
is a precipitous drop in the secretion of estrogens by the Pregnenolone formed in the mitochondria is converted in
ovary that appears to precede ovulation [30–33] and occurs the cytoplasm to progesterone by the enzyme 3β-hydro-
34 to 36 h after the initiation of the LH surge. xysteroid dehydrogenase Δ 5,4 isomerase type 2 (HSD3B2)
Following ovulation, the granulosa and theca cells in the [43]. This microsomal enzyme both catalyzes the conversion
follicle that released the ovum, along with the surrounding of the carbon 3-hydroxyl group of pregnenolone to a keto
stroma, capillaries and connective tissue, undergo morpho- group and isomerizes the Δ 5 to Δ 4 double bond, giving rise
logical changes and evolve into the corpus luteum. This to progesterone.
process is referred to as luteinization. Both the granulosa and 3.5. Conversion of progesterone to androstenedione
theca cells are luteinized and begin secretion as a temporary and testosterone
endocrine gland. During the midluteal phase, the corpus
luteum secretes large amounts of progesterone and lesser Progesterone is then transformed to androstenedione by
amounts of E2 [34]. The functional life span of the corpus the microsomal P450c17 (CYP17A1), an enzyme that has
luteum is normally 14 days, after which it regresses unless both 17α-hydroxylase and the C17,20-lyase activities. In this
F.Z. Stanczyk et al. / Contraception 87 (2013) 706–727 709

HO
Cholesterol (LDL-Cholesterol)

Side Chain Cleavage


Enzyme (CYP11A1) StAR
CH3 CH3
C=O C=O

3β-Hydroxysteroid Dehydrogenase
∆5,4 Isomerase
(HSD3B2)
HO O
Pregnenolone Progesterone
17α -Hydroxylase 17α -Hydroxylase
(CYP17A1) (CYP17A1)
CH3 CH3
C=O C=O
OH OH
3β-Hydroxysteroid Dehydrogenase
∆5,4 Isomerase
(HSD3B2)

HO O
17α-Hydroxypregnenolone 17α-Hydroxyprogesterone

17,20-Lyase 17,20-Lyase
(CYP17A1) (CYP17A1)

O O

3β-Hydroxysteroid Dehydrogenase OH
5,4
∆ Isomerase
(HSD3B2) 17β-HSD
HO O
Dehydroepiandrosterone Androstenedione
O
Aromatase Testosterone
(CYP19A1)
O
Aromatase
(CYP19A1) O

17β-HSD

HO
Estrone HO
17β-Estradiol

Fig. 2. Biosynthesis of steroids from cholesterol in the normal ovary. The steroidogenic genes involved are shown in parentheses; 17β-HSD enzymes are
involved in the interconversion of androstenedione to testosterone and estrone to estradiol, respectively.

conversion, progesterone is first transformed to 17α- (DHEA) by P450c17, and then DHEA is converted to
hydroxyprogesterone followed by the cleavage of the C17– androstenedione by HSD3B2. Androstenedione is converted
C20 side chain to yield the C19 steroid androstenedione. to testosterone via the enzyme 17β-HSD. Testosterone can
Alternatively, pregnenolone can be first converted to 17α- also be formed by another pathway in which DHEA is first
hydroxypregnenolone and to dehydroepiandrosterone converted to 5-androstene-3β,17β-diol by 17β-HSD, and
710 F.Z. Stanczyk et al. / Contraception 87 (2013) 706–727

the latter compound is then converted to testosterone by such as the breast, skin, adipose, brain, bone and muscles
HSD3B2. During the follicular phase, this process is from circulating DHEA, mainly of adrenal origin [47–53].
regulated by a synergistic action of LH and FSH and by E1 can also be metabolized to E2 in extraglandular sites.
paracrine interactions between the granulosa and theca cells, However, only about 5% of E2 formed by this extraglandular
a process that will be discussed later. pathway enters the circulation [54–57].
Based on the pathways just discussed, the plasma
3.6. Conversion of androgens to estrogens concentration, ovarian secretion rate and production rate
(sum of rates of extraglandular and ovarian secretions) of E2,
E1 and E2 are produced from androstenedione and
E1 and progesterone during the menstrual cycle are shown in
testosterone, respectively, by a complex series of enzymatic
Table 1. E2 and E1 secretion and plasma concentrations
reactions catalyzed by a single microsomal P450aro
are lowest during the early follicular phase, highest during
(CYP19A1) enzyme, resulting in aromatization of ring A
the late follicular phase and substantial during the luteal
of the androgen [44,45]. These estrogens are interconvert-
phase [30–33].
ible. E1 is converted to E2 and E2 to E1 through the action of
HSD17B1 and HSD17B2, respectively.
3.8. In vivo transport of estrogen and progesterone
3.7. The two-cell hypothesis
Serum E2 and progesterone reversibly bind to sex
Theca cells in the ovarian follicle produce and secrete the hormone-binding globulin (SHBG) and corticosteroid-bind-
androgens that diffuse into the granulosa cells where they are ing globulin (CBG), respectively. This form of binding of
aromatized to estrogens [44,46]. The hypothesis termed steroid hormones plays an important role in the transport and
the two-cell hypothesis of steroidogenesis proposes that the distribution of these hormones. Circulating E2 and proges-
theca and granulosa cells play synergistic roles in the terone are primarily bound nonspecifically to serum albumin
biosynthesis of E2 by the maturing preovulatory follicle. with low affinity and specifically with high affinity to SHBG
During the follicular phase, LH acting via its receptors in the and CBG [58–60]. Only a small fraction (1%–3%) of these
theca cells stimulates the production of androstenedione and hormones circulate in the unbound or free form [59,60]. It is
testosterone from cholesterol, which then diffuse into the generally recognized that only the free (unbound) form of
granulosa cells where FSH stimulates the aromatization of steroid hormones is able to enter the target cells to exert its
androstenedione and testosterone to E1 and E2, respectively. biological effect, or be further metabolized and excreted.
Granulosa cells lack the ability to produce cholesterol, There is, however, some suggestive evidence that some
pregnenolone or progesterone and depend on the obligatory protein-bound steroids also can enter the cell, but the
estrogen precursor androstenedione produced in the theca physiological significance of this observation remains to be
cells. The corpus luteum formed after ovulation accumulates established [61]. Since steroid hormones bind to serum
large deposits of cholesterol derived primarily from LDL-C. albumin with low affinity (1×10 4–1×10 6 Ka) and high
The theca and granulosa cells, under the influence of capacity, these bound hormones can dissociate to free
gonadotropins, are transformed into theca lutein and hormones readily, and the pool of albumin-bound hormone
granulosa lutein cells, respectively. These luteinized cells is also considered to be available for biological action. It has
are now the major source of sex steroids secreted by the been estimated that, in women with normal menstrual cycles,
ovary postovulation. approximately 60% of the E2 in the circulation is bound to
It is important to note that the conversions of androstene- albumin, and 38% is bound to SHBG (Ka= 1×10 8–1×10 9)
dione to testosterone and E1 to E2 are both reversible, and in [62,63]. In contrast, only about 17% of progesterone is
the ovary during both phases of the menstrual cycle, the bound to CBG; the remainder is bound predominantly to
predominant estrogen secreted is E2. Although, in ovulatory albumin [60].
women, this is the main mechanism for the formation of E2, The levels of SHBG synthesized in the liver are controlled
small amounts of E2 are formed in extraglandular tissues by the levels of various hormones. Thus, testosterone and

Table 1
Plasma concentrations, ovarian secretion rates and total production rates of estradiol, E1 and progesterone during the various phases of the menstrual cycle
Steroid Early follicular Late follicular Midluteal
Plasma Secretion rate Production rate Plasma Secretion rate Production rate Plasma Secretion rate Production
concentration (mg/day) (mg/day) concentration (mg/day) (mg/day) concentration (mg/day) rate (mg/day)
(pg/mL) (pg/mL) (pg/mL)
Estradiol 60 0.07 0.08 330–700 0.4–0.8 0.44–0.94 200 0.25 0.27
E1 50 0.08 0.11 150–300 0.25–0.5 0.33–0.66 110 0.16 0.24
Progesterone 11,300 24 25
Modified from Little and Billiar [30], MacDonald et al. [31], Baird et al. [32], Baird and Guevara [33].
F.Z. Stanczyk et al. / Contraception 87 (2013) 706–727 711

insulin inhibit SHBG synthesis, while E2 and thyroxine leading to carcinogenesis. Although the mechanisms have
stimulate SHBG formation [64]. The oral administration not been delineated, it appears that these hydroxylated
of E2 increases SHBG levels, resulting in a greater metabolites are easily auto-oxidized to semiquinones
percentage of E2 bound to SHBG, and therefore, a lesser and quinones that can covalently bind to DNA, thereby
amount of free hormone will be available for biological resulting in DNA damage [67–72]. To date, there is no direct
action. In contrast, androgen excess, hypothyroidism and evidence that 2- and 4-hydroxyestrogens are carcinogenic in
obesity lower SHBG levels and result in an increase in the vivo in humans. Various proposed hypotheses and mecha-
free fraction of E2 [65]. The extent and degree of binding of nisms are circumstantial.
E2 to serum proteins not only determines the amount of E2 Apart from the metabolism of E1 and E2 to catechol
available for biological action but, more importantly, can estrogens, these estrogens also can undergo 16α-hydroxyl-
have an effect on the overall metabolic clearance rate and ation (P450 3A4) to form estriol, which is rapidly cleared
other pharmacokinetic parameters [38,66], which will be and excreted in urine as estriol glucuronide. In addition to
addressed in subsequent sections. estriol (16α-hydroxyestradiol), 16α-hydroxyestrone also is
formed, and this 16α-hydroxylated estrogen has been
3.9. Metabolism of endogenous estradiol implicated in the oncogenic process [73,74]. The hypothesis
is based on the demonstration that 16α-hydroxyestrone, a
Besides the reversible metabolic interconversions be- major urinary metabolite of E2 and E1, can form large stable
tween E1 and E2, estrogens undergo irreversible metabolism covalent adducts (compounds) with macromolecules such as
in tissues such as the liver and the kidney and, to some DNA and proteins (Fig. 3) [75,76]. It has further been
extent, in target tissues such as the endometrium and brain suggested that this adduct formation plays a role in the
[38]. The major irreversible metabolic transformations of etiology of breast cancer [75,76]. Based on in vitro
estrogens occur in the A and D rings, giving rise to mainly experiments, it has been proposed that the covalent adduct
2-, 4- and 16α-hydroxylated metabolites (Fig. 3). formation between 16α-hydroxyestrone and macromole-
The 2- and 4-hydroxylated metabolites are the so-called cules occurs because of the presence of D ring α-ketol
catechol estrogens. In vivo and in vitro data indicate that (Fig. 3, circled). It also has been hypothesized that estrogen
these metabolites are formed in humans, and, more metabolism favoring 16α-hydroxylation leads to increased
importantly, it has been suggested that the oncogenic cancer risk in postmenopausal women [77]. Based on the
potential of E1 and E2 depends on the extent of their observation that 16α-hydroxyestrone is a potent estrogen
metabolism to extremely labile catechol estrogens. The whereas 2-hydroxyestrone is relatively inactive, it has been
hypothesis, in general, is that the cytochrome P450- proposed that the ratio of urinary 2-hydroxyestrone to 16α-
dependent oxidative metabolism of estrogens leads to hydroxyestrone is a predictor of cancer risk and that this ratio
the formation of metabolites that generate reactive free is inversely correlated with the risk for breast and cervical
radicals that can cause oxidative stress and genomic damage cancers [77,78]. In contrast, a number of well-carried-out

2 and 4-Hydroxy-Catechol Estrogens


OH

O HO
Estradiol

HO Potential Stable Adducts


O
Estrone OH

HO
-Hydroxyestrone

Fig. 3. In vivo and in vitro metabolism of estrone via catechol estrogens and 16α-hydroxylation. The potential for adduct formation with macromolecules is
indicated. The α-ketol is circled. Modified from Bhavnani [66].
712 F.Z. Stanczyk et al. / Contraception 87 (2013) 706–727

studies did not find a significant correlation between the ratio As described earlier, E2 and E1 are metabolized to
of urinary 2-hydroxyestrone and 16α-hydroxyestrone and numerous metabolites. The metabolites that contain a
breast cancer risk in both premenopausal and postmeno- hydroxyl group can be conjugated via sulfuryl or glucuronyl
pausal women, with or without breast cancer [79–82]. transferases to form a variety of hydroxylated and nonhy-
The circulating levels of catechol and 16α-hydroxylated droxylated sulfates and glucuronides of E2 and E1. If we take
estrogens obtained after oral administration of the new into account all the unconjugated and conjugated forms,
E2-based COCs need to be determined. However, one would there are over 100 metabolites of E2 and E1 found in the
expect them to be relatively low owing to their extremely circulation. The estrogen metabolites formed from exoge-
high MCRs. nous E2 are essentially the same as those formed from
endogenous E2 and E1 except that they are present in much
higher concentrations.
4. Pharmacokinetics of E2 and EE E1S is the principal circulating metabolite of E 2,
irrespective of the route of administration. About 15% of
4.1. Pharmacokinetics of E2
orally administered E2 is converted to E1 and about 65% to
4.1.1. Absorption and distribution E1S [90]. E1S appears to form a pool from which E2 can be
E2 is rapidly absorbed following oral administration of regenerated, first by transformation of E1S to E1 through the
therapeutic doses [83]. Circulating E2 levels are dose action of sulfatase and then conversion of E1 to E2 via the
dependent. After a single oral dose administration of 2 mg action of HSD17B1. About 5% of E1 and 1.4% of E1S can be
E2 to young women, maximum plasma E2 levels of 30 to converted back to E2. Another 21% of E1S can be
50 pg/mL were observed within 6 to 10 h [84]. The levels transformed to E1, and the conversion of E1 to E1S is
increased linearly up to an E2 dose of 4 mg, but absorption about 54%.
was incomplete after a dose of 8 mg. Because of the presence of the highly active 17β-HSD in
Although reduced particle size (micronization) of orally the gastrointestinal mucosa, serum levels of E1 are several-
administered E2 has been shown to improve its absorption fold higher than those of E2. Following oral administration of
[85], E2 still has a very low bioavailability. Together with the 1 mg micronized E2 to a postmenopausal woman, serum E2
first-pass metabolism in the liver, about 95% of the levels are in the range of 30 to 50 pg/mL, whereas the
administered E2 is metabolized before entering the systemic corresponding range of E1 levels is 150 to 300 pg/mL [91].
circulation [86]; less than 5% of the administered E2 is The E1 levels are, on average, about five times higher than
bioavailable. It has been shown that, of the administered E2 those of E2 and can be about 10 times higher in some
dose, 15% is absorbed as E1, 25% as E1 sulfate (E1S), 25% as women. The E1/E2 ratio obtained after oral E2 administration
E1 glucuronide and 25% as estradiol glucuronide. differs from the corresponding ratios found in premenopau-
As shown earlier in premenopausal women, approxi- sal women during the menstrual cycle and in postmeno-
mately 38% and 60% of circulating E2 are bound to SHBG pausal women not receiving oral estrogen therapy; these
and albumin, respectively, so that about 2% of E2 is in free ratios are, on average, 0.6:1 and 3:1, respectively. The
form [59]. However, during oral E2 treatment, serum SHBG biologic significance of the different E1/E2 ratios is not
levels increase, resulting in a reduced free E2 fraction. known. In contrast to the oral route of E2 administration, the
E1/E2 ratio is approximately 1 when E2 is administered
4.1.2. Metabolism parenterally (e.g., by transdermal patch).
When E2 is administered orally, it undergoes extensive When E2 is administered orally, serum levels of E1S rise
hepatic first-pass metabolism. A considerable part of the dramatically. The biologic significance of the elevated E1S
administered dose is metabolized in the gastrointestinal levels obtained after oral E2 administration is not known;
mucosa, predominantly through the action of HSD17B2, however, E1S itself has no biologic activity. When the E2
forming E1. Following its absorption, partly metabolized derivative E2V is administered orally, it is rapidly and
and unmetabolized E2 enters the portal vein blood, which completely hydrolyzed to E2 during the hepatic first pass
perfuses the liver where a variety of enzymes, including [92]; however, there may be considerable intersubject
hydroxylases, methyltransferases, and sulfuryl and glu- variability in the E2 concentration obtained. The metabolism
curonyl transferases, transform E2 and E1 to a variety and pharmacokinetics of E2 derived from E2V are the same
of metabolites. as those of E2.
Estrogens are excreted to a significant extent in bile and
are partly reabsorbed from the intestine. The enterohepatic 4.1.3. Excretion
recirculation of E2 and E1 has a delaying effect on their final Following oral administration of 2 mg of E2V to
elimination from the body [87,88]. Because of the large pool premenopausal women, 54% of the dose was recovered in
of estrogen metabolites formed and the enterohepatic urine and only 6% in the feces [93], even though a
recirculation, the terminal half-life of E2 represents a substantial amount of E2 has been shown to be excreted in
composite parameter that is dependent on these processes bile [94]. Thus, it appears that there is a highly efficient
and is in the range of about 13 to 20 h [89]. enterohepatic recirculation of biliary excreted estrogen
F.Z. Stanczyk et al. / Contraception 87 (2013) 706–727 713

metabolites which, following reabsorption, are metabolized 1.77 have been calculated for EE, with a terminal half-life of
further and eventually excreted in urine [89]. After oral 10 to 20 h and a 24-h dosing interval [89]. This means that, in
administration of 3H-E2, the urinary estrogen metabolites are comparison to single-dose EE administration, one can expect
predominantly in the form of glucuronides, but a substantial an accumulation between 23% and 77% during repeated oral
amount of sulfated estrogen is also present [86]. The main EE administration.
urinary estrogens include glucuronides of E1 (13%–30%),
2-hydroxy-E1 (2.6%–10.1%), E2 (5.2%–7.5%), E3 (2.0%- 4.2.2. Metabolism
5.9%) and 16α-hydroxy-E1 (1.0%–2.9%). Oxidation reactions at various carbons of the steroid
nucleus play a major role in the metabolism of EE, similar to
that of E2. Quantitatively, the most important pathways of
4.2. Pharmacokinetics of ethinyl estradiol
E2 metabolism involve 2-hydroxylation and 16α-hydroxyl-
4.2.1. Absorption and distribution ation, as discussed earlier. In contrast, 16α-hydroxylation
Similar to the absorption of E2, EE is rapidly and of EE has not been reported presumably owing to blockage
completely absorbed following oral administration of thera- (steric hindrance) by the ethinyl group at carbon 17. The
peutic doses [95]. After the administration of 30 mcg EE, peak 2-hydroxylation of EE is quantitatively the most important
plasma EE levels of 90 to 130 pg/mL are found within 1 to 2 h pathway of EE metabolism [89]. Hydroxylations of EE
after ingestion [95]. Occasionally, a secondary peak can be at carbons 4, 6 and 16β have been reported but appear
observed about 10 to 14 h following oral EE administration, to contribute only to a small extent to its metabolism.
which is attributed to the enterohepatic circulation of EE. Whether the absence of 16α-hydroxylation of EE compared
Dose proportionality in EE levels is observed with EE doses with E2 is of any pharmacological significance remains to
of 20 to 100 mcg [95]. Although there is considerable be determined.
presystemic metabolism during the hepatic first pass of orally A considerable part of the intersubject variability in EE
administered EE, it is not as extensive as that of E2. The mean pharmacokinetics, which will be discussed later, can be
oral bioavailability of EE is approximately 45%, with a large attributed to differences in extent of 2-hydroxylation of EE,
inter- and intraindividual variability in the range of 20% to which is primarily catalyzed by hepatic cytochrome P450
65% [96]. Owing to the large variability in EE bioavailability, 3A4. A wide variation in levels of this enzyme has been
EE levels produced by 35 mcg EE in one woman may be demonstrated among individuals [101].
indistinguishable from those generated by 50 mcg in another Both EE and its hydroxylated metabolites undergo
woman [97]. extensive conjugation, primarily in the liver. EE is rapidly
Unlike E2, which binds with high affinity to SHBG in conjugated partly to a glucuronide (EE-3-glucuronide and
blood, EE does not bind to SHBG [98]. Following oral EE-17-glucuronide), which is inactive and undergoes rapid
administration of EE, the time course of plasma EE levels renal clearance, and to EE-3-sulfate and EE-17-sulfate,
can be described by a two-compartment model in most cases: which circulate at concentrations that are about 10-fold
a rapid distribution phase followed by a terminal disposition higher than EE itself [102]. During enterohepatic recircula-
phase that is characterized by a half-life in the range of tion, the EE sulfates are partially deconjugated to EE (the
approximately 5 to 30 h [96]. The large range in terminal 17-sulfate to about 12% and the 3-sulfate to about 20%), thus
half-life can be attributed only partly to interpatient providing a relatively small contribution to the circulating
variability. The other important contributing factor pertains unconjugated EE.
to measurement of low levels of EE [89]. When EE is
administered in doses b 50 mcg, plasma EE levels at 24 h are 4.2.3. Excretion
usually at the lower limit of measurement of the EE assays. In contrast to oral administration of E2, considerably more
Therefore, a terminal half-life that is longer than 10 h cannot of the total excreted EE is eliminated in feces compared with
be determined with any accuracy; this is a limitation of most urine. In one study [103], approximately 62% of the total
published half-life data of EE. More sensitive EE assays excreted radioactive EE and its metabolites were eliminated
have to be developed to obtain accurate EE terminal half- in feces and about 38% in urine following oral administration
lives (e.g., by use of mass spectrometry assays). of 3H-EE to women.
During repeated oral administration of EE, an increase in About 6% of the administered oral EE dose appears as
trough serum EE levels can be observed until approximately untransformed EE in urine [104], and the remaining portion
5 to 10 days after initiating treatment, with only minimal includes predominantly conjugated EE metabolites, which
change until the end of the treatment cycle [99,100]. Several are mostly (about 80%) in the form of glucuronides (3-
studies have demonstrated an increase in serum EE levels glucuronides; 17-glucuronides; 3,17-diglucuronides); sulfat-
between 30% and 60% within a treatment cycle with ed forms constitute only about 8% to 10% [89]. About 9% of
different COCs [89]. This large range can be explained by orally administered EE appears as untransformed EE in
the pharmacokinetic accumulation of EE and is determined feces, whereas the conjugated forms are poorly characterized
by calculating the ratio of the terminal half-life of EE and the [89]. However, the predominant forms are EE glucuronide
dosing interval. Accumulation factors ranging from 1.23 to and EE sulfate in bile [105].
714 F.Z. Stanczyk et al. / Contraception 87 (2013) 706–727

5. Mechanism of action of EE and E2 progestin play a role, but the type of regimen also seems to
determine the effect on levels of unfavorable lipoproteins.
Estrogens interact with estrogen receptors (ERs) that are Several short-term clinical studies using various newer
members of a superfamily of nuclear receptors. Estrogen COC formulations consisting of EE b 50 mcg combined with
receptor α (ERα) and estrogen receptor β (ERβ) can be one of a variety of progestins such as levonorgestrel (LNG),
detected in a broad spectrum of tissues. Both receptor types norethindrone, norgestimate, gestodene or desogestrel gen-
are expressed at similar levels in some organs, whereas one erally show an increase in triglycerides, which is most likely
subtype predominates in other organs. In addition, both ascribed to the estrogen component. Variable effects on
receptor subtypes may be present in the same tissue but in LDL-C, HDL-C, apolipoprotein A and apolipoprotein B also
different cell types. ERα is mainly expressed in the uterus, were found. The changes were small and appeared to have no
prostate, ovary, testes, epididymis, bone, breast, various significant impact on lipid profiles after 12 months of use in
regions of the brain, white adipose tissue and liver, whereas most of the studies [116–122].
ERβ is expressed primarily in the colon, prostate, testis, Some of these short-term studies suggested small
ovary, bone marrow, salivary gland, vascular endothelium differences between monophasic and triphasic formulations.
and certain regions of the brain [106]. Both E2 and EE exert It is highly unlikely that subtle changes in lipid profiles can
their biological effects via interactions with estrogen have a significant impact on the incidence of cardiovascular
receptors by the same mechanism that has been extensively disease in young women using the newer lower-dose COCs.
reviewed [107–111] and will not be discussed here. This is in keeping with the rarity of arterial disease diagnosed
Based on this mechanism, it has been shown that E2 is in young women [123] and, more importantly, observations
100-fold less active compared with EE, and it has been that 64% of young women who had sustained a myocardial
recognized for decades that much higher doses of oral E2 are infarction (MI) while they were using COCs did not show
required to bring about the same degree of biological effect angiographic atherosclerosis [124]. Similarly, a review of 14
as by EE [112]. reports of MI and use of COCs showed no evidence of
significant fixed obstruction by arteriography in these
women [125]. The authors [124] concluded that MIs in
6. Pharmacodynamic effects of E2 and EE COCs users may be a discrete disease entity unrelated to
coronary atherosclerosis. A large study by the Royal College
Pharmacodynamic effects of orally administered E2 have of General Practitioners (RCGP) also found no significant
been studied primarily in postmenopausal women, whereas effect of COC use on acute MI in nonsmokers; however, the
pharmacodynamic effects of oral EE have mostly been risk was significantly higher in smokers who were over the
studied in combination with a progestin in premenopausal age of 35 years [126].
women. We provide a general overview of the pharmaco- A review of numerous studies dating from 1980 to 1993
dynamic effects of COCs containing either EE or E2 in including those involving the newer progestins (gestodene,
women in the following sections. norgestimate or desogestrel) combined with 35 or b 35 mcg
of EE indicated that these newer progestins were less
androgenic than the older formulations, specifically those
6.1. Metabolic effects
containing LNG and norethindrone. The new generation of
6.1.1. Lipids progestins has a lower impact on apolipoprotein and
It has been known for over 30 years that the comparison carbohydrate metabolism; however, whether these small
of earlier high-dose COCs with more recent lower-dose differences have any clinical relevance remains to be
formulations shows that the effect of COCs on the levels of established [127]. In a more recent case–control study, a
lipoproteins depends on the ratio of estrogen and progestin in direct comparison of users of more recent formulations of
each formulation [113]. Premenopausal women who used COCs versus older formulations showed that use of newer-
COCs with a high-dose estrogen/low-dose progestin ratio generation COCs was not associated with an excess risk for
were found to have significantly higher concentrations of MI [odds ratio (OR), 0.28; confidence interval (CI), 0.09–
high-density lipoprotein cholesterol (HDL-C) compared 0.86] [128]. The study further showed that the risk was
with nonusers [114]. In contrast, women who used low- ameliorated in smokers who used the newer COCs. A
estrogen/high-progestin COCs had a significantly lower number of studies have shown that smoking is the most
level of HDL-C but higher levels of LDL-C. Whether these important independent risk factor and that concomitant use
effects on lipoprotein cholesterol levels affect cardiovascular of COCs by smokers who were over the age of 35 years
risk is controversial. Some of the metabolic changes such as significantly increased the risk of MI [126,129,130]. These
increased LDL-C, triglycerides and insulin levels and observations and a number of others were considered by a
decreased levels of HDL-C are similar to those observed in consensus panel [131], and their recommendations were that
individuals who are at increased risk of coronary heart patients over 35 years of age who smoked heavily (N 15
disease (CHD) [114]. These findings have been reviewed cigarettes/day) should be “denied the use of oral contracep-
previously [115]. Interestingly, not only does the type of tives.” Their review of COCs containing 20 mcg EE
F.Z. Stanczyk et al. / Contraception 87 (2013) 706–727 715

suggested that this dose may be a safer alternative, even for to two progestin-only formulations, were compared to non-
those who are over 35 years of age and smoke occasionally. COC use. Depending on the type of progestin and its dose in
However, the general consensus is that, in women over 35 the COC users, the plasma glucose levels obtained following
years of age who are heavy smokers, COCs are most a glucose tolerance test were 43% to 61% higher than in
definitely contraindicated, as recommended by the US controls, insulin response was 12% to 40% higher, and the
Centers for Disease Control and Prevention [132]. The C-peptide response was 18% to 45% higher than in nonusers.
results from another large population-based case–control The progestin-only formulations were associated with only
study (3989 patients and 4900 controls) similarly showed minor metabolic effects compared with nonusers. Thus, it
that both current and former smokers had a moderately would appear that insulin sensitivity is mainly affected by the
increased risk of venous thromboembolism (VTE) (OR, estrogenic component, and the magnitude of the response
1.43; CI, 1.28–1.60 in current smokers and OR, 1.23; CI, can be modified by the dose and type of progestin [115].
1.09–1.38 in former smokers) compared with nonsmokers The association between COC use and incidence of type 2
[133]. More importantly, women who were current smokers diabetes [non-insulin-dependent diabetes mellitus
and used COCs had an approximately ninefold higher risk of (NIDDM)] was examined in a large prospective study (the
VTE (OR, 8.79; CI, 5.73–13.49) than nonsmokers who were Nurses' Health Study cohort) [138]. The findings indicate
not COC users. The risk was fivefold higher in women who that current users of COCs did not have an increased risk of
were identified as having a factor V Leiden mutation and NIDDM [relative risk (RR), 0.86; 95% CI, 0.45–1.61]
current smokers, and for those identified with the prothrom- compared with never users. In this study, 98,590 nurses,
bin 20210 A mutation, the risk was sixfold higher compared aged 25 to 42 years and free of diabetes, coronary heart
to women without the mutation [133]. disease, stroke and cancer, were followed for 4 years. No
The effects of an E2-based COC on lipids, carbohydrates significant increase was detected in NIDDM among current
and hemostasis were compared to an EE-based COC in a users or past users of low-dose COC preparations. The
recent study [134]. A total of 121 women, aged 18–50 years, authors also concluded in this study that the number of cases
were randomly assigned to receive E2 (1.5 mg)/NOMAC of NIDDM in COC users was very small (185 cases) during
(2.5 mg) in a 24/4-day regimen (N= 60) or EE (30 mcg)/LNG 352,067 person-years of follow-up, indicating a low absolute
(150 mcg) in a 21/7-day regimen (N= 61) for six cycles. The risk of this disease in young women [138].
E2/NOMAC group showed a favorable lipid profile A large epidemiologic study [139] surveyed data from 89
compared to the EE/LNG group; however, the changes randomly selected locations in the United States. The study
were overall small and within the normal range, and may be findings showed that the vast majority of current COC users
due to the differences in the regimen (24 vs. 21 days) and/or were using newer low-dose estrogen monophasic and
the progestins in the formulations. Detailed long- triphasic formulations. Moreover, the study found that
term clinical studies are needed to evaluate the effects of current COC users did not have adverse carbohydrate-
E2-based COCs on lipid parameters in comparison to EE- related side effects such as elevated levels of hemoglobin
based COCs. A1c, fasting glucose, insulin and C-peptide levels. Similarly,
Because of the rare finding of CHD in young women, the in former users of low-dose COCs, there were no significant
determination of the safety profile of new COC formulations changes in these parameters. Thus, low-dose EE-based
is an ongoing challenge. Whether the subtle minimal COCs are not associated with any adverse changes in
changes in metabolic parameters among different COCs glucose metabolism. Studies that investigated the effect of
have any relevant clinical significance has not yet been E2-based COCs on carbohydrate metabolism in healthy
demonstrated. The sample size in studies required to firmly young premenopausal women are scarce. The short-term
evaluate these associations would be extremely large so as to study by Ågren et al. [134] described above showed minor
make such studies difficult to investigate [123]. changes in carbohydrate metabolism between E2- and EE-
based COCs. Increases in glucose and insulin were
6.2. Carbohydrates significantly greater with the latter formulation. This
finding is similar to that reported in other studies, but it is
Older high-dose COCs were associated with changes in unknown whether these observations have any clinical
carbohydrate metabolism in women. Elevated blood sugar significance [140,141].
and insulin levels along with impaired glucose tolerance
were observed in high-dose COC users [135–137]. Since 6.3. Vasculature
these effects were thought to be mainly due to the estrogen
component of COC preparations, the estrogen dose has Vascular endothelial and smooth muscle cells express
significantly declined over the past 25 years. both ERα and ERβ. Estrogen has both rapid vasodilatory
Another study [115] explored the effects of nine different effects and longer-term actions that inhibit the response to
oral contraceptive formulations on lipid and carbohydrate vascular injury and prevent atherosclerosis [142]. Some of
metabolism; seven COCs with various doses and types of the rapid estrogen signaling in the vasculature may be
progestins combined with 30 mcg to 40 mcg EE, in addition mediated through the activation of G-protein-coupled
716 F.Z. Stanczyk et al. / Contraception 87 (2013) 706–727

estrogen receptors [143]. Estrogen promotes vasodilation by production and breakdown of fibrin. The purpose of the
inducing the production of endothelial nitric oxide (NO) clotting mechanism is to produce thrombin (activated factor
synthase and NO release. NO has strong vasodilatory and II), which converts fibrinogen to a fibrin clot. Thrombin is
antithrombotic properties and triggers the formation of cyclic generated from prothrombin through the action of factor X in
guanosine monophosphate (cGMP). Estrogen also affects the presence of factor V and factor VIII. The coagulation
the production of other endothelial factors, including pathway is initiated by the release of the inhibitor, tissue
products of arachidonic acid, by increasing the activity of factor, which activates factor VII; the latter factor, in turn,
cyclooxygenase and prostacyclin synthase [142]. activates factor X. In addition to tissue factor, other
In postmenopausal women, short- and long-term E2 inhibitors that safeguard against excessive thrombosis
dosing enhances flow-mediated dilation (reactive hyper- include antithrombin III, which inhibits the formation of
emia) [144,145]. However, no studies have explored thrombin, and protein C and protein S, which inhibit the
the effect of EE without concomitant progestin administra- formation of factors V and VIII.
tion on vascular blood flow in women. In premenopausal The coagulation system is balanced by the fibrinolytic
women, basal NO production and release (measured by system, which involves the degradation of fibrin and the
changes in flow-mediated dilation) appeared to be enhanced lysing of clots by plasmin. Plasmin is formed from
by use of COCs containing EE [146]. EE-containing COCs plasminogen through the action of tissue-type plasminogen
also influenced the renal excretion of various vasoactive (t-PA), which is balanced by a specific inhibitor, plasmin-
markers: cGMP excretion was significantly enhanced and ogen activator inhibitor type 1 (PAI-1).
thromboxane metabolite excretion was decreased after 12 A variety of laboratory assays are used to assess
cycles [147]. coagulatory and fibrinolytic activity. Prothrombin fragment
1+2 (F1+2) and the thrombin/antithrombin complex are
6.4. Hypertension determined to evaluate the thrombin generation rate. Tissue
factor antigen, factor VII activity and F1+2 reflect
Blood pressure (BP) varies during the menstrual cycle
coagulatory activity. Antithrombin III, protein C and protein
[148]. A number of studies have shown that use of high-dose
S are important measures of the inhibition of thrombin
COCs, containing at least 50 mcg EE and 1 mg to 4 mg of
generation and thrombin activity. Assays used to assess
progestin, is associated with a moderate increase in BP in
fibrinolytic activity include the plasmin–antiplasmin com-
approximately 5% of women [149]. Some studies have
plex, t-PA and PAI-1. Finally, D-dimer, which is a
assessed the rise in BP according to the estrogen dose and
degradation product of fibrin, is used to assess the net
progestin type used in the formulation. Thus, use of 250 mcg
activity of the hemostatic system.
LNG+50 mcg EE and 1 mg norethisterone (NET)+30 mcg
Studies investigating the effect of EE or E2 alone on the
EE for up to a year was not associated with a significant BP
coagulation and fibrinolytic systems in premenopausal
increase. In contrast, use of 250 mcg LNG+30 mcg EE
women are lacking. Existing studies involving the effects
showed a slight but significant increase [150]. It would
of EE on these systems have been carried out with EE-based
therefore appear that the effect of the progestin to estrogen
COCs with different progestins. Since E2-based COCs are
ratio in COCs may be more important than just the dose on
relatively new, there is a paucity of data on their effects on
BP changes. Another study that compared four COCs
the hemostatic system. Some of these studies with EE- and
containing 1 mg NET, 150 mcg LNG, 150 mcg desogestrel
E2-based COCs will now be discussed.
or 75 mcg gestodene combined with 30 mcg EE showed that
It has been shown that both the estrogenic and
all four formulations were associated with a small but
progestational components of COCs influence hemostasis.
significant increase in BP. No woman in the trial developed
High-dose COCs (50 mcg EE) increase the production of
clinically significant hypertension necessitating discontinu-
clotting factors such as factor V, factor VIII, factor X and
ation of the COC [151].
fibrinogen [152]. Some studies have shown that monophasic
A large prospective cohort study [149] consisting of
and multiphasic low-dose (30–35 mcg EE) COCs have no
68,297 female nurses aged 25 to 42 years found that,
significant clinical impact on the coagulation system and that
compared with women who had never used COCs, the age-
slight increases in thrombin formation are offset by increased
adjusted RR for hypertension was 1.5 (95% CI, 1.2–1.8) for
fibrinolytic activity [153,154]. However, other studies with
current users and 1.1 (95% CI, 0.9–1.2) for past users. The
low-dose COCs show an increase in clotting factors
calculated incidence of hypertension was only 41.5 cases per
associated with an increase in platelet activity [155]. In a
10,000 women per year. This risk decreased soon after
study by the Oral Contraceptive and Hemostasis Study
cessation of COC use, and past users had only a slightly
Group [156], the effects of COCs on 24 hemostatic variables
increased risk.
were determined in 747 healthy nonsmoking women. The
6.5. Hemostasis women were treated for six cycles with one of seven
monophasic COCs containing 20, 30 or 50 mcg EE
The coagulation and fibrinolytic systems are two separate combined with one of four different progestins. The results
but interlinked cascades of proteins that regulate the show that the clotting factor responses were influenced not
F.Z. Stanczyk et al. / Contraception 87 (2013) 706–727 717

only by the progestin component but also by the EE dose (30 combined with LNG (50–125 mcg) during a treatment
vs. 50 mcg). period of seven cycles. Changes in coagulation and
In the six-cycle study by Ågren et al. [134], which fibrinolytic parameter values from baseline to end of
compared E2 (1.5 mg)/NOMAC (2.5 mg) vs. EE (30 mcg)/ treatment were relatively small and remained within the
LNG (150 mcg), both formulations induced minimal range of normal values.
changes from baseline to the end of treatment in procoagu- Studies have shown that COCs can induce activities of the
latory factors II (prothrombin), VIIa and VIII, whereas factor coagulation system, which are attributed to the estrogenic
VIIc was essentially unchanged in the E2/NOMAC group but component of the COC [159]. Activation of the coagulation
decreased significantly by 12.7% in the EE/LNG group. system is especially evident in COC users who smoke. COCs
Results of the anticoagulatory indices showed relatively containing a high dose of EE (50 mcg) induce greater
small changes from baseline in each group (E2/NOMAC vs. changes in coagulation than those containing 30 or 35 mcg
EE/LNG), with significant differences in some but not other EE, which still induce a significant activation of coagulation
indices that included the activated partial thromboplastin parameters [160]. In contrast, COCs with 20 mcg EE appear
time-based activated protein C (APC) sensitivity ratio (3.3% to have a negligible or no effect on these parameters. In
vs. 2.0%; p=.97), antithrombin III (3.9% vs. − 3.6%, p= nonsmoking COC users, any procoagulatory effects that may
.004), protein C (− 3.1% vs. 8.2%; p=.002), and free (13.3% occur are counterbalanced by fibrinolytic effects. However,
vs. 11.9%, p=.97) and total (4.7% vs. − 3.6%, pb.001) in smokers, compensating fibrinolytic effects to offset
protein S. A substantial change from baseline was observed procoagulatory effects observed with COCs containing 30
in the endogenous thrombin potential-based APC sensitivity mcg EE are absent, thereby shifting the hemostatic profile
ratio (60% vs. 146%; p=.001). As for markers of thrombin toward a hypercoagulable state.
turnover, the percent change from baseline to end of Based on the limited data available from the short-term
treatment showed an increase in F1+2 in the EE/LNG studies that compared the effects of E2 with EE, each
group but not in the E2/NOMAC group (− 1.7% vs. 13.5%; combined with a different progestin, on hemostatic param-
p=.085), but D-dimer was essentially unchanged in both eters, it appears that the effects of the E2- and EE-based
groups. However, the D-dimer results are inconclusive formulations are similar. Long-term studies on hemostatic
because more than 50% of the values were below the parameters using different types and doses of progestins
detection limit. A notable observation in the hemostatic data combined with different doses of E2 or EE are needed.
is the large variability of virtually all the indices measured in
each of these two treatment groups. Also, although no 6.6. Bone
reference ranges were reported, it appears that most, if not
all, changes with treatment in the hemostatic parameter During puberty, endogenous estrogen helps to shape the
measures remained within the normal range with both skeleton and leads to growth of the long bones and
contraceptive formulations. epiphyseal closure [161]. The estrogens stimulate chondro-
In the same study, C-reactive protein (CRP), which is a genesis in the epiphyseal growth plate. Oral [162],
marker of cardiovascular risk, was also measured in both transdermal [163–165] and transvaginal [166] E2 increases
treatment groups. The % change from baseline was greater in bone mineral density in postmenopausal women. Although
the EE/LNG group compared to the E2/NOMAC group the mechanism of action of E2 on bone is not understood
(258% vs. 66.7%, respectively; p≤.001). However, the CRP fully, E2 may increase bone mass by decreasing the number
levels in the EE/LNG group are within the normal range of and activity of osteoclasts, thus minimizing bone resorption;
CRP values for premenopausal women [134]. it also may replace lost bone mineral [167]. ERs are found in
A study similar to the one just described compared the osteoblasts (bone forming cells) and may indirectly control
hemostatic effects of E2/NOMAC with those of EE/LNG bone resorption by osteoclasts [168].
using a lower dose of the EE/LNG formulation (20 mcg EE Since EE and, more recently, E2 are administered in
combined with 100 mcg LNG) and a shorter treatment period combination with a progestin in COCs, the specific effects of
(three consecutive cycles instead of six cycles) [157]. The these estrogens alone on bone in premenopausal women are
results show that although some of the changes from baseline not known. However, COCs do not decrease bone density
to end of treatment in the measures of the coagulation and even in adolescents. A review of recent studies of COCs
fibrinolysis parameters differed between the two groups, containing 20 versus 30 mcg EE indicated that the lower
most of the changes were relatively small, and the end of dose may not support peak bone mass acquisition in
treatment values are within the normal range. Also, no adolescents [169]. However, overall, observational evidence
significant between-group differences were found on platelet suggests that EE-based COCs have either a positive or
aggregation at the end of treatment. neutral effect on bone density and are associated with a
In another related study [158], the hemostatic effects of a reduction in most markers of bone turnover [170]. In a 2-year
multiphasic formulation consisting of E2V (1–3 mg) study of women taking either E2 (1.5 mg)/NOMAC (2.5 mg)
combined with dienogest (2–3 mg) were compared with or EE (30 mcg)/LNG (150 mcg), neither treatment
those of a triphasic formulation containing EE (30–40 mcg) negatively impacted bone mineral density [171].
718 F.Z. Stanczyk et al. / Contraception 87 (2013) 706–727

6.7. Nervous system even though high doses of LNG cause acne. Since the
androgenic properties of LNG cause a suppression of SHBG,
ERs are found in parts of the brain stem and limbic system one would expect a higher level of free testosterone with
that control mood, behavior, sex drive and autonomic function LNG-based low-dose COCs. However, free testosterone
[172]. Endogenous estrogens have been linked to several levels achieved by LNG-containing COCs are similar to
movement disorders (including chorea), but the mechanism is those obtained with the nonandrogenic COCs [179]. This
not known [172]. There is evidence linking decreasing E2 effect is due to the strong impact of LNG on the HPO axis,
levels to premenstrual migraine headaches [173]. Estrogen which causes a reduction in ovarian total testosterone
fluctuations represent trigger factors for migraine attacks; production. Consequently, free testosterone concentrations
however, the exact mechanisms and mediators underlying become very low, even though SHBG levels are increased to
these effects are largely unknown [174]. a relatively small extent. Presently, there are insufficient
Estrogens increase seizure activity, whereas progestins data on the effectiveness of the new E2-based COCs on acne
decrease this activity. COCs containing EE, for most and hirsutism.
women, do not influence neurological or psychological
function. Some women may develop mild or moderate
depression or loss of libido [175], while other studies suggest 7. Drug interactions with COCs
an improvement in mood [176].
While it is important to understand the pharmacokinetic
6.8. Liver and pharmacodynamic effects of estrogenic components, it is
essential to remember that they are often used in combination
Earlier studies with high-dose oral contraceptives had
with other drugs. Many drugs are also metabolized/
suggested that the risk of liver tumors may be increased in
detoxified by the same cytochrome P450 enzymes that act
COC users. However, a detailed review of these observations
on endogenous estrogens, and can therefore alter the
indicated that the incidence of liver disease was generally
circulating levels of these estrogens.
low, and, more importantly, studies spanning over 27 years
showed no evidence of an increased risk of serious liver
7.1. Anticonvulsants
disease in both current and former users of COCs [177].
The stimulatory effect of orally administered estrogens on Estrogens are metabolized by the hepatic cytochrome
hepatic globulins and other proteins is well recognized. P450 (CYP) system, specifically CYP3A4 [1]. Many anti-
However, most of the studies on the comparative effects of convulsants (carbamazepine, phenobarbital and phenytoin)
different estrogens have been carried out in postmenopausal can alter the activity of CYP isoenzymes in the liver.
women. Increases in SHBG, corticosteroid-binding globulin, Treatment of premenopausal epileptic women with enzyme-
thyroid-binding globulin and angiotensinogen (also a inducing antiepileptic drugs (AEDs) can result in significant
globulin) are achieved with increasing doses of EE and E2. abnormalities in reproductive hormone concentrations
However, greater increases are found with EE compared [181,182]. In contrast, the concentrations of these agents in
with E2 [178]. When the estrogen is combined with a women who received AEDs that do not alter CYP enzymes
progestin, the stimulatory effect of the estrogen on SHBG (gabapentin or lamotrigine) were no different from non-
levels may be suppressed, depending on the androgenic epileptic controls [181]. Newer AEDs, which do not induce
properties of the progestin. Variable effects of the estrogens CYPs (levetiracetam, zonisamide, gabapentin and tiagabine),
are observed in COC users on hemostatic proteins involved do not appear to alter COC pharmacokinetics or plasma
in coagulation, as discussed earlier. concentrations [183]. Contraceptives containing EE can in
addition induce the activity of uridine-diphosphate glucur-
6.9. Skin onosyltransferase and thus increase the rate of drug
glucuronidation, a mechanism proposed to be responsible
Low-dose COCs are effective in treating acne and
for the observed increase in renal excretion and reduced
hirsutism. This effect is predominantly a consequence of
therapeutic concentrations of the AED lamotrigine in women
decreased circulating levels of free testosterone owing to the
using EE-based contraceptives [184]. Progestin-only con-
pronounced increase in SHBG caused by the EE component
traceptives did not affect lamotrigine serum concentrations.
of COCs. The extent of the SHBG increase depends on the
amount of estrogen and type of progestin in the COC. 7.2. Antibiotics
Formulations containing 30 to 35 mcg EE in combination
with one of the nonandrogenic progestins such as cyproter- Antibiotics do not appear to interfere with COC
one acetate, norgestimate, desogestrel, gestodene or drospir- effectiveness [185,186]. Rifampin is an exception because
enone increase SHBG levels considerably, thereby it increases hepatic metabolism (via CYP450 enzyme) and
decreasing free testosterone levels to very low concentra- thus increases steroid metabolism resulting in decreased
tions [179,180]. COCs containing the androgenic progestin COC hormone levels. Women prescribed rifampin concom-
LNG also have a beneficial effect on acne and hirsutism, itantly with COCs have a significant risk of contraceptive
F.Z. Stanczyk et al. / Contraception 87 (2013) 706–727 719

failure [187]; therefore, women taking rifampin should use 8. Side effects of E2 and EE
other forms of contraception.
Breast tenderness, headache, fluid retention (bloating)
7.3. Smoking and drug–drug interactions and nausea are all recognized minor side effects of
(contraindications) exogenous estrogen used for contraception or menopausal
symptoms. Most documented side effects of estrogenic
Cigarette smoke causes lung cancer and is a significant therapy in young women are derived from studies of COCs.
risk factor for the development of numerous cardiovascular Early reports of EE-related side effects often occurred as a
diseases and diabetes. The estrogenic activity of orally result of the use of very high doses of EE in COCs (initially
administered hormone therapy in postmenopausal women 150 mcg daily, later reduced to 50 mcg daily) [194].
who were smokers may be impaired or even eliminated Lowering the dose of EE from 35 to 20 mcg resulted in
completely [188,189], primarily through the effects of reduced symptoms of breast tenderness, nausea and
smoking on enzymatic activity with resultant increased dizziness [5,195], but was associated with a higher incidence
metabolic clearance of the estrogens. Smoking diminishes of unscheduled bleeding [5] than COCs with a higher EE
the effects of estrogen supplements on hot flashes and dose. In a systematic review, no significant difference in
urogenital symptoms, along with their positive effects on contraceptive efficacy was observed for COCs that contained
lipid metabolism, and also reduces estrogen's ability to 20 or 35 mcg EE [5]. When women who experienced
prevent osteoporosis [189]. Women metabolize nicotine estrogen-related side effects while using COCs switched to
significantly more quickly than men [190]. Nicotine an estrogen-free progestin-only contraceptive, headache,
clearance is also higher in women taking oral contraceptives breast tenderness and nausea improved [196].
than in those who do not. Among oral contraceptive users, There were few differences in side effects in recent trials
nicotine metabolism was accelerated among those taking comparing COCs with EE to those with E2 or E2V. No
COCs and also in postmenopausal women who were using differences were noted in most side effects, including weight
only estrogen for hormone therapy. In women who used gain and headache, in a study comparing E2 (1.5 mg)/
progestin-only contraceptives, nicotine metabolism was not NOMAC (2.5 mg) with EE (30 mcg)/drospirenone (3 mg)
altered. Nicotine metabolism was similar in postmenopausal [21]. Both drugs reduced acne from baseline, although more
women who were not using estrogenic supplements and in participants in the E2/NOMAC arm than in the EE/
men [190]. These studies suggest that smoking can influence drospirenone reported acne (15% vs. 7%, respectively) at
the effect of estrogens and estrogen metabolism, and the end of the 1-year trial. Also, more patients in the E2/
estrogens also can influence the metabolism of other NOMAC arm reported an absence of withdrawal bleeding
compounds. In women who are cigarette smokers, COC ranging from 22% (cycle 4) to 31% (cycle 13) than those in
use can influence the prostacyclin/thromboxane balance the EE/drospirenone arm (3% to 6%, without any particular
negatively compared with nonsmokers [191]. trend). Most side effects reported in a seven-cycle compar-
HIV-infected patients often receive a complex therapeutic ative study of E2V/dienogest and EE/LNG were similar
regimen that includes two or more antiretroviral agents and between groups [18].
several prophylactic drugs for protection against common
opportunistic pathogens. Understanding the interactions
between antiretrovirals and oral contraceptives is important,
as women of childbearing age represent a growing 9. Adverse and serious effects associated with exogenous
proportion of people living with HIV. administration of E2 and EE: risk assessment
Interactions between antiretroviral drugs and hormonal
9.1. Cardiovascular events
contraceptives might alter the safety and effectiveness of
both the hormonal contraceptive and the antiretroviral drug 9.1.1. Stroke and myocardial infarction resulting from
[132]. When healthy volunteers received a single dose of a arterial thromboembolic events
COC containing 50 mcg EE with ritonavir, mean peak Although COCs increase the risk of ischemic stroke
plasma EE concentration decreased by 32% and mean area [197,198] and MI [197], their risk may be overestimated
under the curve decreased by 41% compared with admin- due to underlying disease such as hypertension or to
istration of the COC alone [192], suggesting an increased smoking rather than COC use alone [199,200]. Over 5
risk of oral contraceptive failure. However, EE and LNG years, arterial thromboembolism (e.g., strokes, arterial
levels were higher in HIV-positive women than in HIV- thrombosis) decreased in EE/drospirenone users but not
negative women in Malawi [193], and COCs maintained in EE/LNG users or nonusers [201]. Taken together, these
effectiveness in this small group of women treated with studies imply that there may be an increased risk of arterial
nevirapine. These results suggest that the clinical outcomes thromboembolism with EE, but it is small and may be
of COCs in women taking antiretroviral drugs could be modified by the concomitant use of some progestins. No
different depending on HIV status. The efficacy of E2-based data on the risk of arterial thromboembolism are available
COCs remains to be investigated in these women. for E2-based COCs.
720 F.Z. Stanczyk et al. / Contraception 87 (2013) 706–727

9.1.2. VTE pills and their possible implication in the incidence of breast
VTE risk from COC use is highest in women with known cancer are briefly reviewed here.
cardiovascular risk factors, including smoking, high BP, A number of early epidemiologic studies investigated the
diabetes, obesity, age, history of thrombosis or other effect(s) of COCs on breast cancer risk; some studies
coagulation abnormalities [133,202–204]. Some studies indicated no significant increase in the risk of breast cancer,
have suggested that COC formulations containing newer, while others found the opposite. In general, these findings
less androgenic progestins are associated with higher VTE were reassuring in that the increase in the risk of breast
risk than older progestins [205,206]. However, in studies that cancer was small. Essentially, all of the early studies were
corrected for various confounding factors such as weight, reanalyzed in a very large meta-analysis carried out by the
smoking status, alcohol use, age and duration of use, no Collaborative Group on Hormonal Factors in Breast Cancer.
difference in VTE risk was seen between users of COCs Data from 54 studies were reanalyzed. Over 53,000 women
containing older and newer progestin types [201,207–210]. from 26 countries with breast cancer and over 100,000
Differences between older and newer progestins in COCs women without breast cancer were assessed for the risk of
have been discounted owing to the methodological problems breast cancer and use of various formulations of COCs
with many studies [211,212]. [217,218]. In this analysis, irrespective of the dose of
The effect of progestins on VTE is controversial as of this estrogen (b 50 to N 50 mcg), the RR of breast cancer in ever
writing. As discussed earlier, E2 has less of a stimulatory users of COCs was statistically, albeit slightly, elevated (RR,
effect than EE on the hepatic system to increase the 1.07; CI, 1.03–1.10). More importantly, this small increase
production of procoagulant proteins. COCs containing E2 in the risk of breast cancer was observed in either women
or E2V have not been in widespread use for a long enough who were current COC users or those who had started use at
time to determine whether they have less of an effect on a young age (before age 20 years). Furthermore, in this group
cardiovascular events than EE-based COCs. of women, the risk of metastatic disease compared with
localized tumors was significantly reduced (RR, 0.88; CI,
0.81–0.95). These data also suggest that, after stopping the
9.2. Cancer
intake of COCs, the protective effect against metastatic
9.2.1. Breast cancer disease persists posttreatment.
The potential role of estrogens in breast cancer was first In contrast to the above study, a Canadian case–control
suggested by Lacassagne [213] when he demonstrated that study assessed the relation of use of COCs to the risk of
the administration of E1 to male mice induced mammary breast cancer in women under the age of 70 years and
cancer. This, coupled with the much earlier observation of showed a beneficial effect [219]. The women in this study
Beatson [214] that oophorectomy had a beneficial effect in had used COCs for different durations ranging from less than
two women who developed breast cancer, evoked the 1 year to more than 15 years prior to the study. The results
hypothesis/belief that estrogens (E2) play an etiological indicate that the RR estimated for 5 or more years of use
role in breast cancer in women. The observation that bilateral followed by an interval of more than 15 years was 0.5 (CI,
oophorectomy reduces the risk of breast cancer has been 0.3–0.38) or a 50% reduced risk of breast cancer. A case–
shown in epidemiologic studies [215,216]. This favorable control study of 4575 American women aged 35 to 64 years
effect of oophorectomy on breast cancer has been of with breast cancer and 4682 controls estimated the risk of
continuing interest and concern regarding the role and breast cancer among former and current users of COCs
relationship of endogenous estrogens that may affect the risk [220]. The results show no increased risk in both current
and development of breast cancer. It is well established that, users (RR, 1.0; CI, 0.8–1.3) and past users (RR, 0.9; CI, 0.8–
in the human breast, estrogens increase fat deposition, the 1.0). The RR did not appear to increase with longer duration
development of stromal tissue, the growth of an extensive of COC use or higher dose of estrogen, or in women who had
ductile system and, to a lesser extent, the development of a family history of breast cancer or in those who had initiated
lobules and alveoli. However, progesterone and prolactin use at a young age.
also are involved in the key growth and function of those Since women with a history of benign breast disease
structures. Therefore, it is highly speculative to infer that the (BBD) are thought to be at a higher risk of breast cancer
decrease in breast cancer risk following oophorectomy is [221,222], a number of studies assessed the relationship
specific to levels of estrogen only and to assume that between BBD and COC use. The results from a number of
estrogens are solely involved in the etiology of breast cancer. studies suggest a protective effect of COCs on all types of
In the past decades, the persevering concern about the effect BBD [223,224]. Thus, in a Canadian cohort study
of estrogen on breast cancer risk has led to numerous investigating the association between COC formulations
epidemiologic studies with no firm conclusions and many that were in use 15 to 20 years ago, protection against
elusive unanswered questions. It is important to note that the proliferative BBD was observed. Greater reduction was seen
early high-dose COCs had numerous side effects and have with increasing duration of COC use, with incidence rate
now been replaced by significantly lower-estrogen-dose ratios of 0.64 (CI, 0.47–0.87) [225]. Similarly, a large
formulations. These more recently formulated contraceptive Oxford Family Planning Association Study that included
F.Z. Stanczyk et al. / Contraception 87 (2013) 706–727 721

17,032 women who had used various methods of contra- lysis and in the examples mentioned above, this practice is
ception including COCs containing N 50, 50 and b 50 mcg of common, perhaps in the mistaken belief that such pseudo-
estrogen not only confirmed the previously observed precision somehow confers validity to associations that are
protective effect against BBD with the earlier formulations not, in fact, demonstrably valid.” Thus, the saga continues,
of COCs containing 50 or N 50 mcg of estrogen but also and the valid concerns in the minds of women using COCs
found the same degree of protection with newer formulations will persist.
containing b 50 mcg estrogen [226].
Although the Collaborative Group Study [217,218] 9.2.2. Endometrial and ovarian cancers
discussed above showed that recent users of COCs were EE-based COC use is associated with a 50% decrease in
associated with a small risk increase that was not altered by a the incidence of endometrial cancer risk that persists for at
family history of breast cancer, concern is still prevalent of least 15 years after stopping the COC [234,235]. Further,
an increased risk of breast cancer in COC users with BRCA1 COCs have been shown to be protective for all three major
and BRCA2 mutations. In this regard, one study based on a histologic subtypes of endometrial cancer (adenocarcinoma,
small number of cases of women with these mutations did adenoacanthoma and adenosquamous cancers) [236].
not observe a statistically significant increase in risk of breast EE-based COCs have a protective effect against ovarian
cancer with COCs [227]. In contrast, a large case–control cancer, one of the most lethal of female reproductive tract
study showed that BRCA1 mutation carriers had a small cancers. Compared to non-COC users, the risk of developing
increased risk in women who had used COCs for at least 5 epithelial ovarian cancer of all histologic subtypes is
years before the age of 30 years and in those who developed decreased by 40% in users of COCs [237]. The protective
breast cancer before the age of 40 years [228]. However, a effect increases with duration of use and continues for 20 or
multicountry population-based study of Caucasian women, more years after the treatment is stopped.
consisting of 1156 women with invasive breast cancer cases
9.2.3. Other cancers
diagnosed before the age of 40 years, concluded that users of
In addition to the protective effect of COCs on endometrial
the more recent formulations of COCs, which contained
and ovarian cancers discussed above, EE-based COCs
lower amounts of estrogen, showed no increase in the risk of
generally have a protective or neutral effect on other cancers,
early onset of breast cancer for both BRCA1 and BRCA2
with the exception of cervical cancer. Studies show that the
mutation carriers. Interestingly, they showed that carriers of
risk for dysplasia and carcinoma in situ of the uterine cervix
the BRCA1 mutation had a significantly reduced risk of
increases with the use of COCs for longer than 5 years [238].
breast cancer with COC use with an OR of 0.22 (CI, 0.10–
However, confounding factors pertaining to the risk of
0.49). The OR of breast cancer with COC use in women with
cervical cancer are difficult to control; therefore, conclusions
the BRCA2 mutation was 1.02 (CI, 0.34–3.09), whereas, in
about this risk are not definite.
noncarriers, it was 0.93 (CI, 0.69–1.24). The authors also
A recent large cohort study [239] by the RCGP found that
concluded that current COC formulations did not appear to
the risk of all cancers among ever users of COCs was lower
exacerbate the risk of ovarian cancer in carriers of these two
compared with never users (adjusted relative risk ratio, 0.85;
BRCA mutations [229].
95% CI, 0.78–0.93).
As discussed above, the implication of hormones,
particularly estrogens, in the etiology of breast cancer has 9.2.4. Mortality
been suggested for decades; however, their role is still In spite of the known risks of COCs containing EE, COCs
unclear in spite of numerous studies dealing with COCs. The overall have an excellent safety record. The RCGP found
evidence regarding breast cancer mortality in COC users that COC ever users had a significantly lower death rate from
found in two large British cohort studies shows that there is any cause (RR, 0.88; 95% CI, 0.82–0.93) compared with
no increase in breast cancer mortality [230–232]. never users [239]. Ever users also had significantly lower
Considering the fairly large body of epidemiological data rates of death from all cancers, including large bowel/rectum,
which generally indicate that the risk of breast cancer in uterine body, ovarian and all gynecological cancers,
COC users is not increased, perception to the contrary still compared with never users. Given the recent approval of
persists. This raises an important question as to whether COCs with E2, no large outcome studies have compared
nonrandomized prospective studies can ever provide a COCs with EE to those containing E2. Whether E2-based
definite answer when the magnitude of risk is low. COCs will show the same or an improved safety profile
Interestingly, Shapiro [233] in his detailed review of the remains to be established.
large meta-analysis [218] concluded the following: “Finally,
this exercise serves to demonstrate that relative risk point
estimates derived from nonexperimental studies are, by their 10. Conclusions
nature, crude and imprecise. Given the small amounts of bias
that it takes to distort such estimates, their expression to more Women have been taking COCs containing EE for more
than one decimal place is pseudoprecision. Yet, for low- than 50 years. Overall, EE-containing COCs have a very
magnitude associations such as in the collaborative reana- favorable safety profile; however, there are some health
722 F.Z. Stanczyk et al. / Contraception 87 (2013) 706–727

concerns and side effects that still need to be addressed. To followed by 0.030 mg desogestrel only for 7 days. Contraception
date, reduction in the dose of EE in currently available COCs 1996;54:333–8.
[13] Hirvonen E, Allonen H, Anttila M, et al. Oral contraceptive
has significantly improved COC safety and tolerability, but containing natural estradiol for premenopausal women. Maturitas
efforts to further reduce EE- and COC-related risks continue. 1995;21:27–32.
E2 and E2V in combination with a progestin have, in a [14] Fruzzetti F, Bitzer J. Review of clinical experience with estradiol in
relatively small number of studies, shown to provide combined oral contraceptives. Contraception 2010;81:8–5.
contraceptive efficacy that is similar to low-dose COCs [15] Mueck AO, Sitruk-Ware R. Nomegestrol acetate, a novel progestogen
for oral contraception. Steroids 2011;76:531–9.
with EE. These recent E2 formulations also appear to have [16] Mueck AO, Seeger H, Buhling KJ. Why use of dienogest for the first
acceptable cycle control and an acceptable safety profile. contraceptive pill with estradiol? Gynecol Endocrinol 2009;26:
However, long-term studies are needed to confirm these 109–13.
observations and to determine whether there are any clinical [17] Endrikat J, Parke S, Trummer D, Schmidt W, Duijkers I, Klipping C.
advantages in using E2 versus EE. Ovulation inhibition with four variations of a four-phasic estradiol
valerate/dienogest combined oral contraceptive: results of two
prospective, randomized, open-label studies. Contraception 2008;
78:218–25.
Acknowledgments
[18] Ahrendt H-J, Makalova D, Parke S, Mellinger U, Mansour D.
Bleeding pattern and cycle control with an estradiol-based oral
The authors would like to thank A. Peter Morello III, contraceptive: a seven-cycle, randomized comparative trial of
Ph.D.; Alex Loeb, Ph.D.; and Tove Anderson, Ph.D., of estradiol valerate/dienogest and ethinyl estradiol/levonorgestrel.
Evidence Scientific Solutions for professional editing Contraception 2009;80:436–44.
[19] Mansour D. Qlaira: a ‘natural’ change of direction. J Fam Plann
services which were supported by Merck Sharp & Dohme
Reprod Health Care 2009;35:139–42.
Corp., a subsidiary of Merck & Co., Inc. No funding was [20] Palacios S, Wildt L, Parke S, Machlitt A, Romer T, Bitzer J. Efficacy
provided to the authors for this review. and safety of a novel oral contraceptive based on oestradiol
(oestradiol valerate/dienogest): a Phase III trial. Eur J Obstet Gynecol
Reprod Biol 2010;149:57–62.
References [21] Mansour D, Verhoeven C, Sommer W, et al. Efficacy and tolerability
of a monophasic combined oral contraceptive containing nome-
[1] Goldzieher JW, Stanczyk FZ. Oral contraceptives and individual gestrol acetate and 17β-oestradiol in a 24/4 regimen, in comparison to
variability of circulating levels of ethinyl estradiol and progestins. an oral contraceptive containing ethinylestradiol and drospirenone in
Contraception 2008;78:4–9. a 21/7 regimen. Eur J Contracept Reprod Health Care 2011;16:
[2] Perone N. The progestins. In: Goldzieher JW, Fotherby K, editors. 430–43.
Pharmacology of the contraceptive steroids. New York: Raven Press, [22] Duijkers IJ, Klipping C, Grob P, Korver T. Effects of a monophasic
Ltd.; 1994, pp. 5–9. combined oral contraceptive containing nomegestrol acetate and
[3] Gerstman BB, Piper JM, Tomita DK, Ferguson WJ, Stadel BV, 17beta-oestradiol on ovarian function in comparison to a monophasic
Lundin FE. Oral contraceptive estrogen dose and the risk of deep combined oral contraceptive containing drospirenone and ethinyles-
venous thromboembolic disease. Am J Epidemiol 1991;133:32–7. tradiol. Eur J Contracept Reprod Health Care 2010;15:314–25.
[4] Vessey M, Mant D, Smith A, Yeates D. Oral contraceptives and [23] Chabbert-Buffet N, Chassard D, Ochsenbein E, Thomas JL, Christin-
venous thromboembolism: findings in a large prospective study. Br Maitre S. Inhibition of ovulation by NOMAC/E2, a novel monophasic
Med J (Clin Res Ed) 1986;292:526. oral contraceptive combining nomegestrol acetate and 17β-oestra-
[5] Gallo MF, Nanda K, Grimes DA, Lopez LM, Schulz KF. 20 μg diol: a double-blind, randomised, dose-finding pilot study. Eur J
versus N 20 μg estrogen combined oral contraceptives for contracep- Contracept Reprod Health Care 2011;16:76–84.
tion. Cochrane Database Syst Rev 2011:CD003989. [24] Christin-Maitre S, Serfaty D, Chabbert-Buffet N, Ochsenbein E,
[6] Astedt B, Jeppsson S, Liedholm P, Rannevik G, Svanberg L. Clinical Chassard D, Thomas JL. Comparison of a 24-day and a 21-day pill
trial of a new oral contraceptive pill containing the natural oestrogen regimen for the novel combined oral contraceptive, nomegestrol
17 beta-oestradiol. Br J Obstet Gynaecol 1979;86:732–6. acetate and 17β-estradiol (NOMAC/E2): a double-blind, randomized
[7] Serup J, Bostofte E, Larsen S, Westergaard J, Lebech PE. Natural study. Hum Reprod 2011;26:1338–47.
oestrogens for oral contraception. Lancet 1979;2:471–2. [25] Westhoff C, Kaunitz AM, Korver T, et al. Efficacy, safety, and
[8] World Health Organization Task Force on Oral Contraception. A tolerability of a monophasic oral contraceptive containing nomeges-
randomized, double-blind study of two combined oral contraceptives trol acetate and 17beta-estradiol: a randomized controlled trial. Obstet
continuing the same progestogen, but different estrogens. Contra- Gynecol 2012;119:989–99.
ception 1980;21:445–59. [26] Knobil E. The neuroendocrine control of the menstrual cycle. Recent
[9] Serup J, Bostofte E, Larsen S, Westergaard J. Effectivity and Prog Horm Res 1980;36:53–88.
acceptability of oral contraceptives containing natural and artificial [27] Gougeon A. Dynamics of follicular growth in the human: a model
estrogens in combination with a gestagen. A controlled double-blind from preliminary results. Hum Reprod 1986;1:81–7.
investigation. Acta Obstet Gynecol Scand 1981;60:203–6. [28] Gougeon A. Regulation of ovarian follicular development in
[10] Schubert W, Cullberg G. Ovulation inhibition with 17 beta-estradiol primates: facts and hypotheses. Endocr Rev 1996;17:121–55.
cyclo-octyl acetate and desogestrel. Acta Obstet Gynecol Scand 1987; [29] Schipper I, Hop WC, Fauser BC. The follicle-stimulating hormone
66:543–7. (FSH) threshold/window concept examined by different interventions
[11] Hirvonen E, Stenman UH, Malkonen M, Rasi V, Vartiainen E, with exogenous FSH during the follicular phase of the normal
Ylostalo P. New natural oestradiol/cyproterone acetate oral contra- menstrual cycle: duration, rather than magnitude, of FSH increase
ceptive for pre-menopausal women. Maturitas 1988;10:201–13. affects follicle development. J Clin Endocrinol Metab 1998;83:
[12] Csemiczky G, Dieben T, Coeling Bennink HJ, Landgren BM. The 1292–8.
pharmacodynamic effects of an oral contraceptive containing 3 mg [30] Little B, Billiar R. Progesterone production. In: Gual C, editor.
micronized 17 beta-estradiol and 0.150 mg desogestrel for 21 days, Progress in endocrinology: Excerpta Medica; 1969, pp. 871–9.
F.Z. Stanczyk et al. / Contraception 87 (2013) 706–727 723

[31] MacDonald PC, Grodin CJ, Siiteri PK. The utilization of plasma [52] Naftolin F, Ryan KJ, Petro Z. Aromatization of androstenedione by
androstenedione for estrone production in women. In: Gual C, editor. the diencephalon. J Clin Endocrinol Metab 1971;33:368–70.
Progress in endocrinology: Excerpta Medica; 1969, pp. 770–6. [53] Longcope C, Pratt JH, Schneider SH, Fineberg SE. Aromatization of
[32] Baird D, Horton R, Longcope C, Tait JF. Steroid prehormones. androgens by muscle and adipose tissue in vivo. J Clin Endocrinol
Perspect Biol Med 1968;11:384–421. Metab 1978;46:146–52.
[33] Baird DT, Guevara A. Concentration of unconjugated estrone and [54] Siiteri PK, MacDonald PC. Role of extraglandular estrogen in human
estradiol in peripheral plasma in nonpregnant women throughout the endocrinology. In: Greep RO, Astwood EB, editors. Handbook of
menstrual cycle, castrate and postmenopausal women and in men. physiology. Washington, DC: Williams and Wilkins; 1973,
J Clin Endocrinol Metab 1969;29:149–56. pp. 615–29.
[34] Carr BR, MacDonald PC, Simpson ER. The role of lipoproteins in the [55] Vandewiele RL, Macdonald PC, Gurpide E, Lieberman S. Studies on
regulation of progesterone secretion by the human corpus luteum. the secretion and interconversion of the androgens. Recent Prog
Fertil Steril 1982;38:303–11. Horm Res 1963;19:275–310.
[35] Bulun SE. The physiology and pathology of the female reproductive [56] MacDonald PC, Edman CD, Kerber IJ, Siiteri PK. Plasma precursors
axis. In: Melmed S, Polonsky KS, Larsen PR, Kronenberg HM, of estrogen. III. Conversion of plasma dehydroisoandrosterone to
editors. Williams textbook of endocrinology. Philadelphia: Elsevier; estrogen in young nonpregnant women. Gynecol Invest 1976;7:
2011, pp. 581–660. 165–75.
[36] Bhavnani BR. The saga of the ring B unsaturated equine estrogens. [57] Longcope C, Layne DS, Tait JF. Metabolic clearance rates and
Endocr Rev 1988;9:396–416. interconversions of estrone and 17beta-estradiol in normal males and
[37] Stanczyk FZ, Bretsky P. Biosynthesis, transport, and metabolism of females. J Clin Invest 1968;47:93–06.
steroid hormones. In: Henderson BE, Ponder B, Ross RK, editors. [58] Anderson DC. Sex-hormone-binding globulin. Clin Endocrinol (Oxf)
Hormones, genes, and cancer. New York: Oxford University Press, 1974;3:69–96.
Inc.; 2003, pp. 12–37. [59] Siiteri PK, Murai JT, Hammond GL, Nisker JA, Raymoure WJ, Kuhn
[38] Bhavnani BR, Tam SP, Lu X. Structure activity relationships and RW. The serum transport of steroid hormones. Recent Prog Horm Res
differential interactions and functional activity of various equine 1982;38:457–510.
estrogens mediated via estrogen receptors (ERs) ERalpha and [60] Westphal U. Steroid Binding to Human SBP. In: Westphal U, editor.
ERbeta. Endocrinology 2008;149:4857–70. Steroid-protein interactions II. Monographs on Endocrinology,
[39] Kallen CB, Billheimer JT, Summers SA, Stayrook SE, Lewis M, Vol 27. New York: Springer Verlag; 1986, pp. 250–9.
Strauss III JF. Steroidogenic acute regulatory protein (StAR) is a [61] Pardridge WM. Transport of protein-bound hormones into tissues in
sterol transfer protein. J Biol Chem 1998;273:26285–8. vivo. Endocr Rev 1981;2:103–23.
[40] Miller WL, Strauss III JF. Molecular pathology and mechanism of [62] Wu CH, Motohashi T, Abdel-Rahman HA, Flickinger GL, Mikhail
action of the steroidogenic acute regulatory protein, StAR. J Steroid G. Free and protein-bound plasma estradiol-17 beta during the
Biochem Mol Biol 1999;69:131–41. menstrual cycle. J Clin Endocrinol Metab 1976;43:436–45.
[41] Pollack SE, Furth EE, Kallen CB, et al. Localization of the [63] Pan CC, Woolever CA, Bhavnani BR. Transport of equine estrogens:
steroidogenic acute regulatory protein in human tissues. J Clin binding of conjugated and unconjugated equine estrogens with
Endocrinol Metab 1997;82:4243–51. human serum proteins. J Clin Endocrinol Metab 1985;61:499–507.
[42] Hanley NA, Ikeda Y, Luo X, Parker KL. Steroidogenic factor 1 (SF- [64] Nestler JE. Obesity, insulin, sex steroids and ovulation. Int J Obes
1) is essential for ovarian development and function. Mol Cell Relat Metab Disord 2000;24(Suppl 2):S71–3.
Endocrinol 2000;163:27–32. [65] Siiteri PK. Adipose tissue as a source of hormones. Am J Clin Nutr
[43] Thomas JL, Myers RP, Strickler RC. Human placental 3 beta- 1987;45:277–82.
hydroxy-5-ene-steroid dehydrogenase and steroid 5–4-ene-isomer- [66] Bhavnani BR. Pharmacokinetics and pharmacodynamics of conju-
ase: purification from mitochondria and kinetic profiles, biophysical gated equine estrogens: chemistry and metabolism. Proc Soc Exp Biol
characterization of the purified mitochondrial and microsomal Med 1998;217:6–16.
enzymes. J Steroid Biochem 1989;33:209–17. [67] Yue W, Santen RJ, Wang JP, et al. Genotoxic metabolites of estradiol
[44] Ryan KJ, Smith OW. Biogenesis of estrogens by the human ovary. I. in breast: potential mechanism of estradiol induced carcinogenesis.
Conversion of acetate-1-C-14 to estrone and estradiol. J Biol Chem J Steroid Biochem Mol Biol 2003;86:477–86.
1961;236:705–9. [68] Purdy RH, Goldzieher JW, LeQuesne PW, Abdel-Baky S. Active
[45] Simpson ER, Mahendroo MS, Means GD, et al. Aromatase metabolites and carcinogenesis. In: Kono S, Merriam GR, Brandon
cytochrome P450, the enzyme responsible for estrogen biosynthesis. DD, editors. Catechol estrogens. New York: Raven Press; 1983,
Endocr Rev 1994;15:342–55. pp. 123–40.
[46] Ryan KJ, Petro Z, Kaiser J. Steroid formation by isolated and [69] Li JJ, Li SA. Estrogen carcinogenesis in hamster tissues: a critical
recombined ovarian granulosa and tehcal cells. J Clin Endocrinol review. Endocr Rev 1990;11:524–31.
Metab 1968;28:355–8. [70] Zhu BT, Roy D, Liehr JG. The carcinogenic activity of ethinyl
[47] Schindler AE, Ebert A, Friedrich E. Conversion of androstenedione estrogens is determined by both their hormonal characteristics and
to estrone by human tissue. J Clin Endocrinol Metab 1972;35: their conversion to catechol metabolites. Endocrinology 1993;132:
627–30. 577–83.
[48] Nimrod A, Ryan KJ. Aromatization of androgens by human [71] Bradlow HL, Telang NT, Sepkovic DW, Osborne MP. 2-Hydro-
abdominal and breast fat tissue. J Clin Endocrinol Metab 1975;40: xyestrone: the ‘good’ estrogen. J Endocrinol 1996;150(Suppl):
367–72. S259–65.
[49] Perel E, Killinger DW. The interconversion and aromatization of [72] Cavalieri E, Chakravarti D, Guttenplan J, et al. Catechol estrogen
androgens by human adipose tissue. J Steroid Biochem 1979;10: quinones as initiators of breast and other human cancers: implications
623–7. for biomarkers of susceptibility and cancer prevention. Biochim
[50] Schweikert HU, Milewich L, Wilson JD. Aromatization of Biophys Acta 2006;1766:63–78.
androstenedione by isolated human hairs. J Clin Endocrinol Metab [73] Schneider J, Kinne D, Fracchia A, et al. Abnormal oxidative
1975;40:413–7. metabolism of estradiol in women with breast cancer. Proc Natl Acad
[51] Schweikert HU, Milewich L, Wilson JD. Aromatization of Sci USA 1982;79:3047–51.
androstenedione by cultured human fibroblasts. J Clin Endocrinol [74] Bradlow HL, Hershcopf RJ, Martucci CP, Fishman J. Estradiol 16
Metab 1976;43:785–95. alpha-hydroxylation in the mouse correlates with mammary tumor
724 F.Z. Stanczyk et al. / Contraception 87 (2013) 706–727

incidence and presence of murine mammary tumor virus: a possible [95] Fotherby K. Pharmacokinetics of ethynyloestradiol in humans.
model for the hormonal etiology of breast cancer in humans. Proc Methods Find Exp Clin Pharmacol 1982;4:133–41.
Natl Acad Sci USA 1985;82:6295–9. [96] Orme ML, Back DJ, Ball S. Interindividual variation in the
[75] Yu SC, Fishman J. Interaction of histones with estrogens. Covalent metabolism of ethynylestradiol. Pharmacol Ther 1989;43:251–60.
adduct formation with 16 alpha-hydroxyestrone. Biochemistry 1985; [97] Brody SA, Turkes A, Goldzieher JW. Pharmacokinetics of
24:8017–21. three bioequivalent norethindrone/mestranol-50 micrograms and
[76] Swaneck GE, Fishman J. Covalent binding of the endogenous three norethindrone/ethinyl estradiol-35 micrograms OC formula-
estrogen 16 alpha-hydroxyestrone to estradiol receptor in human tions: are “low-dose” pills really lower? Contraception 1989;40:
breast cancer cells: characterization and intranuclear localization. 269–84.
Proc Natl Acad Sci USA 1988;85:7831–5. [98] Kuhnz W, Pfeffer M, al-Yacoub G. Protein binding of the
[77] Kabat GC, Chang CJ, Sparano JA, et al. Urinary estrogen metabolites contraceptive steroids gestodene, 3-keto-desogestrel and ethinyles-
and breast cancer: a case–control study. Cancer Epidemiol Bio- tradiol in human serum. J Steroid Biochem 1990;35:313–8.
markers Prev 1997;6:505–9. [99] Hammerstein J, Daume E, Simon A, et al. Influence of gestodene and
[78] Muti P, Bradlow HL, Micheli A, et al. Estrogen metabolism and risk desogestrel as components of low-dose oral contraceptives on the
of breast cancer: a prospective study of the 2:16alpha-hydroxyestrone pharmacokinetics of ethinyl estradiol (EE2), on serum CBG and on
ratio in premenopausal and postmenopausal women. Epidemiology urinary cortisol and 6 beta-hydroxycortisol. Contraception 1993;47:
2000;11:635–40. 263–81.
[79] Ursin G, London S, Stanczyk FZ, et al. A pilot study of urinary [100] Kaufman JM, Thiery M, Vermeulen A. Plasma levels of ethinyles-
estrogen metabolites (16alpha-OHE1 and 2-OHE1) in postmeno- tradiol (EE) during cyclic treatment with combined oral contracep-
pausal women with and without breast cancer. Environ Health tives. Contraception 1981;24:589–602.
Perspect 1997;105(Suppl 3):601–5. [101] Guengerich FP. Metabolism of 17 alpha-ethynylestradiol in humans.
[80] Ursin G, London S, Stanczyk FZ, et al. Urinary 2-hydroxyestro- Life Sci 1990;47:1981–8.
ne/16alpha-hydroxyestrone ratio and risk of breast cancer in [102] Goldzieher JW. Pharmacology of contraceptive steroids: a brief
postmenopausal women. J Natl Cancer Inst 1999;91:1067–72. review. Am J Obstet Gynecol 1989;160:1260–4.
[81] Ursin G, London S, Yang D, et al. Urinary 2-hydroxyestrone/ [103] Speck U, Wendt H, Schulze PE, Jentsch D. Bio-availability and
16alpha-hydroxyestrone ratio and family history of breast cancer pharmacokinetics of cyproterone acetate-14C and ethinyloestradiol-
in premenopausal women. Breast Cancer Res Treat 2002;72:139–43. 3H after oral administration as a coated tablet (SH B 209 AB).
[82] Ursin G, Wilson M, Henderson BE, et al. Do urinary estrogen Contraception 1976;14:151–63.
metabolites reflect the differences in breast cancer risk between [104] Reed MJ, Fotherby K, Steele SJ. Metabolism of ethynyloestradiol in
Singapore Chinese and United States African-American and white man. J Endocrinol 1972;55:351–61.
women? Cancer Res 2001;61:3326–9. [105] Cargill DI, Steinetz BG, Gosnell E, et al. Fate of ingested radiolabeled
[83] Fishman J, Goldberg S, Rosenfeld RS, Zumoff B, Hellman L, ethynylestradiol and its 3-cyclopentyl ether in patients with bile
Gallagher TF. Intermediates in the transformation of oral estradiol. fistulas. J Clin Endocrinol Metab 1969;29:1051–61.
J Clin Endocrinol Metab 1969;29:41–6. [106] Dahlman-Wright K, Cavailles V, Fuqua SA, et al. International Union
[84] Kuhnz W, Gansau C, Mahler M. Pharmacokinetics of estradiol, free of Pharmacology. LXIV. Estrogen receptors. Pharmacol Rev 2006;
and total estrone, in young women following single intravenous and 58:773–81.
oral administration of 17 beta-estradiol. Arzneimittelforschung 1993; [107] McDonnell DP. The molecular determinants of estrogen receptor
43:966–73. pharmacology. Maturitas 2004;48(Suppl 1):S7–12.
[85] Kvorning I. Bioavailability of four oestradiol suspensions with [108] Kuiper GG, Enmark E, Pelto-Huikko M, Nilsson S, Gustafsson JA.
different particle sizes - in vivo/in vitro correlation. Drug Dev Ind Cloning of a novel receptor expressed in rat prostate and ovary. Proc
Pharm 1981;7:289–303. Natl Acad Sci USA 1996;93:5925–30.
[86] Longcope C, Gorbach S, Goldin B, Woods M, Dwyer J, Warram J. [109] Evans RM. The steroid and thyroid hormone receptor superfamily.
The metabolism of estradiol; oral compared to intravenous admin- Science 1988;240:889–95.
istration. J Steroid Biochem 1985;23:1065–70. [110] Mangelsdorf DJ, Thummel C, Beato M, et al. The nuclear receptor
[87] Adlercreutz H, Martin F. Biliary excretion and intestinal metabolism superfamily: the second decade. Cell 1995;83:835–9.
of progesterone and estrogens in man. J Steroid Biochem 1980;13: [111] Nilsson S, Gustafsson JA. Biological role of estrogen and estrogen
231–44. receptors. Crit Rev Biochem Mol Biol 2002;37:1–8.
[88] Adlercreutz H, Martin F, Jarvenpaa P, Fotsis T. Steroid absorption [112] Dickson RB, Eisenfeld AJ. 17 Alpha-ethinyl estradiol is more potent
and enterohepatic recycling. Contraception 1979;20:201–23. than estradiol in receptor interactions with isolated hepatic paren-
[89] Kuhnz W, Blode H, Zimmermann H. Pharmacokinetics of exogenous chymal cells. Endocrinology 1981;108:1511–8.
natural and synthetic estrogens and antiestrogens. In: Ottel M, [113] Wahl P, Walden C, Knopp R, et al. Effect of estrogen/progestin
Schillinger E, editors. Estrogens and antiestrogens II. Berlin: potency on lipid/lipoprotein cholesterol. N Engl J Med 1983;308:
Springer-Verlag; 1999, pp. 261–322. 862–7.
[90] Buster JE. Estrogen metabolism. In: Sciarra JJ, editor. Reproductive [114] Crook D, Godsland IF, Wynn V. Oral contraceptives and coronary
endocrinology, infertility and genetics. Philadelphia (Pa): Harper & heart disease: modulation of glucose tolerance and plasma lipid risk
Row; 1987. factors by progestins. Am J Obstet Gynecol 1988;158:1612–20.
[91] Stanczyk FZ. Estrogens used for replacement therapy in postmeno- [115] Godsland IF, Crook D, Simpson R, et al. The effects of different
pausal women. Gynecol Endocrinol 2001;15:17–25. formulations of oral contraceptive agents on lipid and carbohydrate
[92] Dusterberg B, Schmidt-Gollwitzer M, Humpel M. Pharmacokinetics metabolism. N Engl J Med 1990;323:1375–81.
and biotransformation of estradiol valerate in ovariectomized women. [116] Burkman RT, Robinson JC, Kruszon-Moran D, Kimball AW,
Horm Res 1985;21:145–54. Kwiterovich P, Burford RG. Lipid and lipoprotein changes associated
[93] Dusterberg B, Nishino Y. Pharmacokinetic and pharmacological with oral contraceptive use: a randomized clinical trial. Obstet
features of oestradiol valerate. Maturitas 1982;4:315–24. Gynecol 1988;71:33–8.
[94] Sandberg AA, Slaunwhite Jr WR. Studies on phenolic steroids in [117] Notelovitz M, Feldman EB, Gillespy M, Gudat J. Lipid and
human subjects. II. The metabolic fate and hepato-biliary-enteric lipoprotein changes in women taking low-dose, triphasic oral
circulation of C14-estrone and C14-estradiol in women. J Clin Invest contraceptives: a controlled, comparative, 12-month clinical trial.
1957;36:1266–78. Am J Obstet Gynecol 1989;160:1269–80.
F.Z. Stanczyk et al. / Contraception 87 (2013) 706–727 725

[118] Patsch W, Brown SA, Gotto Jr AM, Young RL. The effect of [138] Chasan-Taber L, Willett WC, Stampfer MJ, et al. A prospective study
triphasic oral contraceptives on plasma lipids and lipoproteins. Am J of oral contraceptives and NIDDM among U.S. women. Diabetes
Obstet Gynecol 1989;161:1396–401. Care 1997;20:330–5.
[119] Gevers Leuven JA, Dersjant-Roorda MC, Helmerhorst FM, de Boer [139] Troisi RJ, Cowie CC, Harris MI. Oral contraceptive use and glucose
R, Neymeyer-Leloux A, Havekes L. Estrogenic effect of gestodene- metabolism in a national sample of women in the united states. Am J
or desogestrel-containing oral contraceptives on lipoprotein metab- Obstet Gynecol 2000;183:389–95.
olism. Am J Obstet Gynecol 1990;163:358–62. [140] Endrikat J, Klipping C, Cronin M, et al. An open label, comparative
[120] Kloosterboer HJ, Rekers H. Effects of three combined study of the effects of a dose-reduced oral contraceptive containing 20
oral contraceptive preparations containing desogestrel plus microg ethinyl estradiol and 100 microg levonorgestrel on hemostat-
ethinyl estradiol on lipid metabolism in comparison with two ic, lipids, and carbohydrate metabolism variables. Contraception
levonorgestrel preparations. Am J Obstet Gynecol 1990;163: 2002;65:215–21.
370–3. [141] Skouby SO, Endrikat J, Düsterberg B, et al. A 1-year randomized
[121] Young RL, DelConte A. Effects of low-dose monophasic levonor- study to evaluate the effects of a dose reduction in oral contraceptives
gestrel with ethinyl estradiol preparation on serum lipid levels: a on lipids and carbohydrate metabolism: 20 microg ethinyl estradiol
twenty-four month clinical trial. Am J Obstet Gynecol 1999;181: combined with 100 microg levonorgestrel. Contraception 2005;71:
59–62. 111–7.
[122] Teichmann A. Metabolic profile of six oral contraceptives containing [142] Miller VM, Duckles SP. Vascular actions of estrogens: functional
norgestimate, gestodene, and desogestrel. Int J Fertil Menopausal implications. Pharmacol Rev 2008;60:210–41.
Stud 1995;40(Suppl 2):98–04. [143] Meyer MR, Prossnitz ER, Barton M. The G protein-coupled estrogen
[123] Crook D, Godsland I. Safety evaluation of modern oral contracep- receptor GPER/GPR30 as a regulator of cardiovascular function.
tives. Effects on lipoprotein and carbohydrate metabolism. Contra- Vascul Pharmacol 2011;55:17–25.
ception 1998;57:189–201. [144] Lieberman EH, Gerhard MD, Uehata A, et al. Estrogen improves
[124] Engel HJ, Engel E, Lichtlen PR. Coronary atherosclerosis and endothelium-dependent, flow-mediated vasodilation in postmeno-
myocardial infarction in young women–role of oral contraceptives. pausal women. Ann Intern Med 1994;121:936–41.
Eur Heart J 1983;4:1–6. [145] Sherwood A, Bower JK, McFetridge-Durdle J, Blumenthal JA,
[125] Jugdutt BI, Stevens GF, Zacks DJ, Lee SJ, Taylor RF. Myocardial Newby LK, Hinderliter AL. Age moderates the short-term effects of
infarction, oral contraception, cigarette smoking, and coronary artery transdermal 17beta-estradiol on endothelium-dependent vascular
spasm in young women. Am Heart J 1983;106:757–61. function in postmenopausal women. Arterioscler Thromb Vasc Biol
[126] Croft P, Hannaford PC. Risk factors for acute myocardial infarction in 2007;27:1782–7.
women: evidence from the Royal College of General Practitioners' [146] John S, Jacobi J, Schlaich MP, Delles C, Schmieder RE. Effects of
oral contraception study. BMJ 1989;298:165–8. oral contraceptives on vascular endothelium in premenopausal
[127] Speroff L, DeCherney A. Evaluation of a new generation of oral women. Am J Obstet Gynecol 2000;183:28–33.
contraceptives. The Advisory Board for the New Progestins. Obstet [147] Mueck AO, Seeger H, Petersen G, Schulte-Wintrop E, Wallwiener D.
Gynecol 1993;81:1034–47. Effect of two oral contraceptives with different ethinyl estradiol and
[128] Lewis MA, Heinemann LA, Spitzer WO, MacRae KD, Bruppacher levonorgestrel concentrations on the urinary excretion of biochemical
R. The use of oral contraceptives and the occurrence of acute vasoactive markers. Contraception 2001;64:357–62.
myocardial infarction in young women. Results from the Transna- [148] Kelleher C, Joyce C, Kelly G, Ferriss JB. Blood pressure alters during
tional Study on Oral Contraceptives and the Health of Young the normal menstrual cycle. Br J Obstet Gynaecol 1986;93:523–6.
Women. Contraception 1997;56:129–40. [149] Chasan-Taber L, Willett WC, Manson JE, et al. Prospective study of
[129] Rosenberg L, Kaufman DW, Helmrich SP, Miller DR, Stolley PD, oral contraceptives and hypertension among women in the United
Shapiro S. Myocardial infarction and cigarette smoking in women States. Circulation 1996;94:483–9.
younger than 50 years of age. JAMA 1985;253:2965–9. [150] Kovacs L, Bartfai G, Apro G, et al. The effect of the contraceptive pill
[130] LaCroix AZ, Lang J, Scherr P, et al. Smoking and mortality among on blood pressure: a randomized controlled trial of three progestogen-
older men and women in three communities. N Engl J Med 1991;324: oestrogen combinations in Szeged, Hungary. Contraception 1986;33:
1619–25. 69–77.
[131] Schiff I, Bell WR, Davis V, et al. Oral contraceptives and smoking, [151] Nichols M, Robinson G, Bounds W, Newman B, Guillebaud J. Effect
current considerations: recommendations of a consensus panel. Am J of four combined oral contraceptives on blood pressure in the pill-free
Obstet Gynecol 1999;180:S383–4. interval. Contraception 1993;47:367–76.
[132] Centers for Disease Control and Prevention. U S. medical eligibility [152] Meade TW. Oral contraceptives, clotting factors, and thrombosis. Am
criteria for contraceptive use. MMWR 2010;59:1–8. J Obstet Gynecol 1982;142:758–61.
[133] Pomp ER, Rosendaal FR, Doggen CJ. Smoking increases the risk of [153] Jespersen J, Petersen KR, Skouby SO. Effects of newer oral
venous thrombosis and acts synergistically with oral contraceptive contraceptives on the inhibition of coagulation and fibrinolysis in
use. Am J Hematol 2008;83:97–02. relation to dosage and type of steroid. Am J Obstet Gynecol 1990;
[134] Ågren UM, Anttila M, Mäenpää-Liukko K, et al. Effects of a 163:396–403.
monophasic combined oral contraceptive containing nomegestrol [154] Notelovitz M, Kitchens CS, Khan FY. Changes in coagulation and
acetate and 17β-oestradiol in comparison to levonorgestrel and anticoagulation in women taking low-dose triphasic oral contracep-
ethinylestradiol on haemostasis, lipids and carbohydrate metabolism. tives: a controlled comparative 12-month clinical trial. Am J Obstet
Eur J Contracept Reprod Health Care 2011;16:444–57. Gynecol 1992;167:1255–61.
[135] Waine H, Frieden EH, Caplan HI. Metabolic effects of Envoid in [155] Schlit AF, Grandjean P, Donnez J, Lavenne E. Large increase in
rheumatoid patients. Arthritis Rheum 1963;6:796–800. plasmatic 11-dehydro-TxB2 levels due to oral contraceptives.
[136] Wynn V, Adams PW, Godsland I, et al. Comparison of effects of Contraception 1995;51:53–8.
different combined oral-contraceptive formulations on carbohydrate [156] Oral Contraceptive and Hemostasis Study Group. The effects of
and lipid metabolism. Lancet 1979;1:1045–9. seven monophasic oral contraceptive regimens on hemostatic vari-
[137] Gaspard UJ, Lefebvre PJ. Clinical aspects of the relationship between ables: conclusions from a large randomized multicenter study.
oral contraceptives, abnormalities in carbohydrate metabolism, and Contraception 2003;67:173–85.
the development of cardiovascular disease. Am J Obstet Gynecol [157] Gaussem P, Alhenc-Gelas M, Thomas JL, et al. Haemostatic effects
1990;163:334–43. of a new combined oral contraceptive, nomegestrol acetate/17beta-
726 F.Z. Stanczyk et al. / Contraception 87 (2013) 706–727

estradiol, compared with those of levonorgestrel/ethinyl estradiol. [180] Thorneycroft IH, Stanczyk FZ, Bradshaw KD, Ballagh SA, Nichols
A double-blind, randomised study. Thromb Haemost 2011;105: M, Weber ME. Effect of low-dose oral contraceptives on androgenic
560–7. markers and acne. Contraception 1999;60:255–62.
[158] Junge W, Mellinger U, Parke S, Serrani M. Metabolic and [181] Morrell MJ, Flynn KL, Seale CG, et al. Reproductive dysfunction in
haemostatic effects of estradiol valerate/dienogest, a novel oral women with epilepsy: antiepileptic drug effects on sex-steroid
contraceptive: a randomized, open-label, single-centre study. Clin hormones. CNS Spectr 2001;6(771–2):783–6.
Drug Investig 2011;31:573–84. [182] Pack AM, Morrell MJ, McMahon DJ, Shane E. Normal vitamin D
[159] Fruzzetti F, Ricci C, Fioretti P. Haemostasis profile in smoking and and low free estradiol levels in women on enzyme-inducing
nonsmoking women taking low-dose oral contraceptives. Contracep- antiepileptic drugs. Epilepsy Behav 2011;21:453–8.
tion 1994;49:579–92. [183] Gaffield ME, Culwell KR, Lee CR. The use of hormonal
[160] Fruzzetti F. Hemostatic effects of smoking and oral contraceptive use. contraception among women taking anticonvulsant therapy. Contra-
Am J Obstet Gynecol 1999;180:S369–74. ception 2011;83:16–29.
[161] Nilsson O, Marino R, De Luca F, Phillip M, Baron J. Endocrine [184] Reimers A, Helde G, Brodtkorb E. Ethinyl estradiol, not pro-
regulation of the growth plate. Horm Res 2005;64:157–65. gestogens, reduces lamotrigine serum concentrations. Epilepsia 2005;
[162] Prestwood KM, Kenny AM, Kleppinger A, Kulldorff M. Ultralow- 46:1414–7.
dose micronized 17beta-estradiol and bone density and bone [185] Archer JS, Archer DF. Oral contraceptive efficacy and antibiotic
metabolism in older women: a randomized controlled trial. JAMA interaction: a myth debunked. J Am Acad Dermatol 2002;46:917–23.
2003;290:1042–8. [186] Toh S, Mitchell AA, Anderka M, de Jong-van den Berg LT,
[163] Delmas PD, Pornel B, Felsenberg D, et al. A dose-ranging trial of a Hernandez-Diaz S. Antibiotics and oral contraceptive failure — a
matrix transdermal 17beta-estradiol for the prevention of bone loss in case–crossover study. Contraception 2011;83:418–25.
early postmenopausal women. International Study Group. Bone [187] Dickinson BD, Altman RD, Nielsen NH, Sterling ML. Drug
1999;24:517–23. interactions between oral contraceptives and antibiotics. Obstet
[164] Weiss SR, Ellman H, Dolker M. A randomized controlled trial of four Gynecol 2001;98:853–60.
doses of transdermal estradiol for preventing postmenopausal bone [188] Tansavatdi K, McClain B, Herrington DM. The effects of smoking on
loss. Transdermal Estradiol Investigator Group. Obstet Gynecol estradiol metabolism. Minerva Ginecol 2004;56:105–14.
1999;94:330–6. [189] Mueck AO, Seeger H. Smoking, estradiol metabolism and hormone
[165] Ettinger B, Ensrud KE, Wallace R, et al. Effects of ultralow-dose replacement therapy. Arzneimittelforschung 2003;53:1–1.
transdermal estradiol on bone mineral density: a randomized clinical [190] Benowitz NL, Lessov-Schlaggar CN, Swan GE, Jacob 3rd P. Female
trial. Obstet Gynecol 2004;104:443–51. sex and oral contraceptive use accelerate nicotine metabolism. Clin
[166] Salminen HS, Saaf ME, Johansson SE, Ringertz H, Strender LE. The Pharmacol Ther 2006;79:480–8.
effect of transvaginal estradiol on bone in aged women: a randomised [191] Stanczyk FZ, Ploszaj S, Gentzschein E, Qian D, Mishell Jr DR. Effect
controlled trial. Maturitas 2007;57:370–81. of oral contraceptives containing 20 and 35 micrograms ethinyl
[167] Christiansen C, Lindsay R. Estrogens, bone loss and preservation. estradiol on urinary prostacyclin and thromboxane metabolite levels
Osteoporos Int 1990;1:7–3. in smokers and nonsmokers. Contraception 1999;59:17–23.
[168] Eriksen EF, Colvard DS, Berg NJ, et al. Evidence of estrogen [192] Ouellet D, Hsu A, Qian J, et al. Effect of ritonavir on the
receptors in normal human osteoblast-like cells. Science 1988;241: pharmacokinetics of ethinyl oestradiol in healthy female volunteers.
84–6. Br J Clin Pharmacol 1998;46:111–6.
[169] Agostino H, Di Meglio G. Low-dose oral contraceptives in [193] Stuart GS, Moses A, Corbett A, et al. Combined oral contraceptives
adolescents: how low can you go? J Pediatr Adolesc Gynecol and antiretroviral PK/PD in Malawian women: pharmacokinetics and
2010;23:195–201. pharmacodynamics of a combined oral contraceptive and a generic
[170] Wei S, Winzenberg T, Laslett LL, Venn A, Jones G. Oral combined formulation antiretroviral in Malawi. J Acquir Immune
contraceptive use and bone. Curr Osteoporos Rep 2011;9:6–1. Defic Syndr 2011;58:e40–3.
[171] Sordal T, Grob P, Verhoeven C. Effects on bone mineral density of a [194] Frobenius W. “The rabbits are prepared …” — The development of
monophasic combined oral contraceptive containing nomegestrol ethinylestradiol and ethinyl testosterone. J Reprod Med Endo 2011;
acetate/17beta-estradiol in comparison to levonorgestrel/ethinylestra- 8(Suppl 1):32–57.
diol. Acta Obstet Gynecol Scand 2012;91:1279–85. [195] Rosenberg MJ, Meyers A, Roy V. Efficacy, cycle control, and side
[172] Keefe D. Nervous system. In: Goldzieher JW, Fotherby K, editors. effects of low- and lower-dose oral contraceptives: a randomized trial
Pharmacology of the contraceptive steroids. New York: Raven Press, of 20 micrograms and 35 micrograms estrogen preparations.
Ltd.; 1994, pp. 283–98. Contraception 1999;60:321–9.
[173] Somerville BW. The role of progesterone in menstrual migraine. [196] Ahrendt HJ, Karckt U, Pichl T, Mueller T, Ernst U. The effects of an
Neurology 1971;21:853–9. oestrogen-free, desogestrel-containing oral contraceptive in women
[174] Tassorelli C, Greco R, Allena M, Terreno E, Nappi RE. Transdermal with cyclical symptoms: results from two studies on oestrogen-related
hormonal therapy in perimenstrual migraine: why, when and how? symptoms and dysmenorrhoea. Eur J Contracept Reprod Health Care
Curr Pain Headache Rep 2012;16:467–73. 2007;12:354–61.
[175] Leeton J. The relationship of oral contraception to depressive [197] Lidegaard O, Kreiner S. Contraceptives and cerebral thrombosis: a
symptoms. Aust N Z J Obstet Gynaecol 1973;13:115–20. five-year national case–control study. Contraception 2002;65:
[176] Toffol E, Heikinheimo O, Koponen P, Luoto R, Partonen T. 197–205.
Hormonal contraception and mental health: results of a population- [198] Baillargeon JP, McClish DK, Essah PA, Nestler JE. Association
based study. Hum Reprod 2011;26:3085–93. between the current use of low-dose oral contraceptives and
[177] Hannaford PC, Kay CR, Vessey MP, Painter R, Mant J. Combined cardiovascular arterial disease: a meta-analysis. J Clin Endocrinol
oral contraceptives and liver disease. Contraception 1997;55:145–51. Metab 2005;90:3863–70.
[178] Mashchak CA, Lobo RA, Dozono-Takano R, et al. Comparison of [199] Recent studies confirm the safety of oral contraceptives with respect
pharmacodynamic properties of various estrogen formulations. Am J to stroke. Contracept Rep 1996;7:4–9.
Obstet Gynecol 1982;144:511–8. [200] Petitti DB, Sidney S, Quesenberry CP. Oral contraceptive use and
[179] Arowojolu AO, Gallo MF, Lopez LM, Grimes DA. Combined oral myocardial infarction. Contraception 1998;57:143–55.
contraceptive pills for treatment of acne. Cochrane Database Syst Rev [201] Dinger JC, Heinemann LA, Kühl-Habich D. The safety of a
2012;7:CD004425. drospirenone-containing oral contraceptive: final results from the
F.Z. Stanczyk et al. / Contraception 87 (2013) 706–727 727

European Active Surveillance Study on oral contraceptives based on [220] Marchbanks PA, McDonald JA, Wilson HG, et al. Oral contracep-
142,475 women-years of observation. Contraception 2007;75:344–54. tives and the risk of breast cancer. N Engl J Med 2002;346:2025–32.
[202] Suissa S, Blais L, Spitzer WO, Cusson J, Lewis M, Heinemann L. [221] Ernster VL. The epidemiology of benign breast disease. Epidemiol
First-time use of newer oral contraceptives and the risk of venous Rev 1981;3:184–202.
thromboembolism. Contraception 1997;56:141–6. [222] Wang DY, Fentiman IS. Epidemiology and endocrinology of benign
[203] Pomp ER, le Cessie S, Rosendaal FR, Doggen CJ. Risk of venous breast disease. Breast Cancer Res Treat 1985;6:5–6.
thrombosis: obesity and its joint effect with oral contraceptive use and [223] Brinton LA, Vessey MP, Flavel R, Yeates D. Risk factors for benign
prothrombotic mutations. Br J Haematol 2007;139:289–96. breast disease. Am J Epidemiol 1981;113:203–14.
[204] Girolami A, Spiezia L, Girolami B, Zocca N, Luzzatto G. Effect of [224] Rohan TE, L'Abbe KA, Cook MG. Oral contraceptives and risk of
age on oral contraceptive-induced venous thrombosis. Clin Appl benign proliferative epithelial disorders of the breast. Int J Cancer
Thromb Hemost 2004;10:259–63. 1992;50:891–4.
[205] Lidegaard O, Lokkegaard E, Svendsen AL, Agger C. Hormonal [225] Rohan TE, Miller AB. A cohort study of oral contraceptive use and
contraception and risk of venous thromboembolism: national follow- risk of benign breast disease. Int J Cancer 1999;82:191–6.
up study. Br Med J 2009;339:b2890. [226] Vessey M, Yeates D. Oral contraceptives and benign breast disease:
[206] van Hylckama Vlieg A, Helmerhorst FM, Vandenbroucke JP, an update of findings in a large cohort study. Contraception 2007;76:
Doggen CJ, Rosendaal FR. The venous thrombotic risk of oral 418–24.
contraceptives, effects of oestrogen dose and progestogen type: [227] Ursin G, Henderson BE, Haile RW, et al. Does oral contraceptive
results of the MEGA case–control study. Br Med J 2009;339:b2921. use increase the risk of breast cancer in women with BRCA1/
[207] Suissa S, Spitzer WO, Rainville B, Cusson J, Lewis M, Heinemann L. BRCA2 mutations more than in other women? Cancer Res 1997;57:
Recurrent use of newer oral contraceptives and the risk of venous 3678–81.
thromboembolism. Hum Reprod 2000;15:817–21. [228] Narod SA, Dube MP, Klijn J, et al. Oral contraceptives and the risk of
[208] Farmer RD, Lawrenson RA, Thompson CR, Kennedy JG, Hambleton breast cancer in BRCA1 and BRCA2 mutation carriers. J Natl Cancer
IR. Population-based study of risk of venous thromboembolism Inst 2002;94:1773–9.
associated with various oral contraceptives. Lancet 1997;349:83–8. [229] Milne RL, Knight JA, John EM, et al. Oral contraceptive use and risk
[209] Farmer RD, Todd JC, Lewis MA, MacRae KD, Williams TJ. The risks of early-onset breast cancer in carriers and noncarriers of BRCA1 and
of venous thromboembolic disease among German women using oral BRCA2 mutations. Cancer Epidemiol Biomarkers Prev 2005;14:
contraceptives: a database study. Contraception 1998;57:67–70. 350–6.
[210] Szarewski A, Mansour D, Shulman LP. 50 years of “The Pill”: [230] Vessey MP, Villard-Mackintosh L, McPherson K, Yeates D.
celebrating a golden anniversary. J Fam Plann Reprod Health Care Mortality among oral contraceptive users: 20 year follow up of
2010;36:231–8. women in a cohort study. BMJ 1989;299:1487–91.
[211] Shapiro S, Dinger J. Risk of venous thromboembolism among users [231] Trivers KF, Gammon MD, Abrahamson PE, et al. Oral contraceptives
of oral contraceptives: a review of two recently published studies. and survival in breast cancer patients aged 20 to 54 years. Cancer
J Fam Plann Reprod Health Care 2010;36:33–8. Epidemiol Biomarkers Prev 2007;16:1822–7.
[212] Reid RL, Westhoff C, Mansour D, et al. Oral contraceptives and [232] Wingo PA, Austin H, Marchbanks PA, et al. Oral contraceptives and
venous thromboembolism consensus opinion from an international the risk of death from breast cancer. Obstet Gynecol 2007;110:
workshop held in Berlin, Germany in December 2009. J Fam Plann 793–800.
Reprod Health Care 2010;36:117–22. [233] Shapiro S. Bias in the evaluation of low-magnitude associations: an
[213] Lacassagne MA. Appearance of mammary cancers in male mice empirical perspective. Am J Epidemiol 2000;151:939–45.
subjected to folliculin injections. Comptes Rendus de l'Academie des [234] Huber JC, Bentz EK, Ott J, Tempfer CB. Non-contraceptive
Sciences 1932;195:630–2. benefits of oral contraceptives. Expert Opin Pharmacother 2008;9:
[214] Beatson GT. On the treatment of inoperable cases of carcinoma of the 2317–25.
mamma: suggestions for a new method of treatment with illustrative [235] Mueck AO, Seeger H, Rabe T. Hormonal contraception and risk of
cases. Lancet 1896;2:104–7. endometrial cancer: a systematic review. Endocr Relat Cancer 2010;
[215] Trichopoulos D, MacMahon B, Cole P. Menopause and breast cancer 17:R263–71.
risk. J Natl Cancer Inst 1972;48:605–13. [236] The reduction in risk of ovarian cancer associated with oral-
[216] Colditz GA, Rosner B. Cumulative risk of breast cancer to age 70 contraceptive use. The Cancer and Steroid Hormone Study of the
years according to risk factor status: data from the Nurses' Health Centers for Disease Control and the National Institute of Child Health
Study. Am J Epidemiol 2000;152:950–64. and Human Development. N Engl J Med 1987;316:650–5.
[217] Collaborative Group on Hormonal Factors in Breast Cancer. Breast [237] Speroff L, Fritz MA, editors. Clinical gynecologic endocrinology and
cancer and hormonal contraceptives: further results. Collaborative infertility. Philadelphia, PA: Lippincott Williams & Wilkins; 2005,
Group on Hormonal Factors in Breast Cancer. Contraception 1996; pp. 691.
54:1S–106S. [238] Appleby P, Beral V, Berrington de Gonzalez A, et al. Cervical cancer
[218] Collaborative Group on Hormonal Factors in Breast Cancer. Breast and hormonal contraceptives: collaborative reanalysis of individual
cancer and hormonal contraceptives: collaborative reanalysis of data for 16,573 women with cervical cancer and 35,509 women
individual data on 53 297 women with breast cancer and 100 239 without cervical cancer from 24 epidemiological studies. Lancet
women without breast cancer from 54 epidemiological studies. 2007;370:1609–21.
Lancet 1996;347:1713–27. [239] Hannaford PC, Iversen L, Macfarlane TV, Elliott AM, Angus V, Lee
[219] Rosenberg L, Palmer JR, Clarke EA, Shapiro S. A case–control study AJ. Mortality among contraceptive pill users: cohort evidence from
of the risk of breast cancer in relation to oral contraceptive use. Am J Royal College of General Practitioners' Oral Contraception Study.
Epidemiol 1992;136:1437–44. BMJ 2010;340:c927.

You might also like