You are on page 1of 10

Contraception 82 (2010) 314 – 323

Review article

Metabolism and pharmacokinetics of contraceptive steroids in obese


women: a review
Alison B. Edelmana,⁎, Ganesh Cheralab,c , Frank Z. Stanczykd,e
a
Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, OR 97239, USA
b
College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
c
School of Pharmacy, Oregon Heath and Science University, Portland, OR 97239, USA
d
Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
e
Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
Received 22 February 2010; revised 6 April 2010; accepted 14 April 2010

Abstract

The effect of obesity on drug metabolism and pharmacokinetics is poorly understood, and this is particularly true in regard to
contraceptive steroids. This article will review the known and theoretical physiologic and pharmacologic interactions between obesity and
contraceptive steroids.
© 2010 Elsevier Inc. All rights reserved.

Keywords: Obesity; Contraceptive steroids; Pharmacokinetics; Birth control

1. Introduction itary and ovaries. The pulsatile release of gonadotropin-


releasing hormone (GnRH) from the hypothalamus signals
The exponential growth of obesity since the 1990s has raised the anterior pituitary to release follicle-stimulating hormone
concern regarding the efficacy of medications whose dose is not (FSH) and luteinizing hormone (LH), also in a pulsatile
based on weight or body mass index (BMI) [1–3]. This is fashion [7–11]. These in turn trigger the ovary to produce
particularly true of hormonal contraception where the “one dose estrogen and progesterone. These ovarian hormones also
fits all” approach is standard practice. Most drugs, including play a key role in regulating gonadotropin secretion through
contraceptive steroids, have not been extensively studied in an both negative and positive feedback loops depending on the
obese population [4]. As hormonal contraception is one of the time in the cycle [7–10,12].
most common prescription medications used by reproductive- The main effect of contraceptive steroids is via negative
age women [5] and the consequences of impaired efficacy is feedback inhibition of the hypothalamic–pituitary–ovarian
high (unplanned pregnancy) [6], understanding the interaction (HPO) axis, with supplementary effects on cervical mucus
between obesity and contraceptive steroids is critical. and the endometrium. Pulsatile secretion of FSH and LH is
The present article reviews the mechanism of action, profoundly suppressed by exogenous estrogen [ethinyl
metabolism and pharmacokinetics of contraceptive steroids estradiol (EE)] and progestin, respectively [8,13–15].
and how obesity is known or thought to affect these processes. However, ovulation suppression, thinning of the endometri-
um and cervical mucus thickening is due mostly to progestin
and is dose-dependent [8,11]. It is unclear whether ovulation
2. Mechanism of action of contraceptive steroids suppression occurs at the level of the hypothalamus, the
pituitary or both [7,13]. Pituitary sensitivity to FSH has been
Normal reproductive function is dependent on the found to be reduced by combined hormonal contraceptives
complex interrelationship between the hypothalamus, pitu- as tested by a GnRH challenge test [13] but rapidly returns
with discontinuation [16]. This is also evident with cyclically
⁎ Corresponding author. Tel.: +1 503 494 5949; fax: +1 503 494 3111. dosed hormonal contraceptives [i.e., combined oral contra-
E-mail address: edelmana@ohsu.edu (A.B. Edelman). ceptives (OCs)] when gonadotropins and ovarian hormones
0010-7824/$ – see front matter © 2010 Elsevier Inc. All rights reserved.
doi:10.1016/j.contraception.2010.04.016
A.B. Edelman et al. / Contraception 82 (2010) 314–323 315

are suppressed during active hormone use but reactivate ferases and glucuronyl transferases), which extensively
during the hormone-free interval (placebo week) [13,16]. transform both the unmetabolized and metabolized steroids
These previous studies only focused on the influence of entering the organ. Some of the contraceptive steroid that
contraceptive steroid hormones on the HPO axis in normal was originally ingested remains unmetabolized. The extent
weight and BMI subjects (animals and women) but not in to which a steroid reaches the systemic circulation following
obese subjects. We recently investigated whether increased hepatic first-pass metabolism is referred to as its bioavail-
BMI affects OC suppression of the HPO axis [17]. Ten ability. It is important to realize that the concentration of
ovulatory women of normal BMI (b25 kg/m2) and 10 obese steroid in the portal vein blood is very high and has a
women (BMI N30 kg/m2) received 100 mcg levonorgestrel profound effect on hepatic proteins, including sex hormone-
combined with 20 mcg EE for two cycles. The subjects were binding globulin (SHBG), corticosteroid-binding globulin,
closely studied at the beginning and end of their hormone- thyroid-binding globulin, angiotensinogen, as well as
free interval. Both BMI groups demonstrated hypothalamic– coagulation and fibrinolytic factors.
pituitary activation at the end of the hormone-free interval,
but more obese women also demonstrated ovarian hormonal
changes associated with the recruitment and maturation of a 4. Metabolism and pharmacokinetics of EE in
dominant follicle. nonobese women

The estrogenic component of practically all currently


3. Review of hepatic first-pass metabolism marketed combined oral contraceptives (COCs) consists of
EE. While around 90% of oral EE is absorbed from the
Hepatic first-pass of a contraceptive steroid has a stomach and upper intestine during the first hour after ingestion
profound effect on the metabolism and pharmacokinetics in most women, it can take as long as 2 h in some individuals to
of the steroid (Fig. 1). After its ingestion, the steroid enters fully absorb the compound [18]. Wide variability in the peak
the stomach where it undergoes dissolution. The dissolved blood EE level and the time taken to reach it has been observed
drug then enters the intestine where it undergoes some between individuals, the latter varying from 1 to 2 h in most
transformation by bacterial enzymes and by enzymes in the women and up to as long as 6 h in others [19,20].
intestinal mucosa. The mixture of metabolized and unme- Ethinyl estradiol metabolism follows similar pathways to
tabolized steroids is then absorbed through the intestinal that of the body's endogenous estradiol, namely, oxidation
mucosa and enters the portal vein blood, which perfuses the reactions at various carbons of the steroid nucleus (Fig. 2)
liver. The liver has many steroid metabolizing enzymes [21,22]. The most important of these for estradiol involves 2-
(reductases, dehydrogenases, hydroxylases, sulfuryl trans- hydroxylation, 16α-hydroxylation and conjugation in the
liver. For EE, 2-hydroxylation is quantitatively the most
important metabolic pathway, while 16α-hydroxylation
appears to be compromised by the presence of the ethinyl
group at carbon 17. Additional hydroxylations at the 4, 6 and
16β positions of EE have been reported, but to a much
smaller extent than at the 2 position. Because the 2-
hydroxylation of EE is catalyzed principally by the hepatic
cytochrome P450 (CYP) pathway, specifically CYP3A4 and
CYP2C9, wide variation in levels of these enzymes between
individuals [21] contributes significantly to the intersubject
variability in EE pharmacokinetics.
After absorption, EE is rapidly conjugated in part to a
glucuronide, which is inactive and undergoes renal excre-
tion, and to the 3- and 17-sulfates, which are present in the
circulation at levels about 10 times those of EE itself [23].
The sulfates are partially deconjugated during enterohepatic
recirculation to EE (the 17-sulfate to about 11% and the 3-
sulfate to about 21%), but this adds little to the circulating
unconjugated EE.
Both bioavailability and elimination half-life, which
reflects exposure of a drug to tissues, are important to the
clinician. For both parameters, EE shows wide intersubject
variability, as well as day-to-day variability within the same
individual. The bioavailability of EE, i.e., the amount
Fig. 1. Hepatic first-pass effect of a steroidal drug (oral administration). reaching the systemic circulation following hepatic first-
316 A.B. Edelman et al. / Contraception 82 (2010) 314–323

Fig. 2. Major routes of EE metabolism.

pass metabolism, ranges from about 25% to 65% of the marketed in the near future. Estradiol differs in chemical
amount ingested. The EE elimination half-life, i.e., the time structure from EE only in that it lacks the ethinyl group.
required for its blood level to fall to 50% of the maximal Nevertheless, this difference results in extensive metabolism
value, ranges from 6 to 27 h [18]. of estradiol during its hepatic first-pass, and consequently a
Studies in different ethnic populations have shown that very low bioavailability (b10%). However, this disadvantage
plasma EE levels differ substantially between ethnic groups. of estradiol may be negated by its pharmacodynamic profile
In a study of EE metabolism in women from Nigeria, Sri and relatively low potency.
Lanka, Singapore, Thailand and the United States, Gold-
zieher et al. [24] found the lowest plasma levels of EE in
Nigerian women and the highest in women from Thailand, 5. Metabolism and pharmacokinetics of progestins in
even when corrected for body surface area. Another study nonobese women
involving 83 women in 14 geographically diverse centers
[25] found that the intercenter EE differences were of the Some 24 different synthetic progestogens (progestins) are
same order as the intracenter variability. When the urinary used therapeutically, predominantly for contraception and/or
metabolites of EE in women from these locales were hormone therapy in postmenopausal women to prevent
analyzed [26], remarkable differences were found: Nigerian
women excreted primarily EE conjugates, with little
evidence of oxidative or other metabolism, while in contrast,
diverse oxidative metabolism at the 2, 4, 6 and 16 positions
of the EE molecule, plus other unidentified metabolism, was
noted in women from the United States; metabolism in Sri
Lankan women occurred to an intermediate extent, between
those extremes.
Interestingly, no studies exist regarding the EE metabo-
lism of nonoral routes, but there is one study that compares
the pharmacokinetics of EE administered via transdermal,
oral and vaginal routes [27]. As expected, with oral
administration, there is a peak-trough pattern in serum EE
levels, whereas with transdermal and vaginal dosing, a more
constant level is observed. The area under the concentration
time (AUC) curve of EE is greatest for transdermal
administration, but it is difficult to compare the pharmaco-
kinetics of EE among these three dosing routes since the dose
of EE for each route was not equivalent (Fig. 3).
Although EE has been the principal estrogenic compo-
nent of COCs over the past 50 years, COCs using estradiol Fig. 3. Comparison of EE pharmacokinetics dosed via three different routes
instead of EE have been developed recently and should be (oral, transdermal and vaginal) [27].
A.B. Edelman et al. / Contraception 82 (2010) 314–323 317

endometrial hyperplasia. Progestins can be classified into two etonogestrel, and norgestimate is transformed to levonorges-
groups on the basis of their chemical structure: (a) those trel and norelgestromin.
related to progesterone and (b) those related to testosterone The wide intersubject differences in progestin metabolism
(Fig. 4). Interestingly, of the 12 progestins in the progesterone mean that, following administration, each woman will have a
group, only two are used for contraception, namely, unique steroidal milieu consisting of the individual's
medroxyprogesterone acetate (MPA) and cyproterone ace- endogenous steroid hormones and their metabolites mixed
tate, while all 12 progestins in the testosterone group are used homogeneously with the progestin and its metabolites. The
for contraception. biologic significance of the steroidal milieu is not yet
The 12 progestins structurally related to (but not derived understood but may have some relationship to contraceptive
from) testosterone can be divided into ethinylated (containing effectiveness and side effects. Major metabolic transforma-
an ethinyl group at carbon 17) and nonethinylated progestins. tions include reduction (addition of two hydrogen) by
The latter group is composed of drospirenone and dienogest, reductases at the double-bond and by hydroxysteroid
whereas the ethinylated group is divided into estranes and 13- dehydrogenases at the ketone group in the A ring of the
ethylgonanes (containing an ethyl group at carbon 13). The progestins (Fig. 5). The unreduced and reduced progestins
estrane group consists of norethindrone and its derivatives can also undergo hydroxylation in which a hydroxyl group is
(norethindrone acetate, ethynodiol diacetate norethynodrel, added to carbon 2 or carbon 16 of the molecule, as well as
lynestrenol and norethindrone enanthate), whereas the 13- conjugation to form sulfates or glucuronides at one or more
ethylgonane group contains levonorgestrel and its deriva- hydroxyl groups of the molecules.
tives, including desogestrel, norgestimate and gestodene. Progestins in OCs undergo hepatic first-pass metabolism
Progestins vary widely not only in their chemical described earlier under Section 2 (Fig. 1). They are all well
structures but also in their metabolism and pharmacokinetics. absorbed. However, as stated earlier, there is wide intersubject
In addition, large intersubject variability, and often even variability in their blood levels and pharmacokinetic para-
intrasubject variability, is observed in their metabolism, meters [28]. In general, they reach a maximum concentration
blood levels and pharmacokinetic parameters. Many of (Cmax) within 1 to 3 h [time to maximum concentration
the progestins are prodrugs, which mean they must (Tmax)], and the Cmax and AUC is dose-dependent. Levo-
be metabolized to become active (Table 1). All five norgestrel, gestodene and dienogest have the highest
norethindrone derivatives described earlier are prodrugs, bioavailabilities, generally N90%, whereas the bioavailabil-
and they are readily converted into parent compounds. In ities for other progestins that are structurally related to
addition, desogestrel is converted to the active product, testosterone are about 20% lower [28]. Norethindrone and

Fig. 4. Classification of synthetic progestins.


318 A.B. Edelman et al. / Contraception 82 (2010) 314–323

Table 1 ization has been used for a long time, this approach is purely
Progestin prodrugs and their active components empirical and based on none or very scanty scientific data.
Prodrug Active progestin With the recent alarming rates of increase in obesity
Norethindrone acetate Norethindrone worldwide, there is a greater need for understanding drug
Ethynodiol diacetate dosing in patients of varying body weights and sizes.
Norethynodrel Attempts have been made over the last decade to understand
Lynestrenol
the physiologic changes accompanying obesity that are
Norethindrone enanthate
Desogestrel Etonogestrel (3-ketodesogestrel) relevant for drug pharmacokinetics, but much is still
Norgestimate Norelgestromin (levonorgestrel-3-oxime) and unknown (Table 2).
levonorgestrel

6.1. Absorption
dienogest have relatively low half-lives, generally in the range Many factors that affect oral drug absorption are reported
of 8 to 12 h [28]. Cyproterone acetate appears to have the to be altered in the obese population [29]. In general, cardiac
highest half-life (50–80 h), followed by drospirenone output is higher, leading to increased gut perfusion, which
(approximately 30 h) and the other progestins (12–24 h) [28]. may enhance the rate and amount of drug/nutrient absorbed
Finally, very little is known regarding the metabolism [30,31]. Accelerated gastric emptying has also been
and pharmacokinetics of contraceptive progestins used observed in obese subjects that could lead to faster
parenterally. Also, there is a paucity of data on how absorption and thereby a decrease in Tmax [32–35]. In regard
estrogen and progestin given in combination affect each to intestinal transit time, which is directly related to the total
other's metabolism. amount of drug/nutrient absorbed, no conclusive data are
available in obese patients [36,37].
Enterohepatic recirculation is an important phenomenon
6. General knowledge about the effect of obesity on the of drug absorption for some drugs like COCs. Thus, any
metabolism and pharmacokinetics of drugs alterations in the recirculation could affect the clinical
outcome [38,39]. While human data are absent, rodent
Pharmacokinetics is the study of the passage of a drug models of obesity suggest a significant decrease in bile salt
through the body. It encompasses four physiologic process- secretion and expression of canalicular transporters such as
es, namely, absorption, distribution, metabolism and excre- the bile salt export pump and organic anion transporter 1
tion. Many factors alter one or more of these four processes, [40]. These changes could potentially decrease the secretion
leading to altered drug pharmacokinetics and thereby altered of some drugs, including COCs, into bile duct, which in
drug therapeutics. Some of those factors include sex, age, turn could lead to a decrease in the amount that re-enters
dietary habits, alcohol intake, starvation, coadministered systemic circulation.
drugs, altitude, infection, disease, seasonal and circadian Blood flow to skin, muscle and subcutaneous fat is an
rhythms, pregnancy and body weight. important determinant of drug absorption when administered
Drug dosing based on body weight is a widely used parenterally, subcutaneously or transdermally. While cardiac
method to individualize dose. While such dose individual- output is increased in obese subjects, the blood flow per

Fig. 5. Major routes of metabolism of different active progestins (norethindrone, levonorgestrel, norelgestromin, etonogestrel, gestodene, drospirenone and
dienogest).
A.B. Edelman et al. / Contraception 82 (2010) 314–323 319

Table 2
Physiologic alterations due to obesity and their pharmacokinetic consequences
Physiologic alterations Pharmacokinetic consequences
Absorption ↑ Cardiac output to gut ↑ Rate and amount of drug absorption; ­ Cmax, ↓ in Tmax
Accelerated gastric emptying
Distribution Altered body composition ↑ Volume of distribution
• ↑ Lean body mass • Moderate ­ for hydrophilic drugs
• ↑ Adipose tissue • Relatively greater ­ for lipophilic drugs
• ↑ Organ sizes Alterations in free fraction of drug
• ↑ Cardiac output • ↓ In free fraction of basic drugs
• ↑ In free fraction of highly bound acidic drugs due to
potential displacement
Alterations in protein binding
• ↑ AAG
• ↑ Plasma lipids
• ↑ Free fatty acids
• ↓ SHBG
Metabolism ↑ Splanchnic flow ↓ In specific cytochrome P450 enzyme activities
↑ Number of hepatocytes ↑ In glucuronidation and sulfation
↑ Fatty infiltration Altered biliary metabolism
↑ Cholestasis Altered enterohepatic recirculation
↑ Periportal fibrosis and infiltration
↓ Biliary secretion and transporters
Excretion ↑ Kidney size ↑ In renal clearance in general
↑ Glomerular surface area ↑ Glomerular filtration rate
↑ Renal plasma flow ↑ Filtration and tubular secretion
↓ Urine pH Altered reabsorption (↑ or ↓ depends on drug pKa)
↓ Biliary secretion and transporters Altered biliary excretion
Adapted from Blouin and Warren [4].

gram of fat is significantly less in morbidly obese compared linearly correlates with absolute body weight. On the other
to lean subjects, which may affect the rate and/or extent drug hand, highly hydrophilic drugs tend to prefer lean mass, and
absorption [41]. However, very limited information is accordingly, their Vd correlate to a greater degree with lean
available regarding the effects of obesity on drug absorption body weight. However, for drugs with intermediate
in humans. Propranolol is a lipophilic drug, similar to lipophilicity and hydrophilicity, Vd is a function of both
contraceptive steroids. The systemic bioavailability of ideal and absolute body weights. In addition to ideal and
propranolol is slightly higher, though statistically not absolute body weights, other body size biomarkers such as
significant, in obese subjects [42]. Data from the limited lean body weight, fat free mass and adjusted body weight
number of studies are inconclusive, and therefore, more were also shown to exhibit a correlation to Vd (and other
focused studies are needed before a conclusion can be made pharmacokinetic parameters) [46].
on the general effect of obesity on drug absorption. Drugs administered parenterally (transdermally, subcuta-
neously, etc.) first partition into fat mass at the site of
6.2. Distribution application/administration, and then they are released over
time into the systemic circulation. The site of drug
Distribution of a drug in the body is governed by the administration may be altered due to thickness of subcuta-
amount of fat and lean mass, blood flow to tissues and neous tissue (i.e., drug is deposited in subcutaneous fat
plasma and tissue binding. In obese subjects, absolute fat instead of intramuscularly), which may affect the amounts
and lean mass is increased [43,44]. However, as a and rate of drug release [47]. This larger amount of
percentage of total body weight, lean mass decreases subcutaneous fat also has a detrimental effect on the
while fat mass increases twofold [45]. As mentioned earlier, physiologic status of a subject [48]. Depending on the
cardiac output increases in obese subjects; this in turn lipophilic and hydrophilic properties of a drug, its retention
increases blood flow to various organs, including fat. in subcutaneous fat depots could be altered. A study by
However, the blood flow per amount of fat mass is Wortsman et al. [49] concluded that the lower bioavailability
decreased [41]. The effect of the above alterations in fat and of vitamin D in obese subjects is due to its slower release
lean mass is mainly dependent on the oil/water partition from subcutaneous fat. Furthermore, the decrease in fat
coefficient (or degree of lipophilicity) of a given drug. In blood flow could negatively affect the parenteral absorption
general, highly lipophilic drugs partition into the greater fat of a drug into the systemic circulation.
mass available in obese subjects, and correspondingly, the Another important determinant of drug distribution is
measure of drug distribution (volume of distribution, Vd) plasma protein binding. While plasma levels of the major
320 A.B. Edelman et al. / Contraception 82 (2010) 314–323

drug binding protein, albumin, are unaltered in obese general, expression of hepatic uptake transporters such as
subjects, the lipoproteins that bind to albumin are greatly organic anion transporting polypeptides and sodium/taur-
increased. It is unclear if increased levels of lipoprotein ocholate cotransporting polypeptide is decreased, and on the
would compete for more protein binding sites and thus could other hand, expression of efflux transporters (multidrug
cause drug displacement from albumin binding sites. Plasma resistance-associated proteins) is increased [66]. Similar to
levels of α1-acid glycoprotein (AAG), which primarily binds the liver, expression of the renal transporters is also altered in
basic drugs, are increased in obese subjects. A few studies obese mice. Enhanced expression of the monocarboxylate
also suggest alterations in the binding affinity of AAG transporter (MCT) is found in the brain of obese mice [67].
[50,51]. Sex hormone-binding globulin, though not found in Various drugs containing monocarboxylic acid moieties,
concentrations as high as albumin and AAG, is a plasma such as valproic acid, pravastatin, simvastatin, lovastatin,
protein that binds specific endogenous androgens and etc., are substrates of MCT [68,69]. Some of the undesirable
estrogens, as well as progestins such as levonorgestrel and central nervous system (CNS) side effects observed with
norethindrone. Decreased plasma levels of SHBG were these drugs are attributed to the transport of parent drug and/
reported in obese subjects [52]. One of our recent studies, or its metabolites across the blood–brain barrier by MCTs,
however, failed to show a correlation between serum levels and therefore, it could potentially place obese subjects at
of SHBG and BMI [17]. greater risk of CNS side effects. MCTs are also expressed in
the intestine and thus could affect drug absorption.
6.3. Metabolism Expression of canalicular transporters such as bile salt
export pump and organic anion transporter 1 was also altered
The liver is the major organ of metabolism. Drug [40], with an implication for clearance of drugs that are
metabolism occurs in two phases: Phase 1 (oxidation, predominantly conjugated and excreted via bile.
reduction, hydrolysis, etc.) and Phase 2 (conjugation
reactions such as glucuronidation, sulfation, etc.). The 6.4. Elimination
expression of Phase 1 and 2 enzymes is regulated by a The kidney is the major organ of elimination. Three
host of factors of which cytokines play a significant role. processes, namely, glomerular filtration, tubular secretion
Obesity is considered as a chronic, low-grade inflammatory and reabsorption, are involved in the renal elimination of
state [53,54] with elevated levels of interleukin 6, tumor drugs either as an unchanged moiety or as a metabolite.
necrosis factor α and other proinflammatory cytokines Many morphological and physiologic changes in the kidney
[55,56]. Cytokines are known transcriptional regulators of accompany obesity [70]. Increases in kidney size [71],
expression of the CYP super family of enzymes and other glomerular Bowman's space area and filtration rate [72],
drug-metabolizing enzymes [57–60]. In addition to cyto- tubular secretion [73] and renin plasma flow [74] were
kine-mediated alterations in the expression of enzymes, fatty reported. Body mass index is inversely related to urine pH
infiltration of the liver might affect the function of the drug- [75]. The reabsorption of drugs is mostly a passive process
metabolizing enzymes [61]. and is dependent on unionized form of a drug. The amount of
Studies in obese human subjects, using CYP-specific drug unionized is determined by the pH of urine; thus,
substrates, show a decrease in CYP3A and CYP2E1 altered urinary pH in obesity could impact the reabsorption
activities. No conclusive data are available for other CYP process. Human studies have conclusively demonstrated that
isozymes in obese humans. However, in genetically obese renal clearance of predominantly renal excreted drugs is
rats, elevated levels of CYP2B, CYP2E, CYP3A and increased with BMI [76–79].
CYP4A were reported [62]. In nutritionally induced obese Biliary excretion is another route of drug elimination and
rats, only CYP2E1 activity and expression was shown to be is a significant pathway for many drugs. In obesity, bile
increased [63]. In addition to Phase 1 enzymes, some of the secretion is decreased and, in addition, expression of
Phase 2 conjugation enzymes were also altered in the obese canalicular bile salt export pump and organic anion
rats. In general, glucuronidation is the most significantly transporter 1 are decreased. A net result of these changes
enhanced, while sulfation is moderately enhanced. Drug could be a decrease in the amount and/or rate of biliary
metabolism also occurs in extrahepatic tissues, and a recent excretion of drugs [40].
rodent study suggests that white adipose tissue is one such
tissue [64]. With increasing BMI, it is possible that the
contribution of adipose tissue to drug metabolism increases, 7. Effects of obesity on the metabolism and
mainly for lipophilic drugs. pharmacokinetics of contraceptive steroids
Drug transporters play a vital role in metabolism and
elimination. The interplay between drug-metabolizing In general, the effect of obesity on drug metabolism and
enzymes and transporters determine the overall clearance pharmacokinetics is poorly understood. This is particularly
of the drug [65]. While studies in obese human subjects are true of steroidal contraceptives administered orally, trans-
lacking, limited data from obese animal studies suggest dermally or parenterally, as most studies have excluded
alterations in the expression of various drug transporters. In women over 130% of ideal body weight. Depending on the
A.B. Edelman et al. / Contraception 82 (2010) 314–323 321

drug studied, BMI can affect pharmacokinetics in several a drug [82,83]. In general, we expect a dependable response
different ways, including bioavailability and hepatic clear- to a particular drug. This is certainly true with regard to
ance, renal clearance and volume of distribution. hormonal contraception where a “one dose fits all” approach
To date, there have only been three studies investigating is pervasive. However, genetic differences may cause
the pharmacokinetics of contraceptive steroids in obese clinically significant variations. No studies to date have
women. In our recent study, mentioned earlier, we also been done to determine if a genetic variation in the response
measured serum levels of levonorgestrel and EE following to contraceptive steroids exists. Warfarin, a drug with similar
their oral administration [17]. The results revealed signifi- metabolic pathways to contraceptive hormones through the
cant differences in levonorgestrel half-life, clearance and CYP enzymes, has been found to have genetic variations in
time to reach steady-state between obese and normal BMI response. If pharmacogenetics does play a role in the
women. The levonorgestrel half-life in the obese subjects response to hormonal contraceptives, it will be interesting to
was twice that of the normal BMI subjects, and therefore, the see if this counteracts or compounds the effects that obesity
time to reach steady-state was doubled (10 days in the obese may have.
subjects versus 5 days in the normal BMI subjects).
Consistent with these changes, more obese women demon-
strated hormonal changes associated with recruitment and 9. Summary
maturation of a dominant follicle. This study did not monitor
ovarian activity directly with ultrasound. The obesity epidemic has created new concerns and
In the second study, Westhoff et al. [80] showed that challenges in health care. Little is known about the effects of
following oral administration of 150 mcg levonorgestrel in obesity on drug pharmacokinetics and how this in turn
combination with 30 mcg EE, obese women had a influences drug therapeutics; this is especially true for
significantly lower AUC and maximum plasma concentra- hormonal contraceptives. A greater understanding of hor-
tion for EE than normal BMI women. Area under the monal contraceptive pharmacokinetics and the potential
concentration time and maximum plasma concentration influence that obesity may have is warranted prior to
differences for levonorgestrel between the two groups were translating this information to the bedside.
smaller and not significant. Trough levels for both
levonorgestrel and EE were similar between the groups.
References
Follicular diameters tended to be larger among the obese
women, but the differences were not significant. The authors [1] World Health Report: overweight, obesity, and high body mass. In:
concluded that these pharmacokinetic differences should not http://www.who.int/whr/2002/en/whr02_en.pdf; 2002:60.
affect contraceptive efficacy in obese COC users. A direct [2] Grimes DA, Shields WC. Family planning for obese women:
comparison of these two pharmacokinetics studies cannot be challenges and opportunities. Contraception 2005;72:1–4.
[3] Trussell J, Schwarz EB, Guthrie K. Obesity and oral contraceptive pill
made as this study did not report time to reach steady state.
failure. Contraception 2009;79:334–8.
Additionally, the AUC calculations were based on one half- [4] Blouin RA, Warren GW. Pharmacokinetic considerations in obesity.
life [80] versus two [17], which could also account for the J Pharm Sci 1999;88:1–7.
differences between the two studies. [5] Center for Disease Control. Health, United States. With a special
In the third study, Segall-Gutierrez et al. [81] investigated emphasis on young adults (Table 97). In: http://www.cdc.gov/nchs/
data/hus/hus08.pdf #097; 2008.
the effect of obesity on follicular development and serum
[6] Finer LB, Henshaw SK. Disparities in rates of unintended pregnancy in
MPA levels in women using subcutaneous depo-MPA the United States, 1994 and 2001. Perspect Sex Reprod Health 2006;
(DMPA-SC 104 mg) for contraception during two 12- 38:90–6.
week cycles. Subjects were recruited for three BMI [7] Genazzani AR, Lucchesi A, Stomati M, et al. Effects of sex steroid
categories (five subjects in each group): normal (18.5–24.9 hormones on the neuroendocrine system. Eur J Contracept Reprod
Health Care 1997;2:63–9.
kg/m2), obese (30.0–39.9 kg/m2) and extremely obese (N40
[8] Mahesh VB. The dynamic interaction between steroids and gonado-
kg/m2). Blood samples were obtained at monthly intervals tropins in the mammalian ovulatory cycle. Neurosci Biobehav Rev
for quantifying MPA levels for a total of 26 weeks. Large 1985;9:245–60.
intersubject variability was observed in serum MPA levels. [9] Spies HG, Niswender GD. Effect of progesterone and estradiol on LH
Median MPA levels remained above the level (200 pg/mL) release and ovulation in Rhesus monkeys. Endocrinology 1972;90:
needed to prevent ovulation, but compared to the normal 257–61.
[10] Wildt L, Hausler A, Marshall G, et al. Frequency and amplitude of
weight subjects, the levels were lower among obese subjects gonadotropin-releasing hormone stimulation and gonadotropin secre-
and lowest among extremely obese subjects. tion in the rhesus monkey. Endocrinology 1981;109:376–85.
[11] Speroff L, Fritz M. Clinical gynecologic endocrinology and infertility.
7th ed. Baltimore (Md): Lippincott Williams & Wilkins; 2005.
8. Pharmacogenetics [12] Knobil E, Plant TM, Wildt L, Belchetz PE, Marshall G. Control of the
rhesus monkey menstrual cycle: permissive role of hypothalamic
gonadotropin-releasing hormone. Science 1980;207:1371–3.
A relatively new area of drug research is pharmacoge- [13] Hemrika DJ, Slaats EH, Kennedy JC, de Vries Robles-Korsen TJ,
netics or the role of inheritance in an individual's response to Schoemaker J. Pulsatile luteinizing hormone secretion during the first
322 A.B. Edelman et al. / Contraception 82 (2010) 314–323

and the fourth cycle on two different oral contraceptives containing [40] Geier A, Dietrich CG, Grote T, et al. Characterization of organic anion
gestodene. Acta Endocrinol (Copenh) 1993;129:229–36. transporter regulation, glutathione metabolism and bile formation in
[14] McPherson III JC, Costoff A, Mahesh VB. Influence of estrogen- the obese Zucker rat. J Hepatol 2005;43:1021–30.
progesterone combinations on gonadotropin secretion in castrate [41] Lesser GT, Deutsch S. Measurement of adipose tissue blood flow and
female rats(1). Endocrinology 1975;97:771–9. perfusion in man by uptake of 85Kr. J Appl Physiol 1967;23:621–30.
[15] Schleussner E, Brueckner T, Brautigam J, Michels W. Influence of two [42] Bowman SL, Hudson SA, Simpson G, Munro JF, Clements JA. A
low-dose oral contraceptives on pulsatile gonadotropin secretion. comparison of the pharmacokinetics of propranolol in obese and
Gynecol Endocrinol 2001;15:259–64. normal volunteers. Br J Clin Pharmacol 1986;21:529–32.
[16] Dericks-Tan JS, Koch P, Taubert HD. Synthesis and release of [43] Cheymol G. Effects of obesity on pharmacokinetics implications for
gonadotropins: effect of an oral contraceptive. Obstet Gynecol 1983; drug therapy. Clin Pharmacokinet 2000;39:215–31.
62:687–90. [44] Kjellberg J, Reizenstein P. Body composition in obesity. Acta Med
[17] Edelman AB, Carlson NE, Cherala G, et al. Impact of obesity on oral Scand 1970;188:161–9.
contraceptive pharmacokinetics and hypothalamic–pituitary–ovarian [45] Forbes GB, Welle SL. Lean body mass in obesity. Int J Obes 1983;7:
activity. Contraception 2009;80:119–27. 99–107.
[18] Goldzieher JW. Pharmacokinetics and metabolism of ethinyl estro- [46] Green B, Duffull SB. What is the best size descriptor to use for
gens. In: Goldzieher JW, Fotherby K, editors. Pharmacology of the pharmacokinetic studies in the obese? Br J Clin Pharmacol 2004;58:
contraceptive steroids. New York: Raven Press Ltd; 1994. p. 127–52. 119–33.
[19] Goldzieher JW. Pharmacology of contraceptive steroids: a brief [47] Nisbet AC. Intramuscular gluteal injections in the increasingly obese
review. Am J Obstet Gynecol 1989;160:1260–4. population: retrospective study. BMJ 2006;332:637–8.
[20] Goldzieher JW, Stanczyk FZ. Oral contraceptives and individual [48] Smith SR, Lovejoy JC, Greenway F, et al. Contributions of total body
variability of circulating levels of ethinyl estradiol and progestins. fat, abdominal subcutaneous adipose tissue compartments, and visceral
Contraception 2008;78:4–9. adipose tissue to the metabolic complications of obesity. Metabolism
[21] Guengerich FP. Metabolism of 17 alpha-ethynylestradiol in humans. 2001;50:425–35.
Life Sci 1990;47:1981–8. [49] Wortsman J, Matsuoka LY, Chen TC, Lu Z, Holick MF. Decreased
[22] Roy S, Bernstein L, Stanczyk FZ. Analysis of oral contraceptive risks. bioavailability of vitamin D in obesity. Am J Clin Nutr 2000;72:
In: Runnebaum B, Rabe T, Kiesel L, editors. Female contraception. 690–3.
Berlin (West Germany): Springer-Verlg; 1988. p. 21–55. [50] Cheymol G, Poirier JM, Barre J, Pradalier A, Dry J. Comparative
[23] Goldzieher JW, Mileikowsky G, Newburger J, Dorantes A, pharmacokinetics of intravenous propranolol in obese and normal
Stavchansky SA. Human pharmacokinetics of ethynyl estradiol 3- volunteers. J Clin Pharmacol 1987;27:874–9.
sulfate and 17-sulfate. Steroids 1988;51:63–79. [51] Derry CL, Kroboth PD, Pittenger AL, Kroboth FJ, Corey SE, Smith
[24] Goldzieher JW, Dozier TS, de la Pena A. Plasma levels and RB. Pharmacokinetics and pharmacodynamics of triazolam after two
pharmacokinetics of ethynyl estrogens in various populations. I. intermittent doses in obese and normal-weight men. J Clin Psycho-
Ethynylestradiol. Contraception 1980;21:1–16. pharmacol 1995;15:197–205.
[25] Fotherby K, Akpoviroro J, Abdel-Rahman HA, et al. Pharmacokinetics [52] Hautanen A. Synthesis and regulation of sex hormone-binding
of ethynyloestradiol in women for different populations. Contraception globulin in obesity. Int J Obes Relat Metab Disord 2000;24(Suppl 2):
1981;23:487–96. S64–S70.
[26] Williams MC, Goldzieher JW. Chromatographic patterns of urinary ethynyl [53] Wisse BE. The inflammatory syndrome: the role of adipose tissue
estrogen metabolites in various populations. Steroids 1980;36:255–82. cytokines in metabolic disorders linked to obesity. J Am Soc Nephrol
[27] van den Heuvel MW, van Bragt AJ, Alnabawy AK, Kaptein MC. 2004;15:2792–800.
Comparison of ethinylestradiol pharmacokinetics in three hormonal [54] Lee YH, Pratley RE. The evolving role of inflammation in obesity and
contraceptive formulations: the vaginal ring, the transdermal patch and the metabolic syndrome. Curr Diab Rep 2005;5:70–5.
an oral contraceptive. Contraception 2005;72:168–74. [55] Aygun AD, Gungor S, Ustundag B, Gurgoze MK, Sen Y.
[28] Stanczyk FZ. All progestins are not created equal. Steroids 2003;68: Proinflammatory cytokines and leptin are increased in serum of
879–90. prepubertal obese children. Mediators Inflamm 2005;2005:180–3.
[29] Xing J, Chen JD. Alterations of gastrointestinal motility in obesity. [56] Das UN. Is obesity an inflammatory condition? Nutrition 2001;17:
Obes Res 2004;12:1723–32. 953–66.
[30] Adams JP, Murphy PG. Obesity in anaesthesia and intensive care. Br J [57] Charles KA, Rivory LP, Brown SL, Liddle C, Clarke SJ, Robertson
Anaesth 2000;85:91–108. GR. Transcriptional repression of hepatic cytochrome P450 3A4 gene
[31] Wisen O, Hellstrom PM. Gastrointestinal motility in obesity. J Intern in the presence of cancer. Clin Cancer Res 2006;12:7492–7.
Med 1995;237:411–8. [58] Frye RF, Schneider VM, Frye CS, Feldman AM. Plasma levels of
[32] Dubois A. Obesity and gastric emptying. Gastroenterology 1983;84: TNF-alpha and IL-6 are inversely related to cytochrome P450-
875–6. dependent drug metabolism in patients with congestive heart failure.
[33] Tosetti C, Corinaldesi R, Stanghellini V, et al. Gastric emptying of J Card Fail 2002;8:315–9.
solids in morbid obesity. Int J Obes Relat Metab Disord 1996;20:200–5. [59] Jover R, Bort R, Gomez-Lechon MJ, Castell JV. Down-regulation of
[34] Wright RA, Krinsky S, Fleeman C, Trujillo J, Teague E. Gastric human CYP3A4 by the inflammatory signal interleukin-6: molecular
emptying and obesity. Gastroenterology 1983;84:747–51. mechanism and transcription factors involved. FASEB J 2002;16:
[35] Zahorska-Markiewicz B, Jonderko K, Lelek A, Skrzypek D. Gastric 1799–801.
emptying in obesity. Hum Nutr Clin Nutr 1986;40:309–13. [60] Morgan ET, Goralski KB, Piquette-Miller M, et al. Regulation of drug-
[36] Basilisco G, Camboni G, Bozzani A, Vita P, Doldi S, Bianchi PA. metabolizing enzymes and transporters in infection, inflammation, and
Orocecal transit delay in obese patients. Dig Dis Sci 1989;34:509–12. cancer. Drug Metab Dispos 2008;36:205–16.
[37] Wisen O, Johansson C. Gastrointestinal function in obesity: motility, [61] Sherlock S. Diseases of the liver biliary system. 7th ed. Boston:
secretion, and absorption following a liquid test meal. Metabolism Blackwell Scientific Publications; 1985.
1992;41:390–5. [62] Irizar A, Barnett CR, Flatt PR, Ioannides C. Defective expression of
[38] Bolt HM. Interactions between clinically used drugs and oral cytochrome P450 proteins in the liver of the genetically obese Zucker
contraceptives. Environ Health Perspect 1994;102(Suppl 9):35–8. rat. Eur J Pharmacol 1995;293:385–93.
[39] Dobrinska MR. Enterohepatic circulation of drugs. J Clin Pharmacol [63] Salazar DE, Sorge CL, Corcoran GB. Obesity as a risk factor for drug-
1989;29:577–80. induced organ injury. VI. Increased hepatic P450 concentration and
A.B. Edelman et al. / Contraception 82 (2010) 314–323 323

microsomal ethanol oxidizing activity in the obese overfed rat. [73] Blouin RA, Kolpek JH, Mann HJ. Influence of obesity on drug
Biochem Biophys Res Commun 1988;157:315–20. disposition. Clin Pharm 1987;6:706–14.
[64] Yoshinari K, Okino N, Sato T, Sugatani J, Miwa M. Induction of [74] Ahmed SB, Fisher ND, Stevanovic R, Hollenberg NK. Body mass
detoxifying enzymes in rodent white adipose tissue by aryl index and angiotensin-dependent control of the renal circulation in
hydrocarbon receptor agonists and antioxidants. Drug Metab Dispos healthy humans. Hypertension 2005;46:1316–20.
2006;34:1081–9. [75] Li WM, Chou YH, Li CC, et al. Association of body mass index
[65] Benet LZ, Cummins CL, Wu CY. Unmasking the dynamic interplay and urine pH in patients with urolithiasis. Urol Res 2009;37:
between efflux transporters and metabolic enzymes. Int J Pharm 193–6.
2004;277:3–9. [76] Allard S, Kinzig M, Boivin G, Sorgel F, LeBel M. Intravenous
[66] Cheng Q, Aleksunes LM, Manautou JE, et al. Drug-metabolizing ciprofloxacin disposition in obesity. Clin Pharmacol Ther 1993;54:
enzyme and transporter expression in a mouse model of diabetes and 368–73.
obesity. Mol Pharm 2008;5:77–91. [77] Bauer LA, Edwards WA, Dellinger EP, Simonowitz DA. Influence
[67] Pierre K, Parent A, Jayet PY, Halestrap AP, Scherrer U, Pellerin L. of weight on aminoglycoside pharmacokinetics in normal weight
Enhanced expression of three monocarboxylate transporter isoforms in and morbidly obese patients. Eur J Clin Pharmacol 1983;24:
the brain of obese mice. J Physiol 2007;583:469–86. 643–7.
[68] Saheki A, Terasaki T, Tamai I, Tsuji A. In vivo and in vitro blood– [78] Korsager S. Administration of gentamicin to obese patients. Int J Clin
brain barrier transport of 3-hydroxy-3-methylglutaryl coenzyme A Pharmacol Ther Toxicol 1980;18:549–53.
(HMG-CoA) reductase inhibitors. Pharm Res 1994;11:305–11. [79] Sketris I, Lesar T, Zaske DE, Cipolle RJ. Effect of obesity on
[69] Tsuji A, Saheki A, Tamai I, Terasaki T. Transport mechanism of 3- gentamicin pharmacokinetics. J Clin Pharmacol 1981;21:288–93.
hydroxy-3-methylglutaryl coenzyme A reductase inhibitors at the [80] Westhoff C, Torgal A, Mayeda E, Pike M, Stanczyk F. Pharmacoki-
blood–brain barrier. J Pharmacol Exp Ther 1993;267:1085–90. netics of a combined oral contraceptive in obese and normal weight
[70] Henegar JR, Bigler SA, Henegar LK, Tyagi SC, Hall JE. Functional women. Contraception 2010;81:474–80.
and structural changes in the kidney in the early stages of obesity. J Am [81] Segall-Gutierrez P, Taylor D, Liu X, Stanczyk FZ, Azen S, Mishell
Soc Nephrol 2001;12:1211–7. DR. Follicular development and ovulation in extremely obese women
[71] Pantoja Zuzuarregui JR, Mallios R, Murphy J. The effect of obesity on receiving depo-medroxyprogesterone acetate subcutaneously. Contra-
kidney length in a healthy pediatric population. Pediatr Nephrol ception 2010;81:487–95.
2009;24:2023–7. [82] Garber AM, Tunis SR. Does comparative-effectiveness research
[72] Chagnac A, Weinstein T, Korzets A, Ramadan E, Hirsch J, Gafter U. threaten personalized medicine? N Engl J Med 2009;360:1925–7.
Glomerular hemodynamics in severe obesity. Am J Physiol Renal [83] Weinshilboum R. Inheritance and drug response. N Engl J Med
Physiol 2000;278:F817–22. 2003;348:529–37.

You might also like