You are on page 1of 15

THERAPEUTICS

Drugs in Pregnancy and


48 Lactation
Paula Russell, Laura Yates, Peter Golightly and Sarah Fenner

at some stage of pregnancy (Headley et al., 2004), with a signii-


Key points cant number of women taking medication at the time their preg-
Drugs in pregnancy nancy is detected. Indications for drug use range from chronic
• Assess risk/benefit ratio for the mother–fetus pair. illnesses, such as epilepsy and depression, to those commonly
• Avoid non-essential drugs. associated with pregnancy, such as nausea and vomiting and uri-
• Where drug treatment is clinically indicated, select an effective nary tract infections.
agent with the best safety profile. Approximately 2–3% of all live births are associated with a
• Use the lowest effective dose for the shortest possible time. congenital anomaly. Although exogenous factors such as drugs
• Provide timely and accurate counselling to help avoid may account for only 1–5% of these (affecting <0.2% of all live
unfounded maternal fears about drug safety that may other- births), given that drug-associated malformations are largely pre-
wise result in non-adherence with drug therapy or unnecessary ventable, they remain an important consideration.␣
pregnancy termination.
• Teratogenic risks of a drug may vary with each trimester of
exposure.
• Drug exposure in the second and third trimesters may also
result in fetal harm.
Drugs as teratogens
Drugs in lactation
A teratogen is deined as any agent that results in structural or
• Avoid unnecessary use of drugs.
functional abnormalities in the fetus, or in the child after birth,
• Maternal therapy only rarely constitutes a reason to avoid as a consequence of maternal exposure during pregnancy.
breastfeeding.
Examples of drugs that are known to be human teratogens are
• Assess the risk/benefit ratio for both mother and infant.
listed in Box 48.1. The teratogenic mechanism for most drugs
• Monitor the infant for unusual signs or symptoms.
remains unclear but may be due to the direct effects of the drug
• Avoid use of new drugs if there is a therapeutic equivalent for on the fetus and/or as a consequence of indirect physiological
which data on use in lactation are available.
changes in the mother or fetus. Perhaps the best known and most
widely studied teratogen is thalidomide, a mild sedative that
was widely marketed as a remedy for pregnancy-related nau-
Drugs in pregnancy sea and vomiting. In 1961 thalidomide was withdrawn from the
UK market after numerous reports of severe anatomical birth
The use of both prescription and over-the-counter drugs in defects in infants of mothers who took the drug in early preg-
pregnancy presents a number of challenges to those asked to nancy. Whereas external congenital anomalies such as limb
provide advice to women either pre-conceptually or during abnormalities, spina biida and hydrocephalus may be obvious
pregnancy. This is in part due to the fact that no two cases are at birth, some defects may take many years to manifest clinically
the same, and that each enquiry ideally requires an individual or be identiied. Examples of delayed effects of teratogens are
risk assessment that takes into account what is known about the behavioural and neurodevelopmental disorders associated
the drug and its effects on the developing fetus, as well as the with in utero sodium valproate exposure and the development of
woman’s personal medical and family history. It is now well clear-cell vaginal cancer in young women after maternal intake
recognised that most medicines and/or their metabolites readily of diethylstilboestrol, used irst in the 1930s for the prevention
cross the placenta, and that some are teratogenic, resulting in of miscarriage and preterm delivery (Herbst et al., 1971).
harm to the fetus. Robust scientiic human data on the effects
of many drugs, particularly newer preparations, are, however,
Critical periods in human fetal development
frequently lacking.
It is therefore generally accepted that maternal pharmacother- The human gestation period is approximately 40 weeks from the
apy should be avoided if not necessary, or minimised where the irst day of the last menstrual period (38 weeks post-conception)
maternal condition allows. Nevertheless, it has been estimated and is conventionally divided into the irst, second and third
820 that more than 80% of expectant mothers take three or four drugs trimesters, each lasting 3 calendar months. Another method
DRUGS IN PREGNANCY AND LACTATION 48
Box 48.1 Examples of drugs considered to be human Fetal stage: weeks 9–38 post-conception
teratogens
During the fetal stage, the fetus continues to develop, grow
Angiotensin-converting enzyme inhibitors and mature and, importantly, remains susceptible to some drug
Androgens effects. This is especially true for the CNS, which can be dam-
Carbamazepine aged by exposure to certain drugs, for example, ethanol, at
Carbimazole any stage of pregnancy. The external genitalia also continue
Cytotoxics (some) to form from the seventh week until term; consequently, dan-
Danazol azol, which has weak androgenic properties, can cause virilisa-
Diethylstilboestrol
Ethanol
tion of a female fetus if given in any trimester after the eighth
Lithium week of pregnancy when the androgen receptors begin to form
Methotrexate (Rosa, 1984).
Misoprostol Further examples include the angiotensin-converting
Mycophenolate mofetil enzyme inhibitors, which if given in the second and third tri-
Tetracyclines mesters can result in fetal renal dysfunction and subsequent
Thalidomide
oligohydramnios, that is, reduced amniotic luid volume
Valproic acid
Vitamin A and derivatives, e.g. isotretinoin
(Sedman et al., 1995). The non-steroidal anti-inlammatory
Warfarin drugs (NSAIDs) are another important group of drugs that may
cause problems, speciically in the third trimester. These drugs
inhibit prostaglandin synthesis in a dose-related fashion and,
when given late in pregnancy, may result in premature clo-
for classifying the stage of pregnancy is according to the stage sure of the fetal ductus arteriosus and fetal renal impairment
of embryo-fetal development. This is a more useful approach (Koren et al., 2006). NSAIDs should therefore be avoided dur-
when assessing the potential risks associated with drug use in ing the third trimester.␣
pregnancy.
Principles of teratogenesis
Pre-embryonic stage: weeks 0–2 post-conception
In 1959 James Wilson, co-founder of The Teratology Society,
The irst 2 weeks post-conception are regarded as the pre- proposed several principles of teratogenesis that have since
embryonic stage and describe the period up to implantation been expanded and modiied but remain fundamental in assess-
of the fertilised ovum. Teratogenic exposure during the pre- ing whether a drug or chemical exposure during pregnancy
embryonic stage, sometimes referred to as the ‘all-or-nothing’ is likely to be associated with reproductive or developmen-
period, is thought to carry a low risk of fetal harm, with most expo- tal toxicity. A basic understanding of these factors is essential
sures leading either to death of the embryo or complete recovery and to both the interpretation of preclinical (animal) reproductive
normal development of the fetus. Fetal malformations after drug toxicity studies and to enable accurate risk assessment in clini-
exposure during this period are therefore thought to be unlikely, cal practice. A subset of Wilson’s principles is discussed as
but robust scientiic data to support a low risk of adverse fetal out- follows.
come are lacking. Risk to the fetus may also be increased where
the half-life of the drug is suficient to extend exposure into the
Timing of exposure
embryonic stage. A good example of the latter is isotretinoin and
related vitamin A derivatives that have half-lives up to a week, The stage of pregnancy at which a drug exposure occurs is
and which when used systemically, for example, for the treat- key to determining the likelihood, severity or nature of any
ment of acne and psoriasis, are recognised teratogens (Nulman adverse effect on the fetus. Risk both between and within tri-
et al., 1998).␣ mesters may be variable. For example, folic acid antagonists
such as trimethoprim have been associated with an increased
risk of neural tube defects if exposure occurs before neural
Embryonic stage: weeks 3–8 post-conception
tube closure (third to fourth week post-conception), but not
Organogenesis occurs predominantly during the embryonic after this period (Hernandez-Diaz et al., 2001). It has also been
stage and, with the exception of the central nervous system suggested that trimethoprim should be avoided after 32 weeks’
(CNS), eyes, teeth, external genitalia and ears, is complete gestation in view of the theoretical risk of severe jaundice in
by the end of the 10th week of pregnancy. Exposure to drugs the neonate as a result of bilirubin displacement from protein
during this critical period therefore represents the greatest binding, although clinical evidence to support this is lacking
risk of major birth defects. For this reason, women are often (Dunn, 1964). Unfortunately, the precise period of teratogenic
advised to avoid or minimise all drug use in the irst trimester risk is known for very few substances. One drug for which this
whenever possible. It is important to bear in mind, however, period has been established is thalidomide, where exposure
that very few drugs are in fact proven teratogens, and that between days 20 and 36 post-conception is associated with a
exposure in the second and third trimesters may still result in high risk of congenital malformation (Lenz, 1988; Newman,
fetal harm.␣ 1986).␣ 821
48 THERAPEUTICS

Drug dose Table 48.1 Examples of drugs with pharmacological effects


on the fetus or neonate
A threshold dose above which drug-induced malformations or
other adverse effects are more likely to occur has now been dem- Possible adverse
onstrated for certain teratogenic compounds, although for most a Drug pharmacological effect Notes
‘safe dose’ has not been conclusively determined. The likelihood
Angiotensin- Fetal and neonatal hy- Monitor fetus if
of a dose relationship underlies the recommendation to use the
converting poxia, hypotension, renal long-term therapy
lowest effective dose in pregnancy. For this reason more frequent enzyme dysfunction, oligohydram- in the second or
monitoring of drug levels may be recommended for certain drugs inhibitors nios and intra-uterine third trimester
during pregnancy.␣ growth retardation

β-Blockers, Neonatal bradycardia, Neonatal symptoms


Species e.g. atenolol hypotension and hyper- are usually mild and
glycaemia improve within
Teratogenicity of a drug may be species dependent. Interestingly, 48 h; no
preclinical thalidomide studies in mice and rats did not result long-term effects
in congenital malformation in the offspring (Breitkreutz and
Anderson, 2008; Miller et al., 2009; Vorhees et al., 2001). Birth Benzodiaz- ‘Floppy infant syndrome’ Risk if regular use in
defects or other adverse reproductive outcomes observed in ani- epines Withdrawal reactions third trimester
mal studies cannot therefore be simply extrapolated to the human Neonatal observa-
tion recommended
situation. Further, the drug dose and route of administration used
in early animal studies may not be comparable with clinical use Corticosteroid Fetal adrenal suppression Dependent on
in humans.␣ (high dose) dose and treatment
interval
Genotype and environmental interaction Non-steroidal Premature closure of the Avoid repeated use
Not all fetuses exposed to known teratogenic drugs show evi- anti- ductus arteriosus (affect- after week 28; if
inflammatory ing fetal circulation) and unavoidable, fetal
dence of having been affected in utero. It remains undetermined
drugs fetal renal impairment circulation moni-
whether this variable susceptibility to teratogenic drugs is a (decreased urine output) tored regularly
result of genetic differences in the exposed mothers, the fetal
genotype, modifying environmental factors or a combination of Opioids Neonatal withdrawal Risk if used long-
all three. Malformations are reported to occur in only 20–50% symptoms term
of infants born to mothers exposed to thalidomide during the Respiratory depression Risk if used near
period of greatest risk of embryopathy, that is, days 20–36 post- term
fertilisation (Lenz, 1966; Newman, 1985). Similarly, maternal
Phenothi- Neonatal withdrawal and Observation for at
treatment with systemic isotretinoin during the irst trimester azines transient extrapyramidal least 48 h;
results in fetal malformation in only 18–35% of the live born symptoms symptoms may last
infants, with a further 30% of children exhibiting developmental for several weeks
delay in the absence of physical deformity (Benke, 1984; Braun
et al., 1984; Hill, 1984).␣ Tricyclic and Neonatal withdrawal Risk if used long-
selective sero- symptoms term and/or near
tonin reuptake term; observation
Pharmacological effect inhibitor anti- for at least 48 h
depressants
Pharmacological effects on the fetus are by far the most common
drug effects during pregnancy, and the consequences are often Adapted from Schaefer et al. (2014).
minor and reversible compared with the idiosyncratic effects
that can lead to major irreversible anomalies. Pharmacological
effects are usually dose related and to some extent predict- The capacity of the neonate to eliminate drugs is reduced,
able. Drugs may adversely affect the fetus via effects on the and this can result in signiicant accumulation of some drugs,
maternal circulation, or they may cross the placenta and exert leading to toxicity. Neonatal withdrawal effects may require
a direct pharmacological effect on the fetus. Equally, drugs are treatment.
sometimes administered to pregnant women to treat fetal dis- Idiosyncratic drug effects in the fetus and neonate are possible
orders; for example, lecainide has been used to resolve fetal but occur rarely compared with pharmacological effects.␣
tachycardia.
The neonate can also be adversely affected by maternal drug
Maternal pharmacokinetic changes
therapy (Table 48.1). It is generally only at birth that signs of
fetal distress are observed due to in utero drug exposure or the Physiological changes in pregnancy induce profound changes to
effects of abrupt discontinuation of the maternal drug supply. the pharmacokinetic properties of many drugs. These changes,
822
DRUGS IN PREGNANCY AND LACTATION 48
peaks at term (36–42 L/h). There is also a mean increase of 8 L in
Table 48.2 Summary of pharmacokinetic changes during
body water (60% to placenta, fetus and amniotic luid and 40% to
pregnancy
maternal tissues). As a consequence, there may be increased dos-
Change during pregnancy age requirements for some drugs to achieve the same therapeutic
effect, provided these effects are not offset by other pharmaco-
Absorption kinetic changes. Both the total plasma and the free-drug concen-
Gastro-intestinal motility ↓
trations of phenytoin, carbamazepine and valproic acid decrease
during pregnancy, but the free-drug fraction (ratio of free to total
Lung function ↑ plasma concentration) may increase (Pavek et al., 2009).
Protein binding. Albumin is the main plasma protein respon-
Skin blood circulation ↑ sible for binding acidic drugs such as phenytoin and salicylates,
whereas α1-acid glycoprotein predominantly binds basic drugs,
Distribution
including β-blockers and opioid analgesics. As pregnancy pro-
Plasma volume ↑ gresses the plasma volume increases at a greater rate than the
increase in albumin which results in hypoalbuminaemia. In addi-
Body water ↑ tion, steroid and placental hormones occupy the protein-binding
sites. This leads to an increase in the fraction of unbound drug.
Plasma protein ↓ Clinical effect is related to the concentration of unbound drug,
which usually remains unchanged even though the total (bound
Fat deposition ↑
plus unbound) plasma concentration is decreased. Thus, a decline
Metabolism in the total plasma concentration does not usually require an
increase in dose. The α1-acid glycoprotein concentrations remain
Liver activity ↑↓ the same as those in non-pregnancy.
Excretion
Phenytoin is bound to albumin and exhibits the effects
described earlier, but the situation is further complicated by
Glomerular filtration ↑ increased hepatic metabolism that may necessitate a dose
increase. Consequently, therapy can only be reliably guided by
Renal tubule secretion ↑ clinical assessment or measurement of unbound rather than total
Adapted from Schaefer et al. (2014).
plasma concentration.␣

Metabolism
summarised in Table 48.2, affect absorption, distribution, metabolism,
and excretion of drugs, and may also affect their pharmacodynamic The metabolic activity of cytochrome P450 isoenzymes CYP3A4,
properties. Some of the changes that occur in maternal organ systems CYP2D6, CYP2A6 and CYP2C9 and uridine 5′-diphosphate
are secondary to pregnancy-induced hormonal changes, whereas glucuronosyltransferase (UGT) isoenzymes (UGT1a1, UGT1A4
others occur to support the pregnant woman and the developing fetus. and UGT2B7) is increased during pregnancy. Drugs metabolised
by these isoenzymes may therefore require dose adjustment. This
may decrease the amount of the drug available for transfer across
Absorption
the placenta, and thereby inluence fetal exposure. In contrast,
Gastric and intestinal emptying time increases by 30–40% in the sec- the metabolic activity of CYP1A2 and CYP2C19 is decreased
ond and third trimesters (Pavek et al., 2009) and could be important during pregnancy, and drugs metabolised by these isoenzymes
in delaying absorption and time to onset of action for some drugs may need dose reduction to minimise toxicity. These changes
(Loebstein et al., 1997). There is also a reduction in gastric acid and their extent depend on the stage of pregnancy, so there may
secretion in the irst and second trimesters, and an increase in mucous be clinically signiicant changes in drug concentrations between
secretion. As a consequence of the increase in gastric pH, the ionisa- trimesters (Table 48.3) (Anderson, 2005; Deligiannidis et al.,
tion, and hence absorption, of weak acids and bases can be affected. 2014). In general the effects on individual drugs are inconsistent
Cardiac output and respiratory volume increase during pregnancy, and dificult to predict, but knowledge of the effect of pregnancy
leading to hyperventilation and increased pulmonary blood perfusion. on isoenzymes may inform decisions about possible monitoring
These changes cause higher pulmonary absorption of anaesthetics, and/or dose alterations.␣
bronchodilators, pollutants, cigarette smoke and other volatile drugs.␣
Excretion
Distribution
Within the irst few weeks of pregnancy the glomerular iltration
The volume of distribution of drugs may be altered because of rate (GFR) increases by approximately 50%. Consequently, those
an increase of up to 50% in blood (plasma) volume and a 30% drugs which are excreted primarily unchanged by the kidneys,
increase in cardiac output. Renal blood low increases by up to 50% for example, lithium, digoxin and penicillin, show enhanced
at the end of the irst trimester, and uterine blood low increases and elimination and lower steady-state concentrations. The following
823
48 THERAPEUTICS

and maternal proteins. For example, fetal and newborn plasma


Table 48.3 Summary of pregnancy-induced effects on
proteins appear to bind ampicillin and benzylpenicillin with
hepatic metabolism of some drugs
less afinity and salicylates with greater afinity than maternal
Effect on clearance proteins. Maternal albumin gradually decreases during preg-
nancy and fetal albumin concentrations increase, so different
Drugs/ First Second Third fetal/maternal albumin concentrations occur at different stages
Isoenzyme probes trimester trimester trimester
of pregnancy. The degree of protein binding of any drug is an
CYP1A2 Caffeine ↓ 33% ↓ 50% ↓ 65% important determinant of its movement across the placenta.
Drugs that are highly protein bound tend to achieve higher
CYP2A6 Nicotine ND ↑ 54% maternal and lower fetal concentrations. Drugs with very large
(combined molecular weights such as insulin and heparin have negligible
data) transfer. Lipophilic, un-ionised drugs cross the placenta more
CYP2C9 Phenytoin No effect No effect ↑ 20%
easily than polar drugs, and weakly basic drugs may become
‘trapped’ in the fetal circulation because of the slightly lower pH
CYP2C19 Proguanil ND ↓ 50% ↓ 50% compared with maternal plasma.
Some other factors such as enzymes or drug eflux transporters
CYP2D6 Dextro- in the placenta may facilitate or restrict the transfer of a drug to
methorphana ND ND ↑ 50% the fetus. The inluence of transporter activity on placental trans-
Metoprolol
fer is increasingly recognised. For example, the accumulating
CYP3A4 Cortisola
evidence about transport mechanisms operating at the placental
ND ND ↑ Variableb barrier is expected to be useful in developing techniques to con-
Nifedipine
trol fetal and placental transfer of antiviral drugs, thus achieving
UGT1A4 Lamotrigine ↑ 200% ↑ 200% ↑ 300% maximum therapeutic beneit while minimising potential fetal
monother- toxicity (Tomi et al., 2011).␣
apy
Polytherapy ↑ 65% ↑ 65% ↑ 90%
Maternal pharmacodynamic changes
UGT2B7 Morphine ND ND ↑ Variableb
(limited Pharmacodynamic changes in pregnancy are less well stud-
data) ied than pharmacokinetic changes, but increases in blood pres-
sure and blood glucose associated with corticosteroids may be
aDextromethorphan and cortisol used as probes of CYP2D6 and CYP3A4
activity.
more common in pregnancy. It is thought that impairment of
bExtent variable depending on the drug studied. cell-mediated immunity affects the eficacy of vaccination, par-
ND, Not detectable. ticularly late in pregnancy. Sensitivity to the bradycardic effects
Adapted from Anderson (2005). of β-receptor blocking drugs such as propranolol may also be
increased in pregnancy (Rubin et al., 1987). There is currently
not a good understanding of pharmacodynamic changes and their
clinical implications in pregnancy.␣

drugs have shown pregnancy-induced increases of 20–65% on


Drug dosing in pregnancy
their renal elimination (Anderson, 2005):
• ampicillin, As a general principle, the dose of a drug given at any stage of
• cefuroxime, pregnancy should be as low as possible to minimise potential
• ceftazidime, toxic effects to the fetus. Drug therapy that is considered essential
• cefazolin, can be tapered to the lowest effective dose either before concep-
• piperacillin, tion (ideally) or at the time the pregnancy is diagnosed. Where
• atenolol, drug exposure during the third trimester is predicted to have an
• sotalol, adverse effect on the neonate postpartum, consideration may
• digoxin, be given to slowly reducing the dose towards term to minimise
• lithium, the risks to the baby. Such decisions are, however, not always
• dalteparin sodium, straightforward. Recommendations to wean an expectant mother
• enoxaparin sodium.␣ off antidepressants and antipsychotics to reduce the likelihood
of neonatal withdrawal syndrome (characterised by jitteriness,
altered muscle tone, poor feeding and irritability) and, in the case
Fetal–placental transfer
of the selective serotonin reuptake inhibitors (SSRIs), avoid the
Most drugs have a molecular weight less than 600 and diffuse possible increased risk of persistent pulmonary hypertension of
easily across the placenta to enter the fetal circulation to some the newborn are now being challenged. Not only are there insuf-
extent. In general, the fetal/maternal drug concentration ratio is icient data to conclusively demonstrate neonatal beneit or an
824 less than 1. Drugs differ in the extent to which they bind to fetal optimal time of weaning, but also the increased risk of psychiatric
DRUGS IN PREGNANCY AND LACTATION 48
problems and relapse in the mother’s immediate postpartum the beneits outweigh the risks’. This is understandable from a
period needs to be taken into account. medico-legal point of view but offers little in terms of risk assess-
Many physiological changes occur during pregnancy that ment. The primary literature is frequently inadequate because
may affect the way the body handles drugs. Knowledge of these ethical and legal restraints mean that randomised controlled trials
changes can allow some prediction of the impact on pharma- are rarely undertaken in pregnant women. Often the only informa-
cotherapy while remaining aware that there is variability in the tion that is available is conined to retrospective studies, voluntary
extent of these changes during the pregnancy and high inter- reporting schemes and/or animal studies. The rate of anomalies
individual variability. The need for changes in dosages is inlu- in retrospective studies and voluntary reporting databases may
enced by whether the drug is excreted unchanged by the kidneys be erroneously elevated due to preferential reporting of abnor-
or which metabolic isoenzymes are involved in its elimination. mal outcomes. Individual case reports are also dificult to inter-
Women who are taking drugs with enhanced clearance and for pret because the denominator of drug exposure is unknown, and
which a good correlation between plasma levels and therapeu- an abnormal outcome may be coincidental to the drug exposure.
tic effect exists should have their plasma concentrations closely More recently, prospective controlled trials have been utilised
monitored and dose adjusted to reduce the risk of suboptimal where the pregnancy outcomes of a deined cohort of women
therapy, for example, with phenytoin, carbamazepine, lithium exposed to the drug are compared with outcomes of a matched
and digoxin. Similarly, highly protein-bound drugs may require control group. Complete follow-up of each pregnancy and postna-
free-drug concentration monitoring. However, there is no clear tal monitoring is an essential feature of this type of investigation.␣
guidance for adjusting doses during pregnancy, and for most
drugs, the concentration of free drug, and therefore the effect of
Pre-conception advice
that drug, is unchanged.
Pregnancy itself can cause a temporary worsening or Drug use during the irst trimester, in particular, the embryonic
improvement of some diseases and in that way inluence drug stage, carries the greatest risk of malformation because this
dosages.␣ is when the fetal organs are being formed. Ideally all unneces-
sary drug therapy should be discontinued prior to conception.
However, inadvertent drug exposure frequently occurs, because
Drug selection in pregnancy
approximately half of all pregnancies are unplanned. It is thus
Although there are few, if any, drugs for which safe use in preg- critical to make careful drug choices when prescribing for women
nancy can be absolutely assured, only a handful of drugs in cur- of reproductive potential.
rent clinical use have been conclusively shown to be teratogenic. Women with chronic illnesses who require drug treatment
In general, drugs that have been used extensively in pregnant should be offered specialist counselling before conception, and
women without apparent problems are recommended in prefer- the options explored to reduce or change drug therapy to a safer
ence to new drugs for which there is less experience of use. For agent if necessary. It is also important to note that many preg-
example, methyldopa is used rarely to treat hypertension in the nant women become less adherent to their drug therapy out of
non-pregnant state but has historically been preferred in preg- concern about possible harm to their infant. In many cases, such
nancy because of a long history of safe use (National Institute as asthma, inlammatory bowel disease and epilepsy, inadequate
for Health and Care Excellence [NICE], 2011). However, older treatment of the underlying disease may be more detrimental to
drugs may be less effective in terms of controlling maternal ill- the mother–fetus pair than the drugs used to treat the condition.
ness and are often associated with an increased side-effect risk It is thus essential that women of reproductive potential are given
proile. clear and up-to-date information so that unrealistic fears about
In most cases the decision whether to commence or continue the risks to their baby do not result in unnecessary pregnancy
with a medication in pregnancy will depend on the risk–beneit termination or disease relapse.
analysis for that speciic mother–infant pair. A frequent error All women who are planning a pregnancy should be offered
made by health professionals was to apply the recently discon- general advice to minimise the risk of congenital anomalies. This
tinued US Food and Drug Administration (FDA) pregnancy risk includes avoidance of recreational drugs, ‘natural’ or herbal rem-
categories (A [no demonstrable risk]; B, C, D and X [teratogenic edies, alcohol, smoking and vitamin A products; minimisation of
agents that are considered to be completely contraindicated in caffeine consumption; and beginning daily supplementation with
pregnancy]) when considering whether to prescribe a drug in at least 400 micrograms of folic acid to reduce the risk of neural
pregnancy. It is now widely accepted that these categories were tube defects (NICE, 2014). It is recommended that the daily dose
oversimpliied and of little practical help in a clinical setting. of folic acid be increased to around 5 mg daily in women who
More detailed information sheets containing a summary of the have epilepsy (NICE, 2016) or who have had a previous child
fetal risks and the additional maternal factors that need to be taken with a neural tube defect (NICE, 2014). Some infectious diseases
into consideration are now being used. Consideration also needs may carry important fetal consequences if contracted during
to be given to the quality and quantity of both human and animal pregnancy. For example, rubella infection in the irst 20 weeks
data, the study design, dose exposure and any reported congenital of pregnancy is associated with an increased risk of miscarriage
malformations and/or adverse events (see http://www.fda.gov for and a syndrome comprising problems such as deafness, cardiac
up-to-date information). defects and mental retardation in more than 20% of pregnancies.
It is worth noting that standard literature sources often contain Women who lack immunity to rubella should be immunised prior
unhelpful information such as ‘do not use in pregnancy unless to conception.␣ 825
48 THERAPEUTICS

Post-conception advice
for this period. Beneits of breastfeeding include protection of
It is important to draw distinction between advice given to women the infant against infectious diseases (gastro-intestinal, upper
pre-conceptually and that provided to a pregnant woman who has and lower respiratory tract, otitis media and urinary tract) and
already been exposed to a drug. In the former setting, it may be rec- reduction in rates of obesity, types 1 and 2 diabetes, inlamma-
ommended that an alternative preparation be considered or that a tory bowel disease, childhood leukaemia, asthma, coeliac disease
drug treatment be stopped where clinically appropriate. This advice and atopic disease (Eidelman and Schanler, 2012; Kramer and
often hinges on the lack of deinitive safety data and does not auto- Kakuma, 2012). There is evidence to show that adults who were
matically translate to exposure to that drug in pregnancy being an breastfed as infants have a predisposition to lower blood pres-
indication for discontinuing the drug, additional fetal monitoring sure and cholesterol levels (Horta et al., 2007). Maternal beneits
or termination of the pregnancy on the basis of the exposure. Any include reduced risk of development of postpartum depression,
change to the woman’s medication should be based on a careful and cardiovascular disease, diabetes (Eidelman and Schanler, 2012),
individual risk assessment, and include a discussion with the woman premenopausal breast and ovarian cancer, and delayed resump-
to provide her with accurate, up-to-date, evidence-based advice. In tion of menstrual cycle (Department of Health, 2003; Eidelman
many such cases the woman can be reassured that a healthy baby is and Schanler, 2012). Breastfeeding strengthens the mother–
the most likely outcome, or where appropriate be offered additional infant bond.
prenatal investigation to screen for congenital malformation where There are few contraindications to breastfeeding; these include
the risk to the fetus is considered to be signiicant.␣ galactosaemia in an infant and active untreated tuberculosis and
HIV infection in developed countries. However, in the devel-
oping world, where mortality is increased in non-breastfeeding
Teratology information services and pregnancy
infants from a combination of malnutrition and infectious dis-
registries
eases, breastfeeding may outweigh the risk of acquiring HIV
It is dificult to keep up to date with the published literature. infection from human milk.
There is an increasing need for summary documents that include Reasons for early discontinuation of breastfeeding include prob-
and critically appraise all available data, and which enable health- lems latching, sore/cracked nipples, engorgement, inconvenience
care providers to have a balanced and informed discussion with (return to work, feeding in public, lifestyle choices), concerns
patients regarding the risks and beneits of a certain therapy in about inadequate lactation and nutritional adequacy, personal ill-
pregnancy. This is evidenced by the ongoing debate surrounding ness and medication, and infant factors (Li et al., 2008).
the teratogenic potential of various antidepressants with conlict- Breastfeeding mothers frequently require treatment with pre-
ing opinion even amongst experts in the ield. scription medicines or may self-medicate with over-the-counter
Teratology Information Services have been established in sev- preparations, nutritional supplements or herbal medicines. It is
eral countries across the world and provide evidence-based, up- important for health professionals to understand the principles of
to-date information and individual case-based risk assessments. safe use of medications during lactation to provide appropriate
In addition to reviewing published literature on drugs, teratology advice.
services also have access to specialist online databases and dis- There are two main goals to consider when formulating advice
cussion forums. A number routinely collect pregnancy outcome for nursing mothers. These are to protect the infant from mater-
data on the women about whom they receive an enquiry, to enable nal drug-related adverse effects and to allow, whenever possible,
surveillance for potential teratogens. necessary maternal medication (Berlin et al., 2009).
For some new drugs, pregnancy registries have been initiated
that record all reported drug exposures and follow up the out-
Transfer of drugs into breast milk
come of the pregnancy. These registries are cumulative and work
on the basis that speciic anomalies would be identiied relatively Most drugs pass into breast milk to some degree, although
quickly, and that there will eventually be suficient statistical transfer is usually low. The drug ‘dose’ ingested by the infant
power to detect the magnitude of any increased risk relative to the via breast milk only rarely causes adverse effects. Examples of
general population. These registries may be held by a Teratology adverse effects observed in breastfed infants exposed to com-
Information Service or by independent groups with an interest in monly used medicines via breast milk are given in Table 48.4.
a deined area. The 2009 A/H1N1 inluenza pandemic provided These examples are of reports that are considered to have a high
an example of teratology services across the globe responding probability of a direct association between infant exposure in
to the urgent need for safety data by establishing registries on breast milk and maternal ingestion, although not all of these are
antiviral and pandemic vaccine exposure during the pandemic.␣ proven to be directly caused by the drug ingested via breast milk.
There are a signiicant number of reports of possible adverse reac-
tions in breastfed infants due to maternal medication in which the
Drugs in lactation association is less clear, unsubstantiated or anecdotal (Anderson
et al., 2003, 2016).
Breast milk is the best form of nutrition for young infants. It pro- Almost all drugs enter milk by passive diffusion of unionised,
vides all the energy and nutrients required for the irst 6 months of unbound drug through the lipid membranes of the alveolar cells
life. The World Health Organization (WHO, 2001) and the United of the breast, according to the pH partitioning theory. Several
Nations Children’s Fund recommend exclusive breastfeeding factors inluence the rate and extent of passive diffusion.
826
DRUGS IN PREGNANCY AND LACTATION 48
Milk differs from blood in that it has a lower pH (7.2 vs. 7.4),
Table 48.4 Adverse reactions reported in breastfed infants
less buffering capacity and higher fat content. The following drug
(Anderson et al., 2003, 2016)
parameters affect the extent of transfer into milk:
Drug Adverse reaction • pKa: This is a measure of the fraction of the drug that is ion-
ised at a given pH, for example, physiological pH. Highly
Acebutolol Hypotension, bradycardia, tachypnoea ionised drugs tend not to concentrate in milk. For basic drugs
(e.g. erythromycin), a greater fraction will be ionised at an
Aspirin Metabolic acidosis, thrombocytopenic
purpura acidic pH, so the milk compartment will tend to ‘trap’ weak
bases. In contrast, acidic drugs (e.g. penicillins) are more ion-
Atenolol Bradycardia, cyanosis, hypotension ised at higher pH values and will be ‘trapped’ in the plasma
compartment. Drugs with higher pKa values generally have
Ciprofloxacin Pseudomembranous colitis higher milk/plasma ratios.
• Protein binding: Drugs that are highly bound to plasma pro-
Codeine Apnoea, bradycardia, death
teins (e.g. warfarin) are likely to be relatively retained in
Cyclophosphamide Neutropenia, thrombocytopenia maternal plasma because there is a lower total protein content
in the milk. High protein binding essentially restricts the drug
Dapsone Haemolytic anaemia to the plasma compartment because only the free fraction of
the drug crosses the biological membrane. Milk concentra-
Diazepam Lethargy, sedation, poor suckling tions of highly protein-bound drugs are usually low.
• Lipophilicity: The alveolar epithelium of the breast is a lipid
Doxepin Sedation and respiratory arrest
barrier. Transfer of water-soluble drugs and ions is inhibited
Erythromycin Pyloric stenosis by this barrier. CNS active drugs usually have the characteris-
tics required to pass into milk.
Fluoxetine Colic, irritability, sedation • Molecular weight: Drugs with low molecular weights (<200)
readily pass into milk through small pores in the cell walls of
Indometacin Seizures alveolar cells. Drugs with higher molecular weights cross cell
membranes by dissolving in the lipid layer which may sub-
Lamotrigine Apnoea, cyanosis
stantially reduce milk concentrations. Proteins such as heparin
Lithium Cyanosis, lethargy, T-wave abnormalities, or insulin with very large molecular weights greater than 6000
thyroid-stimulating hormone elevation are virtually excluded from milk.
The proile of a drug that passes minimally into milk would
Mesalazine Watery diarrhoea therefore be an acidic drug that is highly protein bound and has
low-to-moderate lipophilicity (e.g. most NSAIDs). In contrast, a
Naproxen Prolonged bleeding, haemorrhage, weakly basic drug that has low plasma protein binding and is rel-
anaemia
atively lipophilic will achieve higher concentrations in the milk
Oxycodone Sedation, hypothermia, poor feeding compartment (e.g. sotalol).
In the irst few days of life, large gaps exist between the
Paroxetine Agitation, feeding difficulty alveolar cells. These permit enhanced passage of drugs into
milk. By the end of the irst week these gaps close under the
Phenindione Postoperative haemorrhage inluence of prolactin (Lawrence and Lawrence, 2016). There
is greater passage of drugs into colostrum (early milk) than in
Phenytoin Methaemoglobinaemia
mature milk because the former contains more protein and less
Sulfasalazine Bloody diarrhoea fat. There is also some variation in fat and protein content of
milk between the beginning and end of a feed, but these changes
Theophylline Irritability, poor sleep have less inluence on drug passage than the physicochemical
properties of the drug.␣
Topiramate Diarrhoea

Yellow fever vaccine Fever, irritability, convulsions, meningo- Milk/plasma concentration ratio
encephalitis
Several methods have been proposed to determine the amount
of drug transferred to breast milk. The milk/plasma (M/P) ratio
is often used as a measure of the extent of drug transfer into
These include maternal plasma level, physiological differences breast milk. It is usually obtained from case reports or small
between plasma and milk, and the physicochemical properties clinical studies and may be based on paired concentrations or
of the drug. Differences in composition between blood and milk full area under the concentration–time curve (AUC) analysis.
determine which physicochemical characteristics inluence M/P ratios that are based on a pair of milk and plasma sam-
diffusion. ples collected simultaneously may be inaccurate because they
827
48 THERAPEUTICS

a 20
75

Cumulative infant dose


Concentration

50
10
b
c
25
Milk
0
Plasma 0 4 8 12 16 20 24
0 Time (hours)
0 6 12 18 24
Time (hours) Fig. 48.2 The cumulative dose received by the infant is plotted
Fig. 48.1 Diagrammatic representation of a drug concentration– against time. The arrows represent the maternal dose times. This type
time profile in the maternal plasma and milk phases after a single oral of study is undertaken by the mother expressing all the milk, from
dose. The points a, b and c illustrate simultaneous sampling times both breasts, at usual feeding times. An aliquot of milk is taken and
from both phases. Sampling at (a) would yield an estimated milk/ assayed for the drug. The volume of milk is measured, and the total
plasma (M/P) ratio of 0.2, at (b) a value of 1.0, and at (c) a value of amount of drug at each time is calculated as the product of concen-
9.3. All can be misleading. The true M/P ratio calculated by the area tration and volume. This type of study must be undertaken at steady
under the concentration–time curve method is 1.2 when extrapolated state.
to infinity.

When using quantitative data from milk analyses, the most


assume that the concentrations of drug in milk and plasma are accurate estimation of the infant ‘dose’ is from studies in which
in parallel, which may not be the case. It is better to collect the milk is collected over a complete dose interval at steady state
multiple samples of plasma and milk across a dosing interval, and the total dose is calculated (Fig. 48.2). Unfortunately, these
or until the drug is cleared from both phases after a single dose, studies are seldom performed. Therefore, information must be
for determination of a M/P ratio based on the respective AUCs obtained from less than ideal conditions.
(M/PAUC). Fig. 48.1 demonstrates the markedly different esti- If the M/P ratio is known from published studies, the likely
mates of M/P ratio that can be obtained via both sampling meth- infant dose (Dinf) can be calculated as follows, with some
ods. The true M/P ratio may vary signiicantly during the same assumptions:
episode of breastfeeding. = × ×
If human-derived M/P ratios are lacking for a particular drug,
it may be possible to predict the extent of transfer using known where Cpmat is the average maternal plasma concentration, and
physicochemical properties, for example, pKa, and a published the M/PAUC should be used in preference to a ratio based on paired
predictive model (Atkinson and Begg, 1990; Begg et al., 1992; concentrations when available. The volume of milk ingested
Zhao et al., 2006). M/P ratios obtained from animal studies (Vmilk) is not known but is generally assumed to be around 150 mL
should not be used for clinical decision making because they may of milk per kilogram of body weight per day. The above equation
not correlate well with human M/P ratios. simpliies if the actual milk concentration data are available:
Studies in humans demonstrate that most drugs have an = ×
M/P ratio less than 1.0, with the range of reported ratios
being from around 0.1–5.0. It is often thought that drugs with The likely infant plasma drug concentration (Cpinf) can be cal-
high M/P ratios (e.g. 5.0) are unsafe because the concentra- culated by:
tion in milk exceeds that in plasma, while those with low = ×
ratios (<1.0) are believed to be safe. This is not always the
case because the M/P ratio often fails to correlate with the where F is oral availability and Clinf is the infant clearance.
‘dose’ of drug the infant ingests via milk. The amount of drug Unfortunately, neither F nor Clinf is known accurately for infants,
transferred into milk is principally determined by the mater- so estimation of the likely steady-state average plasma drug con-
nal plasma level. Thus, even where the M/P ratio is high, if centration will be very approximate. Weight-adjusted Clinf values
the maternal plasma level is low, drug transfer is still low. (i.e. L/h/kg) are often signiicantly less than adult values in the
Therefore, the M/P ratio must never be used as the sole mea- early stages of life (Table 48.5).
sure of drug safety in breastfeeding. However, it can be used Given the dificulty in estimating infant plasma drug concen-
to estimate the ‘dose’ ingested via milk, which is a better pre- trations, the relative infant dose (RID), for example, compared
dictor of safety.␣ with a therapeutic infant dose, is often used as a surrogate of
exposure. To give some basis for comparison, the likely infant
dose from milk can be compared with an infant therapeutic dose.
Calculating the infant ‘dose’ ingested via milk
This is reasonable for drugs such as paracetamol that are usually
Infant plasma drug levels are the most accurate indicator of drug administered to infants, but is unsuitable for drugs such as anti-
828 exposure, but these are seldom available. depressants that are not. In the absence of a clearly deined range
DRUGS IN PREGNANCY AND LACTATION 48
Table 48.5 Approximate drug clearance by age as percentage Box 48.2 Factors that affect infant risk from maternal therapy
of maternal value (Begg, 2000)
• Drug adverse reaction profile
Post-conceptual age (weeks) Approximate drug clearance (%) • Relative infant dose
• Oral bioavailability
24–28 5 • Active metabolites
• Half-life
28–34 10 • Gestational age of the infant
• Full maternal drug regimen
34–40 33 • Pharmacogenetic factors

40–44 50

44–68 66 Assessing the risk to the infant


Many factors must be considered when assessing the risk of
>68 100
maternal drug therapy to the breastfeeding infant (Box 48.2).

Pharmacokinetic and pharmacodynamic factors


of infant doses, the weight-adjusted maternal dose expressed
as a percentage (% dose) is widely used to indicate infant drug Inherent toxicity of the drug will be a main factor in determin-
exposure. ing infant safety. Thus, antineoplastic drugs, radionuclides and
( ) iodine-containing compounds would be of concern. Multiple
( )= × maternal therapy with drugs with similar side-effect proiles,
( )
for example, psychotropic drugs or anticonvulsants, is likely to
Dinf represents dose in infant via milk, and Dmat represents dose increase the risk to the infant. Oral bioavailability is an indicator
in mother. of the drug’s ability to reach the systemic circulation after oral
For the great majority of drugs, this calculation yields infant administration. Drugs with a low oral bioavailability are either
doses in the order of 0.1–5.0% of the weight-adjusted maternal poorly absorbed from the gastro-intestinal tract, broken down in
dose expressed as a percentage (% dose). It is generally thought the gut or undergo extensive ‘irst-pass’ metabolism in the liver
that RID values of less than 10% of the maternal dose are prob- before entering plasma.
ably safe. However, the inherent toxicity of the drug should be The presence of active metabolites (e.g. desmethyldiazepam)
taken into account when using this igure. may prolong infant drug exposure and lead to drug accumulation,
To calculate the daily infant drug intake via milk, the standard especially where drug clearance is low, such as in the neonatal
milk intake of 150 mL/kg/day is multiplied by the concentration period. Similarly, drugs with long half-lives (e.g. luoxetine) may
of the medication in milk. Some authors use the peak concentra- be problematic at this time. Drug clearance by the infant does not
tion in milk to indicate the maximum infant intake. reach adult values until 6–7 months (see Table 48.5). A premature
infant of 30 weeks’ gestational age has a drug clearance value of
Estimated daily infant intake = drug concentration in milk about 10% of the maternal value. It is important to distinguish
micrograms/day micrograms/L × 0. 15 × between gestational age and time after delivery. A 2-week-old
infant weight kg ␣ infant born at 28 weeks will have a gestational age of 30 weeks.
The maternal drug regimen can affect infant risk. Single doses
or short courses seldom present problems, whereas chronic ther-
Variability
apy can be problematic. Topical or inhalation therapy usually
To further complicate matters, there will be signiicant variabil- results in much lower plasma drug levels and, therefore, lower
ity between and within individuals in the values used to estimate passage into milk. Multiple maternal medications increase the
infant exposure (i.e. Dinf, F, Clinf, where Dinf is itself a function of risk to the infant.␣
the estimated parameters Cpmat, M/P and volume of milk). Some
of this variability will change over time because of developing
Pharmacogenetic factors
organ function in the maturing baby, and some will be unex-
plained variability. In addition to this pharmacokinetic variabil- Recently, attention has been focused on the possible role of phar-
ity, there will be variability in response of the infant to any given macogenetic factors in affecting the safety of breastfed infants
concentration of the drug. It is fortunate that most drugs seem to exposed to drugs via milk (Madadi et al., 2009). Sedation (and
fall readily into a safe range (RID <10%) based on expected expo- one death) occurred in infants of mothers with rare genotypes of
sure. However, care should be taken when using these values to cytochrome P450 CYP2D6. leading to ultrarapid metabolism of
assess drug safety in lactation, when variability in the estimates of codeine to morphine. The incidence of these genotypes varies
the parameters used may impact on the accuracy of prediction of amongst different populations. The overall percentage of Western
their safety. This is especially true for those circumstances when Europeans with the CYP2D6 ultrarapid metaboliser phenotype is
initial estimates of these parameters are less precise, for example, 5.4% (Ingelman-Sundberg, 2005). Higher percentages have been
in neonates.␣ reported in populations from northeast Africa and the Middle East.␣ 829
48 THERAPEUTICS

Reducing risk to the breastfed infant Box 48.3 Measures to ensure the safety of the breastfed infant

A number of strategies may be adopted to reduce the risk of drug-


• Avoid unnecessary maternal drug use.
related side effects in the breastfed infant. One technique that has • Seek professional advice on the suitability of over-the-counter
been recommended for reducing infant exposure is to give the mater- products.
nal dose immediately after the infant has been fed with the aim of • Avoid herbal products because of lack of data to support
avoiding feeding at peak milk concentrations. However, this is often safe use.
impractical, especially where young infants are feeding frequently • Assess the risk/benefit ratio for both mother and infant.
up to 2 hourly. In addition, accurate data on times of peak levels in • Use the minimum clinically effective dose for the shortest
possible time.
milk are often unavailable, and it cannot be assumed that times of • Review maternal therapy towards the end of pregnancy.
peak milk levels mirror those in plasma. This technique should be • Choose a drug regimen and route of administration which
used selectively, where the drug has a short half-life and where peak presents the minimum of drug to the infant via breast milk.
and trough levels are predictable (e.g. antibiotics, anaesthetics). • Avoid drugs known to cause serious toxicity in adults or
Where a single dose of a drug known to be hazardous is given children.
to a breastfeeding mother, for example, a radiopharmaceutical, • Monitor the infant for unusual signs or symptoms.
• Avoid new drugs if there is a therapeutic equivalent with data
it will usually be possible to resume breastfeeding after a suit-
to support safe use in lactation.
able washout period, calculated as ive times the half-life. Where • Recognise risk factors, e.g. prematurity, infant
the half-life is very long, the washout period necessary to avoid morbidity and multiple maternal medications.
hazardous exposure to the infant may exceed the period of sus-
tainable lactation.
Breastfeeding mothers should be advised to avoid self-medi-
cation. Where drug use is clearly indicated, the lowest effective Table 48.6 Examples of commonly used drugs thought to be
dose should be used for the shortest possible time. Use of topical safe for use in breastfeeding mothers of full-term,
healthy infants
therapy, such as eye/nasal drops for hay fever, would reduce drug
exposure in comparison with oral antihistamines. Drug groups Individual drugs
The maternal regimen should be simpliied wherever possible.
A review of therapy before delivery will help reduce risks to the Antacids Cetirizine
neonate. New drugs are best avoided if a therapeutic equivalent is Bulk laxatives Chloroquine/proguanil
available for which data on safe use in lactation exists. All infants Cephalosporins Clotrimazole
Contrast media Cromoglycate
exposed to drugs via breast milk should be monitored for any
Inhaled medications (e.g. Diclofenac
untoward effects. Measures to ensure the safety of the breastfed salbutamol, beclometasone) Fluconazole
infant are summarised in Box 48.3. Insulins Heparin (including low
Some commonly used drugs thought to be safe to use in moth- Penicillins molecular weight)
ers of full-term, healthy infants are listed in Table 48.6.␣ Progestogen-only contracep- Ibuprofen
tives Iron supplements
Topical formulations (except Loratadine
Special situations when applied to breast) Lactulose
Vaccines (except yellow fever) Levothyroxine
Neonates and premature infants Vitamins (except high-dose Nystatin
A and D) Omeprazole
Neonates and premature infants are at greater risk of develop- Paracetamol
ment of adverse effects after exposure to drugs via breast milk. Paroxetine/sertraline
Gastric emptying time is signiicantly prolonged and, in some Warfarin
cases, may alter absorption kinetics. Protein binding is decreased This table is to be used as a guide only. Expert advice is required when the
and values for total body water are higher than for adults. Renal maternal dose is high and if the infant is premature, has renal or hepatic
function is limited because the kidney is anatomically and func- disease, or has glucose-6-phosphate dehydrogenase deficiency.
tionally immature (van den Anket, 1996). The neonate’s capacity
to conjugate drugs in the liver is often deicient.␣
Breastfeeding should be avoided or a safer alternative chosen
if the infant has a known or suspected G6PD deiciency and
Glucose-6-phosphate dehydrogenase deficiency
the mother is taking drugs that have been reported to cause
Infants with glucose-6-phosphate dehydrogenase (G6PD) oxidative stress (e.g. nitrofurantoin, dapsone).␣
deiciency are susceptible to adverse effects even when only
small amounts of certain drugs are present in milk. G6PD is Allergy
an enzyme present in erythrocytes that is responsible for main-
taining the antioxidant compound glutathione in its active The theoretical possibility exists for an allergic reaction in an
form. Deiciency of this enzyme makes the erythrocyte more infant exposed to a drug in breast milk. Even minimal exposure
susceptible to oxidative stress, resulting in haemolysis. Only to the drug could cause an allergic response. However, in prac-
830 small amounts of drug are needed to cause such a reaction. tice, such reactions are rare, and only if the infant has already
DRUGS IN PREGNANCY AND LACTATION 48
experienced an allergic reaction to a particular drug should an unlicensed (‘off-label’) indication. However, it is appropriate
maternal use be discouraged or breastfeeding avoided.␣ for the recommendations from the review to apply to its use as
a galactagogue. Because there are limited alternative options for
the stimulation of lactation, the use of domperidone can be con-
Recreational drug use
sidered provided non-pharmacological options have been unsuc-
Accurate background data of maternal use of recreational drugs cessful. A maternal dosage of 30 mg daily for 1 week should not
may be dificult to obtain. Usage may be chronic or sporadic. usually be exceeded, and it should not be used if the mother or
The role of the healthcare professional in ensuring the safety of infant:
the breastfed infant is important, and the advice should be that • has conditions where cardiac conduction is, or could be,
substances such as cannabis, LSD and cocaine should be avoided impaired;
whilst breastfeeding. • has underlying cardiac diseases such as congestive heart
Signiicant amounts of alcohol pass into milk, although it is failure;
not normally harmful to the infant if the quantity and duration of • is receiving other medications known to prolong QT interval
intake are limited. The occasional consumption of a small alco- or are potent CYP3A4 inhibitors;
holic beverage is acceptable if breastfeeding is avoided for about • has severe hepatic impairment.
2 hours after the drink. Chronic or heavy consumers of alcohol If treatment is still required after 1 week, the risks of continuing
should not breastfeed. High intakes of alcohol decrease milk let- domperidone must be considered carefully. Alternatively, meto-
down and disrupt nursing until maternal levels decrease. Heavy clopramide can be used; however, only for up to 5 days because
maternal use may cause infant sedation, luid retention and hor- of the risk of neurological side effects (European Medicines
mone imbalances in breastfed infants. Agency, 2013).
Nicotine has been suggested to decrease basal prolactin produc- Other drugs may affect lactation as an unwanted side effect, for
tion, although effects may be variable. Ideally mothers should be example, diuretics causing dehydration. When these are used on a
encouraged not to smoke whilst breastfeeding. Nicotine and its long-term basis, infant weight gain should be monitored.␣
metabolite, cotinine, are both present in milk. Undertaking smok-
ing cessation with a nicotine patch is a safer option than continued
smoking. Whilst transdermal nicotine patches produce a sustained Case studies
lower nicotine plasma level, nicotine gums produce large varia-
tions in peak levels. A 2- to 3-hour washout period is recom-
mended before breastfeeding after maternal use of a nicotine gum.
Case 48.1
Caffeine appears in breast milk rapidly after maternal intake. Ms LP is 6 weeks’ pregnant and has been diagnosed with depres-
Fussiness, jitteriness and poor sleep patterns have been reported sion that warrants pharmacological intervention. She wishes to
in infants of mothers with very high caffeine intakes equivalent recommence venlafaxine, which has been helpful in the past. Ms
to about 10 or more cups of coffee daily. Preterm and newborn LP is also anxious that the ethanol she consumed around the time
infants metabolise caffeine very slowly and are at increased risk of conception may have harmed her baby.
of adverse effects.␣

Questions
Drug effects on lactation
1. What are the safest antidepressants in the first trimester of
Drugs that affect dopamine activity are the main cause of pregnancy?
effects on milk production, mainly mediated by effects on 2. Is it reasonable for Ms LP to commence venlafaxine?
prolactin. Early postpartum use of oestrogens may reduce the 3. Is there any risk from the ethanol ingestion?␣
volume of milk, but the effect is variable and depends on the
dose and the individual response. Progestogen only contracep-
Answers
tives are preferred.
Drugs may occasionally be used therapeutically for their effect 1. The concept of ‘safe’ needs to take into account the risks of
on lactation. Dopamine agonists such as cabergoline decrease inadequate treatment of the mother, as well as those to the fetus
milk production and this effect may be utilised, for example, after through in utero exposure to a particular antidepressant. Choice
of antidepressant in pregnancy will therefore depend on the
an infant death. Dopamine agonists should not be used routinely
severity of the maternal depression, the presence of associated
for relief of the symptoms of postpartum pain or engorgement symptoms such as anxiety or obsessive-compulsive behaviors,
that can be managed with simple analgesics or breast support. availability and suitability of non-pharmaceutical interventions
Dopamine antagonists such as domperidone may be used in and, finally, any prior treatment regimens that have been suc-
cases of inadequate lactation that have not responded to irst-line cessfully or unsuccessfully used. SSRIs are currently the most
methods such as improved technique or milk expression by hand commonly prescribed antidepressant group and the most stud-
or pump. However, a European and UK regulatory review has ied in pregnancy. However, human pregnancy data will vary for
individual SSRIs. Although not without risk to the fetus, SSRIs are
recommended restricted use of domperidone because of the risk
generally prescribed as first-line therapy in preference to tricy-
of cardiac adverse effects (Medicines and Healthcare Products clic antidepressants which carry a higher risk of maternal toxicity,
Regulatory Agency, 2014). The use of domperidone to enhance especially in overdose, and which are less well studied in human
lactation is not speciically covered by the review because it is pregnancy. 831
48 THERAPEUTICS

2. Experience with the use of venlafaxine and many other antide- dose’ has been defined, if this patient needs to remain taking
pressants (e.g. moclobemide) in pregnancy is limited. However, valproate, it may be possible to gradually reduce her daily dose
in some instances it may be necessary to use an agent such as under specialist review. General preconception advice could be
venlafaxine. For example, if the mother had a history of severe given, that is, commence folic acid, optimise weight and gen-
depression that did not respond to multiple trials of other anti- eral health, etc. It is important that Ms AD understands that
depressants, then venlafaxine may be considered the most folic acid 5 mg daily is advised for all women with epilepsy
appropriate choice of antidepressant for that woman. In this who are planning a pregnancy (NICE, 2016), but that at pres-
case any potential risks associated with venlafaxine are likely to ent there is no evidence that it prevents or reduces the risk
be less than those associated with inadequately treated depres- of birth defects or neurodevelopmental impairment caused by
sion. Discussion of the available data and its limitations with Ms valproate.␣
LP is, however, necessary to enable her to make an informed
decision.
3. Ethanol is a human teratogen. Fetal alcohol spectrum disorders
are characterised by low birth weight, facial dysmorphogenesis
Case 48.3
and/or delayed development. A ‘safe limit’ for alcohol con- A mother, Ms TF, with a history of depression has relapsed and
sumption in pregnancy has not been defined, and best prac- requires treatment with an antidepressant. Ms TF is fully breast-
tice is therefore to avoid all alcohol exposure in pregnancy. In feeding a 2-week old, full-term, fit and healthy baby. In the past
this case Ms LP ingested ethanol at the time of conception. She she has taken fluoxetine, but stopped this when she found out she
should be reassured that this is regarded as a relatively low-risk was pregnant.
period for fetal harm, but that further ethanol ingestion should
be avoided.␣
Questions
Case 48.2 1. What is the antidepressant of choice during breastfeeding?
2. Ms TF has responded well to fluoxetine previously, so can this be
A 30-year-old woman with epilepsy, Ms AD, is currently taking val- used during breastfeeding?
proic acid 1500 mg daily. She wishes to conceive but is concerned 3. What are the potential risks of using fluoxetine?␣
about the possibility of birth defects caused by valproate expo-
sure in pregnancy. Ms AD’s seizures have been difficult to control
with alternative anticonvulsants. Answers
1. Sertraline and paroxetine are the preferred SSRIs for use during
Questions breastfeeding because they have shorter half-lives, lower pas-
sage into milk and a larger evidence base compared with the
1. What are the risks associated with valproate treatment in other SSRIs. However, it is important that Ms TF has an antide-
pregnancy? pressant that works for her, so choice of agent should primarily
2. What advice should Ms AD be given?␣ be based on suitability for the patient rather than safety during
breastfeeding.
2. If a woman has been successfully treated with an SSRI during
Answers pregnancy and requires continued therapy in the postnatal period
there is no need to change therapy, provided the infant is full
1. Valproate is a human teratogen which has been associated with
term, fit and healthy, and can be adequately monitored. Although
both structural and lasting neurodevelopmental effects. These
fluoxetine is not a preferred SSRI during lactation, due to its long
may include neural tube defects, craniofacial anomalies such as
half-life, because it has proved effective for Ms TF in the past,
cleft lip and palate, heart defects, autism, attention deficit hyper-
the balance of benefit to risk will favour use of fluoxetine in this
activity disorder and learning difficulties. Recommendations
mother.
from a European-wide review in 2014 (European Medicines
3. Because fluoxetine has a very long half-life there is a risk of accu-
Agency, 2014) advise against valproate use in female children, in
mulation and, therefore, adverse effects in the breastfed baby. The
female adolescents, in women of childbearing potential and in
baby should be monitored for drowsiness, poor feeding, irritability
pregnant women unless other treatments are ineffective or not
and behavioural effects.␣
tolerated. Women of childbearing potential in whom valproate
treatment is considered necessary for the treatment of epilepsy
are advised to remain on effective contraception during treat-
ment. The MHRA has produced a number of guidance docu-
Case 48.4
ments for healthcare professionals in the UK, including an openly A mother, Ms CG, is breastfeeding her 20-day-old, full-term, fit
accessible toolkit for women (https://www.gov.uk/government/- and healthy baby. She has postpartum hypertension and has been
publications/toolkit-on-the-risks-of-valproate-medicines-in- started on labetalol.
female-patients). Labetalol does not control Ms CG’s hypertension and she is
2. It is imperative that Ms AD be advised to request referral to, started on nifedipine.
or review by, a neurologist, ideally with expertise in the treat-
ment of women with epilepsy who become pregnant, and that
she continues to use effective contraception until the risks Questions
and options available to her have been fully reviewed and
discussed. New anticonvulsant therapies that do not appear 1. Comment on the suitability of labetalol as a suitable β-blocker for
to carry the same risks as valproate may have become avail- use during breastfeeding.
able since Ms AD was last reviewed. Valproate teratogenicity 2. Are there any monitoring requirements?
appears to increase with increasing dose. Although no ‘safe 3. Can Ms CG still continue to breastfeed?␣
832
DRUGS IN PREGNANCY AND LACTATION 48
She was given a course of fluconazole 200 mg daily for 10 days
Answers for a presumed candidal infection but expressed concern that the
medication might affect the infant.
1. In general, β-blockers that are considered to pose less risk to a
breastfeeding infant have, or are predicted to have, lower levels
in breast milk, due to a high degree of plasma protein binding,
Questions
low lipid solubility and a short half-life, and relatively low renal
excretion. The risks of available β-blockers vary widely due to 1. Is this regimen safe to use in lactation?
these features. Although propranolol is considered to be the 2. What other therapeutic measures should be taken?␣
β-blocker of choice in breastfeeding, because clinical properties
of β-blockers vary widely, propranolol may not always be a suitable
clinical choice. Metoprolol and labetalol are also considered to Answers
pose low risk. Labetalol would be suitable to use in this situation,
with appropriate monitoring. 1. Topical miconazole is effective in treating superficial candidal infec-
2. The breastfed infant should be monitored for signs of β-blockade, tions, but oral therapy with fluconazole is needed when the infection
especially bradycardia. Also consider monitoring for hypotension spreads to the milk ducts. Fluconazole, after a 200 mg oral dose,
and hypoglycaemia. produces levels in breast milk similar to those found in maternal
3. Because there will be low levels of nifedipine in breastmilk, plasma. Fluconazole is recommended for use in the treatment of
amounts ingested by the baby will be small. Nifedipine, includ- neonates with fungal infections at a dose starting at 3 mg/kg every
ing modified-release preparations, is considered compatible 72 hours. The calculated dose of fluconazole ingested by an infant
with breastfeeding. Therefore, Ms CG can continue to breast- feeding at times of peak milk levels of fluconazole would be approx-
fed, but the risk of additive adverse effects in the baby must be imately 0.6 mg/kg/day, which is 60% of the neonatal dose and 20%
remembered.␣ of the dose for infants aged ≥1 month. Ms SV may be reassured that
the amount of drug reaching the infant via milk is a fraction of the
dose that would be used to treat an infection in the infant.
Case 48.5 2. Candidal infections are easily passed between the mother and the
infant, and both should receive treatment. Infants can be treated
A breastfeeding mother, Ms SV, returned to see her midwife with nystatin suspension or oral miconazole gel. Miconazole gel is
4 weeks after delivery of a full-term, healthy infant. She is com- not licensed for use in children younger than 4 months because of
plaining of bilateral nipple pain during and after breastfeeding, a the small risk of choking in young babies; this risk can be reduced
problem that was constant for the past 4 days. Ms SV was advised by using smaller quantities of the gel, applying the gel with a clean
to use miconazole cream 2% to the nipples after each feed. This finger rather than a spoon and being careful not to touch the back
provided initial relief, but symptoms returned after a few days. of the infant’s throat when applying the gel.

Acknowledgements
The contributions of Stephen B. Duffull, Sharon J. Gardiner, David K. Woods and Elena Grant to previous versions of this chapter which
appeared in earlier editions of the textbook are gratefully acknowledged.

References
Anderson, G., 2005. Pregnancy-induced changes in pharmacokinetics: a Department of Health, 2003. Infant feeding recommendation. Department of
mechanistic-based approach. Clin. Pharmacokinet. 44, 989–1008. Health, London. Available at: http://webarchive.nationalarchives.gov.uk/
Anderson, P.O., Pochop, S.L., Manoguerra, A.S., 2003. Adverse drug reactions 20130107105354/http:/www.dh.gov.uk/prod_consum_dh/groups/
in breastfed infants: less than imagined. Clin. Pediatr. (Phila.) 42, 325–340. dh_digitalassets/@dh/@en/documents/digitalasset/dh_4096999.
Anderson, P.O., Manoguerra, A.S., Pochop, S.L., 2016. A review of adverse pdf.
reactions in infants from medications in breastmilk. Clin. Pediatr. (Phila.) Dunn, P.M., 1964. The possible relationship between the maternal
55, 236–244. administration of sulphamethoxypyridazine and hyperbilirubinaemia
Atkinson, H.C., Begg, J., 1990. Prediction of drug distribution into human milk in the newborn. J. Obstet. Gynaecol. Br. Commonw. 71, 128–131.
from physicochemical characteristics. Clin. Pharmacokinet. 18, 151–167. Eidelman, A.I., Schanler, R.J., 2012. American Academy of Pediatrics –
Begg, E.J., 2000. Clinical Pharmacology Essentials. The Principles Behind section on breastfeeding. Breastfeeding and the use of human milk.
the Prescribing Process. Adis International, Auckland. Pediatrics 129, e827–e841.
Begg, E.J., Atkinson, E.J., Duffull, S.B., 1992. Prospective evaluation of a model European Medicines Agency (EMA), 2013. European Medicines Agency
for the prediction of milk:plasma drug concentrations from physicochemical recommends changes to the use of metoclopramide. Changes aim mainly
characteristics. Br. J. Clin. Pharmacol. 33, 501–505. to reduce the risk of neurological side effects. EMA, London. Available at:
Benke, P.J., 1984. The isotretinoin teratogen syndrome. JAMA 251, 3267–3269. http://www.ema.europa.eu/docs/en_GB/document_library/Press_release/
Berlin, C.M., Paul, I.M., Vesell, E.S., 2009. Safety issues of maternal drug 2013/07/WC500146614.pdf.
therapy during breastfeeding. Clin. Pharmacol. Ther. 85, 20–22. European Medicines Agency (EMA), 2014. CMDh agrees to strengthen
Braun, J.T., Franciosi, R.A., Mastri, A.R., et al., 1984. Isotretinoin dysmorphic warnings on the use of valproate medicines in women and girls.
syndrome. Lancet 1, 506–507. Women to be better informed of risks of valproate use in pregnancy
Breitkreutz, I., Anderson, K.C., 2008. Thalidomide in multiple myeloma: and need for contraception. EMA, London. Available at: http://
clinical trials and aspects of drug metabolism and toxicity. Exp. Opin. www.ema.europa.eu/docs/en_GB/document_library/Press_release/2014/11/
Drug Metab. Toxicol. 4, 973–985. WC500177638.pdf.
Deligiannidis, K.M., Byatt, N., Freeman, M.P., 2014. Pharmacotherapy Headley, J., Northstone, K., Simmons, H., et al., 2004. Medication use
for mood disorders in pregnancy: a review of pharmacokinetic changes during pregnancy: data from the Avon longitudinal study of parents and
and clinical recommendations for therapeutic drug monitoring. J. Clin. children. Eur. J. Clin. Pharmacol. 60, 355–361.
Psychopharmacol. 34 (2), 244–255. 833
48 THERAPEUTICS

Herbst, A.L., Ulfelder, H., Poskanzer, D.C., 1971. Adenocarcinoma of the National Institute for Health and Care Excellence (NICE), 2014. Maternal and
vagina. Association of maternal stilbestrol therapy with tumor appearance child nutrition. Public health guideline (PH11). NICE, London. Available
in young women. N. Engl. J. Med. 15, 878–881. at: https://www.nice.org.uk/guidance/PH11/.
Hernandez-Diaz, S., Werler, M.M., Walker, A.M., et al., 2001. Neural tube National Institute for Health and Care Excellence (NICE), 2016. Epilepsies:
defects in relation to use of folic acid antagonists during pregnancy. Am. diagnosis and management. Clinical guidance (CG137). NICE, London.
J. Epidemiol. 153, 961–968. Available at: https://www.nice.org.uk/guidance/cg137/.
Hill, R.M., 1984. Isotretinoin teratogenicity. Lancet 1, 1465. Newman, C.G., 1985. Teratogen update: clinical aspects of thalidomide
Horta, B.L., Bahl, R., Martines, J.C., et al., 2007. Evidence On The Long- embryopathy: a continuing preoccupation. Teratology 32, 133–144.
Term Effects Of Breastfeeding. WHO, Geneva. Newman, C.G., 1986. The thalidomide syndrome: risks of exposure and
Ingelman-Sundberg, M., 2005. Genetic polymorphisms of cytochrome spectrum of malformations. Clin. Perinatol. 13, 555–573.
P450 2D6 (CYP2D6): clinical consequences, evolutionary aspects and Nulman, I., Berkovitch, M., Klein, J., et al., 1998. Steady-state pharma-
functional diversity. Pharmacogenomics J. 5, 6–13. cokinetics of isotretinoin and its 4-oxo metabolite: implications for fetal
Koren, G., Florescu, A., Costei, A.M., et al., 2006. Non-steroidal anti- safety. J. Clin. Pharmacol. 38, 926–930.
inlammatory drugs during third trimester and the risk of premature Pavek, P., Ceckova, M., Staud, F., 2009. Variation of drug kinetics in pregnancy.
closure of the ductus arteriosus: a meta-analysis. Ann. Pharmacother. 40, Curr. Drug Metab. 10, 520–529.
824–829. Rosa, F.W., 1984. Virilization of the female fetus with maternal danazol
Kramer, M.S., Kakuma, R., 2012. Optimal duration of exclusive breastfeeding. exposure. Am. J. Obstet. Gynecol. 149, 99–100.
Cochrane Database Syst. Rev. 8, CD003517. https://doi.org/10.1002/ Rubin, P.C., Butters, L., McCabe, R., et al., 1987. The inluence of pregnancy
14651858.CD003517.pub2. on drug action; concentration-effect modelling with propranolol. Clin. Sci.
Lawrence, R.A., Lawrence, R.M., 2016. Breastfeeding. A Guide for the 73, 48–52.
Medical Profession, eighth ed. Elsevier, Philadelphia. Schaefer, C., Peters, P., Miller, R.K. (Eds.), 2014. Drugs during Pregnancy
Lenz, W., 1966. Malformations caused by drugs in pregnancy. Am. J. Dis. and Lactation: Treatment Options and Risk Assessment, third ed. Elsevier,
Child. 112, 99–106. Amsterdam.
Lenz, W., 1988. A short history of thalidomide embryopathy. Teratology 38, Sedman, A.B., Kershaw, D.B., Bunchman, T.E., 1995. Recognition and
203–215. management of angiotensin converting enzyme inhibitor fetopathy. Pediatr.
Li, R., Fein, S.B., Chen, J., et al., 2008. Why mothers stop breastfeeding: Nephrol. 9, 382–385.
mothers’ self reported reasons for stopping during the irst year. Pediatrics Tomi, M., Nishimura, T., Nakashima, E., 2011. Mother-to-fetus transfer of
122 (Suppl. 2), S69–S76. antiviral drugs and the involvement of transporters at the placental barrier.
Loebstein, R., Lalkin, A., Koren, G., 1997. Pharmacokinetic changes during J. Pharm. Sci. 100, 3708–3718. https://doi.org/10.1002/jps.22642.
pregnancy and their clinical relevance. Clin. Pharmacokinet. 33, 328–343. van den Anket, J.N., 1996. Pharmacokinetics and renal function in preterm
Madadi, P., Ross, C.J., Hayden, M.R., et al., 2009. Pharmacogenetics of neona- infants. Acta Pediatr. 85, 1393–1399.
tal opioid toxicity following maternal use of codeine during breastfeeding: a Vorhees, C.V., Weisenburger, W.P., Minck, D.R., 2001. Neurobehavioral
case control study. Clin. Pharmacol. Ther. 85, 31–35. teratogenic effects of thalidomide in rats. Neurotoxicol. Teratol. 23,
Medicines and Healthcare Products Regulatory Agency, 2014. Domperidone: 255–264.
risks of cardiac side effects – indication restricted to nausea and vomiting, World Health Organization (WHO), 2001. 54th World Health Assembly
new contraindications, and reduced dose and duration of use. Drug Saf. Global Strategy for Infant and Young Child Feeding. The Optimal Duration
Update 7 (10), A1. of Exclusive Breastfeeding. WHO, Geneva. Available at: http://apps.who.int/
Miller, M.T., Ventura, L., Stromland, K., 2009. Thalidomide and gb/archive/pdf_iles/WHA54/ea54id4.pdf.
misoprostol: ophthalmologic manifestations and associations both Zhao, C., Zhang, H., Zhang, X., et al., 2006. Prediction of milk/plasma drug
expected and unexpected. Birth Defects Res. A Clin. Mol. Teratol. 85, concentration (M/P) ratio using support vector machine (SVM) method.
667–676. Pharm. Res. 23, 41–48.
National Institute for Health and Care Excellence (NICE), 2011. Hypertension
in pregnancy: diagnosis and management. Clinical guidance (CG107). NICE,
London. Available at: https://www.nice.org.uk/guidance/cg107/.

Further reading
Hale, T.W., Rowe, H.E., 2014. Medications and Mothers’ Milk, sixteenth ed. Schaefer, C., Peters, P., Miller, R.K. (Eds.), 2014. Drugs During Pregnancy
Hale Publishing, Amarillo, TX. and Lactation: Treatment Options and Risk Assessment, third ed. Elsevier,
Lee, A., 2006. Adverse Drug Reactions, second ed. Pharmaceutical Press, Amsterdam.
London. Thomas, S.H., Yates, L.M., 2012. Prescribing without evidence – pregnancy.
Br. J. Clin. Pharmacol. 74, 691–697.

Useful websites
Bumps (Patient Information Lealets and Online Reporting Facility by UKTIS): MHRA Toolkit on the Risks of Valproate Medicines in Female Patients: https://
Best Use of Medicines in Pregnancy: http://www.medicinesinpregnancy.org www.gov.uk/government/publications/toolkit-on-the-risks-of-valproate-
UK Teratology Information Service (UKTIS): http://www.uktis.org medicines-in-female-patients
TOXBASE: http://www.toxbase.org LactMed, National Library of Health: http://toxnet.nlm.nih.gov/
European Network of Teratology Information Services: https://www.entis- cgi-bin/sis/htmlgen?LACT
org.eu/ Medication & Mother’s Milk Online, Hale Publishing: http://
American Organisation of Teratology Information Services: OTIS/ www.medsmilk.com/ (subscription only).
MotherToBaby: http://mothertobaby.org/ UK Drugs in Lactation Advisory Service (UKDILAS): https://www.sps.nhs.
Motherisk (Canadian Teratology information service): http://www.motherisk.org uk/articles/ukdilas/

834

You might also like