SSRI - menyusui

download SSRI - menyusui

of 14

Transcript of SSRI - menyusui

  • 7/24/2019 SSRI - menyusui

    1/14

    Arch Womens Ment Health (2007) 10: 3951

    DOI 10.1007/s00737-007-0173-0

    Printed in The Netherlands

    Review

    SSRIs during breastfeeding: spotlight on milk-to-plasma ratio

    S. Gentile1, A. Rossi2, and C. Bellantuono3

    1 Department of Mental Health ASL Salerno 1, Mental Health Center n.4, Cava de Tirreni (Salerno), Italy2 Medical Department Eli Lilly Italia, Sesto Fiorentino (FI), Italy3 Section of Psychiatry and Clinical Psychology, Department of Medicine and Public Health, University of Verona, Verona, Italy

    Received October 1, 2006; accepted January 13, 2007

    Published online February 12, 2007 # Springer-Verlag 2007

    Summary

    Objective: To investigate the usefulness of the milk-to-plasma (M=P)ratio for assessing the risks for the breastfed infant associated with the

    maternal use of SSRIs.

    Data sources: Medline, Toxnet, Embase, Current Contents, and

    PsycInfo indexed articles from 1980 to September 2006.

    Study selection and data extraction:All studies reporting the M=Pratio in mothers taking SSRIs while breastfeeding or studies which such

    an information could be calculated from data reported in the article.

    Data synthesis: Higher M=P ratios were rarely associated with a

    clinically significant impact on the babies during the early phases ofbreastfeeding.

    Conclusions: So far no evidence-based information seems to support

    the hypothesis that SSRIs characterized by a M=P ratio

  • 7/24/2019 SSRI - menyusui

    2/14

    metabolic, and atopic diseases (Allen & Hector, 2005).

    Breastfeeding also provides a number of benefits for the

    nursing mother, such reduced risks of both ovarian and

    breast cancer (Martin et al, 2005; Riman et al, 2004).

    Several methods and parameters have been proposed

    and utilized to establish the amount of psychoactive

    drugs transferred to maternal milk, in order to reduce

    the infants exposure as much as possible. Some inves-

    tigators actually examined the size, lipid solubility, and

    protein binding of the molecule in an attempt to forecast

    the extent of passage into maternal milk, also taking into

    account the bioavailability of the drug and the infants

    competence to excrete the drug and its metabolites (Pons

    et al, 1994; Hale, 2002). Another important value is the

    theoretic infant dose. It is an estimate of the maximumlikely dose per kilogram per day an infant would get

    trough breast milk (Malone et al, 2004). The limitations

    of such methods for estimating the infant exposure, how-

    ever, are beyond the scope of this article.

    One of the parameters most frequently used, however,

    is the milk-to-plasma (M=P) ratio, which represents the

    ratio of drug concentration in breast milk to drug con-

    centrations in maternal plasma. Hence, an M=P ratio less

    than 1.0 should indicate that the drug transfer into breast

    milk is relatively low, the preferred situation (Malone

    et al, 2004). An M=P ratio of 1.0 or more indicates that

    the drug may be present in breast milk at higher levels

    than in the mothers plasma; such a situation might be

    associated with a relatively higher amount of drug trans-

    ferred to infants and, consequently, with a theoretical

    higher risk of inducing iatrogenic unwanted events.

    Indeed, maternal breast milk level seems to be signi-

    ficantly correlated with infant plasma level for most

    SSRIs, such as citalopram, fluoxetine, and paroxetine

    (Weissman et al, 2004). Since milk drug concentrations

    depend on several factors (such as time to peak milk

    concentration, the dosing schedule, breast modifications

    during pregnancy, maternal bodyweight and metabo-

    lism) and also fluctuate over the whole duration of lacta-

    tion as well as during the course of the day, the best

    method for estimating the M=P ratio may be based on

    an average of several milk samples.

    Moreover, the M=P ratio is also utilized in:

    a) calculating the Exposure Index (as percent of weight

    adjusted maternal dose), which is directly proportional

    to the M=P ratio but inversely proportional to the rate

    of clearance of the drug by the infant (Ito, 2000);

    b) estimating the infant dose after importing the value

    of the M=P ratio in specific equations, such as the

    Atkinson formula.

    Despite an M=P ratio of>1.0 does not necessarily mean

    that the drug is contraindicated during lactation, but

    just that is less desirable than a drug characterized by

    an M=P ratio

  • 7/24/2019 SSRI - menyusui

    3/14

    ratio, SSRIs. A separate search was also run to complete the

    electronic search for the six SSRIs: fluoxetine, fluvoxamine,

    sertraline, paroxetine, citalopram, and escitalopram. The studies

    retrieved were examined for additional references. 135 studies

    were recognized.

    Inclusion criteria were however represented by the articles

    which reported the M=P ratio for this class of antidepressants

    or those where the drug concentrations in maternal plasma andmilk were reported, permitting the calculation of this parameter.

    Hence, 99 articles were excluded. 34 and 2 studies respectively

    meeting the first and the second inclusion criterion were con-

    versely reviewed.

    Results

    Fluoxetine

    Preliminary information about the excretion of the med-

    ication in human milk and the M=P ratios of both fluox-

    etine and its active metabolite became available in 1990:

    Isenberg evaluated a woman suffering from dysthymic

    disorder often complicated by major depressive episodes

    who was treated with fluoxetine while breastfeeding.

    The M=P ratios for fluoxetine and norfluoxetine were

    both lower than 1.0. The mother and the infants paedia-

    trician did not notice any drug-related adverse event.

    The infants exposure was not analysed.

    A further study by Burch & Wells (1992) conducted

    on a woman requiring antidepressant treatment because

    of bipolar depression also failed to show adverse behav-

    ioral or developmental outcome in the infant. Also in

    this study, the M=P ratios for the parent drug and itsmetabolite were lower than 1.0. However, an estimate

    of the babys dose, albeit indirect, was possible only by

    assuming theoretically that the baby received 0.15 L=kg

    of milk per day.

    Six year later, four mothers (one affected by obses-

    sive-compulsive disorder and three by major depressive

    episodes) were studied by Yoshida et al (1998). No ad-

    verse events were recorded during the early postpartum

    period, despite in some women the M=P ratios for fluox-

    etine and its metabolite overcame the notional level of

    concern of 1.0. Moreover, the development of the four

    infants exposed to fluoxetine until they were 12 months

    old and repeatedly assessed by the Bayley Scales of

    Infant Development was normal.

    A further study was specifically designed to charac-

    terize the M=P ratio and the infant dose for fluoxetine

    and norfluoxetine in a relatively large number of breast-

    feeding women taking the medication for the treatment

    of their depression (Kristensen et al, 1999). The absolute

    infant dose, calculated as fluoxetine equivalent, was de-

    termined by 2 different methodologies (both assuming

    an oral availability of 100% and an average milk intake

    of 0.15Lkg1 day). Fluoxetine was detected in 5 of

    the 9 infants from whom samples were collected, where-

    as norfluoxetine was detected in 7. Four infants showed

    adverse events. In three of these cases characterized by

    the occurrence of unwanted reactions the M=P ratios

    was lower than 1.0. Of note, such adverse events wereassessed by interviewing the mother and=or the pae-

    diatrician. No specific assessment tools were used.

    Moreover, methadone maternal use may have been a

    contributing factor in the withdrawal phenomena in

    one infant.

    Drug-related adverse events requiring hospitalisation

    were also described in an infant born to a depressed

    mother who took fluoxetine throughout her pregnancy

    (Hale et al, 2001). In this study, however, mothers

    serum and breast samples were collected at different

    days. Hence, no information can be drawn on the true

    M=P ratio. In the infants serum, no detectable fluoxetine

    levels were recorded; norfluoxetine levels conversely

    ranged from 86 to 142 ng=mL.

    In the study by Hendrick et al (2001) it was demon-

    strated that fluoxetine and norfluoxetine concentrations

    in the maternal serum positively correlate with infant

    norfluoxetine concentrations in the infants serum. The

    authors also demonstrated that peak milk concentrations

    occurred approximately 8 h after maternal dosing and

    also influenced norfluoxetine levels in the infants se-

    rum. Mothers were questioned about potential unwanted

    sequelae to their infants and did not report any suchfindings, despite in some of these women the M=P ratio

    was broadly higher than 1.0 for both the drugs and its

    metabolite.

    Such results were confirmed in a further study con-

    ducted by the same group of researchers. This study also

    matched 3 different methods of estimating the daily dose

    to nursing infants (Suri et al, 2002): it was concluded

    that the Mathematical Model (consisting of determining

    the gradient of excretion of medication into breast milk

    at a specified time after the maternal dose, applying such

    a gradient to each nursing collection, and finally sum-ming the values for 24 h), rather than the Babys Total

    Daily Dose Model or the Atkinson Model, seems to

    reflect the infants serum concentration with an higher

    degree of accuracy. However, also the Mathematical

    Model provides an indirect estimation of the amount

    of drug ingested by the infant.

    To investigate the pharmacokinetics of fluoxetine and

    norfluoxetine during pregnancy, delivery, and lactation, a

    number of mothers suffering from depression or panic

    SSRIs during breastfeeding 41

  • 7/24/2019 SSRI - menyusui

    4/14

    Table2.Fluoxetine(FLX)=norfluoxetine(NFX)

    Study=sample

    size

    Maternalbody

    weight(kg)

    Maternal

    dose(mg)

    Postpartum

    time

    Antidepressant

    treatmentduration

    Timepost

    maternal

    dose

    M=Pratios

    Repercussionson

    theclinicaloutcome

    Isenberg(1990)

    N=A

    20

    3months

    2months

    N=A

    FLX0.28

    irritability

    (n

    1)

    NFX0.21

    Burch&Wells(1992)

    N=A

    20

    2months

    53days

    4h

    FLX0.49

    no

    (n

    1)

    NFX0.34

    Yoshidaetal(1998)

    N=A

    2040

    118days

    1252weeks

    N=A

    FLX:0.521.45

    no

    (n

    4)

    NFX0.081.1

    Kristensenetal(1999)

    mean67.3

    2080

    N=A

    1375days

    variable

    FLX0.520.84

    2casesofcolic

    (n

    14)

    NFX0.350.77

    2casesofwithdrawal

    syndrome

    Haleetal(2001)

    (n

    1)

    N=A

    20

    day11

    themotherstookthe

    compoundduringthe

    entirepregnancyduration

    N=A

    mothersserumandmilk

    sampleswerecollected

    atdifferenttimes

    somnolence,lethargy,

    fever,unresponsiveness

    Surietal(2002)

    (n

    10)

    N=A

    2060

    5weeks3months

    themothersstartedFLX

    treatmentduring

    breastfeeding(aftera

    minimumof6weeks

    treatment)

    N=A

    FLX0.13.92

    NFX0.51.4

    no

    Hendricketal(2001)

    (n

    19)

    N=A

    1060

    534days

    throughoutpregnancy

    today4after

    parturition

    N=A

    FLX0.656.09

    NFX0.372.08

    no

    Heikkinenetal(2003)

    (n

    11)

    N=A

    2040(mean20)

    pregnancy,delivery,

    day2,day4,

    week2,month2

    6womenusedFLX

    duringpregnancy;

    5startedtakingFLX

    laterinpregnancy

    (week2235)

    N=A

    FLX0.32.2

    NFX0.11.7

    no

    Kimetal(2005)

    (n

    9)

    mean74.1

    1030

    mean3.7months

    themotherstookthe

    compoundduring

    pregnancyfora

    minimumof3weeks

    priordelivery

    variable

    R-FLX

    mean0.84

    S-FLX

    mean0.54

    R-NFX

    mean0.75

    S-NFX

    no

    mean0.55

    Berleetal(2004)

    (n

    1)

    N=A

    20

    N=A

    85days

    immediatelybefore

    administration

    ofthe

    nextdose

    0.31

    no

    1

    Forthesumoffluoxetineandnorfluoxetine.

    42 S. Gentile et al

  • 7/24/2019 SSRI - menyusui

    5/14

    disorder were recruited in a relatively recent study

    (Heikkinen et al, 2003). Common clinical doses of the

    medication results in lower trough plasma concentra-

    tions of fluoxetine and fluoxetine plus norfluoxetine than

    those detected in a clinical setting with similar doses in

    the nonpregnant state, because of increased demethyla-

    tion of fluoxetine by cytochrome P450 (CYP) 2D6.Indeed, it has been reported that pregnancy specifically

    increases CYP2D6 activity (Wadelius et al, 1997).

    Nevertheless, also in this study both fluoxetine and its

    metabolite were likely to cause M=P ratios higher than

    1.0. The infant exposure to fluoxetine through breast

    milk was calculated by the Atkinsons model (Atkinson

    et al, 1998). The infants evaluated in this study showed

    normal growth and development up to 1 year of age:

    however, no specific assessment tools were used for the

    infants examination.

    A recent and well-designed study by Berle et al

    (2004) demonstrated that the levels of fluoxetine in a

    breastfed infant (as percentage of maternal serum con-

    centration) was 6.4, despite a M=P ratio of 0.3. The

    baby, however, showed no unwanted reactions.

    On the other hand, it was very recently confirmed that

    the infant dose is mainly determined by the concentra-

    tions of the maternal drug, which in part depend on the

    maternal dosage (Kim et al, 2005).

    Table 2 shows specific details from the studies on

    fluoxetine and norfluoxetine M=P ratios.

    Paroxetine

    Information about the M=P ratios of paroxetine became

    available since 1996. A case report described a woman

    who started paroxetine treatment 3 days after delivery

    because of exacerbation of her depressive and obsessive

    symptoms (Spigset et al, 1996). The M=P ratio was con-

    siderably lower than 1.0. The relative infant dose was

    calculated indirectly on the basis of information derived

    from previously published literature (Isemberg, 1990;

    Lester et al, 1993). No adverse events were observed

    in the infant during the breastfeeding period.

    The amount of paroxetine excreted in breast milk was

    also quantified by OOhman et al (1999). The mean par-

    oxetine concentrations in hindmilk (the milk secreted at

    the end of the feed) were 78% higher than in foremilk

    (the milk secreted at the start of the feed), and the in-

    crease corresponded to the increase in milk triglyceride

    levels. The M=P ratio was close to 1.0. No adverse

    events were however recorded in this infant, whose par-

    oxetine dose per kg body weight was calculated by a

    specific equation.

    In the key-study by Begg et al (1999) two different

    evaluations were performed. The first involved nursing

    mothers who were evaluated over a 24 h dose interval at

    steady-state: the total amount of paroxetine in the milk

    was calculated which represented the dose to the infant.

    The second evaluation involved a different subgroup

    of nursing mothers who were studied at steady-state,around a normal feeding time. The relative infant dose

    ranged from 0.5 to 1.7 (cumulative excretion in milk,

    expressed as percentage of the weight-adjusted maternal

    dose) and from 0.38 to 2.24% in the first and second

    evaluation, respectively. No adverse events were ob-

    served in any infant, despite the paroxetine M=P ratio

    widely overcame the notional limit of concern of 1.0.

    In a study conducted on a relatively large sample of

    women, paroxetine concentrations were present in all

    breast milk samples; both paroxetine M=P ratios and its

    levels in the infants (quantified by the Mathematical

    model) were widely variable: the highest values were 1.3

    and 101ng=ml, respectively (Stowe et al, 2000). This

    study, however, showed no untoward events in the infants.

    Such results were substantially confirmed by Misri

    et al (2000). Other relevant findings of the study were

    represented by the absence of both detectable paroxetine

    levels in all of the infant serum samples and adverse

    events in the babies.

    No detectable levels of paroxetine were also found

    in the breast milk of a woman requiring antidepressant

    treatment because affected by panic disorder with agora-

    phobia (Hendrick et al, 2000).Berle et al (2004) conversely found that appreciable

    paroxetine levels in breast milk (ranging from 18 to

    152 nmol=L) are not associated with detectable levels

    of the drug in the infants serum.

    Further information on the excretion of paroxetine in

    breast milk became available in the pooled analysis by

    Weissman et al (2004). Table 3 summarizes the most

    relevant findings on the M=P ratio for paroxetine.

    Sertraline

    Altshuler et al (1995) found that sertraline levels in

    breast milk varied widely over 24 h, with the peak occur-

    ring between hours 1 and 9 after the drug intake. The

    M=P ratio was lower than 1.0. Sertraline was also quan-

    titated in the infant serum specimens: neither appreci-

    able sertraline levels in the infants serum nor adverse

    events were demonstrated. However, the desmethyl-

    metabolite was not measured.

    In the study by Stowe et al (1997) sertraline showed a

    gradient from foremilk to hindmilk; the peak in breast-

    SSRIs during breastfeeding 43

  • 7/24/2019 SSRI - menyusui

    6/14

    Table3.Paroxetine(PAR)

    Study=sample

    size

    Maternalbody

    weight(kg)

    Maternal

    dose(mg)

    Postpartumtime

    Antidepressant

    treatmentduration

    Timepost

    maternaldose

    M=Pratios

    Repercussionson

    theclinicaloutcome

    Spigsetetal(1996)

    (n

    1)

    60

    20

    milklevelswereobtained

    10daysafterparturition,

    whereasserumlevels

    wereobtainedlater

    (afterthematernaldose

    hadbeenincreasedto

    40mg=day)

    milklevelswereobtained

    7daysafterparturition,

    whereasserumlevels

    wereobtainedlater

    (aftermaternaldose

    hadbeenincreasedto

    40mg=day)

    milklevelswerecalculated

    4hafterdrugintake

    0.09

    no

    OOhmanetal(1999)

    (n

    6)

    5267

    2040

    N=A

    atleast8days

    firstevaluation:47h

    afterdrugintake

    mean

    SD

    no

    lastevalua

    tion:24h

    afterdrugintake

    0.69

    0.29

    Beggetal(1999)

    (n

    10)

    5386

    1030

    N=Ainbothevaluations

    atleast2weeksatthe

    samedosingschedule

    (firstevaluation)

    firstevaluation:atatime

    asclose

    aspossibleto

    thedrugadministration

    0.323.33

    no

    N=Ainthesecond

    evaluation

    secondevaluation:Around

    anormalinfantfeedingtime

    Stoweetal(2000)

    N=A

    1050

    455.2weeks

    longerthan10days

    15h

    0.0561.3

    no

    (n

    16)

    Misrietal(2000)

    (n

    24)

    N=A

    1040

    110.5months

    N=A

    about6h

    0.111.67

    no

    Hendricketal(2000)

    (n

    1)

    N=A

    10

    3weeks

    3weeks

    immediate

    lyafter

    drugintake

    breastmilklevels

    werebelowthe

    quantificationlimits

    no

    Berleetal(2004)

    (n

    6)

    N=A

    1030

    N=A

    atleast14days

    immediate

    lybefore

    adminis

    trationofthe

    nextdose

    0.60.9

    no

    Weissmanetal(2004)

    (n

    3)

    66.9135.8

    1030

    7.328.1weeks

    2156weeks

    810h

    0.580.79

    no

    44 S. Gentile et al

  • 7/24/2019 SSRI - menyusui

    7/14

    Table4.Sertraline(SER)anditsmetabolites

    Study=sample

    size

    Maternalbody

    weight(kg)

    Maternal

    dose(mg)

    Postpartum

    time

    Antidepressant

    treatmentduration

    Timep

    ost

    maternaldose

    M=Pratios

    Repercussionson

    theclinicaloutcome

    Altshuleretal(1995)

    (n

    1)

    N=A

    100a

    3weeks

    themotherstarted

    antidepressanttreatment

    duringpregnancy

    12h(m

    aternal

    serum)

    SER0.53

    no

    Stoweetal(1997)

    (n

    11)

    N=A

    25150

    4141

    3mothersstarted

    antidepressanttreatment

    duringpregnancy,9after

    parturition

    N=A

    SER2.3

    1.3

    desmethylsertraline1.4

    0.8

    no no

    Stoweetal(2003)

    (n

    26)

    N=A

    123.9

    62.8

    436weeks

    12mothersstarted

    antidepressanttreatment

    duringpregnancy,8after

    parturition.For1womanthe

    informationwasunavailable

    N=A

    0.484.81(forbothSER

    anddesmethylsertraline)

    no

    Kristensenetal(1998)

    (n

    8)

    mean71.1

    50200

    1.814.2months

    1.184.25months

    at0,1,2,3,4,6,

    8,12

    ,and24h

    SER1.93

    0.16

    desmethylsertraline1.640.19

    no

    Doddetal(2000)

    N=A

    N=A

    N=A

    morethan2weekson

    426h

    SER1.76

    1.72

    no

    (n

    10)

    fixeddose

    Berleetal(2004)

    (n

    6)

    N=A

    50100

    N=A

    atleast14days

    immediatelybefore

    administration

    ofth

    enextdose

    SER1.23.5

    desmethylsertraline0.64.7

    no

    Weissmanetal(2004)

    (n

    19)

    52.498.0(d

    ata

    availablefor4=19

    women)

    50100

    4.019.6weeks

    N=A

    127h

    SER0.543.0

    norsertraline0.381.66

    (dataavailable

    for4=19women)

    no

    a

    Themotheralsotooknortriptyline,125mg=day.

    SSRIs during breastfeeding 45

  • 7/24/2019 SSRI - menyusui

    8/14

    milk occurred however later, between 7 and 10 hours

    after dosing; M=P ratios for both the drug and its meta-

    bolite were higher than 1.0 without interfering, however,

    with the infants well-being. Also in this study, the

    Mathematical Model was used for estimating the 24-h

    infant medication dose. Such results were substantially

    confirmed in a more recent study performed by the sameresearch group (Stowe et al, 2003).

    In a further study, neither sertraline nor its metabo-

    lite were detected in plasma samples from a few infants

    breastfed by mothers who took the compound during the

    postpartum period, despite M=P ratios higher than 1.0

    for both sertraline and desmethylsertraline. In addition,

    these infants showed no acute adverse events and all

    achieved normal developmental milestones (Kristensen

    et al, 1998). The methodologies used for estimating the

    infant dose were analogue to those described in a trial

    above reported. (Kristensen et al, 1999).

    Reassuring results also emerged from the study by

    Dodd et al (2000) conducted on nursing women affected

    by major depression. The average dose to the infants

    (estimated by the Atkinson model) was less than 2% of

    the maternal daily dose, despite the M=P ratio for sertral-

    ine was found to be higher than 1.0.

    No detectable levels of sertraline were found in 6

    infants whose mothers took the compound while breast-

    feeding (Berle et al, 2004).

    A recent study also provided additional information

    on sertraline excretion in breast milk (Weissman et al,

    2004). Table 4 highlights the studies documenting theM=P ratios for both sertraline and its metabolite.

    Citalopram and escitalopram

    Data on the excretion of citalopram into breast milk have

    been available since 1997. OOhman et al (1997) found that

    the estimated dose to the infant ranged from 5 to 9% of

    the weight-adjusted maternal dose for M=P ratios higher

    than 1.0 for both citalopram and demethylcitalopram.

    In a more detailed study, the average infant dose

    was 4.8% of the maternal dose (Jensen et al, 1997).

    Citalopram reached a peak concentration in breast milk

    6 hours after the drug intake: the resulting M=P ratio was

    higher than 1.0 for both the parent drug and its meta-

    bolite. However, despite the amount of citalopram trans-

    ferred to the baby was determined by theoretically

    assuming a milk volume of 0.15 L=kg baby, the volume

    of milk produced in 24 h was not determined accurately.

    No adverse events occurred in the infants.

    Widely ranging values for the relative infant dose

    (1.85.9%, calculated by a specific equation) associated

    with an M=P ratio higher than 1.0 were also reported by

    Spigset et al (1997). No complications were reported in

    the infants.

    In contrast, a mild adverse event in an infant receiving

    5.4% of the maternal dose was reported in a subsequent

    study (Schmidt et al, 2000). In this study, the M=P ratio

    was found to be more than double the notional limit ofconcern of 1.0.

    Rampono et al (2000) demonstrated that peak milk

    concentration occurs earlier for citalopram (3.9 h) than

    for desmethylcitalopram (5.7 h). The clinical significance

    of such a finding, however, remains unknown, as well as

    the very high M=P ratios of the drug and its metabolite.

    Citalopram was detected in three infants, whereas de-

    methylcitalopram in two. Such infants showed normal

    development for age as assessed by the Denver develop-

    mental screening test.

    Relatively low values of relative infant dose (ranging

    from 0.2 to 0.3% at the ages of 2 months and 2 weeks,

    respectively) was found in a study examining the effi-

    cacy and safety of citalopram in relation to concentra-

    tions of the drug and its metabolites during pregnancy

    and lactation (Heikkinen et al, 2002), despite an M=P

    ratio close to 5.0 for didesmethylcitalopram. Citalopram

    and desmethylcitalopram also showed high M=P ratios.

    The infant exposure to citalopram was quantified by the

    Atkinson model. One case of transient neurodevelop-

    mental delay (spontaneously resolved without any fur-

    ther problem) was reported.

    In the study by Berle et al (2004), the ingestion ofcitalopram by the lactating woman was associated with

    detectable levels in the infants. However, the clinical

    repercussions of such a finding remain unclear.

    Other data on the excretion of citalopram into hu-

    man milk became available in the pooled analysis by

    Weissman et al (2004).

    Very recently, Franssen et al (2006) described the case

    of an infant who developed irregular breathing, sleep

    disorders, and hypotonia evolving into hypertonia after

    exposure to citalopram through the placenta and mater-

    nal milk. However, all the symptoms disappeared spon-taneously within 3 weeks. The M=P ratio of citalopram

    was higher than 1.0 in three consecutive observations,

    whereas the levels of the medication in the infants

    serum were in the range of 3.77.1 nM.

    So far, only one study is available on escitalopram,

    the latest SSRI introduced onto the market (Rampono

    et al, 2006). No adverse events were observed in 8

    infants who showed a relative dose (as a percent of the

    maternal weight-adjusted dose) ranging from 4.2 to

    46 S. Gentile et al

  • 7/24/2019 SSRI - menyusui

    9/14

    Table5.Citalopram(CIT),itsmetabolites,andescitalopram(ESC)

    Drug=study=sample

    size

    Maternal

    body

    weight(kg)

    Maternal

    dose(mg)

    Postpartum

    time

    Antidepressant

    treatmentduration

    Timepost

    maternald

    ose

    M=Pratios

    Repercussionson

    theclinicaloutcome

    OOhmanetal(1997)

    N=A

    2060

    N=A

    N=A

    3.56.5h

    CIT1.812.24

    no

    CIT(n

    2)

    demethylcitalopram

    2.052.91

    Jensenetal(1997)

    CIT(n

    1)

    61

    20

    N=A

    15days

    N=A

    3forboththedrug

    anditsmetabolite

    no

    Spigsetetal(1997)

    CIT(n

    3)

    6470

    2040

    210months

    N=A

    beforethe

    daily

    doseand4hlater

    in2=3ofwomena

    CIT1.161.88

    no

    Schmidtetal(2000)

    CIT(n

    1)

    40

    40

    N=A

    10days

    N=A

    CIT2.07

    uneasysleep,resolved

    aftermaternaldose

    reduction

    Ramponoetal(2000)

    CIT(n

    7)

    mean62.6

    mean36

    mean4.1months

    mean97days

    at0,2,4,

    6,8,

    12,and

    24h

    CIT1.23.0

    demethylcitalopram

    1.02.5

    no

    Heikkinenetal(2002)

    CIT(n

    11)

    N=A

    2040

    N=A

    10womenstarted

    CITatthetimeof

    conception,1at20

    weeksofpregnancy

    justbefore

    takingthedrug

    CIT1.23.3

    desmethylcitalopram

    1.34.1

    didesmethylcitalopram

    1.14.6

    no

    Berleetal(2004)

    CIT(n

    9)

    N=A

    2050

    N=A

    atleast14days

    immediate

    lybefore

    administration

    ofthen

    extdose

    CIT1.14.3

    desmethylcitalopram

    0.96.3

    no

    Weissmanetal(2004)

    65.373.4

    20

    1.611.7months

    1226weeks

    910h

    CIT0.931.79

    no

    CIT(n

    2)

    desmethylcitalopram

    1.361.56

    didesmethylcitalopram

    1.832.0

    Franssenetal(2006)

    CIT(n

    1)

    55

    40

    at12,25,46,

    and53days

    afterdelivery

    thewomanstarted

    takingthedrug

    duringpregnancy

    N=A

    CIT2.02.8

    sleepandrespiratory

    disturbances,

    hypo=hypertonia

    Ramponoetal(2006)

    ESC(n

    8)

    5865

    1020

    N=A

    23240days

    justbefore

    themorning

    doseandat2,4,

    and6h

    afterdose

    ESC1.72.7

    desmethylescitalopram

    1.83.1

    no

    a

    Inthethirdwoman(ahealthyvolunte

    er)serumsampleswereobtainedafter1,2,3

    ,4,5,6,8,10,12,24,48,and72h.Breastm

    ilksampleswereobtainedafter2,4,6,8,14,24,48,72h.

    SSRIs during breastfeeding 47

  • 7/24/2019 SSRI - menyusui

    10/14

    6.4% for escitalopram plus desmethylcitalopram. Both

    compounds, however, showed M=P ratios higher than

    1.0. Data on the excretion of both compounds are avail-

    able in Table 5.

    Fluvoxamine

    The first study evaluating the excretion of fluvoxamine

    in breast milk of a woman affected by post-natal depres-

    sion was performed by Wright et al (1991). The M=P

    ratio was lower than 1.0. Fluvoxamine concentrations in

    the infants serum, however, were quantified indirectly

    by the Atkinson model (Atkinson et al, 1998). The

    maternal antidepressant intake induced no untoward ef-

    fects in this infant.

    These results were replicated in a further case report;the infant showed neither acute toxic effects of fluvox-

    amine nor neurodevelopmental repercussions at month

    21 as assessed by using the Bayley Scales for the Infant

    Development, despite the fact that this baby was born by

    forceps delivery at week 41 of gestation (Yoshida et al,

    1997). The study showed a relatively low M=P ratio.

    Fluvoxamine concentrations in the infants serum were

    quantified indirectly: indeed, the estimate of the babys

    dose was possible only by assuming theoretically that

    the baby received 0.15 L=kg1 of milk per day.

    In contrast, significantly higher M=P ratios were

    reported by Haagg et al (2000), although no unusual reac-

    tions were observed in the infant.

    An M=P ratio higher than 1.0 (without any complica-

    tions in the infants) was also reported in two breastfeed-

    ing women (Kristensen et al, 2002). The study did not

    include the analysis of serum infant samples.

    Further data on the excretion of fluoxetine into hu-

    man milk became available in the pooled analysis by

    Weissman et al (2004). Table 6 reassumes the available

    information on M=P ratio values for fluvoxamine.

    Discussion

    Human milk is a suspension of protein and fat globules

    in a carbohydrate-based suspension (Briggs et al, 1994).

    Most drugs are transferred into maternal milk by passive

    diffusion, reaching a concentration equilibrium with the

    concentration in the blood.

    Hence, almost without exception, as the level of the

    medication in the mothers plasma begins its rise, the

    concentration in milk begins its rise as well. Maternal

    blood concentration largely depends on maternal body-

    weight and medication dose. (Texas Tech University,

    Mechanisms of Drug Entry into Human Milk, Accessed:

    July 2, 2006).

    However, the antidepressant agents enter milk also

    by secretory methods (Hale, 2002). Indeed, medicationsalso may be transferred into breastmilk incorporated

    with fat globules or bound to proteins, primarily casein

    and lactalbumin (Pediatric Pharmacotherapy, 1996. No

    Authors listed).

    Morphological changes of the breast occurring during

    the first weeks after parturition also influence the amount

    of drug excreted in maternal milk. During the first 4 to

    10 days of life, large gaps between alveolar cells exist.

    These gaps permit an enhanced drug-access to the milk.

    Soon after the first week, the alveolar cells swell, sub-

    sequently closing the intracellular gaps and limiting

    access to the milk (Malone et al, 2004; Whitby & Smith,

    2005). Moreover, the composition of breastmilk varies

    from the initial colostrum (which shows a relatively

    higher protein concentration) to mature milk; thus, drug

    concentrations change after the first 3 to 4 days of lacta-

    tion (Crisholm and Kuller, 1997).

    Furthermore, milk composition varies even during a

    single breastfeeding session, with milk expressed to-

    wards the end of a feeding having greater fat contents

    (Pediatric Pharmacotherapy, 1996. No Authors listed).

    Table 6. Fluvoxamine (FVX)

    Study=sample size Maternal bodyweight (kg)

    Maternal

    dose (mg)

    Postpartum

    time

    Antidepressant

    treatment duration

    Time post

    maternal dose

    M=Pratios

    Repercussions on

    the clinical outcome

    Wright et al (1991)

    (n1)

    70 200 14 weeks 2 weeks 4 h and 45 min 0.29 no

    Yoshida et al (1997)

    (n

    1)

    N=A 100 17 weeks 2 weeks 3 h 0.29 no

    Haagg et al (2000)

    (n1)

    90 200 3 months 25 weeks every hour during

    a 12-h period

    1.061.59 no

    Kristensen et al

    (2002) (n2)

    5470 50150 0.7526.5

    months

    3 6 weeks at 0, 1, 2, 3, 4, 6,

    8, 12, and 24 h

    mean 1.01 no

    Weisman et al

    (2004) (n1)

    62.4 250 29.1 weeks 94 weeks 9 h 1.02 no

    48 S. Gentile et al

  • 7/24/2019 SSRI - menyusui

    11/14

    These variations have specific repercussions on the

    levels of drugs in maternal milk.

    On the part of the infant, sucking patterns, feeding

    duration, and volume ingested also determine the amount

    of drug ingested. Finally, once a drug has entered the

    mothers milk and has been ingested by the infant, it must

    traverse through the infants gastrointestinal tract and beabsorbed (Texas Tech University. Mechanisms of Drug

    Entry into Human Milk. Accessed: July 2, 2006). Also,

    the percentage of babies showing detectable serum drug

    levels and, consequently, the magnitude of such levels is

    also associated with the individual timing of the infants

    hepatic maturation, which usually occurs not before the

    third month of life (Warner, 1986).

    Hence, a large number of factors regulate the amount

    of drug transferred to milk. It should primarily be taken

    into consideration, however, that the M=P ratio shows

    intrinsic limitations: the primary drawback is that it

    relies a one-point determination and does not reflect

    the other variables interfering with the drug transfer.

    The true M=P ratio also varies significantly during the

    same episode of breastfeeding too (Pediatric Pharma-

    cotherapy, 1996. No Authors listed).

    In addition, most of such factors (and, especially,

    maternal bodyweight, maternal dose, specific portion

    of breastfeeding, timing of evaluation after parturition,

    and timing of antidepressant treatment before evalua-

    tion) were widely inhomogeneous in nearly the totality

    of the studies focused to estimate the M=P ratio value.

    This bias could explain the wide ranges of M=P ratioscalculated in different studies conducted on the same

    SSRI.

    Thus, it not surprising that the M=P ratio seems to be

    associated with no or minimal clinical relevance: indeed,

    in several cases appreciable concentrations of antide-

    pressant agents in the breast milk were associated with

    no detectable levels in infant plasma.

    Moreover, because in more than a few studies the

    M=P ratio was derived from data obtained at single time

    point, its usefulness in attempting to establish the safety

    of SSRIs for the breastfed infant was limited and oftenmisleading because it deviated significantly from the

    time-averaged value.

    Furthermore, all published information was supported

    by findings emerging from very small sample sizes: for

    this reason, this information is applicable to only a few

    women and, hence, is not really generalizable to the

    breastfeeding population.

    In addition, both studies documenting fluoxetine-

    related untoward events in the suckling infants reported

    an M=P ratio lower than 1.0, and their results were also

    contaminated by two potential confounding factors: ma-

    ternal co-medications and=or previous exposure of the

    baby through the placenta. A similar number of unsafe

    reports were reported for citalopram, whose M=P ratio

    ranges from 0.93 to 4.6; however, the baby who suffered

    from the most serious adverse event was also exposed tocitalopram during fetal life. Therefore, it is possible that

    such events could to be not related to the exposure to the

    drug via maternal milk, but to toxic events due to pla-

    cental exposure which was prolonged during the first

    days after parturition. In addition, in the vast majority

    of cases the infant evaluation was based on empirical

    observations.

    Hence, so far no evidence-based information seems

    to support the hypothesis that SSRIs characterized by a

    M=P ratio

  • 7/24/2019 SSRI - menyusui

    12/14

    quantification of the levels of antidepressant in both

    maternal and infant serum.

    In any case, if the mother wishes to breastfeed her

    infant while taking an SSRI, the baby should be closely

    monitored in order to detect any iatrogenic event as soon

    as possible.

    Acknowledgments

    The authors would like to acknowledge Anna Maria Desiati,

    Information Specialist, Scientific Information Service, Eli Lilly

    Italia SpA, and her colleague Leonardo Pavese. Their support in

    the electronic searching and for obtaining full-text articles was

    highly valuable and professional. No sources of funding were

    used for the preparation of manuscript.

    Disclosure of interests

    Salvatore Gentile is on the speaker-bureau for Eli Lilly Italia

    SpA. In the last 5 years, he also received travel support from

    Astra Zeneca, Janssen-Cilag, Lundbeck, GlaxoSmithKline,

    Bristol-Meyer Squibb, Pfizer, Novartis, and Recordati.

    Andrea Rossi is employed in the Medical Department of Eli

    Lilly Italy.

    Cesario Bellantuono has received educational grants from Eli

    Lilly, Astra Zeneca, Janssen-Cilag, Lundbeck, GlaxoSmithKline,

    Bristol-Meyer Squibb.

    References

    Allen J, Hector D (2005) Benefits of breastfeeding. New South Wales

    Public Health Bull 16 (34): 46.

    Altshuler LL, Burt VK, McMullen M, Hendrick V (1995) Breastfeedingand sertraline: a 24-hour analysis. J Clin Psychiatry 56: 243245.

    American Academy of Pediatrics (1997) Work Group on Breastfeeding:

    Breastfeedingand the use of human milk. Pediatrics 100:10351039.

    Atkinson HC, Begg EJ, Darlow BA (1998) Drugs in human milk: clini-

    cal pharmacokinetic consideration. Clin Pharmacokinet 23: 139147.

    Beck CT (2006)Postpartumdepression: it isnt justthe blues.Am J Nurs

    106(5): 4050.

    Begg EJ, Atkinson HC, Duffull SB (1992) Prospective evaluation of a

    model for the prediction of milk: plasma drug concentrations from

    physicochemical characteristics. J Clin Pharmacol 33: 501505.

    Begg EJ, Duffull SB, Saunders DA, Buttimore RC, Ilett KF, Hackett LP,

    Yapp P, Wilson DA (1999) Paroxetine in human milk. Br J Clin

    Pharmacol 48: 142147.

    Berle J, Steen VM, Aamo TO, Breilid H, Zahlsen K, Spigset O (2004)

    Breastfeeding during maternal antidepressant treatment with seroto-nin reuptake inhibitors: infant exposure, clinical symptoms, and

    cytochrome P450 genotypes. J Clin Psychiatry 65: 12281234.

    Briggs GG, Freeman RK, Yaffe SJ (1994) Drugs in pregnancy and

    lactation. 4th edn. William and Wilkins, Baltimore.

    Boerner RJ, Mooller HJ (1999) The importance of new antidepressants in

    the treatment of anxiety=depressive disorders. Pharmacopsychiatry32: 119126.

    Burch KJ, Wells BG (1992) Fluoxetine=norfluoxetine concentrations inhuman milk. Pediatrics 88: 676677.

    Burt VK, Suri R, Altshuler L, Stowe ZN, Hendrick VC, Muntean E

    (2001) The use of psychotropic medications during breastfeeding.

    Am J Psychiatry 158: 10011009.

    Citalopram. Drug Monograph. Available on: http:==www.mentalhealth.com=drug=p30-c04.html. Accessed July 2, 2006.

    Crisholm CA, Kuller JA (1997) A guide to the safety of CNS-active

    agents during breastfeeding. Drug Saf 17(2): 127142.

    Dodd S, StockiA, Buist A, Burrows GD,Maguire K, NormanTR (2000)

    Sertraline in paired blood plasma and breast-milk samples for nursing

    mothers. Hum Psychopharmacol Clin Exper 15: 261264.

    Eberhard-Gran M, Eskild A, Opjordsmoen A (2006) Use of psycho-

    tropic medications in treating mood disorders during lactation:practical recommendations. CNS Drugs 20(3): 187198.

    Fluoxetine Drug Monograph. Available on: http:==www_mentalhealth.com=drug=p30-p05.hclm. Accessed: July 2, 2006.

    Fluvoxamine Drug Monograph. Available on: http:==www.mentalhealth.com=drug=p30-l05.html. Accessed: July 2, 2006.

    Franssen EJF, Meijs V, Ettaher F, Valerio PG, Keesen M, Lameijer W

    (2006) Citalopram serum and milk levels in mother and infant during

    lactation. Ther Drug Monitor 28: 24.

    Freeman MP, Anthony M (2004) Focus on womens mental health

    [Editorial]. J Clin Psychiatry 65: 11641165.

    Gale S, Harlow BL (2003) Postpartum mood disorder: a review of

    clinical and epidemiological factors. J Psychosom Obstet Gynaecol

    24(4): 257266.

    Gentile S (2005a) SSRIs in pregnancy and lactation. Emphasis on

    neurodevelopmental outcome. CNS Drugs 19: 623633.Gentile S (2005b) The safety of newer antidepressants in pregnancy and

    breastfeeding. Drug Safety 28: 135152.

    Gentile S (2007) Use of contemporary antidepressants during breastfeed-

    ing.A proposal for a specific safety index. Drug Saf 30(20):107 121.

    GlaxoSmithKline. Paroxetine US Prescribing Information. Avail-

    able on: http:==GSK.com=products=assets=us_paxil.pdf. AccessedSeptember 22, 2006.

    Haagg S, Granberg K, Carleborg L (2000) Excretion of fluvoxamine into

    breast milk [Letter]. Br J Clin Pharmacol 49: 286287.

    Hale TW (2002) Medications and mothers milk, 10th edn. Pharmasoft

    Medical, Amarillo, Texas.

    Hale TW, Shum S, Grossberg M (2001) Fluoxetine toxicity in a

    breastfed infant. Clin Pediatrics 40: 681684.

    Heikkinen T, Ekblad U, Kero P, Ekblad S, Laine K (2002) Citalopram in

    pregnancy and lactation. Clin Pharmacol Ther 72: 184191.Heikkinen T, Ekblad U, Palo P, Laine K (2003) Pharmacokinetics

    of fluoxetine and norfluoxetine in pregnancy and lactation. Clin

    Pharmacol Ther 73: 330337.

    Hendrick V, Stowe ZN, Altshuler LL, Lori L, Hostetter A, Fukuchi A

    (2000) Paroxetine use during breastfeeding [Letter]. J Clin Psycho-

    pharmacol 20: 587589.

    Hendrick V, Stowe ZN, Altshuler LL, Mintz J, Hwang S, Hostetter A

    et al (2001) Fluoxetine and norfluoxetine concentrations in nursing

    infants and breast milk. Biol Psychiatry 50: 775782.

    Isenberg KE (1990) Excretion of fluoxetine in human breast milk

    [Letter]. J Clin Psychiatry 51: 169.

    Ito S (2000) Drug therapy for breast-feeding infants. New Engl J Med

    343: 118127.

    Jensen PN, Olesen OV, Bertelsen A, Linnet K (1997) Citalopram and

    desmethylcitalopram concentrations in breast milk and in serum ofmother and infant. Ther Drug Monitor 19: 236239.

    Kim J, Riggs KW, Misri S, Kent N, Oberlander TF, Grunau RE et al

    (2005) Stereoselective disposition of fluoxetine and norfluoxetine

    during pregnancy and breastfeeding. Br J Clin Pharmacolol 61:

    155163.

    Kristensen JH, Ilett KF, Dusci LJ, Hackett LP, Yapp P, Wojnar-

    Horton RE et al (1998) Distribution and excretion of sertraline

    and N-desmethylsertraline in human milk. Br J Clin Pharmacol

    45: 453457.

    Kristensen JH, Ilett KF, Hackett LP, Yapp P, Paech M, Begg EJ (1999)

    Distribution and excretion of fluoxetine and norfluoxetine in human

    milk. Br J Clin Pharmacol 48: 521527.

    50 S. Gentile et al

  • 7/24/2019 SSRI - menyusui

    13/14

    Kristensen JH, Hackett PL, Kohan R, Paech M, Ilett KF (2002) The

    amount of fluvoxamine in milk is unlikely to be a cause of adverse

    effects in breastfed infants. J Hum Lactat 18: 139143.

    Larsen LA, Ito S, Koren G (2003) Prediction of milk=plasma ratio ofdrugs. Ann Pharmacother 37(9): 12991306.

    Lexa Lexapro FAQs. Available on: http:==www.lexapro.com=english=faq=currently_taking.aspx#qo12. Accessed July 2, 2006.

    Malone K, Papagni K, Ramini S, Keltner NL (2004) Antidepressants,

    antipsychotics, benzodiazepines, and the breastfeeding dyad. PerspectPsych Care 40: 7385.

    Martin RM, Middleton N, Gunnell D, Owen CG, Smith GD (2005)

    Breast-feeding and cancer: the Body Orr cohort and a systematic

    review with meta-analysis. J Natl Cancer Inst 97: 14461457.

    Misri S, Kim J, Riggs K, Kostaras X (2000) Paroxetine levels in

    postpartum depressed women, breast milk, and infant serum. J Clin

    Psychiatry 61: 828832.OOhman I, Wikner-Norsted B, Vitols S (1997) Citalopram and metabolite

    levels in plasma and breast milk in two nursing women [Abstract].

    Eur J Clin Pharmacol 52 (Suppl): A179.OOhman R, Haagg S, Carleborg L, Spigset O (1999) Excretion of

    paroxetine into breast milk. J Clin Psychiatry 60: 519523.

    Pediatric Pharmacotherapy (1996) Determining the degree of drug

    transfer. Available on: www.healthsystem.virginia.edu=internet=

    pediatrics=pharma-news=April96.PDF.Pons G, Rey E, Matheson I (1994) Excretion of psychoactive drugs into

    breast milk. Clin Pharmacokin 27: 270289.

    Rampono J, Kristensen JH, Hackett LP, Paech M, Kohan R, Ilett KF

    (2000) Citalopram and demethylcitalopram in human milk; distribu-

    tion, excretion and effects in breast fed infants. Br J Clin Pharmacol

    50: 263268.

    Rampono J, Hackett LP, Kristensen JH, Kohan R, Page-Sharp M,

    Ilett KF (2006) Transfer of escitalopram and its metabolite de-

    methylescitalopram into breast milk. Br J Clin Pharmacol 62(3):

    316322.

    Riman T, Dickman PW, Nilsson S, Correia N, Nordlinder H,

    Magnusson CM et al (2004) Risk factors for invasive epithelial

    ovarian cancer: results from a Swedish case-control study. Am J

    Epidemiol 156: 763773.

    Schmidt K, Olesen OV, Jensen PN (2000) Citalopram and breast-feeding: serum concentration and side effects in the infant. Biol

    Psychiatry 47: 164165.

    Sertraline. Drug Monograph. Available on: http:==www.mentalhealth.com=drug=p30-z02.html. Accessed: July 2, 2006.

    Spigset O, Carleborg L, Norstroom A, Sandlund M (1996) Paroxetine

    levels in breast milk [Letter]. J Clin Psychiatry 57: 39.

    Spigset O, Carleborg L, OOhman R, Nostroom A (1997) Excretion of

    citalopram in breast milk. Br J Clin Pharmacol 44: 295298.

    Stowe ZN, Owens MJ, Landry JC, Kilts CD, Ely T, Llewellyn A et al

    (1997) Sertraline and desmethylsertraline in human breast milk and

    nursing infants. Am J Psychiatry 154: 12551260.

    Stowe ZN,Cohen LS,HostetterA, Ritchie JC,Owens MJ,NemeroffCB

    (2000) Paroxetine in human breast milk and nursing infants. Am J

    Psychiatry 157: 185189.

    Stowe ZN,Hostetter AL,Owens MJ, Ritchie JC, Stemberg K, Cohen LS

    et al (2003) The pharmacokinetics of sertraline excretion into human

    breast milk: determinants of infant serum concentrations. J ClinPsychiatry 64: 7380.

    Suri R, Stowe ZN, Hendrick V, Hostetter A, Widawski M, Altshuler LL

    (2002) Estimates of nursing infant daily dose of fluoxetine through

    breast milk. Biol Psychiatry 52: 446451.

    Texas Tech University. Mechanisms of Drug Entry into Human Milk.

    Available on: www.ncbi.nlm.nih.gov=entrez=query.fcgi?cmdRetrieve&dbPubMed&list_uids9233209&dopt Abstract.

    Accessed: July 2, 2006.

    Wadelius M, Darj E, Frenne G, Rane A (1997) Induction of CYP2D6 in

    pregnancy. Clin Pharmacol Ther 26: 400407.

    Warner A (1986) Drug use in the neonate: interrelationships of phar-

    macokinetics, toxicity, and biochemical maturity. Clin Chemistry 32:

    721727.

    Weissman AM, Levy BT, Hartz AJ, Bentler S, Donohue M, Ellingrod

    VL et al (2004) Pooled analysis of antidepressant levels in lactatingmothers, breast milk, and nursing infants. Am J Psychiatry 161:

    10661078.

    Whitby DH, Smith KL (2005) The use of tricyclic antidepressants and

    selective serotonin reuptake inhibitors in women who are breastfeed-

    ing. Pharmacother 25: 411425.

    Wisner KL, Parry BL, Piontek CM (2002) Postpartum depression. New

    Engl J Med 347: 194199.

    Wolf AW, De Andraca I, Lozoff B (2002) Maternal depression in three

    Latin American samples. Soc Psychiatry Psychiatr Epidemiol 37:

    169176.

    Wright S, Dawling S, Ashford JJ (1991) Excretion of fluvoxamine in

    breast milk [Letter]. Br J Clin Pharmacolol 31: 209.

    Yoshida K, Smith B, Channi Kumar R (1997) Fluvoxamine in breast

    milk and infant development [Letter]. Br J Clin Pharmacolol 44:

    210211.Yoshida K, Smith B, Craggs M, Channi Kumar R (1998) Fluoxetine in

    breast-milk and developmental outcome of breast-fed infants. Br J

    Psychiatry 172: 175179.

    Correspondence: Dr. Salvatore Gentile, Department of

    Mental Health ASL Salerno 1, Mental Health Center n.4,

    Piazza Galdi 84013, Cava de Tirreni (Salerno), Italy; e-mail:

    [email protected]

    SSRIs during breastfeeding 51

  • 7/24/2019 SSRI - menyusui

    14/14