Sie sind auf Seite 1von 12

THEMED ARTICLE y Breast Cancer Review

For reprint orders, please contact reprints@expert-reviews.com

Motherhood after breast


cancer: searching for
la dolce vita
Expert Review of Anticancer Therapy Downloaded from informahealthcare.com by Washington University Library on 12/28/14

Expert Rev. Anticancer Ther. 11(2), 287–298 (2011)

Hatem A Azim Jr†1, Advances in the field of adjuvant therapy in breast cancer have led to significant improvements
Fedro A Peccatori2, in breast cancer survival. This has resulted in a progressive decline in breast cancer-related
Evandro de Azambuja1 mortality, such that in 2010 there were estimated to be 400,000 breast cancer survivors under
the age of 40 in the USA. Hence, enquiry into the feasibility of fertility preservation, subsequent
and Martine J Piccart1
pregnancy and breastfeeding is increasingly encountered. Fertility counseling remains suboptimal
1
Department of Medical Oncology,
in breast cancer clinics, and there is a wide perception that pregnancy could worsen the
Jules Bordet Institute, Boulevard de
Waterloo, 121, 1000 Brussels, Belgium prognosis of young breast cancer survivors, despite the lack of evidence supporting this notion.
2
Department of Medicine, Division of In addition, fertility preservation by means of embryo or oocyte cryopreservation requires
Hematology Oncology, European ovarian stimulation, which is associated with a significant rise in estradiol levels and might delay
Institute of Oncology, Milan, Italy

Author for correspondence:
initiation of therapy. All these factors, and others, have influenced the quality of fertility
Tel.: +32 2541 3099 counseling offered to young breast cancer patients. In this article, we will critically analyze the
Fax: +32 2541 3477 available clinical and biological evidence on the safety and feasibility of pregnancy and
For personal use only.

hatem.azim@bordet.be
breastfeeding following breast cancer. In addition, we will discuss the different fertility-
preservation techniques available for these patients.

Keywords : breastfeeding after breast cancer • fertility counseling • fertility preservation • pregnancy after
breast cancer

Breast cancer in young women tends to be more strong evidence on one hand, and conceptual
aggressive compared with their older counter- fears that subsequent pregnancy and breastfeed-
parts [1–4] . Young breast cancer patients are more ing could worsen breast cancer outcome on the
often diagnosed with node-positive and triple- other, interfere with the wishes of many breast
negative disease, and consequently adjuvant cancer survivors to start or complete their fami-
chemotherapy is more frequently considered lies [10] . Hence, there is indeed an urgent need to
in these patients [1,2] . However, advances in the provide more robust recommendations on how
field of adjuvant therapies have improved breast to properly counsel these women [8] . In this arti-
cancer survival and significantly reduced the risk cle, we will critically discuss in detail the safety
of recurrence, such that there were an estimated and feasibility of pregnancy and breastfeeding
400,000 breast cancer survivors below the age following completion of breast cancer therapy.
of 40 years in the USA in 2010 [5] . We will also touch on advances in the field of
Given the rising trend of delaying pregnancy fertility preservation.
to later in life [6] , more women are diagnosed
with breast cancer before completing their fami- Risk of amenorrhea following adjuvant
lies. Therefore, patient enquiry into fertility-pres- systemic therapies
ervation techniques, chances of future concep- Normally, there is a progressive loss of oocytes
tion, pregnancy and breastfeeding is increasingly from fetal life through menopause [11] . At the
encountered nowadays in breast cancer clinics. age of 37 years, there is accelerated atresia of the
An internet-based survey has pointed out that oocytes, resulting in an average age of meno-
more than 50% of young women have fertility pause of around 51 years in Western nations
concerns upon being diagnosed with breast can- [11,12] . The situation is quite different in breast
cer [7] . However, unfortunately, fertility counsel- cancer patients, because adjuvant systemic
ing for these women remains unsatisfactory and anticancer therapy, particularly chemotherapy,
lacks a standardized approach [8,9] . The lack of significantly increases the risk of developing

www.expert-reviews.com 10.1586/ERA.10.208 © 2011 Expert Reviews Ltd ISSN 1473-7140 287


Review Azim Jr, Peccatori, de Azambuja & Piccart

early menopause [13] . Earlier work by Partridge and colleagues than 800,000 parous women [26] . At a mean period of 16 years,
involving 767 breast cancer patients under the age of 40 years there was a short-term increase in the risk of breast cancer in
has shown that those who received one or six cycles of cyclo- the 3 years after a full-term pregnancy, which was followed by
phosphamide, methotrexate and 5-flourouracil, and resumed a long-lasting decrease in risk. Similar results were obtained in
their menses afterwards developed menopause at 44 and 41 years smaller studies, with an increased breast cancer risk ranging from
of age, respectively (p < 0.0001) [14] . The same group went on 3 to 5 years following pregnancy [27–29] . The duration of this
to show that those women whose menses resumed had reduced risk depends on several factors, including parity and age at first
ovarian reserve when compared with population age-matched delivery, with younger first-time mothers showing the shortest
controls [15] . duration of increased risk. Hence, pregnancy seems to exert a
Chemotherapy induces temporary amenorrhea in the major- bidirectional time-dependent effect on breast cancer: it is pro-
Expert Review of Anticancer Therapy Downloaded from informahealthcare.com by Washington University Library on 12/28/14

ity of patients. Menses are resumed in some, with resumption tective in the long term, but in the short term tumor incidence
occurring within 6 months following chemotherapy cessation is increased.
for approximately 60% of those patients, and within 12 months Several hypotheses have been described that try to explain the
for 90% [16] . Thus, amenorrhea for more than 1 year follow- time-dependent effect of pregnancy on the risk of breast cancer.
ing chemotherapy cessation could be regarded as permanent, It has been suggested that the short-term increase in breast cancer
chemotherapy-induced menopause. The main determinant of risk following pregnancy could be secondary to the process of
developing permanent menopause following chemotherapy is postnatal breast involution, which is a tissue-remodeling pro-
age [13] . It is estimated that approximately 50–90% of women cess analogous to wound healing in which angiogenesis, inflam-
above the age of 40 years will develop permanent menopause mation and extracellular matrix alterations are activated  [30,31] .
secondary to chemotherapy, which will vary according to the Asztalos and colleagues showed that TGFb3, which is important
number of chemotherapy cycles and the drugs used, particularly for extracellular matrix stability, is downregulated in women
the cumulative dose of cyclophosphamide. The risk is significantly without breast cancer who have been pregnant within the pre-
lower in younger women (i.e., <40 years of age), ranging from ceding 2 years, which was not the case in those who had been
15 to 45% [17–19] . Newer regimens utilizing taxanes, dose-dense pregnant 5–10  years ago or in nulliparous women [32] . The
For personal use only.

administrations and trastuzumab did not appear to increase the effect of pregnancy on the mammary stem cells has also been
risk of amenorrhea observed with older regimens [16,20] . Table 1 proposed as a potential hypothesis behind the associated short-
summarizes the risk of amenorrhea associated with different term risk. In this study, an Australian group recently found in
­chemotherapy regimens. animal models a transient 11-fold increase in mammary stem
cell (MaSC) numbers following pregnancy [33] . It is suggested
Pregnancy & its relationship with breast cancer that pregnancy could stimulate MaSCs to grow via the effect
There is a strong body of evidence linking pregnancy to the risk of growth hormone (GH) of pregnancy on the MaSCs, which
of developing breast cancer [21] . This started centuries ago, when express GH receptors [34–36] .
it was noted that nuns had an elevated breast cancer risk [22] . By contrast, pregnancy-related hormonal changes seem to be
This observation led to the hypothesis that pregnancy is protec- more related to the long-term protective effect than the short-
tive against breast cancer, which was subsequently supported by term transient increase in risk. In a study conducted in nullipa-
numerous epidemiological studies [23–25] . One outcome of these rous women and two groups of parous women (2 and 10 years
studies was the finding that the protective effect is not immedi- following pregnancy), there was reduced expression of estrogen
ate and that there is a transient increase in breast cancer risk for receptor (ER)a (p = 0.006) and increased expression of ERb
a few years following pregnancy. This was clearly demonstrated (p = 0.003) in both parous groups compared with nulliparous
in a large population-based study from Norway involving more women [32] .

Table 1. Risk of chemotherapy-induced amenorrhea Pregnancy following breast cancer:


according to age. understanding the risks
Several case–control and population-based studies
Chemotherapy Age <30 years Age 30–40 Age >40 years Ref.
have been conducted to try to address the safety of
regimen (%) years (%) (%)
pregnancy following breast cancer diagnosis (Table 2)
AC × 4 13 57–63 [15,17]
[37–52] . None of these studies have shown a detrimen-
CMF × 6 19 31–38 76–96 [12,13] tal effect of pregnancy on breast cancer outcome.
CAF/CEF × 6 23–47 23–47 80–90 [16,17] Moreover, a recent study demonstrated that the mean
[19]
10‑year cumulative mortality of breast cancer patients
FEC 100 × 6 †
76.3 76.3
with subsequent pregnancy was within the range of
FEC 100 × 3 – D × 3 †
63.5 63.5 [19]
the 10‑year mortality predicted by ‘Adjuvant!Online’

Values provided are for premenopausal patients and are not age specific. for women with T1 N0 tumors in perfect health [52] .
AC: Doxorubicin, cyclophosphamide; CAF: Cyclophosphamide, doxorubicin, 5-fluorouracil;
CEF: Cyclophosphamide, epirubicin, 5-fluorouracil; CMF: Cyclophosphamide, methotrexate, Nevertheless, available evidence suggests that breast
5-fluorouracil; D: Docetaxel; FEC: 5-fluorouracil, epirubicin, cyclophosphamide. cancer survivors are frequently counseled against

288 Expert Rev. Anticancer Ther. 11(2), (2011)


Expert Review of Anticancer Therapy Downloaded from informahealthcare.com by Washington University Library on 12/28/14
For personal use only.

Table 2. Studies comparing survival in women who got pregnant following breast cancer diagnosis with breast cancer controls.
Study (year) Patients who Patients who did Age at Study design Overall survival Matching criteria for Ref.
got pregnant not get pregnant diagnosis relative risk (95% CI) choosing controls
after BC (n) after BC (n) (years)
Cooper et al. (1970) 28 56 ≤40 Matched CC 0.64 (0.31–1.31) Age, stage, nodal status [37]

www.expert-reviews.com
Mignot et al. (1986) 68 136 ≤45 Matched CC 0.86 (0.34–2.18) Age, year of tumor treatment, [38]
stage, histology
Ariel et al. (1989) 46 900 22–45 Population based 0.85 (0.55–1.33) NA [39]

Sankila et al. (1994) 91 471 <40 Matched CC 0.21 (0.10–0.45) Age, year of diagnosis, stage [40]

Malamos et al. (1996) 21 222 <35 Hospital based 0.55 (0.39–0.77) NA [41]

Lethaby et al. (1996) 14 334 <45 Population based 0.78 (0.58–1.05) NA [42]

Velentgas et al. (1999) 53 265 <45 Matched CC 0.80 (0.30–2.30) Stage [43]

Birgisson et al. (2000) 14 33 <50 Matched CC 0.54 (0.25–1.13) Year of diagnosis, tumor size, [44]
nodal status
Gelber et al. (2001) 94 188 <53 Matched CC 0.44 (0.21–0.96) Sage, year of diagnosis, nodal [45]
status, tumor size
Mueller et al. (2003) 328 2002 <45 Matched CC 0.54 (0.41–0.71) Age, race/ethnicity, year of [46]
diagnosis, stage
Blakely et al. (2004) 47 323 <35 Hospital based 0.47 (0.27–0.82) NA [47]

Ives et al. (2007) 123 2416 <45 Population based 0.59 (0.37–0.95) NA [48]

Kroman et al. (2008) 199 10037 <45 Population based 0.73 (0.54–0.99) NA [49]

Largillier et al. (2009) 118 762 <35 Hospital based 0.23 (0.10–0.52) NA [50]
† [51]
Kranick et al. (2010) 107 344 <45 Matched CC 1.2 (0.8–2.0) Age, year of diagnosis, stage
Verkooijen et al. (2010) 492 8529 <40 Population based Not provided‡ NA [52]

Hazard ratio.

Provided mortality rates that were lower in the pregnancy group compared with the non-pregnancy group 16.8 (95% CI: 13.3–20.9).
BC: Breast cancer; CC: Case–control; NA: Not applicable; NR: Not reported.
Motherhood after breast cancer: searching for la dolce vita
Review

289
Review Azim Jr, Peccatori, de Azambuja & Piccart

future conception. In a study by Gelber and colleagues, 23 out 15% nonsignificant reduction in the risk of death (hazard ratio:
of 33 induced abortions (69%) were recorded as having been 0.85; 95% CI: 0.53–1.35). This result further suggests that preg-
recommended by the treating physician [45] . Similar observations nancy does not confer a detrimental effect on breast cancer sur-
were encountered in other studies as well [10] . This highlights vival. It also points out that pregnancy might exert a protective
a wide perception within the medical community that preg- effect that could be restricted to a certain breast cancer subtype
nancy worsens the prognosis of these women. It is postulated (e.g., hormone receptor positive). Unfortunately, all of these stud-
that the hormonal milieu of pregnancy could stimulate breast ies included patients treated a long time ago, and thus none of
cancer recurrence. them provided information regarding hormone receptor status.
In an attempt to refine our understanding in this area, we To address this point, we are currently conducting a multicentric
recently conducted a meta-ana­lysis addressing the safety of preg- case–control study to address the effect of pregnancy on different
Expert Review of Anticancer Therapy Downloaded from informahealthcare.com by Washington University Library on 12/28/14

nancy following breast cancer diagnosis and its effect on overall breast cancer subtypes. It is important to note that promoting
survival [53] . In this study, we analyzed 14 case–control, popu- the possible protective effect of pregnancy in these patients is
lation-based and hospital-based studies published between 1970 not of high clinical relevance, as pregnancy is not meant to be a
and 2009. The primary objective was to examine the effect of therapeutic intervention against breast cancer. However, we need
pregnancy on overall survival in the whole population. We then to ensure that pregnancy does not adversely affect breast cancer
performed sensitivity analyses using published and individual outcome, and thus improve the counseling of women wishing
patient data from three studies to better characterize the effect of to conceive.
pregnancy on different subgroups. The ana­lysis included nearly The timing of pregnancy following breast cancer diagnosis is
20,000 patients, of whom 1417 patients got pregnant following another point that deserves emphasis. To address this, we per-
breast cancer diagnosis and were compared with breast cancer formed a sensitivity ana­lysis by separately analyzing patients who
patients who did not get pregnant. Seven studies had matched got pregnant within 6–24 months following breast cancer diag-
controls and matching criteria were mainly age, stage and year of nosis and those who got pregnant at a later time. We found that
diagnosis. The primary ana­lysis showed that patients who became pregnancy 2 years following breast cancer diagnosis conferred a
pregnant had a 41% reduced risk of death compared with breast significant reduction in overall mortality, with no evidence of het-
For personal use only.

cancer controls (hazard ratio: 0.59; 95% CI: 0.50–0.70). This erogeneity (pooled relative risk: 0.55; 95% CI: 0.36–0.84; p-value
apparent protective effect was seen mainly in patients under the for heterogeneity = 0.16). The same findings were observed in
age of 35 years with lymph node-negative disease. women becoming pregnant within 6–24 months of diagnosis,
The major drawback of this ana­lysis remains in the selection yet with significant heterogeneity (I 2 : 71.5%; p  =  0.007). In
of controls. It was previously suggested that patients who became this context, it is important to point out the bidirectional, time-
pregnant following breast cancer diagnosis are possibly healthier, dependent effect of pregnancy on breast cancer observed in the
what was described as the ‘healthy mother effect’ [40] . Recently, epidemiological studies of the general population [26–28] . As noted
a group from the University of California (CA, USA) evalu- earlier, biological evidence suggests that pregnancy might serve as
ated the association between mental and physical health scores, a fertile soil for incipient lesions in the breast in the short term [31] .
and childbearing after breast cancer [54] . This study compared In addition, a recent population-based study from Singapore has
27 women having children after breast cancer and 54 age and shown that the risk of death decreases significantly with increas-
stage-matched breast cancer controls. The assessments were car- ing interval between diagnosis and subsequent childbirth [52] .
ried out before the pregnancy events took place. Interestingly, Hence, owing to the reassuring results of patients who got preg-
they demonstrated that mental but not physical health scores nant more than 2 years following breast cancer diagnosis, and
were marginally different between cases and controls (p = 0.08). the possible adverse effect of pregnancy and high incidence of
Thus, according to this study, the perception that breast can- tumor recurrence during the first 2 years [55] , a minimum period
cer patients who subsequently became pregnant were physically of 2 years following diagnosis should be allowed before attempting
healthier does not seem to be entirely true. However, in most of to get pregnant. This would also allow patients to recover from
the previous studies, selection of controls did not account for the chemotherapy-induced ovarian toxicity.
time to relapse, as relapsing patients would not consider getting For patients with a history of ER-positive breast cancer, 5 years
pregnant, and thus the observed protective effect of pregnancy of adjuvant hormonal therapy remains the current standard of
could reflect selection bias. care [56] , and women should complete their treatment before con-
In the meta-ana­lysis, a sensitivity ana­lysis involving four stud- sidering pregnancy. A conflict will always remain for those women
ies was carried out, in which patients who got pregnant follow- with ER-positive disease who are willing to interrupt hormonal
ing breast cancer diagnosis were compared with controls who therapy to become pregnant. These women should be counseled
were known to be free of relapse at a time interval equivalent that the interruption of hormonal therapy could be detrimen-
to that elapsing between breast cancer diagnosis and pregnancy tal for their breast cancer outcome, and this should therefore
in the investigational group [38,43,45,49] . The ana­lysis included be discouraged. Nevertheless, an interesting research question
414 patients who became pregnant following breast cancer diag- remains open in whether or not women interrupting their adju-
nosis and were compared with 10,626 breast cancer controls. The vant endocrine therapy and becoming pregnant would benefit
results showed that subsequent pregnancy is associated with a from ­restarting hormonal treatment after childbirth.

290 Expert Rev. Anticancer Ther. 11(2), (2011)


Motherhood after breast cancer: searching for la dolce vita Review

It is frequently quoted that pregnancy in women with a his- hormonal therapy were prone to the in utero toxic effects of these
tory of breast cancer is associated with a higher risk of abor- agents. Unfortunately, the Swedish trial did not provide detailed
tion compared with the general population [57] . Caution should ­information in this regard.
be taken when interpreting this data. In most of the studies On the other hand, only four neonatal deaths were reported,
reported, no clear distinction was made between spontaneous with no differences observed between the two groups. The higher
and induced abortion, the latter being frequently advocated by incidence of women above the age of 35 years in the breast can-
treating physicians as mentioned earlier [8] . However, on review- cer group (53 vs 11%) could partly explain the increased risk of
ing the results of studies that provided detailed information on complications; however, this was also observed in the Danish
abortion, the risk of spontaneous abortion does not appear to study (49 vs 9%) [59] , with no differences observed in the rate
be significantly higher in breast cancer survivors compared with of pregnancy complications. Another explanation could be the
Expert Review of Anticancer Therapy Downloaded from informahealthcare.com by Washington University Library on 12/28/14

the general population. In the second largest population-based fact that more breast cancer survivors in the Swedish study had
study reported by the Danish group, which involved 371 women a history of infertility for more than 1 year compared with the
with a history of breast cancer, only 15 women (4%) experienced control arm (OR: 2.53; 95% CI: 1.6–3.9).
spontaneous abortion [49] . This compares very favorably to the
risk in the general population, which is in the range of 15% [58] . Breastfeeding & breast cancer risk
In the same study, 157 patients (42%) were subjected to induced Breastfeeding is known to have immediate and long-lasting
abortion. The results of this study clearly reflect the potential advantages for both infant and mother. Breast-fed newborns
bias we might face on interpreting the data on reported abor- suffer lower rates of neonatal infection, autoimmune disease and
tions without specifying the reason (i.e., spontaneous or induced), subsequent obesity, and benefit from better neuropsychological
which results in overestimating the risk of spontaneous abortion and cognitive development [61–63] . For mothers, lactation facili-
in breast cancer survivors. The same was observed in a large tates maternal weight loss following delivery, controls postpartum
Australian population-based study involving 175 pregnancies bleeding and reduces blood glucose levels in women experienc-
in women with previous history of breast cancer [48] . In this ing gestational diabetes mellitus [63,64] . Importantly, it provides
study 12% experienced spontaneous abortion, while 34% had a unique opportunity for the mother to naturally bond with her
For personal use only.

elective termination. newborn, which could have a considerably positive psychological


Two large studies have assessed the fetal outcome in women effect, particularly for a woman with a history of breast cancer [65] .
with a history of breast cancer [59,60] . In a large Danish study, In 2002, the Collaborative Group on Hormonal Factors and
216  women who became pregnant following breast cancer Breastfeeding conducted the largest ana­lysis evaluating the effect
diagnosis were compared with a large cohort of 10,453 women of breastfeeding on breast cancer incidence in healthy women [66] .
without breast cancer [59] . The mean gestational age at deliv- They analyzed 47 case–control and cohort studies from 30 dif-
ery was 39 weeks in both groups, with similar risks of preterm ferent countries, which included at least 100 women with subse-
delivery, low birth weight and congenital anomalies (6.5, 1.5 quent breast cancer. The relative risk reduction of breast cancer
and 3.4%, respectively). By contrast, a larger Swedish popula- incidence was 4.3% (95% CI: 2.9–5.8; p < 0.0001) for each year
tion-based study has suggested that pregnancy in women with of breastfeeding. Subgroup analyses found no differences related
a history of breast cancer should be regarded as ‘high risk’ [60] . to age, ethnicity, parity, age at first delivery, or BMI. The mag-
In this study, 331 women who first gave birth at a mean time nitude of benefit appears to be even higher in BRCA1 carriers.
of 3 years following breast cancer diagnosis were identified out Jernstrom and colleagues have shown that women with BRCA1
of a total number of 2,870,932 births registered in the Swedish mutation who breastfed their babies for more than 1 year had a
database between 1973 and 2002. The large majority (~91%) 45% reduction in the risk of developing breast cancer compared
of births from women previously treated for breast cancer had with women who never breast fed (OR: 0.55; 95% CI: 0.38–0.80;
no adverse events. However, this group was associated with an p = 0.001) [67] .
increased risk of delivery complications (odds ratio [OR]: 1.5; Several hypotheses have been postulated to explain the protec-
95% CI: 1.2–1.9), cesarean section (OR: 1.3; 95% CI: 1.0–1.7), tive effect of breastfeeding on breast cancer incidence. Some stud-
very preterm birth (<32  weeks; OR: 3.2; 95%  CI: 1.7–6.0) ies have linked the reduced risk of developing breast cancer with
and low birth weight (<1500 g; OR: 2.9; 95% CI: 1.4–5.8). the duration of lactation amenorrhea [68] . Others have pointed
Despite this, it is important to point out that the large majority to the role of prolactin, which is a key hormone involved in both
of infants were born at full term, with Apgar scores at 5 min mammary development and lactation. The effect of prolactin on
of seven or more. A slight increase in the incidence of congeni- breast cancer development has been the object of considerable con-
tal malformations were encountered in the breast cancer group troversy, although evidence currently favors the view that elevated
compared with the general population (7 vs 4%, respectively). prolactin levels could be protective against breast cancer [69] . Early
The reason for this difference, which was not observed in other anecdotal evidence suggested breast cancer regression following
studies, is not very clear. This study included patients who got pituitary stalk section [70] . Later studies in the adjuvant setting
pregnant as early as 7 months following breast cancer surgery, have shown that high postoperative serum prolactin levels and
with a mean period of approximately 3 years. It is possible that the presence of activated Stat5, which is a prolactin ­transcription
those women who got pregnant shortly after chemotherapy or factor, are associated with better prognosis [71–73] .

www.expert-reviews.com 291
Review Azim Jr, Peccatori, de Azambuja & Piccart

Breastfeeding: safety & feasibility following breast of the remaining 18 women, lactation occurred in ten (56%), did
cancer diagnosis not occur in seven (39%) and was unknown in one (5%). Breast
There are no epidemiological data regarding breastfeeding fol- volume was reported to be significantly diminished in 80% of
lowing breast cancer. To our knowledge, only two reports have treated breasts. This observation is consistent with that of our
addressed the effect of breastfeeding on breast cancer outcome. group and is probably related to radiotherapy-induced fibrosis.
The first was conducted on 94 patients who got pregnant follow- The proximity of the incision to the areola and nipple, the loca-
ing breast cancer diagnosis and were previously enrolled in an tion of the tumor, and the dose and type of radiotherapy are all
International Breast Cancer Study Group case–control study [45] . factors contributing to lactation success from the treated breast
Data on breastfeeding were reported by the referring oncolo- [80] . However, it is important to point out that none of the previ-
gist and lacked details about duration, exclusiveness and site of ously reported studies described compromised milk production
Expert Review of Anticancer Therapy Downloaded from informahealthcare.com by Washington University Library on 12/28/14

lactation in cases of breast-conserving surgery. Nonetheless, 27 from the contralateral breast.


of the 94 women were reported as having breastfed their babies, In a recent exploratory ana­lysis on the experience of breast-
25 were reported as having bottle-fed, while 42 had unknown feeding in breast cancer survivors, Gorman and colleagues dem-
nursing status. Breastfeeding was not considered in the original onstrated that a number of themes emerged from their inter-
survival ana­lysis, but subsequent review suggested that breast- views  [65] . These included uncertainty regarding the possibility
feeding was not detrimental, but was rather associated with bet- of breastfeeding, worries about nursing from one side only and
ter survival [74] . At a median follow-up of 10 years from breast the challenge of not having sufficient milk production. Patients
cancer diagnosis, only one patient died out of 27 women who and physicians should be informed that the milk produced by one
breastfed their babies (3%), compared with six patients out of breast is sufficient for the nutritional needs of a newborn [81] . The
25 women who did not breastfeed (24%). Those with unknown experience of mothers who chose to use exclusively one breast for
nursing status had a survival somewhere in between (four out breastfeeding validates this notion [82] , together with the historical
of 42 women; 9%). While this ana­lysis is very small and lacks habit of wet nursing more than one child [83] .
much vital information regarding lactation habits and duration, Counseling remains a crucial point that could have a signifi-
it emphasizes that breastfeeding does not seem to worsen breast cant impact on the success of breastfeeding in women who have
For personal use only.

cancer outcome. had breast cancer. We observed in our survey that the main rea-
We recently published a study addressing the pattern of breast- sons for not initiating breastfeeding were uncertainty regarding
feeding in a series of 20 patients who got pregnant following maternal safety and a priori unfeasibility expressed either by the
the completion of adjuvant systemic therapy for breast cancer. ­obstetrician or by the oncologist [75] .
We also investigated the effect of breastfeeding on breast cancer By contrast, we have also observed that proper lactation coun-
outcome [75] . In this study, ten women (50%) initiated breast- seling had a positive effect on the success of breastfeeding and in
feeding, four (20%) stopped within 1 month and six (30%) had facing the different challenges highlighted earlier. In our study,
long-term success with a median period of 11 months (range: five out of 20 women were offered qualified lactation counseling
7–17 months). At a median follow-up of 48 months following [75] . All five women – including a woman who breastfed twice –
delivery, all 20 women were alive with two relapses occurring, enjoyed successful prolonged breastfeeding with a median dura-
one in each group (i.e., lactating and nonlactating). Although tion of 12 months (range: 7–17 months). These results underscore
we acknowledge the small size of the study, it confirms the previ- the need for and importance of proper counseling.
ous observation regarding the lack of the apparent detrimental
effects of breastfeeding on breast cancer outcome. This is further Fertility preservation
endorsed by the Society of Obstetricians and Gynecologists of In the previous sections, we critically analyzed available evidence
Canada, which clearly states that “women previously treated for on the feasibility and safety of pregnancy and breastfeeding in
breast cancer who do not show any evidence of residual tumor breast cancer survivors. Despite the limitations of the available
should be encouraged to breastfeed their children” [76] . data, it appears that these women can safely get pregnant and con-
However, it is evident that breast cancer survivors are faced with sider breastfeeding. However, as mentioned earlier, a significant
considerable challenges when they attempt breastfeeding. Several proportion of breast cancer patients develop permanent amenor-
studies have reported that milk production is significantly reduced rhea secondary to adjuvant chemotherapy and are thus denied
in the breast that was previously subjected to a conservative sur- this opportunity. In addition, the ovarian reserve of those who
gery and radiotherapy [77–79] . This is in addition to nipple pain experienced temporary amenorrhea is compromised [15] . Hence,
and discomfort during latching [75] . An early study has shown it is vital to try to preserve the ovarian function of women who
that only four out of ten patients were able to lactate from the are considering pregnancy following the completion of breast
treated breast, while no similar problems were encountered in cancer therapies. Several methods have been proposed to preserve
the contralateral breast [77] . Later on, Moran and colleagues ret- their fertility. These include ovarian function suppression, in vitro
rospectively analyzed over 3000 patients treated in their hospital fertilization with embryo cryopreservation, ovarian tissue freez-
from 1965 to 2003, and were able to identify 29 pregnancies in ing and oocyte cryopreservation [84] . For the sake of this article,
21 patients (one patient had bilateral breast cancer) [78] . Four we will focus only on the former two techniques, as the latter
women elected pharmacological suppression of lactation and out ­methods remain highly investigational for the time being.

292 Expert Rev. Anticancer Ther. 11(2), (2011)


Motherhood after breast cancer: searching for la dolce vita Review

Table 3. Phase II studies of luteinizing hormone-releasing hormone agonists to preserve fertility in


breast cancer.
Author (year) Patients Median age in LHRH agonist Chemotherapy used Resuming Pregnancies Ref.
(n) years (range) menses (n) (%) (n)
Fox et al. (2003) 24 35 (23–42) Leuprolide AC, AC-T, CAF, AC-CMF 23 (96%) 6 [85]

Del Mastro et al. 30 38 (29–47) Goserelin CEF × 6 21 (72%) NR [86]


(2006)
Recchia et al. 100 43 (27–50) Goserelin CMF/FEC ± T, E-CMF ± T, 67 (67%) 3 [87]
Expert Review of Anticancer Therapy Downloaded from informahealthcare.com by Washington University Library on 12/28/14

(2006) E-HDCT-CMF
Urruticoechea 51 34† (20–34) Goserelin AC/FEC ± T 45 (90%) 8 [88]
et al. (2008)

Mean age.
AC: Doxorubicin, cyclophosphamide; CAF: Cyclophosphamide, doxorubicin, 5-fluorouracil; CMF: Cyclophosphamide, methotrexate, 5-fluorouracil; E: Epirubicin;
FEC: 5-fluorouracil, epirubicin, cyclophosphamide; HDCT: High-dose chemotherapy; LHRH: Luteinizing hormone-releasing hormone; NR: Not reported; T: Taxane.

Several Phase II studies have been published addressing the women with breast cancer. The first randomized trial was pre-
role of luteinizing hormone-releasing hormone (LHRH) agonists sented at the Annual Meeting of the American Society of Clinical
in preserving the fertility of young women undergoing adjuvant Oncology (ASCO) 2008 and included only 49 patients (median
systemic chemotherapy (Table 3) [85–88] . These studies were nonran- age: 39 years) who were randomized to chemotherapy with or
domized, without a control group and involved a small number of without triptorelin [89] . No difference was found between the two
patients (range: 24–100 patients). Only one study used the ability treatment arms with respect to the risk of chemotherapy-related
to conceive and live birth as end points. In this study, six pregnan- amenorrhea at 18 months of follow-up. The median time to return
cies occurred in 23 women [85] . It should be noted that this study of menses was 6.1 months in the triptorelin arm, compared with
For personal use only.

was first presented in 2003 and was not published until now. The 4.7 months in the control arm (p = 0.79).
remaining studies had as their main end point the percentage The second study was recently published by an Egyptian group
of patients who resumed menstruation following chemotherapy, in which 78 patients were randomized to six cycles of standard
which ranged from 67 to 100%. This is a major limitation because chemotherapy (5-fluorouracil, doxorubicin, cyclophosphamide)
resuming menstruation is not a good surrogate for ovarian reserve with or without goserelin [90] . In this study, the goserelin group
and ability to conceive, as highlighted earlier [15] . had a significantly lower incidence of premature ovarian failure
Recently, three randomized trials were conducted to address (11 vs 66%; p < 0.001), a higher incidence of ovulation (69 vs
the potential role of LHRH in preserving the fertility of young 25%; p < 0.001) and higher E2 levels (279 vs 75; p < 0.001).

Table 4. Ongoing randomized studies investigating the role of luteinizing hormone-releasing hormone in
fertility preservation of patients with invasive breast cancer.
Sponsor Target Design Primary objective Secondary objective
accrual (n)
Southwest Oncology 416 Phase III; adjuvant (neo) Rate of premature Ovarian reserve rate
Group cyclophosphamide-based regimen ovarian failure Pregnancy rate
± goserelin in HR- Ovarian dysfunction rate
Anglo Celtic Cooperative 400 Phase III; adjuvant (neo) Resuming menses within Incidence of menopausal
Oncology Group cyclophosphamide and/or 1 year symptoms
anthracycline-based regimen QoL
± goserelin BMD loss
Hormone levels
Pregnancy rate
MD Anderson 148 Phase III; adjuvant (neo) Resuming menses within Pregnancy rate
Cancer Center chemotherapy ± goserelin in HR- 1 year QoL
DFS, OS
German Breast Group 62 Phase II; adjuvant (neo) Resuming menses within Treatment delay
anthracycline-based regimen 8 months Toxicity
± goserelin in HR- QoL
Pregnancy rate
BMD: Bone mineral density; DFS: Disease-free survival; HR-: Hormone receptor negative; OS: Overall survival; QoL: Quality of life.
Data taken from [202].

www.expert-reviews.com 293
Review Azim Jr, Peccatori, de Azambuja & Piccart

While both trials are very small, the second study was larger breast cancer outcome. Ethical and social considerations also exist.
in size (78 vs 49 patients in the former) with a younger patient This procedure requires a partner, and the fate of the cryopreserved
population (median age: 29 vs 39 years in the former). These fac- embryo, if available, remains a problem in case the patient died
tors could partly explain the different results obtained. However, before implantation. These latter two disadvantages to embryo
the latter trial suffered some limitations that should be taken preservation could therefore represent a potential ­advantage for
into consideration [91,92] . Only 33% of the patients in the control ovarian tissue freezing and oocyte cryopreservation.
arm resumed their menses, which is very low compared with Oktay and colleagues have conducted a series of studies to
approximately 70% in previous reports for patients of the same address some of these concerns [96,99–102] . They first used high-
age treated with similar regimens. Moreover, patients were fol- dose tamoxifen (60 mg) for ovarian stimulation, which resulted
lowed up for only 8 months, which does not allow for adequate in a 2.5-fold increase in embryo yield compared with natural-
Expert Review of Anticancer Therapy Downloaded from informahealthcare.com by Washington University Library on 12/28/14

ovarian function recovery of the control group, which could take cycle IVF, with no breast cancer recurrences observed in the short
up to 12 months. Finally, the design of the study and its short term [96] . Later, they investigated the role of letrozole for ovarian
follow-up did not allow for investigation of the fertility rate in stimulation and compared it with tamoxifen [100] . In this study,
both groups, which remains the main question. In the 2010 60 patients with primary breast cancer were enrolled in which
ASCO meeting, Del Mastro presented on behalf of the Gruppo 29 patients underwent ovarian stimulation using three different
Italiano Mammella the results of the largest randomized Phase III regimens: tamoxifen; tamoxifen plus low-dose follicle-stimulating
randomized clinical trial, which investigated the effect of adding hormone (FSH); and letrozole plus low-dose FSH. Embryo yield
triptorelin to modern chemotherapy on preserving the ovarian was compared across the three regimens, and recurrence rates
function [93] . This study included 281 patients with a median were compared with the remaining 31 patients who elected not
age of 39 years (range: 18–45 years) who received a median of to undergo IVF. In this study, the letrozole arm was associated
six adjuvant chemotherapy cycles. The triptorelin arm was asso- with the highest embryo yield (5.3 ± 0.8 embryos) compared with
ciated with a 19% absolute reduction in the risk of amenorrhea tamoxifen–FSH (3.8 ± 0.8 embryos; p < 0.05) and tamoxifen alone
(p = 0.0002). Premenopausal estrogen levels were more frequently (1.3 ± 0.2  embryos; p < 0.001). Furthermore, the letrozole arm
observed in those who received triptorelin compared with those was associated with the lowest estradiol peak (380 ± 57 pg/ml),
For personal use only.

who received chemotherapy alone (77 vs 58%; p = 0.006). This which was significantly lower than the other two arms (p < 0.05).
study is not yet published, and it is still too early to comment At a median follow-up of 18 months, there was no difference in the
on the fertility rates in both treatment groups. However, if these rate of recurrence between the IVF and the control groups (three
observations led to an improvement in fertility rates, and con- out of 29 vs three out of 31 patients). Given the appealing results
firmed by other ongoing trials this could certainly place LHRH observed with letrozole, Oktay’s group more recently conducted
agonists as a favored intervention to preserve fertility of young a prospective study involving 215 patients to determine the effect
women with breast cancer. Currently, several randomized clinical of controlled ovarian stimulation using letrozole plus FSH on
trials are investigating the role of LHRH in fertility preservation disease-free survival in women undergoing embryo or oocyte cryo-
to confirm these results (Table 4) . preservation prior to starting adjuvant chemotherapy [102] . A total
On the other hand, embryo cryopreservation has been an estab- of 215 women were prospectively evaluated, of whom 79 elected
lished and widely available method in treating infertility since the to undergo controlled ovarian stimulation and the rest served as
early 1980s. In the USA, the delivery rate is approximately 30 and controls. The results showed that the time to starting adjuvant
16% per embryo thaw in patients younger than 35 years and older chemotherapy was longer for patients who underwent controlled
than 40 years of age, respectively [201] . In vitro fertilization (IVF) ovarian stimulation (45 vs 33.4 days; p < 0.01). However, at a
in cancer patients for embryo ‘banking’ for future use has been median time of almost 2 years, there were no differences observed
reported by several groups [94–96] . As patients undergoing IVF in terms of disease-free and overall survival (p = 0.36). When we
with embryo cryopreservation are generally not infertile, preg- consider that it is impossible to have a randomized trial in this
nancy rates might well be higher than those achieved by couples setting, this study represents a real step forward in evaluating the
who potentially have poor oocytes and sperm quality. risk of ovarian stimulation on breast cancer outcome. However,
However, it is important to take into account several points when longer follow-up is needed to better define such risk.
considering IVF for breast cancer patients. The use of traditional
ovarian-stimulation regimens result in a significant increase in Expert commentary
estradiol levels (ten- to 20-times the levels in a regular menstrual Based on the available evidence, women with a history of success-
cycle) [97] . Despite this lasting for only a few days (~10 days), fully treated breast cancer should not be denied the opportunity
oncologists fear that the estrogenic milieu could stimulate breast to get pregnant or to breastfeed their children. While treatment
cancer progression or relapse. To address this point, some patients modalities used to treat breast cancer (surgery, radiotherapy and
were offered IVF during unstimulated cycles (natural-cycle IVF); chemotherapy) could reduce the chances of subsequent conception
however, this technique is associated with low embryo yield [96,98] . and breastfeeding, once encountered, they do not adversely affect
In addition, the fact that ovarian stimulation takes place prior to breast cancer prognosis. Thus, physicians should take this informa-
commencing adjuvant chemotherapy could result in a significant tion into account when counseling women with a history of breast
delay in initiating therapy, which could have a detrimental effect on cancer and enquiring into the safety of subsequent pregnancy and

294 Expert Rev. Anticancer Ther. 11(2), (2011)


Motherhood after breast cancer: searching for la dolce vita Review

breastfeeding. In addition, and as stressed in the recent ASCO The coming few years will reveal much about the role of
guidelines, they should be considered for fertility preservation [103] . LHRH in fertility preservation. Several large randomized tri-
To date, there is no established method to preserve the fertility of als have completed their accrual, and their results are eagerly
young women undergoing chemotherapy. However, data about awaited. These studies will provide sufficient evidence on the effect
LHRH agonists and embryo cryopreservation is promising. of LHRH on ovarian function, ovarian reserve and rate of preg-
Fertility counseling needs to be better addressed in breast can- nancy, despite being tested as secondary end points. In parallel,
cer clinics. Denying breast cancer survivors the opportunity to one can expect progressive improvements in ovarian tissue freezing
become pregnant and/or breastfeed remains unjustified in the and oocyte cryopreservation techniques, which would help over-
absence of supporting evidence. This would indeed improve the come the ethical and social considerations associated with embryo
quality of life of these women and help them restore normalcy ­cryopreservation for breast cancer patients.
Expert Review of Anticancer Therapy Downloaded from informahealthcare.com by Washington University Library on 12/28/14

in their lives.
Financial & competing interests disclosure
Five-year view Hatem A Azim Jr acknowledges a translational research fellowship from
Several questions remain unanswered regarding the impact of the European Society for Medical Oncology (ESMO) and a PHD grant
pregnancy on breast cancer outcome in women with a history of from the Universite libre de Bruxelles (ULB). The authors have no other
breast cancer. For example, although it appears that pregnancy relevant affiliations or financial involvement with any organization
following breast cancer is generally safe, too little is known about or entity with a financial interest in or financial conflict with the sub-
the timing of pregnancy and its effect according to breast cancer ject  matter or materials discussed in the manuscript apart from
subtypes. In this regard, we are currently conducting a multi- those disclosed.
centric case–control study, which will be adequately powered to The authors would like to thank Carolyn Straehle for her editorial
address these points. ­assistance during the preparation of the manuscript.

Key issues
For personal use only.

• All published studies mentioned in this article and a recently conducted meta-ana­lysis confirm that pregnancy does not worsen the
survival rates of women with a history of breast cancer.
• There is a lack of sufficient evidence on the timing of pregnancy and outcome according to the endocrine responsiveness of a tumor.
However, waiting for a minimum of 2 years following completion of breast cancer therapy in patients with estrogen receptor-negative
tumors is a reasonable option. For those with estrogen receptor-positive breast cancer, 5 years of hormonal therapy should be
completed before attempting to become pregnant.
• Data on the safety of breastfeeding is very scarce; however, available evidence does not show any detrimental effects. The challenges
and fears faced by the patient entails offering them proper lactation counseling.
• The success rate of breastfeeding from the ipsilateral breast in women previously subjected to breast-conserving surgery and
radiotherapy is low (~4%), which is secondary to distortion in the nipple/areola complex and radiotherapy-induced fibrosis.
• The coadministration of luteinizing hormone-releasing hormone with chemotherapy appears to reduce the risk of permanent
amenorrhea secondary to chemotherapy. Whether or not this would result in improving the ovarian reserve and increasing the
pregnancy rate remains to be determined.
• Significant progress has been made in ovarian stimulation and embryo cryopreservation in breast cancer patients who are willing to
become pregnant afterwards. This approach could be considered in centers with a high experience dealing with such cases.

HER2-positive breast cancer have a 6 Berkowitz GS, Skovron ML, Lapinski RH,
References
poorer prognosis when compared with Berkowitz RL. Delayed childbearing and
Papers of special note have been highlighted as:
• of interest older patients with a similar breast the outcome of pregnancy. N. Engl. J. Med.
•• of considerable interest cancer subtype. 322(10), 659–664 (1990).

1 Fredholm H, Eaker S, Frisell J et al. Breast 3 Swanson GM, Lin CS. Survival patterns 7 Partridge AH, Gelber S, Peppercorn J et al.
cancer in young women: poor survival among younger women with breast cancer: Web-based survey of fertility issues in
despite intensive treatment. PLoS One the effects of age, race, stage, and young women with breast cancer. J. Clin.
4(11), e7695 (2009). treatment. J. Natl Cancer Inst. Monogr. 16, Oncol. 22(20), 4174–4183 (2004).
69–77 (1994). 8 Peccatori FA, Azim HA Jr. Pregnancy in
2 Cancello G, Maisonneuve P, Rotmensz N
et al. Prognosis and adjuvant treatment 4 Colleoni M, Rotmensz N, Robertson C breast cancer survivors: a need for proper
effects in selected breast cancer subtypes of et al. Very young women (<35 years) with counseling. Breast (Edinb., Scotland) 18(6),
very young women (<35 years) with operable breast cancer: features of disease at 337–338 (2009).
operable breast cancer. Ann. Oncol. 21(10), presentation. Ann. Oncol. 13(2), 273–279 9 Rippy EE, Karat IF, Kissin MW.
1974–1981 (2010). (2002). Pregnancy after breast cancer: the
5 Jemal A, Siegel R, Ward E et al. Cancer importance of active counselling and
• Large study demonstrating that young
statistics, 2009. CA Cancer J. Clin. 59(4), planning. Breast (Edinb., Scotland) 18(6),
women younger than 35 years of age with
225–249 (2009). 345–350 (2009).
triple-negative, luminal-B and

www.expert-reviews.com 295
Review Azim Jr, Peccatori, de Azambuja & Piccart

10 Peccatori F, Cinieri S, Orlando L, Bellettini cancer treatment: a prospective 32 Asztalos S, Gann PH, Hayes MK et al.
G. Subsequent pregnancy after breast study. J. Clin. Oncol. 24(7), 1045–1051 Gene expression patterns in the human
cancer. Recent Results Cancer Res. 178, (2006). breast after pregnancy. Cancer Prev. Res.
57–67 (2008). 20 Berliere M, Dalenc F, Malingret N et al. (Phila., PA) 3(3), 301–311 (2010).
11 Faddy MJ, Gosden RG, Gougeon A, Incidence of reversible amenorrhea in 33 Asselin-Labat ML, Vaillant F, Sheridan JM
Richardson SJ, Nelson JF. Accelerated women with breast cancer undergoing et al. Control of mammary stem cell
disappearance of ovarian follicles in adjuvant anthracycline-based function by steroid hormone signalling.
mid-life: implications for forecasting chemotherapy with or without docetaxel. Nature 465(7299), 798–802 (2010).
menopause. Hum. Reprod. (Oxford, Engl.) BMC Cancer 8, 56 (2008). 34 Dontu G, Abdallah WM, Foley JM et al.
7(10), 1342–1346 (1992). 21 Nechuta S, Paneth N, Velie EM. Pregnancy In vitro propagation and transcriptional
Gougeon A, Ecochard R, Thalabard JC. characteristics and maternal breast cancer profiling of human mammary stem/
Expert Review of Anticancer Therapy Downloaded from informahealthcare.com by Washington University Library on 12/28/14

12
Age-related changes of the population of risk: a review of the epidemiologic progenitor cells. Genes Dev. 17(10),
human ovarian follicles: increase in the literature. Cancer Causes Control 21(7), 1253–1270 (2003).
disappearance rate of non-growing and 967–989 (2010). 35 Dontu G, Al-Hajj M, Abdallah WM, Clarke
early-growing follicles in aging women. 22 Mustacchi P. Ramazzini and Rigoni-Stern MF, Wicha MS. Stem cells in normal breast
Biol. Reproduct. 50(3), 653–663 (1994). on parity and breast cancer. Clinical development and breast cancer. Cell Prolif.
13 Partridge AH, Gelber S, Peppercorn J et al. impression and statistical corroboration. 36(Suppl. 1), 59–72 (2003).
Fertility and menopausal outcomes in Arch. Int. Med. 108, 639–642 (1961). 36 Laban C, Bustin SA, Jenkins PJ. The
young breast cancer survivors. Clin. Breast 23 Albrektsen G, Heuch I, Tretli S, Kvale G. GH–IGF-I axis and breast cancer. Trends
Cancer 8(1), 65–69 (2008). Breast cancer incidence before age 55 in Endocrinol. Metab. 14(1), 28–34 (2003).
14 Partridge A, Gelber S, Gelber RD et al. Age relation to parity and age at first and last 37 Cooper DR, Butterfield J. Pregnancy
of menopause among women who remain births: a prospective study of one million subsequent to mastectomy for cancer of the
premenopausal following treatment for Norwegian women. Epidemiology breast. Ann. Surg. 171(3), 429–433 (1970).
early breast cancer: long-term results from (Cambridge, Mass.) 5(6), 604–611 (1994).
38 Mignot L, Morvan F, Berdah J et al.
International Breast Cancer Study Group 24 MacMahon B, Cole P, Lin TM et al. Age at [Pregnancy after treated breast cancer.
Trials V and VI. Eur. J. Cancer 43(11), first birth and breast cancer risk. Bull. World Results of a case–control study]. Presse
For personal use only.

1646–1653 (2007). Health Organ. 43(2), 209–221 (1970). Med. 15(39), 1961–1964 (1986).
15 Partridge AH, Ruddy KJ, Gelber S et al. 25 Rosner B, Colditz GA, Willett WC. 39 Ariel IM, Kempner R. The prognosis of
Ovarian reserve in women who remain Reproductive risk factors in a prospective patients who become pregnant after
premenopausal after chemotherapy for study of breast cancer: the Nurses’ Health mastectomy for breast cancer. Int. Surg.
early stage breast cancer. Fertil. Steril. study. Am. J. Epidemiol. 139(8), 819–835 74(3), 185–187 (1989).
94(2), 638–644 (2010). (1994).
40 Sankila R, Heinavaara S, Hakulinen T.
•• Demonstrates that ovarian reserve is poor 26 Albrektsen G, Heuch I, Hansen S, Kvale G. Survival of breast cancer patients after
in women who resume menstruation Breast cancer risk by age at birth, time subsequent term pregnancy: ‘healthy
following chemotherapy, highlighting since birth and time intervals between mother effect’. Am. J. Obstet. Gynecol.
that menses is an imperfect surrogate for births: exploring interaction effects. Br. J. 170(3), 818–823 (1994).
prediction of future fertility. Cancer 92(1), 167–175 (2005).
41 Malamos NA, Stathopoulos GP,
16 Abusief ME, Missmer SA, Ginsburg ES, 27 Lambe M, Hsieh C, Trichopoulos D et al. Keramopoulos A, Papadiamantis J,
Weeks JC, Partridge AH. The effects of Transient increase in the risk of breast Vassilaros S. Pregnancy and offspring after
paclitaxel, dose density, and trastuzumab cancer after giving birth. N. Engl. J. Med. the appearance of breast cancer. Oncology
on treatment-related amenorrhea in 331(1), 5–9 (1994). 53(6), 471–475 (1996).
premenopausal women with breast cancer. 28 Peterson NB, Huang Y, Newcomb PA et al. 42 Lethaby AE, O’Neill MA, Mason BH,
Cancer 116(4), 791–798 (2010). Childbearing recency and modifiers of Holdaway IM, Harvey VJ. Overall survival
•• Large study examining the effects premenopausal breast cancer risk. Cancer from breast cancer in women pregnant or
of different chemotherapy regimens Epidemiol. Biomarkers Prev. 17(11), lactating at or after diagnosis. Auckland
and trastuzumab on inducing 3284–3287 (2008). Breast Cancer Study Group. Int. J. Cancer
permanent amenorrhea. 29 Daling JR, Malone KE, Doody DR, 67(6), 751–755 (1996).
17 Burstein HJ, Winer EP. Primary care for Anderson BO, Porter PL. The relation of 43 Velentgas P, Daling JR, Malone KE et al.
survivors of breast cancer. N. Engl. J. Med. reproductive factors to mortality from Pregnancy after breast carcinoma:
343(15), 1086–1094 (2000). breast cancer. Cancer Epidemiol. Biomarkers outcomes and influence on mortality.
Prev. 11(3), 235–241 (2002). Cancer 85(11), 2424–2432 (1999).
18 Fornier MN, Modi S, Panageas KS, Norton
L, Hudis C. Incidence of chemotherapy- 30 Bemis LT, Schedin P. Reproductive state of 44 Birgisson H, Tryggvadottir L, Tulinius H.
induced, long-term amenorrhea in patients rat mammary gland stroma modulates [The effect of pregnancy on the survival of
with breast carcinoma age 40 years and human breast cancer cell migration and women diagnosed with breast cancer].
younger after adjuvant anthracycline and invasion. Cancer Res. 60(13), 3414–3418 Laeknabladid 86(7/8), 495–498 (2000).
taxane. Cancer 104(8), 1575–1579 (2005). (2000).
45 Gelber S, Coates AS, Goldhirsch A et al.
19 Petrek JA, Naughton MJ, Case LD et al. 31 Polyak K. Pregnancy and breast cancer: the Effect of pregnancy on overall survival after
Incidence, time course, and determinants other side of the coin. Cancer Cell 9(3), the diagnosis of early-stage breast cancer.
of menstrual bleeding after breast 151–153 (2006). J. Clin. Oncol. 19(6), 1671–1675 (2001).

296 Expert Rev. Anticancer Ther. 11(2), (2011)


Motherhood after breast cancer: searching for la dolce vita Review

46 Mueller BA, Simon MS, Deapen D et al. 57 Gerber B, Dieterich M, Muller H, Reimer 69 Bercovich D, Goodman G. Pregnancy and
Childbearing and survival after breast T. Controversies in preservation of ovary lactation after breast cancer elevate plasma
carcinoma in young women. Cancer 98(6), function and fertility in patients with prolactin, do not shorten and may prolong
1131–1140 (2003). breast cancer. Breast Cancer Res. Treat. survival. Medical Hypoth. 73(6), 942–947
47 Blakely LJ, Buzdar AU, Lozada JA et al. 108(1), 1–7 (2008). (2009).
Effects of pregnancy after treatment for 58 Mills JL, Simpson JL, Driscoll SG et al. 70 Turkington RW, Underwood LE,
breast carcinoma on survival and risk of Incidence of spontaneous abortion among van Wyk JJ. Elevated serum prolactin
recurrence. Cancer 100(3), 465–469 normal women and insulin-dependent levels after pituitary-stalk section in
(2004). diabetic women whose pregnancies were man. N. Engl. J. Med. 285(13), 707–710
48 Ives A, Saunders C, Bulsara M, Semmens J. identified within 21 days of conception. (1971).
Pregnancy after breast cancer: population N. Engl. J. Med. 319(25), 1617–1623 Nevalainen MT, Xie J, Torhorst J et al.
Expert Review of Anticancer Therapy Downloaded from informahealthcare.com by Washington University Library on 12/28/14

71
based study. Br. Med. J. 334(7586), 194 (1988). Signal transducer and activator of
(2007). 59 Langagergaard V, Gislum M, Skriver MV transcription-5 activation and
49 Kroman N, Jensen MB, Wohlfahrt J, et al. Birth outcome in women with breast breast cancer prognosis. J. Clin. Oncol.
Ejlertsen B. Pregnancy after treatment of cancer. Br. J. Cancer 94(1), 142–146 22(11), 2053–2060 (2004).
breast cancer – a population-based study on (2006). 72 Wang DY, Hampson S, Kwa HG et al.
behalf of Danish Breast Cancer 60 Dalberg K, Eriksson J, Holmberg L. Birth Serum prolactin levels in women with
Cooperative Group. Acta Oncol. 47(4), outcome in women with previously treated breast cancer and their relationship to
545–549 (2008). breast cancer – a population-based cohort survival. Eur. J. Cancer Clin. Oncol. 22(4),
50 Largillier R, Savignoni A, Gligorov J et al. study from Sweden. PLoS Med. 3(9), e336 487–492 (1986).
Prognostic role of pregnancy occurring (2006). 73 Tran TH, Utama FE, Lin J et al. Prolactin
before or after treatment of early breast 61 Agostoni C, Trojan S, Bellu R et al. inhibits BCL6 expression in breast cancer
cancer patients aged <35 years: a GET(N)A Developmental quotient at 24 months and through a Stat5a-dependent mechanism.
Working Group analysis. Cancer 115(22), fatty acid composition of diet in early Cancer Res. 70(4), 1711–1721 (2010).
5155–5165 (2009). infancy: a follow up study. Arch. Dis. Child. 74 Azim HA Jr, Bellettini G, Gelber S,
51 Kranick JA, Schaefer C, Rowell S et al. 76(5), 421–424 (1997). Peccatori FA. Breast-feeding after
For personal use only.

Is pregnancy after breast cancer safe? 62 Anderson JW, Johnstone BM, Remley DT. breast cancer: if you wish, madam.
Breast J. 16(4), 404–411 (2010). Breast-feeding and cognitive development: Breast Cancer Res. Treat. 114(1), 7–12
52 Verkooijen HM, Lim GH, Czene K et al. a meta-analysis. Am. J. Clin. Nutr. 70(4), (2009).
Effect of childbirth after treatment on 525–535 (1999). • Comprehensive review on the safety and
long-term survival from breast cancer. Br. 63 Zembo CT. Breastfeeding. Obstet. Gynecol. feasibility of breastfeeding in breast
J. Surg. 97(8), 1253–1259 (2010). Clin. North Am. 29(1), 51–76 (2002). cancer survivors including an original
53 Azim HA Jr, Santoro L, Pavlidis N et al. 64 Kjos SL, Henry O, Lee RM, Buchanan TA, survival analysis – the only in literature
Safety of pregnancy following breast Mishell DR Jr. The effect of lactation on – comparing the overall survival of
cancer diagnosis: a meta-analysis of glucose and lipid metabolism in women women who became pregnant following
14 studies. Eur. J. Cancer 47(1), 74–83 with recent gestational diabetes. Obstet. breast cancer diagnosis according to
(2011). Gynecol. 82(3), 451–455 (1993). breastfeeding status.
•• A large meta-analysis involving 65 Gorman JR, Usita PM, Madlensky L, 75 Azim HA Jr, Bellettini G, Liptrott SJ et al.
approximately 20,000 patients showing Pierce JP. A qualitative investigation of Breastfeeding in breast cancer survivors:
the safety of pregnancy in women with a breast cancer survivors’ experiences with pattern, behaviour and effect on breast
history of breast cancer diagnosis. breastfeeding. J. Cancer Surviv. 3(3), cancer outcome. Breast 19(6), 527–531
181–191 (2009). (2010).
54 Gorman JR, Roesch SC, Parker BA et al.
Physical and mental health correlates 66 Collaborative Group on Hormonal Factors 76 Helewa M, Levesque P, Provencher D et al.
of pregnancy following breast in Breast Cancer. Breast cancer and Breast cancer, pregnancy, and
cancer. Psychooncology 19(5), 517–524 breastfeeding: collaborative reanalysis of breastfeeding. J. Obstet. Gynaecol. Can.
(2010). individual data from 47 epidemiological 24(2), 164–180; quiz 181–164 (2002).
studies in 30 countries, including
55 Pagani O, Price KN, Gelber RD et al. 77 Higgins S, Haffty BG. Pregnancy and
50302 women with breast cancer and
Patterns of recurrence of early breast cancer lactation after breast-conserving therapy for
96973 women without the disease. Lancet
according to estrogen receptor status: a early stage breast cancer. Cancer 73(8),
360(9328), 187–195 (2002).
therapeutic target for a quarter of a century. 2175–2180 (1994).
Breast Cancer Res. Treat. 117(2), 319–324 67 Jernstrom H, Lubinski J, Lynch HT et al.
78 Moran MS, Colasanto JM, Haffty BG
(2009). Breast-feeding and the risk of breast cancer
et al. Effects of breast-conserving
in BRCA1 and BRCA2 mutation carriers.
56 Early Breast Cancer Trialists’ Collaborative therapy on lactation after pregnancy.
J. Natl Cancer Inst. 96(14), 1094–1098
Group (EBCTCG). Effects of Cancer J. (Sudbury, Mass.) 11(5), 399–403
(2004).
chemotherapy and hormonal therapy for (2005).
early breast cancer on recurrence and 68 McNeilly AS, Tay CC, Glasier A.
79 Tralins AH. Lactation after conservative
15-year survival: an overview of the Physiological mechanisms underlying
breast surgery combined with radiation
randomised trials. Lancet 365(9472), lactational amenorrhea. Ann. NY Acad. Sci.
therapy. Am. J. Clin. Oncol. 18(1), 40–43
1687–1717 (2005). 709, 145–155 (1994).
(1995).

www.expert-reviews.com 297
Review Azim Jr, Peccatori, de Azambuja & Piccart

80 Schnitt SJ, Goldwyn RM, Slavin SA. 90 Badawy A, Elnashar A, El-Ashry M, 98 Omland AK, Fedorcsak P, Storeng R et al.
Mammary ducts in the areola: implications Shahat M. Gonadotropin-releasing Natural cycle IVF in unexplained,
for patients undergoing reconstructive hormone agonists for prevention of endometriosis-associated and tubal factor
surgery of the breast. Plast. Reconstr. Surg. chemotherapy-induced ovarian damage: infertility. Hum. Reprod. (Oxford, Engl.)
92(7), 1290–1293 (1993). prospective randomized study. Fertil. Steril. 16(12), 2587–2592 (2001).
81 Camune B, Gabzdyl E. Breast-feeding after 91(3), 694–697 (2009). 99 Oktay K. Further evidence on the safety
breast cancer in childbearing women. 91 Peccatori F, Demeestere I. GnRH analogue and success of ovarian stimulation with
J. Perinat. Neonatal Nurs. 21(3), 225–233 for chemotherapy-induced ovarian damage: letrozole and tamoxifen in breast cancer
(2007). too early to say? Fertil. Steril. 92(2), e33; patients undergoing in vitro fertilization to
82 Ing R, Petrakis NL, Ho JH. Unilateral author reply e34 (2009). cryopreserve their embryos for fertility
breast-feeding and breast cancer. Lancet Oktay K, Sonmezer M. Questioning preservation. J. Clin. Oncol. 23(16),
Expert Review of Anticancer Therapy Downloaded from informahealthcare.com by Washington University Library on 12/28/14

92
2(8029), 124–127 (1977). GnRH analogs for gonadal protection in 3858–3859 (2005).
83 Riordan J, Gill-Hopple K, Angeron J. cancer patients. Fertil. Steril. 92(2), e32; 100 Oktay K, Buyuk E, Libertella N, Akar M,
Indicators of effective breastfeeding and author reply e34 (2009). Rosenwaks Z. Fertility preservation in
estimates of breast milk intake. J. Hum. 93 Del Mastro L, Boni L, Michelotti A et al. breast cancer patients: a prospective
Lact. 21(4), 406–412 (2005). Role of luteinizing hormone-releasing controlled comparison of ovarian
hormone analog (LHRHa) triptorelin (T) stimulation with tamoxifen and letrozole
84 Anchan RM, Ginsburg ES. Fertility
in preserving ovarian function during for embryo cryopreservation. J. Clin. Oncol.
concerns and preservation in younger
chemotherapy for early breast cancer 23(19), 4347–4353 (2005).
women with breast cancer. Crit. Rev.
Oncol. Hematol. 74(3), 175–192 (2010). patients: results of a multicenter Phase III 101 Oktay K, Hourvitz A, Sahin G et al.
trial of Gruppo Italiano Mammella (GIM) Letrozole reduces estrogen and
85 Fox KR, Scialla J, Moore H. Preventing
group. J. Clin. Oncol. 28(15 Suppl.), (2010) gonadotropin exposure in women with
chemotherapy-related amenorrhea using
(Abstract 528). breast cancer undergoing ovarian
leuprolide during adjuvant chemotherapy
•• Largest study to date evaluating the stimulation before chemotherapy. J. Clin.
for early-stage breast cancer. Proc. Am. Soc.
effect of luteinizing hormone-releasing Endocrinol. Metab. 91(10), 3885–3890
Clin. Oncol. 22 (2003) (Abstract 50).
(2006).
86 Del Mastro L, Catzeddu T, Boni L et al. hormone agonists, demonstrating an
For personal use only.

apparent positive effect on preserving 102 Azim AA, Costantini-Ferrando M, Oktay


Prevention of chemotherapy-induced
ovarian function in young women K. Safety of fertility preservation by
menopause by temporary ovarian
receiving chemotherapy. ovarian stimulation with letrozole
suppression with goserelin in young, early
and gonadotropins in patients with
breast cancer patients. Ann. Oncol. 17(1), 94 Azem F, Amit A, Merimsky O, Lessing JB. breast cancer: a prospective controlled
74–78 (2006). Successful transfer of frozen–thawed study. J. Clin. Oncol. 26(16), 2630–2635
87 Recchia F, Saggio G, Amiconi G et al. embryos obtained after subtotal (2008).
Gonadotropin-releasing hormone colectomy for colorectal cancer
and before fluorouracil-based •• Study demonstrating the safety and
analogues added to adjuvant chemotherapy
chemotherapy. Gynecologic Oncol. 93(1), feasibility of using letrozole and
protect ovarian function and improve
clinical outcomes in young women with 263–265 (2004). gonadotropins in ovarian stimulation of
early breast carcinoma. Cancer 106(3), 95 Ginsburg ES, Yanushpolsky EH, Jackson young breast cancer patients prior to
514–523 (2006). KV. In vitro fertilization for cancer patients starting adjuvant chemotherapy.
88 Urruticoechea A, Arnedos M, Walsh G, and survivors. Fertil. Steril. 75(4), 705–710 103 Lee SJ, Schover LR, Partridge AH et al.
Dowsett M, Smith IE. Ovarian protection (2001). American Society of Clinical Oncology
with goserelin during adjuvant 96 Oktay K, Buyuk E, Davis O et al. Fertility recommendations on fertility preservation
chemotherapy for pre-menopausal women preservation in breast cancer patients: in cancer patients. J. Clin. Oncol. 24(18),
with early breast cancer (EBC). Breast IVF and embryo cryopreservation after 2917–2931 (2006).
Cancer Res. Treat. 110(3), 411–416 (2008). ovarian stimulation with tamoxifen. Hum.
89 Ismail-Khan R, Minton S, Cox C et al. Reprod. (Oxford, Engl.) 18(1), 90–95
(2003). Websites
Preservation of ovarian function in young
women treated with neoadjuvant 97 Ayhan A, Salman MC, Celik H et al. 201 Center for Disease Control and Prevention
chemotherapy for breast cancer: a Association between fertility drugs and (CDC): 2004 Assisted Reproductive
randomized trial using the GnRH agonist gynecologic cancers, breast cancer, Technology (ART) Report
(triptorelin) during chemotherapy. J. Clin. and childhood cancers. Acta Obstet. www.cdc.giv/art/ART2004/section3.htm
Oncol. 26(20 Suppl.), (2008) Gynecol. Scand. Suppl. 83(12), 1104–1111 202 ClinicalTrials.gov
(Abstract 524). (2004). www.clinicaltrials.gov

298 Expert Rev. Anticancer Ther. 11(2), (2011)

Das könnte Ihnen auch gefallen