CC BY-NC-ND 4.0 · Geburtshilfe Frauenheilkd 2019; 79(06): 618-625
DOI: 10.1055/a-0880-9295
GebFra Science
Review/Übersicht
Georg Thieme Verlag KG Stuttgart · New York

Trastuzumab in the Treatment of Pregnant Breast Cancer Patients – an Overview of the Literature

Article in several languages: English | deutsch
Sophia S. Goller
Universitätsklinikum Jena, Klinik für Geburtsmedizin, Placenta-Labor, Jena, Germany
,
Udo R. Markert
Universitätsklinikum Jena, Klinik für Geburtsmedizin, Placenta-Labor, Jena, Germany
,
Karolin Fröhlich
Universitätsklinikum Jena, Klinik für Geburtsmedizin, Placenta-Labor, Jena, Germany
› Author Affiliations
Further Information

Correspondence/Korrespondenzadresse

Prof. Dr. Udo R. Markert
Universitätsklinikum Jena
Klinik für Geburtsmedizin, Placenta-Labor
Am Klinikum 1
07747 Jena
Germany   

Publication History

received 29 July 2018
revised 08 February 2019

accepted 20 March 2019

Publication Date:
14 June 2019 (online)

 

Abstract

Breast cancer is one of the most common malignancies which appear during pregnancy. Since women are increasingly not giving birth until they are at a more advanced age, it can be assumed that the incidence of pregnancy-related breast cancers will continue to increase in the future. Because of pregnancy-induced changes and conservative diagnosis, these carcinomas are frequently not detected until they are at an advanced stage and thus generally require systemic adjuvant therapy. The available data on optimal chemotherapeutic management are limited. Particularly for the use of the target agent trastuzumab which could crucially contribute to improving the prognosis in the therapy of HER2-overexpressing breast cancer in non-pregnant women, there is a lack of definitive information regarding the profile of action and safety in pregnancy as well as with regard to any long-term effects on the child. Thirty-eight pregnancies on trastuzumab for the treatment of breast cancer were able to be analysed in the literature currently available. Information can be gained from this and conclusions can be drawn which can individualise and decisively improve therapeutic options in the future for the pregnant breast cancer patient.


#

Introduction

By definition, pregnancy-associated breast cancer (PABC) occurs during pregnancy or within one year after childbirth [1]. Based on all breast cancer diagnoses, fewer than 1% in Europe occur during pregnancy [2]. Nonetheless, along with carcinoma of the cervix and diseases of the hematopoietic system, breast cancer is one of the most frequently occurring malignancies during this time [1], [3], [4]. Breast cancer represents approximately 25% of all malignant diseases which occur during pregnancy [5].

The incidence of PABC is indicated in industrialised nations with values between 1 : 1000 and 1 : 3000 pregnancies, and many authors state a figure of 1 : 10 000 pregnancies [5], [6], [7], [8], [9], [10], [11], [12]. A steady increase in the incidence has been able to be recorded in recent decades. The reasons for this development include the increased occurrence of malignant diseases overall as well as the fact that women in industrialized nations more and more frequently do not decide on pregnancy until a more advanced age and a positive correlation with the incidence of breast cancer should be noted in this regard [10], [13], [14], [15]. The mean age of the woman at diagnosis is 33 years; the mean gestational age is 21 weeks [16].


#

Characteristics of Pregnancy-Associated Breast Cancer

Genetics

Approximately 5% of all breast cancers can be attributed to an autosomal-dominant inherited genetic disease [17]. Mutations in the genes BRCA1 and BRCA2 (breast cancer 1 and 2) are of primary importance for the hereditary breast cancer variants. In comparison to sporadically occurring malignancies, these manifest at a significantly younger age [18]. Overall, women with pregnancy-associated breast cancer more frequently carry these genetic predispositions than non-pregnant women from comparative populations [19].


#

Pathology

Histopathologically, PABC does not significantly differ from breast cancer in the non-pregnant woman. Many different types of breast cancer occur with similar frequency in pregnant as well as non-pregnant women [19], [20]. Accounting for 70 – 90% of all cases, invasive ductal breast cancer is the most common type, followed by invasive lobular carcinoma. By contrast, inflammatory breast cancer occurs rather rarely [21], [22].


#

Potential for metastasis

Just as in the case of breast cancer in the non-pregnant woman, PABC also metastasises most frequently to the lung, liver and skeletal system [23]. However, for pregnant women, in comparison to non-pregnant women from comparative collectives, there is a likelihood 2.5 times as great of being diagnosed at an already metastatic stage [24].

Pregnancy-induced changes to the breast tissue as well as the conservative use of diagnostic measures in pregnancy may be causes for the frequent delay in diagnosis which subsequently determines the often already advanced tumour stage of the pregnant women [4], [12], [25].


#

Diagnosis

PABC presents most frequently as painless nodules in the breast and is discovered by the patient herself [26]. Therefore it is important that any abnormal findings on palpation during pregnancy which persist more than two to four weeks be clarified by means of an additional clinical examination, imaging, and tissue biopsy [23]. The literature indicates average figures for delay of diagnosis in pregnant women between two and fifteen months [27].


#

Staging

Imaging methods for the staging of breast cancer during pregnancy are indicated if they have an impact on subsequent therapeutic strategies. If the risk of metastasis is estimated as being low, there is fundamentally also the option of not performing staging examinations until after delivery and thus avoiding exposure of the foetus to any unnecessary radiation risks [4], [22].


#

Prognosis

Just as in the case of breast cancer in non-pregnant women, the prognosis of pregnancy-associated breast cancer depends above all on the size of the tumour, the differentiation, and the involvement of axillary lymph nodes [28]. The question as to whether the pregnancy per se has an influence on patientsʼ prognosis was the subject of controversial debate in the 1980s and 1990s. Nowadays, it is largely assumed that the pregnancy per se should not be considered to be an indicator of a poor prognosis [2], [28], [29], [30].


#
#

Trastuzumab in the Breast Cancer Therapy of Pregnant Patients

General

Drugs which attack certain sites of the cellular signalling cascade in a targeted manner on the molecular level are becoming more important in the treatment of malignant diseases in general. These so-called target agents thus also play an ever larger role in the treatment of breast cancer. The considerations here focus on HER2, the human epidermal growth factor receptor 2, which has a crucial impact on the regulation of cell growth as well as cell differentiation. It is overexpressed in about 20 – 30% of all breast cancers [31]. The monoclonal antibody trastuzumab, among others, is targeted against this; this antibody has already significantly improved the prognosis of HER2-positive tumours to date and led to a considerable survival advantage for affected patients with HER2-overexpressing tumours [32], [33].

Trastuzumab has been approved in Germany since August 2000 for the treatment of HER2-overexpressing metastatic breast cancer, and since May 2006, it has also been approved for (neo-)adjuvant treatment in HER2-overexpressing tumours following surgery and standard chemotherapy as well as radiation therapy, if applicable [32], [34].

The exact molecular mechanism of action of this human monoclonal antibody is not yet entirely understood [35], [36], [37]. Mechanisms of action on an extracellular as well as an intracellular level are discussed. According to current data, the antineoplastic mode of action of the antibody, on the extracellular level, is based in particular on cell-mediated cytotoxicity [38], [39]. By contrast, the intracellular mechanisms of action of trastuzumab are controversially described [31].

Another monoclonal antibody is pertuzumab, which is used together with trastuzumab and docetaxel in the first-line therapy of inoperable metastatic breast cancer [40]. It binds HER2 at another site than trastuzumab and as a result, it prevents the dimerisation of HER2 with other receptors of the HER family [41]. Various studies have already been able to prove the superiority of dual HER2 inhibition with trastuzumab and pertuzumab in non-pregnant women in comparison to the use of one of the two antibodies alone [42], [43]. However, there are still no data on the use of pertuzumab in pregnancy.

In general, antibodies are used in pregnancy only very cautiously. This is due in particular to the currently limited data as well as to preclinical studies which are critical of the use of these active substances in pregnant women [44]. Thus a transplacental transfer of trastuzumab was also able to be demonstrated in earlier studies [9].


#

Indications

A precondition for HER2 antibody therapy is initially the quality-controlled determination of the HER2 status. Verification of the HER2 gene is necessary and this is done using fluorescence in situ hybridization (FISH) [34].

(Neo-)adjuvant therapy with trastuzumab is fundamentally indicated in patients with nodal-positive tumours and nodal-negative tumours ≥ 1 cm in diameter with HER2 overexpression. Treatment lasts one year and is generally carried out simultaneously or consecutively in combination with standard chemotherapy. Thus in this setting, the patients generally receive an anthracycline, followed by a taxane in combination with trastuzumab. Even if smaller tumours with a diameter of < 1 cm and HER2 overexpression are present, patients can benefit from therapy with trastuzumab following a benefit/risk assessment beforehand. Patients with HER2-overexpressing carcinomas which are already metastatic should also be treated with antibody therapy [34]. In these patients, a long, progression-free survival as well as overall survival were able to be demonstrated on trastuzumab [45]. As first-line therapy, in the metastatic stage, dual HER-2 blockade should take place in combination with a taxane.


#

Patient collectives

Breast cancer patients whose tumours demonstrate HER2 overexpression benefit from therapeutic strategies targeted against HER2 [32], [33]. When selecting the patient collectives benefiting from this therapy, it should be borne in mind that the HER2 status may differ between the primary tumour as well as any metastases in up to 25% of cases, which is why metastases which subsequently occur should also be tested for possible HER2 positivity [46], [47].


#

Courses of pregnancy on trastuzumab

[Table 1] provides an overview of currently available data on the use of trastuzumab for the treatment of breast cancer during pregnancy. These data are based at this time primarily on case reports in which 22 pregnancies, including one twin pregnancy, are described [no. 1 – 18, 20 – 23]. Additional information was able to be obtained from the results of the international, multicentre, randomised phase III study called HERceptin Adjuvant (HERA). Among other things, it investigated the outcome of 16 children of patients who became pregnant during or up to three months after the administration of trastuzumab [no. 19]. In most cases, the antibody was administered to patients who were already in metastatic stages of the disease. Pregnant patients in early non-metastatic stages received trastuzumab more rarely. The case reports include 14 cases of exposure in the first trimester [no. 2, 3, 6 – 12, 14, 16, 18, 22, 23], whereby the vast majority of breast cancer patients on trastuzumab therapy became pregnant on an unplanned basis. In four patients, the treatment was discontinued after the pregnancy was discovered. Thirteen patients were treated only with trastuzumab, while in the remaining cases, double or triple combination regimens with vinorelbin, paclitaxel, docetaxel, tamoxifen, goserelin and carboplatin were used. One pregnant patient with cerebral metastases additionally received dexamethasone as well as cranial radiotherapy [no. 21]. Oligo- or anhydramnios were diagnosed in 13 of the total of 20 pregnancies [no. 2, 4, 5, 8 – 10, 12, 15, 17, 18, 20, 22, 23]; eight women were treated in the first trimester, seven received trastuzumab as monotherapy. In addition, there was also foetal intrauterine growth retardation (IUGR) in two cases; in one case in each instance, there was additional premature placental abruption, vaginal bleeding, premature rupture of membranes or foetal renal failure. Fifteen children were delivered via Caesarean section, on average in the 34th gestational week. Apart from one exception [no. 11], their birth weight was below 2700 g. In ten cases, there were neonatal complications; this included eight children of patients who developed oligo- or anhydramnios during pregnancy. All of these children suffered from respiratory complications [no. 4, 9, 10, 12, 13, 15, 16, 18, 23]. Three children concomitantly had renal failure [no. 4, 9, 12]. Four children died within the first four months of life [no. 9, 10, 12, 15]. The other children demonstrated normal development at follow-up examinations after an average of 22 months (2 – 84 months). One ectopic pregnancy was electively terminated. No pregnancy complications or anomalies were reported in the 16 patients of the HERA study [no. 19]. Nonetheless, a total of eleven abortions were recorded. Four of these were spontaneous abortions, seven were performed electively. However, the latter were not attributed to pregnancy complications, but instead to a particular degree of uncertainty and anxiety with regard to foetal trastuzumab exposure on the part of the patients as well as the attending physicians [48]. The five live births weighed an average of 3485 g and all demonstrated normal development. It should be noted overall that many complications which occurred could also be attributed to the preterm births of the children as such and cannot be attributed to the cytotoxic therapy alone.

Table 1 Trastuzumab for the treatment of PABC, overview of international case reports.

No.

Reference

n

Therapeutic regimen

Trimester of exposure

Receptors/
metastasis

Pregnancy complications/anomalies

Delivery

Birth weight

Neonatal status/complications

Follow-up children

EF: Ejection fraction; IUGR: Intrauterine growth restriction; Y: Years; Mo: Months; MOF: Multiorgan failure; NI: Not indicated; ND: Normal development; NRF: Neonatal respiratory failure; P: Percentile; RT: Radiation therapy; Section: Caesarean section; W: Gestational week

1

[75]

1

Trastuzumab + vinorelbin

3

ER−/PR−/HER2+, metastatic

None

Vaginal, W 34

2270 g (20th P)

None

6 mo: ND

2

[76]

1

Trastuzumab

Preconception, 1, 2

NI, metastatic

Anhydramnios (W 23)

Vaginal, W 37.5

2960 g (40th P)

None

6 mo: ND

3

[77]

1

Trastuzumab

Preconception, 1

NI, metastatic

None

Vaginal, NI

NI

None

NI

4

[78]

1

Trastuzumab + paclitaxel

2, 3

ER−/PR+/HER2+, metastatic

Anhydramnios, IUGR

Section, W 32

1460 g (10th P)

Bacteraemia, transient NRF, transient renal failure

3 mo: ND

5

[68]

1

Trastuzumab + docetaxel

2, 3

NI, metastatic

Anhydramnios (W 30), IUGR

Section, W 36

2230 g

None

NI

6

[79]

1

Trastuzumab

Preconception, 1, 2

ER−/PR−/HER2+, metastatic

None

Section, W 37

2600 g (10th P)

None

2 mo: ND

7

[80]

1

Trastuzumab

Preconception, 1

ER−/PR−/HER2+, metastatic

Ectopic pregnancy, elective abortion

8

[73]

1

Trastuzumab

Preconception, 1, 2, 3

ER−/PR−/HER2+, metastatic

Oligohydramnios

NI, W 32

1810 g

None

60 mo: ND

9

[81]

1

Trastuzumab

Preconception, 1, 2

NI, metastatic

Oligohydramnios, premature placental abruption

Section, W 27

NI

Decreased renal perfusion, renal dys-/hypoplasia, NRF

4 mo: death

10

[82]

1

Trastuzumab

Preconception, 1, 2, 3

ER+/PR−/HER2+, metastatic

Oligohydramnios, vaginal bleeding

Section, W 27

1015 g (57th P)

NRF, capillary leak syndrome, necrotising enterocolitis

21st week: Death due to MOF

11

[83]

1

Trastuzumab

Preconception

ER−/PR−/HER2+, metastatic

None

Section, W 39

3550 g

None

14 mo: ND

12

[84]

1

Trastuzumab + tamoxifen (+ methadone)

Preconception, 1, 2

ER+/HER2+, NI

Anhydramnios, premature rupture of membranes

Section, W 31

Twins

A: 1590 g

B: 1705 g

A: NRF, renal failure

B: transient NRF

A: 3 mo: Death due to respiratory failure

13

[85]

1

Trastuzumab

2

ER−/PR−/HER2+, metastatic

None

Section, W 29

1220 g

NRF

36 mo: ND

14

[85]

1

Trastuzumab

Preconception, 1

ER−/PR−/HER2+, not metastatic

2 gestational sacs with a viable foetus in only one of them

Vaginal, W 39

2940 g

None

24 mo: ND

15

[86]

1

Trastuzumab + tamoxifen + goserelin

2

ER+/PR−/HER2+, NI

Anhydramnios

Section, W 37

2690 g

Severe pulmonary hypoplasia, atelectasis

40 min: death

16

[87]

1

Trastuzumab

Preconception, 1, 2

ER−/PR−/HER2+, metastatic

Mother: Decrease in cardiac EF by up to 40%

Vaginal, W 37

3200 g

Mild transient tachypnoea

NI

17

[88]

1

Trastuzumab + carboplatin + docetaxel

2, 3

ER+/PR+/HER2+, not metastatic

Anhydramnios, foetal renal failure (W 21), IUGR

Section, W 34

NI

None

NI: ND

18

[89]

1

Trastuzumab (+ dexamethasone)

Preconception, 1, 2, 3

ER−/PR−/HER2+, metastatic

Oligohydramnios

Vaginal, W 37

3060 g

Transient tachypnoea

28 mo: ND

19

[48]

16

Trastuzumab

Pregnancies which occurred during or up to 3 months after the administration of trastuzumab

HER2+, not metastatic

None

11 abortions (4 spontaneous, 7 elective), 5 live births: NI, W 40 on average

3485 g on average

None

NI, ND

20

[90]

1

Trastuzumab + vinorelbin

3

ER−/PR−/HER2+, metastatic

Anhydramnios

Section, W 33

1990 g

None

13 mo: ND

21

[91]

1

Trastuzumab + paclitaxel + dexamethasone + RT

2, 3

ER−/PR−/HER2+, metastatic

None

Section, W 38

NI

None

6 mo: ND

22

[92]

1

Trastuzumab

1, 2

NI, metastatic

Oligohydramnios (W 24)

Section, W 36

NI

None

3 Y: ND

23

[93]

1

Trastuzumab

1, 2

ER−/PR−/HER2+, not metastatic

Anhydramnios (W 28)

Section, W 32

1655 g

NRF, pulmonary infection

7 Y: ND


#

Influence of the start of therapy

The data on the maternal outcome in breast cancer during pregnancy are not very comprehensive overall. In most studies, the success of treatment as well as the long-term prognoses of the affected children were primarily investigated. However, it can be said that there is presently a predominant consensus that pregnancy per se should not be considered to be an indicator for a worse prognosis of breast cancer. It was able to be demonstrated in several studies that pregnant breast cancer patients have a similar prognosis compared to non-pregnant women of the same age and tumour stage [2], [28], [29], [30]. Compared to non-pregnant patients, they are disproportionally frequently not diagnosed until an advanced tumour stage and for this reason, the outcome of these patients frequently appears to be worse overall [21], [49].

Based on this, it can thus be argued that postponing therapy in pregnant women would likely also have comparable negative effects as in the case of non-pregnant women. There is consensus that an existing pregnancy in general should not be a reason for postponing any indicated antineoplastic therapy [50]. Rather, the immediate introduction of therapy appropriate for the stage and tumour after diagnosis appears to be crucial for the outcome of the mother and, subsequently, also of the child, indirectly. Therefore any modifications to the recommended therapy to protect the unborn child should, if possible, be kept to a minimum.

However, it should also be noted at this point that this generally involves individual case-by-case decisions and the expectant motherʼs wishes should also be incorporated in the decision-making process [51]. HER2 inhibition with trastuzumab improves both progression-free survival as well as the overall survival in non-metastatic HER2-positive carcinomas as well as in stages which are already metastatic, and for this reason it can be assumed overall that postponing therapy for pregnant women, just as for non-pregnant women with comparable tumour characteristics, may have negative effects on these parameters [45], [52], [53].


#

Potential impact on the child

The concentration of a foreign substance which passes from the mother to the child largely depends on the placental transfer and, to a lesser extent, also on the placental as well as the foetal metabolism itself. The transplacental exchange of medicinal substances is affected by many factors. On the one hand, biochemical properties of the substance, such as the degree of ionisation, lipophilia, the degree of protein binding as well as the molecular weight, play a crucial role. On the other hand, the transfer is determined by characteristics relating to the placenta, such as blood flow, substance concentration gradients over the membrane, pH differences, an increasing exchange area with advancing pregnancy, or medicinal-product-metabolising enzymes which develop [54].

Most active substances overcome the placental barrier by means of passive diffusion. In this case, placental circulation, the pH difference between the motherʼs and childʼs blood as well as the biochemical substance properties and the degree of protein binding in each case represent in particular determining or limiting factors for the exchange substances [55], [56]. The facilitated diffusion as well as endocytosis are rarer means of transplacental medicinal substance transfer [57]. A paracellular exchange of substances, in particular of hydrophilic and charged substances via certain channel proteins, is possible [58], [59], [60].

By means of the mechanisms indicated, most drugs on the childʼs side of the placenta reach concentrations between 20 and 80% of the maternal concentration [61], [62]. However, the thickness of the syncytiotrophoblasts continues to decrease with advancing pregnancy and also the cytotrophoblast, stromal and endothelial cells which follow become thinner and more permeable, and for this reason the exchange of substances between mother and child increasingly becomes easier [63].

Trastuzumab is a humanised monoclonal IgGk1 antibody [64]. Since immunoglobulins are large hydrophilic molecules with a molecular weight of about 150 kD, they overcome cell membranes via active transport by channel proteins [65]. The transplacental transport of IgG ultimately takes place via at least two membranes, that of the syncytiotrophoblast as well as that of the foetal capillary endothelium [63], [66].

HER2 receptors were identified in placental tissue in advanced pregnancy as well as in foetal renal tissue. As already described above, it is suspected that the interaction of trastuzumab with these receptors plays a role with regard to the more frequent occurrence of oligo- and anhydramnios on therapy [67]. The time of application of the antibody as well as the duration of therapy appear to have an influence here. Overall, a toxic effect of the active substance on foetal renal cells is considered to be the cause of the decrease in amniotic fluid. It is interesting that the amount of amniotic fluid increases once again after discontinuing the antibody and this is therefore a reversible effect. Growth factor receptors of the HER2 family which, as already mentioned, are expressed to an increased degree in human nephrocytes during the foetal period, induce DNA synthesis processes and promote cell division. In experimental studies as well, it was able to be shown that a blockade of these receptors led to decreased division activity of the nephrogenic cells [68], [69], [70]. That trastuzumab causes damage to foetal renal cells but not, however, maternal renal cells or adult renal cells is very likely due to the different protein structure of the growth receptor factor. This is therefore present in foetal renal cells as heterodimers, while in adults, it has developed in the form of a homodimer [71].

Moreover, trastuzumab is also involved in foetal neuronal and myocardial development. To date, however, no case of damage on trastuzumab to the paediatric nervous system or heart has been reported [72].


#
#

Conclusion

Numerous applications of trastuzumab were documented and the corresponding cases were published. Several low-complication pregnancies but also significant adverse effects were seen in some cases, and thus a general recommendation for use in pregnancy still cannot initially be made. Therefore a decision should be made in individual cases by a team of experts as to whether trastuzumab therapy is indicated.

Oligo- or anhydramnios occurs comparatively frequently during therapy. Interestingly, the amount of amniotic fluid increases once again after the antibody is stopped. This effect thus appears to be reversible. A toxic effect on the renal cells of the child is therefore presumed [68]. To date, the molecular mechanism by which trastuzumab causes a decrease in amniotic fluid has not been able to be clarified, however there are hypotheses which refer to the involvement of epidermal growth factor receptors in foetal tissue [73].

Growth factors of the HER2 family are expressed to an increased degree in human nephrocytes during the foetal period. They induce DNA synthesis processes there and promote cell division. In experimental studies, it was able to be shown that a blockade of these receptors led to decreased division activity of the nephrogenic cells. Similarly, trastuzumab could play a role in the HER2 blockade [69], [70]. In addition, HER2 is also involved in foetal neuronal and myocardial development. To date, no case of damage to the paediatric nervous system or heart on trastuzumab is known, however this potential risk should not be disregarded. In the case of administration of anthracyclines and the subsequent administration of trastuzumab, caution is called for in particular with regard to possible adverse cardiotoxic effects [74].


#
#

Conflict of Interest/Interessenkonflikt

The authors declare that they have no conflict of interest./Die Autoren geben an, dass kein Interessenkonflikt besteht.

Acknowledgements

The placenta laboratory received research funding from the Wilhelm Sander Foundation for the subject of breast cancer in pregnancy (project number 2014.152.1). Karolin Fröhlich was funded by a Ph. D. grant from Evangelisches Bildungswerk Villigst (Villigst Protestant Training Institute). Udo R. Markert receives research funding from the German Research Foundation (DFG, MA 1550/12-1).

  • References/Literatur

  • 1 Pirvulescu C, Mau C, Schultz H. et al. Breast Cancer during Pregnancy: An Interdisciplinary Approach in Our Institution. Breast Care (Basel) 2012; 7: 311-314
  • 2 Stensheim H, Moller B, van Dijk T. et al. Cause-specific survival for women diagnosed with cancer during pregnancy or lactation: a registry-based cohort study. J Clin Oncol 2009; 27: 45-51
  • 3 Vetter G, Zimmermann F, Bruder E. et al. Aggressive Breast Cancer during Pregnancy with a Rare Form of Metastasis in the Maternal Placenta. Geburtsh Frauenheilk 2014; 74: 579-582
  • 4 Amant F, Loibl S, Neven P. et al. Breast cancer in pregnancy. Lancet 2012; 379: 570-579
  • 5 Dow KH. Pregnancy and breast cancer. J Obstet Gynecol Neonatal Nurs 2000; 29: 634-640
  • 6 Pavlidis NA. Coexistence of pregnancy and malignancy. Oncologist 2002; 7: 279-287
  • 7 Barnes DM, Newman LA. Pregnancy-associated breast cancer: a literature review. Surg Clin North Am 2007; 87: 417-430 x
  • 8 Keinan-Boker L, Lerner-Geva L, Kaufman B. et al. Pregnancy-associated breast cancer. Isr Med Assoc J 2008; 10: 722-727
  • 9 Loibl S. New Therapeutic Options for Breast Cancer during Pregnancy. Breast Care (Basel) 2008; 3: 171-176
  • 10 Loibl S. Adjuvant therapy in patients with breast cancer during pregnancy. Cancer Treat Res 2009; 151: 317-328
  • 11 Asgeirsson KS. Pregnancy-associated breast cancer. Acta Obstet Gynecol Scand 2011; 90: 158-166
  • 12 Salani R, Billingsley CC, Crafton SM. Cancer and pregnancy: an overview for obstetricians and gynecologists. Am J Obstet Gynecol 2014; 211: 7-14
  • 13 Voulgaris E, Pentheroudakis G, Pavlidis N. Cancer and pregnancy: a comprehensive review. Surg Oncol 2011; 20: e175-e185
  • 14 Cardoso F, Loibl S, Pagani O. et al. The European Society of Breast Cancer Specialists recommendations for the management of young women with breast cancer. Eur J Cancer 2012; 48: 3355-3377
  • 15 Loibl S, Schmidt A, Gentilini O. et al. Breast Cancer Diagnosed During Pregnancy: Adapting Recent Advances in Breast Cancer Care for Pregnant Patients. JAMA Oncol 2015; 1: 1145-1153
  • 16 Van Calsteren K, Heyns L, De Smet F. et al. Cancer during pregnancy: an analysis of 215 patients emphasizing the obstetrical and the neonatal outcomes. J Clin Oncol 2010; 28: 683-689
  • 17 Rich TA, Woodson AH, Litton J. et al. Hereditary breast cancer syndromes and genetic testing. J Surg Oncol 2015; 111: 66-80
  • 18 Sheikh A, Hussain SA, Ghori Q. et al. The spectrum of genetic mutations in breast cancer. Asian Pac J Cancer Prev 2015; 16: 2177-2185
  • 19 Woo JC, Yu T, Hurd TC. Breast cancer in pregnancy: a literature review. Arch Surg 2003; 138: 91-98 discussion 99
  • 20 Madaras L, Kovacs KA, Szasz AM. et al. Clinicopathological features and prognosis of pregnancy associated breast cancer – a matched case control study. Pathol Oncol Res 2014; 20: 581-590
  • 21 Pavlidis N, Pentheroudakis G. The pregnant mother with breast cancer: diagnostic and therapeutic management. Cancer Treat Rev 2005; 31: 439-447
  • 22 Loibl S, Han SN, Amant F. Being Pregnant and Diagnosed with Breast Cancer. Breast Care (Basel) 2012; 7: 204-209
  • 23 Amant F, Deckers S, Van Calsteren K. et al. Breast cancer in pregnancy: recommendations of an international consensus meeting. Eur J Cancer 2010; 46: 3158-3168
  • 24 Zemlickis D, Lishner M, Degendorfer P. et al. Maternal and fetal outcome after breast cancer in pregnancy. Am J Obstet Gynecol 1992; 166: 781-787
  • 25 Froehlich K, Stensheim H, Markert UR. et al. Breast carcinoma in pregnancy with spheroid-like placental metastases-a case report. APMIS 2018; 126: 448-452
  • 26 Molckovsky A, Madarnas Y. Breast cancer in pregnancy: a literature review. Breast Cancer Res Treat 2008; 108: 333-338
  • 27 Puckridge PJ, Saunders CM, Ives AD. et al. Breast cancer and pregnancy: a diagnostic and management dilemma. ANZ J Surg 2003; 73: 500-503
  • 28 Aebi S, Loibl S. Breast cancer during pregnancy: medical therapy and prognosis. Recent Results Cancer Res 2008; 178: 45-55
  • 29 Halaska MJ, Pentheroudakis G, Strnad P. et al. Presentation, management and outcome of 32 patients with pregnancy-associated breast cancer: a matched controlled study. Breast J 2009; 15: 461-467
  • 30 Cardonick E, Dougherty R, Grana G. et al. Breast cancer during pregnancy: maternal and fetal outcomes. Cancer J 2010; 16: 76-82
  • 31 Nami B, Maadi H, Wang Z. Mechanisms Underlying the Action and Synergism of Trastuzumab and Pertuzumab in Targeting HER2-Positive Breast Cancer. Cancers (Basel) 2018; DOI: 10.3390/cancers10100342.
  • 32 Slamon DJ, Leyland-Jones B, Shak S. et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med 2001; 344: 783-792
  • 33 Paik S, Bryant J, Tan-Chiu E. et al. Real-world performance of HER2 testing–National Surgical Adjuvant Breast and Bowel Project experience. J Natl Cancer Inst 2002; 94: 852-854
  • 34 Leitlinienprogramm Onkologie (Deutsche Krebsgesellschaft, Deutsche Krebshilfe, AWMF). S3-Leitlinie Früherkennung, Diagnose, Therapie und Nachsorge des Mammakarzinoms, Version 4.1, 2018. AWMF Registernummer: 032-045OL. Online: http://www.leitlinienprogramm-onkologie.de/leitlinien/mammakarzinom/ last access: 28.05.2018
  • 35 Spector NL, Blackwell KL. Understanding the mechanisms behind trastuzumab therapy for human epidermal growth factor receptor 2-positive breast cancer. J Clin Oncol 2009; 27: 5838-5847
  • 36 Fiszman GL, Jasnis MA. Molecular Mechanisms of Trastuzumab Resistance in HER2 Overexpressing Breast Cancer. Int J Breast Cancer 2011; 2011: 352182
  • 37 Nuti M, Bellati F, Visconti V. et al. Immune effects of trastuzumab. J Cancer 2011; 2: 317-323
  • 38 Duong MN, Cleret A, Matera EL. et al. Adipose cells promote resistance of breast cancer cells to trastuzumab-mediated antibody-dependent cellular cytotoxicity. Breast Cancer Res 2015; 17: 57
  • 39 Maadi H, Nami B, Tong J. et al. The effects of trastuzumab on HER2-mediated cell signaling in CHO cells expressing human HER2. BMC Cancer 2018; 18: 238
  • 40 Niikura N, Liu J, Hayashi N. et al. Loss of human epidermal growth factor receptor 2 (HER2) expression in metastatic sites of HER2-overexpressing primary breast tumors. J Clin Oncol 2012; 30: 593-599
  • 41 Franklin MC, Carey KD, Vajdos FF. et al. Insights into ErbB signaling from the structure of the ErbB2-pertuzumab complex. Cancer Cell 2004; 5: 317-328
  • 42 Baselga J, Cortes J, Kim SB. et al. Pertuzumab plus trastuzumab plus docetaxel for metastatic breast cancer. N Engl J Med 2012; 366: 109-119
  • 43 Gianni L, Pienkowski T, Im YH. et al. Efficacy and safety of neoadjuvant pertuzumab and trastuzumab in women with locally advanced, inflammatory, or early HER2-positive breast cancer (NeoSphere): a randomised multicentre, open-label, phase 2 trial. Lancet Oncol 2012; 13: 25-32
  • 44 Lambertini M, Peccatori FA, Azim jr. HA. Targeted agents for cancer treatment during pregnancy. Cancer Treat Rev 2015; 41: 301-309
  • 45 Wong Y, Raghavendra AS, Hatzis C. et al. Long-Term Survival of De Novo Stage IV Human Epidermal Growth Receptor 2 (HER2) Positive Breast Cancers Treated with HER2-Targeted Therapy. Oncologist 2019; 24: 313-318 doi:10.1634/theoncologist.2018-0213
  • 46 Wolff AC, Hammond ME, Hicks DG. et al. Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline update. J Clin Oncol 2013; 31: 3997-4013
  • 47 Niikura N, Tomotaki A, Miyata H. et al. Changes in tumor expression of HER2 and hormone receptors status after neoadjuvant chemotherapy in 21,755 patients from the Japanese breast cancer registry. Ann Oncol 2016; 27: 480-487
  • 48 Azim jr. HA, Metzger-Filho O, de Azambuja E. et al. Pregnancy occurring during or following adjuvant trastuzumab in patients enrolled in the HERA trial (BIG 01-01). Breast Cancer Res Treat 2012; 133: 387-391
  • 49 Petrek JA, Dukoff R, Rogatko A. Prognosis of pregnancy-associated breast cancer. Cancer 1991; 67: 869-872
  • 50 Amant F, Vandenbroucke T, Verheecke M. et al. Pediatric Outcome after Maternal Cancer Diagnosed during Pregnancy. N Engl J Med 2015; 373: 1824-1834
  • 51 Shlensky V, Hallmeyer S, Juarez L. et al. Management of Breast Cancer during Pregnancy: Are We Compliant with Current Guidelines?. AJP Rep 2017; 7: e39-e43
  • 52 Shen Y, Fujii T, Ueno NT. et al. Comparative efficacy of adjuvant trastuzumab-containing chemotherapies for patients with early HER2-positive primary breast cancer: a network meta-analysis. Breast Cancer Res Treat 2018; DOI: 10.1007/s10549-018-4969-6.
  • 53 Niraula S, Gyawali B. Optimal duration of adjuvant trastuzumab in treatment of early breast cancer: a meta-analysis of randomized controlled trials. Breast Cancer Res Treat 2018; DOI: 10.1007/s10549-018-4967-8.
  • 54 Poulsen MS, Rytting E, Mose T. et al. Modeling placental transport: correlation of in vitro BeWo cell permeability and ex vivo human placental perfusion. Toxicol In Vitro 2009; 23: 1380-1386
  • 55 Pacifici GM, Nottoli R. Placental transfer of drugs administered to the mother. Clin Pharmacokinet 1995; 28: 235-269
  • 56 Audus KL. Controlling drug delivery across the placenta. Eur J Pharm Sci 1999; 8: 161-165
  • 57 Syme MR, Paxton JW, Keelan JA. Drug transfer and metabolism by the human placenta. Clin Pharmacokinet 2004; 43: 487-514
  • 58 Robinson NR, Atkinson DE, Jones CJ. et al. Permeability of the near-term rat placenta to hydrophilic solutes. Placenta 1988; 9: 361-372
  • 59 Sibley CP. Review article: mechanisms of ion transfer by the rat placenta: a model for the human placenta?. Placenta 1994; 15: 675-691
  • 60 St-Pierre MV, Ugele B, Gambling L. et al. Mechanisms of drug transfer across the human placenta-a workshop report. Placenta 2002; 23 (Suppl. A) S159-S164
  • 61 Loebstein R, Lalkin A, Koren G. Pharmacokinetic changes during pregnancy and their clinical relevance. Clin Pharmacokinet 1997; 33: 328-343
  • 62 Juchau MR. Bioactivation in chemical teratogenesis. Annu Rev Pharmacol Toxicol 1989; 29: 165-187
  • 63 van der Aa EM, Peereboom-Stegeman JH, Noordhoek J. et al. Mechanisms of drug transfer across the human placenta. Pharm World Sci 1998; 20: 139-148
  • 64 Wong WM. Drug update: trastuzumab: anti-HER2 antibody for treatment of metastatic breast cancer. Cancer Pract 1999; 7: 48-50
  • 65 Saji F, Samejima Y, Kamiura S. et al. Dynamics of immunoglobulins at the feto-maternal interface. Rev Reprod 1999; 4: 81-89
  • 66 Takizawa T, Anderson CL, Robinson JM. A novel Fc gamma R-defined, IgG-containing organelle in placental endothelium. J Immunol 2005; 175: 2331-2339
  • 67 Pentsuk N, van der Laan JW. An interspecies comparison of placental antibody transfer: new insights into developmental toxicity testing of monoclonal antibodies. Birth Defects Res B Dev Reprod Toxicol 2009; 86: 328-344
  • 68 Sekar R, Stone PR. Trastuzumab use for metastatic breast cancer in pregnancy. Obstet Gynecol 2007; 110: 507-510
  • 69 Chailler P, Briere N. Mitogenic effects of EGF/TGF alpha and immunolocalization of cognate receptors in human fetal kidneys. Biofactors 1998; 7: 323-335
  • 70 Press MF, Cordon-Cardo C, Slamon DJ. Expression of the HER-2/neu proto-oncogene in normal human adult and fetal tissues. Oncogene 1990; 5: 953-962
  • 71 Robinson AA, Watson WJ, Leslie KK. Targeted treatment using monoclonal antibodies and tyrosine-kinase inhibitors in pregnancy. Lancet Oncol 2007; 8: 738-743
  • 72 Coelingh Bennink HJ, Holinka CF, Diczfalusy E. Estetrol review: profile and potential clinical applications. Climacteric 2008; 11 (Suppl. 01) 47-58
  • 73 Pant S, Landon MB, Blumenfeld M. et al. Treatment of breast cancer with trastuzumab during pregnancy. J Clin Oncol 2008; 26: 1567-1569
  • 74 Meattini I, Curigliano G, Terziani F. et al. SAFE trial: an ongoing randomized clinical study to assess the role of cardiotoxicity prevention in breast cancer patients treated with anthracyclines with or without trastuzumab. Med Oncol 2017; 34: 75
  • 75 Fanale MA, Uyei AR, Theriault RL. et al. Treatment of metastatic breast cancer with trastuzumab and vinorelbine during pregnancy. Clin Breast Cancer 2005; 6: 354-356
  • 76 Watson WJ. Herceptin (trastuzumab) therapy during pregnancy: association with reversible anhydramnios. Obstet Gynecol 2005; 105: 642-643
  • 77 Waterston AM, Graham J. Effect of adjuvant trastuzumab on pregnancy. J Clin Oncol 2006; 24: 321-322
  • 78 Bader AA, Schlembach D, Tamussino KF. et al. Anhydramnios associated with administration of trastuzumab and paclitaxel for metastatic breast cancer during pregnancy. Lancet Oncol 2007; 8: 79-81
  • 79 Shrim A, Garcia-Bournissen F, Maxwell C. et al. Favorable pregnancy outcome following Trastuzumab (Herceptin) use during pregnancy–Case report and updated literature review. Reprod Toxicol 2007; 23: 611-613
  • 80 Berveiller P, Mir O, Sauvanet E. et al. Ectopic pregnancy in a breast cancer patient receiving trastuzumab. Reprod Toxicol 2008; 25: 286-288
  • 81 Weber-Schoendorfer C, Schaefer C. Untitled. Reprod Toxicol 2008; 25: 390-391
  • 82 Witzel ID, Muller V, Harps E. et al. Trastuzumab in pregnancy associated with poor fetal outcome. Ann Oncol 2008; 19: 191-192
  • 83 Azim jr. HA, Peccatori FA, Liptrott SJ. et al. Breast cancer and pregnancy: how safe is trastuzumab?. Nat Rev Clin Oncol 2009; 6: 367-370
  • 84 Beale JM, Tuohy J, McDowell SJ. Herceptin (trastuzumab) therapy in a twin pregnancy with associated oligohydramnios. Am J Obstet Gynecol 2009; 201: e13-e14
  • 85 Goodyer MJ, Ismail JR, OʼReilly SP. et al. Safety of trastuzumab (Herceptin) during pregnancy: two case reports. Cases J 2009; 2: 9329
  • 86 Warraich Q, Smith N. Herceptin therapy in pregnancy: continuation of pregnancy in the presence of anhydramnios. J Obstet Gynaecol 2009; 29: 147-148
  • 87 Roberts NJ, Auld BJ. Trastuzamab (Herceptin)-related cardiotoxicity in pregnancy. J R Soc Med 2010; 103: 157-159
  • 88 Gottschalk I, Berg C, Harbeck N. et al. Fetal Renal Insufficiency Following Trastuzumab Treatment for Breast Cancer in Pregnancy: Case Report und Review of the Current Literature. Breast Care (Basel) 2011; 6: 475-478
  • 89 Mandrawa CL, Stewart J, Fabinyi GC. et al. A case study of trastuzumab treatment for metastatic breast cancer in pregnancy: fetal risks and management of cerebral metastases. Aust N Z J Obstet Gynaecol 2011; 51: 372-376
  • 90 El-Safadi S, Wuesten O, Muenstedt K. Primary diagnosis of metastatic breast cancer in the third trimester of pregnancy: a case report and review of the literature. J Obstet Gynaecol Res 2012; 38: 589-592
  • 91 Gupta S, Jain P, McDunn S. Breast cancer with brain metastases in pregnancy. J Community Support Oncol 2014; 12: 378-380
  • 92 Rasenack R, Gaupp N, Rautenberg B. et al. [Case Report on Treatment of Metastatic Breast Cancer with Trastuzumab during Pregnancy]. Z Geburtshilfe Neonatol 2016; 220: 81-83
  • 93 Andrade JM, Brito LG, Moises EC. et al. Trastuzumab use during pregnancy: long-term survival after locally advanced breast cancer and long-term infant follow-up. Anticancer Drugs 2016; 27: 369-372

Correspondence/Korrespondenzadresse

Prof. Dr. Udo R. Markert
Universitätsklinikum Jena
Klinik für Geburtsmedizin, Placenta-Labor
Am Klinikum 1
07747 Jena
Germany   

  • References/Literatur

  • 1 Pirvulescu C, Mau C, Schultz H. et al. Breast Cancer during Pregnancy: An Interdisciplinary Approach in Our Institution. Breast Care (Basel) 2012; 7: 311-314
  • 2 Stensheim H, Moller B, van Dijk T. et al. Cause-specific survival for women diagnosed with cancer during pregnancy or lactation: a registry-based cohort study. J Clin Oncol 2009; 27: 45-51
  • 3 Vetter G, Zimmermann F, Bruder E. et al. Aggressive Breast Cancer during Pregnancy with a Rare Form of Metastasis in the Maternal Placenta. Geburtsh Frauenheilk 2014; 74: 579-582
  • 4 Amant F, Loibl S, Neven P. et al. Breast cancer in pregnancy. Lancet 2012; 379: 570-579
  • 5 Dow KH. Pregnancy and breast cancer. J Obstet Gynecol Neonatal Nurs 2000; 29: 634-640
  • 6 Pavlidis NA. Coexistence of pregnancy and malignancy. Oncologist 2002; 7: 279-287
  • 7 Barnes DM, Newman LA. Pregnancy-associated breast cancer: a literature review. Surg Clin North Am 2007; 87: 417-430 x
  • 8 Keinan-Boker L, Lerner-Geva L, Kaufman B. et al. Pregnancy-associated breast cancer. Isr Med Assoc J 2008; 10: 722-727
  • 9 Loibl S. New Therapeutic Options for Breast Cancer during Pregnancy. Breast Care (Basel) 2008; 3: 171-176
  • 10 Loibl S. Adjuvant therapy in patients with breast cancer during pregnancy. Cancer Treat Res 2009; 151: 317-328
  • 11 Asgeirsson KS. Pregnancy-associated breast cancer. Acta Obstet Gynecol Scand 2011; 90: 158-166
  • 12 Salani R, Billingsley CC, Crafton SM. Cancer and pregnancy: an overview for obstetricians and gynecologists. Am J Obstet Gynecol 2014; 211: 7-14
  • 13 Voulgaris E, Pentheroudakis G, Pavlidis N. Cancer and pregnancy: a comprehensive review. Surg Oncol 2011; 20: e175-e185
  • 14 Cardoso F, Loibl S, Pagani O. et al. The European Society of Breast Cancer Specialists recommendations for the management of young women with breast cancer. Eur J Cancer 2012; 48: 3355-3377
  • 15 Loibl S, Schmidt A, Gentilini O. et al. Breast Cancer Diagnosed During Pregnancy: Adapting Recent Advances in Breast Cancer Care for Pregnant Patients. JAMA Oncol 2015; 1: 1145-1153
  • 16 Van Calsteren K, Heyns L, De Smet F. et al. Cancer during pregnancy: an analysis of 215 patients emphasizing the obstetrical and the neonatal outcomes. J Clin Oncol 2010; 28: 683-689
  • 17 Rich TA, Woodson AH, Litton J. et al. Hereditary breast cancer syndromes and genetic testing. J Surg Oncol 2015; 111: 66-80
  • 18 Sheikh A, Hussain SA, Ghori Q. et al. The spectrum of genetic mutations in breast cancer. Asian Pac J Cancer Prev 2015; 16: 2177-2185
  • 19 Woo JC, Yu T, Hurd TC. Breast cancer in pregnancy: a literature review. Arch Surg 2003; 138: 91-98 discussion 99
  • 20 Madaras L, Kovacs KA, Szasz AM. et al. Clinicopathological features and prognosis of pregnancy associated breast cancer – a matched case control study. Pathol Oncol Res 2014; 20: 581-590
  • 21 Pavlidis N, Pentheroudakis G. The pregnant mother with breast cancer: diagnostic and therapeutic management. Cancer Treat Rev 2005; 31: 439-447
  • 22 Loibl S, Han SN, Amant F. Being Pregnant and Diagnosed with Breast Cancer. Breast Care (Basel) 2012; 7: 204-209
  • 23 Amant F, Deckers S, Van Calsteren K. et al. Breast cancer in pregnancy: recommendations of an international consensus meeting. Eur J Cancer 2010; 46: 3158-3168
  • 24 Zemlickis D, Lishner M, Degendorfer P. et al. Maternal and fetal outcome after breast cancer in pregnancy. Am J Obstet Gynecol 1992; 166: 781-787
  • 25 Froehlich K, Stensheim H, Markert UR. et al. Breast carcinoma in pregnancy with spheroid-like placental metastases-a case report. APMIS 2018; 126: 448-452
  • 26 Molckovsky A, Madarnas Y. Breast cancer in pregnancy: a literature review. Breast Cancer Res Treat 2008; 108: 333-338
  • 27 Puckridge PJ, Saunders CM, Ives AD. et al. Breast cancer and pregnancy: a diagnostic and management dilemma. ANZ J Surg 2003; 73: 500-503
  • 28 Aebi S, Loibl S. Breast cancer during pregnancy: medical therapy and prognosis. Recent Results Cancer Res 2008; 178: 45-55
  • 29 Halaska MJ, Pentheroudakis G, Strnad P. et al. Presentation, management and outcome of 32 patients with pregnancy-associated breast cancer: a matched controlled study. Breast J 2009; 15: 461-467
  • 30 Cardonick E, Dougherty R, Grana G. et al. Breast cancer during pregnancy: maternal and fetal outcomes. Cancer J 2010; 16: 76-82
  • 31 Nami B, Maadi H, Wang Z. Mechanisms Underlying the Action and Synergism of Trastuzumab and Pertuzumab in Targeting HER2-Positive Breast Cancer. Cancers (Basel) 2018; DOI: 10.3390/cancers10100342.
  • 32 Slamon DJ, Leyland-Jones B, Shak S. et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med 2001; 344: 783-792
  • 33 Paik S, Bryant J, Tan-Chiu E. et al. Real-world performance of HER2 testing–National Surgical Adjuvant Breast and Bowel Project experience. J Natl Cancer Inst 2002; 94: 852-854
  • 34 Leitlinienprogramm Onkologie (Deutsche Krebsgesellschaft, Deutsche Krebshilfe, AWMF). S3-Leitlinie Früherkennung, Diagnose, Therapie und Nachsorge des Mammakarzinoms, Version 4.1, 2018. AWMF Registernummer: 032-045OL. Online: http://www.leitlinienprogramm-onkologie.de/leitlinien/mammakarzinom/ last access: 28.05.2018
  • 35 Spector NL, Blackwell KL. Understanding the mechanisms behind trastuzumab therapy for human epidermal growth factor receptor 2-positive breast cancer. J Clin Oncol 2009; 27: 5838-5847
  • 36 Fiszman GL, Jasnis MA. Molecular Mechanisms of Trastuzumab Resistance in HER2 Overexpressing Breast Cancer. Int J Breast Cancer 2011; 2011: 352182
  • 37 Nuti M, Bellati F, Visconti V. et al. Immune effects of trastuzumab. J Cancer 2011; 2: 317-323
  • 38 Duong MN, Cleret A, Matera EL. et al. Adipose cells promote resistance of breast cancer cells to trastuzumab-mediated antibody-dependent cellular cytotoxicity. Breast Cancer Res 2015; 17: 57
  • 39 Maadi H, Nami B, Tong J. et al. The effects of trastuzumab on HER2-mediated cell signaling in CHO cells expressing human HER2. BMC Cancer 2018; 18: 238
  • 40 Niikura N, Liu J, Hayashi N. et al. Loss of human epidermal growth factor receptor 2 (HER2) expression in metastatic sites of HER2-overexpressing primary breast tumors. J Clin Oncol 2012; 30: 593-599
  • 41 Franklin MC, Carey KD, Vajdos FF. et al. Insights into ErbB signaling from the structure of the ErbB2-pertuzumab complex. Cancer Cell 2004; 5: 317-328
  • 42 Baselga J, Cortes J, Kim SB. et al. Pertuzumab plus trastuzumab plus docetaxel for metastatic breast cancer. N Engl J Med 2012; 366: 109-119
  • 43 Gianni L, Pienkowski T, Im YH. et al. Efficacy and safety of neoadjuvant pertuzumab and trastuzumab in women with locally advanced, inflammatory, or early HER2-positive breast cancer (NeoSphere): a randomised multicentre, open-label, phase 2 trial. Lancet Oncol 2012; 13: 25-32
  • 44 Lambertini M, Peccatori FA, Azim jr. HA. Targeted agents for cancer treatment during pregnancy. Cancer Treat Rev 2015; 41: 301-309
  • 45 Wong Y, Raghavendra AS, Hatzis C. et al. Long-Term Survival of De Novo Stage IV Human Epidermal Growth Receptor 2 (HER2) Positive Breast Cancers Treated with HER2-Targeted Therapy. Oncologist 2019; 24: 313-318 doi:10.1634/theoncologist.2018-0213
  • 46 Wolff AC, Hammond ME, Hicks DG. et al. Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline update. J Clin Oncol 2013; 31: 3997-4013
  • 47 Niikura N, Tomotaki A, Miyata H. et al. Changes in tumor expression of HER2 and hormone receptors status after neoadjuvant chemotherapy in 21,755 patients from the Japanese breast cancer registry. Ann Oncol 2016; 27: 480-487
  • 48 Azim jr. HA, Metzger-Filho O, de Azambuja E. et al. Pregnancy occurring during or following adjuvant trastuzumab in patients enrolled in the HERA trial (BIG 01-01). Breast Cancer Res Treat 2012; 133: 387-391
  • 49 Petrek JA, Dukoff R, Rogatko A. Prognosis of pregnancy-associated breast cancer. Cancer 1991; 67: 869-872
  • 50 Amant F, Vandenbroucke T, Verheecke M. et al. Pediatric Outcome after Maternal Cancer Diagnosed during Pregnancy. N Engl J Med 2015; 373: 1824-1834
  • 51 Shlensky V, Hallmeyer S, Juarez L. et al. Management of Breast Cancer during Pregnancy: Are We Compliant with Current Guidelines?. AJP Rep 2017; 7: e39-e43
  • 52 Shen Y, Fujii T, Ueno NT. et al. Comparative efficacy of adjuvant trastuzumab-containing chemotherapies for patients with early HER2-positive primary breast cancer: a network meta-analysis. Breast Cancer Res Treat 2018; DOI: 10.1007/s10549-018-4969-6.
  • 53 Niraula S, Gyawali B. Optimal duration of adjuvant trastuzumab in treatment of early breast cancer: a meta-analysis of randomized controlled trials. Breast Cancer Res Treat 2018; DOI: 10.1007/s10549-018-4967-8.
  • 54 Poulsen MS, Rytting E, Mose T. et al. Modeling placental transport: correlation of in vitro BeWo cell permeability and ex vivo human placental perfusion. Toxicol In Vitro 2009; 23: 1380-1386
  • 55 Pacifici GM, Nottoli R. Placental transfer of drugs administered to the mother. Clin Pharmacokinet 1995; 28: 235-269
  • 56 Audus KL. Controlling drug delivery across the placenta. Eur J Pharm Sci 1999; 8: 161-165
  • 57 Syme MR, Paxton JW, Keelan JA. Drug transfer and metabolism by the human placenta. Clin Pharmacokinet 2004; 43: 487-514
  • 58 Robinson NR, Atkinson DE, Jones CJ. et al. Permeability of the near-term rat placenta to hydrophilic solutes. Placenta 1988; 9: 361-372
  • 59 Sibley CP. Review article: mechanisms of ion transfer by the rat placenta: a model for the human placenta?. Placenta 1994; 15: 675-691
  • 60 St-Pierre MV, Ugele B, Gambling L. et al. Mechanisms of drug transfer across the human placenta-a workshop report. Placenta 2002; 23 (Suppl. A) S159-S164
  • 61 Loebstein R, Lalkin A, Koren G. Pharmacokinetic changes during pregnancy and their clinical relevance. Clin Pharmacokinet 1997; 33: 328-343
  • 62 Juchau MR. Bioactivation in chemical teratogenesis. Annu Rev Pharmacol Toxicol 1989; 29: 165-187
  • 63 van der Aa EM, Peereboom-Stegeman JH, Noordhoek J. et al. Mechanisms of drug transfer across the human placenta. Pharm World Sci 1998; 20: 139-148
  • 64 Wong WM. Drug update: trastuzumab: anti-HER2 antibody for treatment of metastatic breast cancer. Cancer Pract 1999; 7: 48-50
  • 65 Saji F, Samejima Y, Kamiura S. et al. Dynamics of immunoglobulins at the feto-maternal interface. Rev Reprod 1999; 4: 81-89
  • 66 Takizawa T, Anderson CL, Robinson JM. A novel Fc gamma R-defined, IgG-containing organelle in placental endothelium. J Immunol 2005; 175: 2331-2339
  • 67 Pentsuk N, van der Laan JW. An interspecies comparison of placental antibody transfer: new insights into developmental toxicity testing of monoclonal antibodies. Birth Defects Res B Dev Reprod Toxicol 2009; 86: 328-344
  • 68 Sekar R, Stone PR. Trastuzumab use for metastatic breast cancer in pregnancy. Obstet Gynecol 2007; 110: 507-510
  • 69 Chailler P, Briere N. Mitogenic effects of EGF/TGF alpha and immunolocalization of cognate receptors in human fetal kidneys. Biofactors 1998; 7: 323-335
  • 70 Press MF, Cordon-Cardo C, Slamon DJ. Expression of the HER-2/neu proto-oncogene in normal human adult and fetal tissues. Oncogene 1990; 5: 953-962
  • 71 Robinson AA, Watson WJ, Leslie KK. Targeted treatment using monoclonal antibodies and tyrosine-kinase inhibitors in pregnancy. Lancet Oncol 2007; 8: 738-743
  • 72 Coelingh Bennink HJ, Holinka CF, Diczfalusy E. Estetrol review: profile and potential clinical applications. Climacteric 2008; 11 (Suppl. 01) 47-58
  • 73 Pant S, Landon MB, Blumenfeld M. et al. Treatment of breast cancer with trastuzumab during pregnancy. J Clin Oncol 2008; 26: 1567-1569
  • 74 Meattini I, Curigliano G, Terziani F. et al. SAFE trial: an ongoing randomized clinical study to assess the role of cardiotoxicity prevention in breast cancer patients treated with anthracyclines with or without trastuzumab. Med Oncol 2017; 34: 75
  • 75 Fanale MA, Uyei AR, Theriault RL. et al. Treatment of metastatic breast cancer with trastuzumab and vinorelbine during pregnancy. Clin Breast Cancer 2005; 6: 354-356
  • 76 Watson WJ. Herceptin (trastuzumab) therapy during pregnancy: association with reversible anhydramnios. Obstet Gynecol 2005; 105: 642-643
  • 77 Waterston AM, Graham J. Effect of adjuvant trastuzumab on pregnancy. J Clin Oncol 2006; 24: 321-322
  • 78 Bader AA, Schlembach D, Tamussino KF. et al. Anhydramnios associated with administration of trastuzumab and paclitaxel for metastatic breast cancer during pregnancy. Lancet Oncol 2007; 8: 79-81
  • 79 Shrim A, Garcia-Bournissen F, Maxwell C. et al. Favorable pregnancy outcome following Trastuzumab (Herceptin) use during pregnancy–Case report and updated literature review. Reprod Toxicol 2007; 23: 611-613
  • 80 Berveiller P, Mir O, Sauvanet E. et al. Ectopic pregnancy in a breast cancer patient receiving trastuzumab. Reprod Toxicol 2008; 25: 286-288
  • 81 Weber-Schoendorfer C, Schaefer C. Untitled. Reprod Toxicol 2008; 25: 390-391
  • 82 Witzel ID, Muller V, Harps E. et al. Trastuzumab in pregnancy associated with poor fetal outcome. Ann Oncol 2008; 19: 191-192
  • 83 Azim jr. HA, Peccatori FA, Liptrott SJ. et al. Breast cancer and pregnancy: how safe is trastuzumab?. Nat Rev Clin Oncol 2009; 6: 367-370
  • 84 Beale JM, Tuohy J, McDowell SJ. Herceptin (trastuzumab) therapy in a twin pregnancy with associated oligohydramnios. Am J Obstet Gynecol 2009; 201: e13-e14
  • 85 Goodyer MJ, Ismail JR, OʼReilly SP. et al. Safety of trastuzumab (Herceptin) during pregnancy: two case reports. Cases J 2009; 2: 9329
  • 86 Warraich Q, Smith N. Herceptin therapy in pregnancy: continuation of pregnancy in the presence of anhydramnios. J Obstet Gynaecol 2009; 29: 147-148
  • 87 Roberts NJ, Auld BJ. Trastuzamab (Herceptin)-related cardiotoxicity in pregnancy. J R Soc Med 2010; 103: 157-159
  • 88 Gottschalk I, Berg C, Harbeck N. et al. Fetal Renal Insufficiency Following Trastuzumab Treatment for Breast Cancer in Pregnancy: Case Report und Review of the Current Literature. Breast Care (Basel) 2011; 6: 475-478
  • 89 Mandrawa CL, Stewart J, Fabinyi GC. et al. A case study of trastuzumab treatment for metastatic breast cancer in pregnancy: fetal risks and management of cerebral metastases. Aust N Z J Obstet Gynaecol 2011; 51: 372-376
  • 90 El-Safadi S, Wuesten O, Muenstedt K. Primary diagnosis of metastatic breast cancer in the third trimester of pregnancy: a case report and review of the literature. J Obstet Gynaecol Res 2012; 38: 589-592
  • 91 Gupta S, Jain P, McDunn S. Breast cancer with brain metastases in pregnancy. J Community Support Oncol 2014; 12: 378-380
  • 92 Rasenack R, Gaupp N, Rautenberg B. et al. [Case Report on Treatment of Metastatic Breast Cancer with Trastuzumab during Pregnancy]. Z Geburtshilfe Neonatol 2016; 220: 81-83
  • 93 Andrade JM, Brito LG, Moises EC. et al. Trastuzumab use during pregnancy: long-term survival after locally advanced breast cancer and long-term infant follow-up. Anticancer Drugs 2016; 27: 369-372