Skip to main content
Erschienen in: BMC Medicine 1/2021

Open Access 01.12.2021 | Correspondence

Reproductive issues in carriers of germline pathogenic variants in the BRCA1/2 genes: an expert meeting

verfasst von: Barbara Buonomo, Claudia Massarotti, Miriam Dellino, Paola Anserini, Alberta Ferrari, Maria Campanella, Mirosa Magnotti, Cristofaro De Stefano, Fedro Alessandro Peccatori, Matteo Lambertini

Erschienen in: BMC Medicine | Ausgabe 1/2021

Abstract

Background

Healthy individuals and patients with cancer who are carriers of germline pathogenic variants in the BRCA1/2 genes face multiple reproductive challenges that require appropriate counseling and specific expertise.

Main body

On December 5th–7th, 2019, patient advocates and physicians with expertise in the field of reproductive medicine, fertility preservation, and oncology were invited to “San Giuseppe Moscati” Hospital in Avellino (Italy) for a workshop on reproductive management of women with germline pathogenic variants in the BRCA1/2 genes. From the discussion regarding the current evidence and future prospective in the field, eight main research questions were formulated and eight recommendations were developed regarding fertility, fertility preservation, preimplantation genetic testing, and pregnancy in healthy carriers and patients with cancer.

Conclusion

Several misconceptions about the topic persist among health care providers and patients often resulting in a discontinuous and suboptimal management. With the aim to offer patient-tailored counseling about reproductive issues, both awareness of current evidences and research should be promoted.
Hinweise
Barbara Buonomo and Claudia Massarotti contributed equally and share first authorship.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
BC
Breast cancer
OC
Ovarian cancer
FP
Fertility preservation
POI
Premature ovarian insufficiency
DSBs
Double strands breaks
AMH
Anti-Müllerian hormone
RRSO
Risk reducing salpingo-oophorectomy
COS
Controlled ovarian stimulation
GnRHa
Gonadotropins releasing hormone analog
ET
Endocrine therapy
PGT-M
Preimplantation genetic diagnosis for monogenic diseases
IVF
In vitro fertilization
OTC
ovarian tissue cryopreservation
IVM
In vitro maturation
ESHRE
European Society for Human Reproduction and Embryology
HBOC
Hereditary breast and ovarian cancers
ASRM
American Society for Reproductive Medicine

Background

About 5% to 10% of all cancers are related to germline pathogenic variants in cancer-susceptibility genes [1]. The most frequent predisposing genetic alterations are in the BRCA1 and BRCA2 genes, with a cumulative lifetime risk for breast cancer (BC) of 72% and 69% for carriers of pathogenic variants in BRCA1 and BRCA2, respectively [2]. The cumulative ovarian cancer (OC) risk is 44% for BRCA1 and 17% for BRCA2 carriers [2]. In the majority of health systems, the access to genetic testing depends on a family-history model [3]; thus, the detection of BRCA1/2 carriers before a cancer diagnosis remains limited. However, their identification has become more common thanks to several factors such as the therapeutic implications of genetic testing in some diseases (including epithelial OC, BC, pancreatic cancer, and prostate cancer) as well as the development and widespread use of multi-gene panel sequencing technologies [4, 5]. Therefore, there is a frequent need of expert counseling not only for patients with BRCA-related cancers, but also for the so-called “previvors” (i.e., individuals that have a BRCA pathogenetic variant without being affected by cancer). This counseling includes a discussion on risk-reducing interventions (including risk-reducing salpingo-oophorectomy [RRSO] at the age of 35–40 years for BRCA1 carriers and between 40 and 45 years for BRCA2 carriers) which temporarily or permanently impact on lifestyle, body image, endocrinological status, and reproductive choices [6]. Thus, previvors and BRCA1/2-mutated patients with cancer face multiple concerns in terms of reproductive challenges that should be separately addressed [7].
In the present manuscript, we summarize and discuss controversial issues in the field of reproductive medicine in young women carrying germline pathogenic variants in the BRCA1/2 genes and report recommendations from a multidisciplinary group of experts.

Main text

On December 5th–7th, 2019, patient advocates and physicians with expertise in the field of reproductive medicine, fertility preservation (FP), and oncology were invited to “San Giuseppe Moscati” Hospital in Avellino (Italy) to participate in a workshop on the reproductive management of women with germline pathogenic variants in the BRCA1/2 genes. The invited experts represented different disciplines related to the topic including oncologists, gynecologists, geneticists, surgeons, and bioethic specialists. Starting from patients’ needs voiced by the advocates present at the workshop, a total of 8 controversial issues were discussed. Experts were asked to present an up-to-date overview of the preclinical and clinical literature available on these topics. On the basis of the data presented, 8 statements were developed.

Question 1: Are pregnancy and breastfeeding safe in BRCA pathogenic variant carriers?

Several studies investigated the effect of parity on BC risk in healthy BRCA pathogenic variant carriers [8]. This topic remains controversial for the reported differences in BRCA1 and BRCA2 pathogenic variant carriers and for the different effects of pregnancy on breast cancer risk according to age also in the general non-BRCA carrier population. A large prospective study showed that women with BRCA1 pathogenic variants who had two, three, four, or more full-term pregnancies were at 21%, 30%, and 50% decreased risk of BC compared to women with a single full-term pregnancy [9]. On the contrary, women with BRCA2 pathogenic variants with multiple pregnancies had a significantly increased risk of developing BC [10]. In the general population, healthy women have a transient increased risk of BC after a pregnancy and the increased risk is higher for women with a family history of BC and for women with a pregnancy at a later age [11, 12]. This increased risk has been attributed to the growth-promoting effect of the endocrinological milieu of pregnancy on existing pre-malignant or malignant breast cancer lesions that occur more frequently at an advanced age [11]. It is possible, even if it has never been proved, that BRCA2 mutation carriers harbor more pre-malignant estrogen receptor-positive lesions and that repeated pregnancies might increase the risk of developing breast cancer [13].
The same differences between BRCA1 and BRCA2 carriers are also apparent regarding the effect of breastfeeding on breast cancer risk. In BRCA1 pathogenic variant carriers, breastfeeding for at least 1 year reduces BC risk (OR = 0.68; 95% CI 0.52 to 0.91; P = 0.008), while no effect has been described for healthy BRCA2 carriers [14]. Thus, the timing of risk-reducing bilateral mastectomy in healthy BRCA carriers remains a matter of discussion. On one hand, removing the breasts before pregnancy consistently reduces the risk of subsequent BC and the need of careful monitoring during pregnancy and breastfeeding. On the other hand, many women highly value the advantages of breastfeeding their infants [15] and are reluctant to undergo surgery before pregnancy. For the above-mentioned considerations, it appears clear that the counseling of young BRCA pathogenic variant carriers seeking pregnancy is complex and should consider risk estimations according to family history, age, and breast density, but also the personal values on reproductive choices, that remain extremely sensitive and personal. If the woman chooses to maintain her breasts and postpone risk-reducing bilateral mastectomy, a careful monitoring with breast ultrasound during pregnancy and with breast ultrasound and mammogram during breastfeeding should be planned [16]. To reduce diagnostic delay, it is of utmost importance that women who are BRCA pathogenic variant carriers and their physicians are aware of the possibility of breast cancer occurrence also during pregnancy and breastfeeding.
To further complicate the issue of pregnancy and breastfeeding in BRCA pathogenic variant carriers, we should consider the available data on the effect of subsequent pregnancy in the population of BRCA carriers already affected by BC. In this group of patients, current data show that subsequent pregnancy does not increase breast cancer-related events [17, 18]. A large international study has recently shown that, independently of the receptor status and especially for BRCA1 pathogenic variants carriers, pregnancy after BC seems to be safe without negative consequences on maternal prognosis or fetal outcomes [19]. Even if this study included a significant number of patients, it had some limitations including short-term follow-up (~ 4 year follow-up since the pregnancy) and limited power to detect differences particularly in BRCA2 carriers [19]. Moreover, the retrospective nature of this and other studies represents an important limitation that does not allow to derive definitive and strong conclusions [20]. In BC patients that need 5 to 10 years of adjuvant endocrine therapy (ET) including BRCA carriers, an international clinical trial, which has recently completed the target accrual, is assessing the safety and feasibility of a temporary interruption of ET after at least 18 months, in order to allow pregnancy [21].
Recommendation 1: In healthy BRCA mutation carriers, the impact of pregnancy and breastfeeding remains controversial. Women should be encouraged to complete childbearing at early age and discuss thorough breast follow-up during pregnancy and breastfeeding if they decide to maintain their breasts. In BRCA-mutated BC patients, subsequent pregnancy following adequate treatment and follow-up does not seem to increase the risk of BC recurrence and should not be discouraged.

Question 2: Does carrying a BRCA pathogenic variant impact ovarian reserve and reproductive potential in healthy women?

A major concern among BRCA1/2 pathogenic variant carriers is the potential higher risk of premature ovarian insufficiency (POI) [22, 23]. Most of the available preclinical evidence suggests that BRCA mutations could directly accelerate ovarian aging, reducing the ovarian reserve both quantitatively and qualitatively [2429]. BRCA1 and 2 are known to be involved in DNA repair mechanism, through ATM-mediated regulation of the DNA double-strand breaks (DSBs) repair [23, 30]. DNA DBS repair mechanisms have a relevant role in ovarian aging; a decrease in their efficiency causes not only an accelerated apoptotic loss of follicles with lethal mutations, but also an increase in meiotic errors and reduced oocyte quality, with an increased number of aneuploidies [23]. There is preclinical evidence that transgenic mice with defective BRCA genes have a reduced ovarian response to stimulation and a diminished reproductive potential [30].
In humans, data about fertility in BRCA pathogenic variant carriers remain controversial. The majority of the available case-control studies did not report a significant difference in fertility outcomes (i.e., spontaneous abortions and parity) among BRCA carriers and non-carriers [3134]. Fewer studies reported differences in favor of non-carriers [35, 36] with different limitations and confounders (e.g., use/not use of hormonal contraceptives, younger age, and study design). Levels of anti-Müllerian hormone (AMH) are considered a quantitative marker of ovarian reserve, although not predictive of chances of spontaneous pregnancy [37]. In some studies, the levels of AMH were found to be significantly lower in women carrying pathogenic variants in BRCA1 [2426, 30] or BRCA2 [27, 29] or both genes [28], while other studies reported no significant difference with controls [36, 38]. Clinical studies describing a decreased oocyte quality in human carriers of BRCA pathogenic variants (i.e., an increase in aneuploidies) are still lacking, while age at natural menopause among BRCA carriers is difficult to ascertain, because of various types of selection bias, diverse control groups, and the small population of the studies [3942]. A further issue related to the shortened window of reproductive opportunity is the recommendation of RRSO at a young age [6]. Although salpingectomy with delayed oophorectomy has been suggested as an option to preserve ovarian function, this strategy is not the gold standard and it should not be recommended [43]. Given these different issues, healthy carriers should be advised not to delay pregnancy beyond 35 years of age [44].
Notably, data about fertility parameters in male carriers of BRCA pathogenic variants are very scarce. Spermatogenesis is different from oogenesis. DNA repair mechanisms have the ability to correct DNA alterations only at the germ cell level [45]. Simhadri et al. reported how a mutant PALB2 protein unable to bind BRCA1 in male mice reduced fertility, due to impaired meiosis and increased germ cells apoptosis [46]. However, semen parameters and gonadal function have been never specifically studied in this population.
Recommendation 2: A potential negative impact of BRCA pathogenic variants on women ovarian reserve and reproductive potential cannot be excluded and should be discussed; however, no strong conclusions can be drawn from existing data. Healthy carriers should be advised to not delay pregnancy beyond 35 years.

Question 3: Does carrying a BRCA pathogenic variant affect ovarian reserve and reproductive potential in BC patients?

Limited data exist about fertility outcomes in BRCA-mutated BC patients [47]. Oktay et al. first described in 2010 a diminished ovarian response and lower number of oocytes for FP in BRCA-mutated cancer patients [48]. Since then, a few more studies found a worse quantitative response to controlled ovarian stimulation (COS) in this cohort [4850], while others reported no difference with not mutated BC patients [5153]. Similarly, some studies reported lower AMH levels at BC diagnosis in BRCA-mutated patients [30, 50, 54], while others did not report a significant difference [52, 53, 55, 56]. A reduced ovarian reserve and a reduced quantitative response to ovarian stimulation would have strong implications for the risk of reduced efficacy of emergency oocytes retrieval and cryopreservation. More research efforts are needed in this field to provide clearer evidence on the need to personalize the oncofertility counseling of women with breast cancer carrying a germline pathogenic variant in BRCA genes.
Recommendation 3: A potential negative impact of BRCA pathogenic variants on women ovarian reserve and reproductive potential including a diminished response to ovarian stimulation in BC patients cannot be excluded and should be discussed during the oncofertility counseling; however, no strong conclusions can be drawn from existing data.

Question 4: Are BRCA-mutated patients with cancer at higher risk of treatment-induced gonadotoxicity as compared to cancer patients without BRCA pathogenic variants?

Young cancer patients requiring cytotoxic drugs are at risk of a negative impact on their ovarian function and reserve that depends on their age at the time of treatment, dosage, and chemotherapy regimen [57]. Due to both the potential high risk of POI in BRCA-mutated patients due to a possible impairment of their ovarian reserve even before starting anticancer therapies and the key role of DNA damage-induced follicle death [58], it can be hypothesized that these patients would be particularly sensitive to the gonadotoxic effect of antineoplastic drugs. Facing this critical issue during oncofertility counseling of BRCA-mutated patients who are candidates for chemotherapy at a young age is mandatory but very demanding because of the lack of clear evidence. Valentini et al. conducted a survey of 1954 BRCA1/2-mutated women who were treated for BC to assess the impact of chemotherapy on the risk of developing treatment-induced POI in this specific subgroup of patients [59]. Chemotherapy-induced POI was defined as ≥ 2 years of amenorrhea commencing within 2 years after initiating chemotherapy. The authors reported a statistically significantly higher proportion of chemotherapy-induced amenorrhea among BRCA2 pathogenic variant carriers than BRCA1 ones. Even excluding patients taking tamoxifen (most numerous among the BRCA2 cohort), the likelihood of chemotherapy-induced amenorrhea remained significantly different compared to patients who did not undergo chemotherapy. Anyway, for neither subgroup, the probability of amenorrhea was higher than that of BC patients without BRCA pathogenic variants [59].
A study by Lambertini et al. in patients receiving anthracycline- and cyclophosphamide-based chemotherapy investigating AMH levels up to 3 years after diagnosis showed no additional detrimental effect by the presence of a deleterious germline BRCA pathogenic variant on the reduction in AMH levels following cytotoxic therapy [55].
Currently, in BRCA-mutated BC patients, platinum agents are often added to standard anthracycline- and taxane-based chemotherapy regimens [60, 61]. Moreover, PARP inhibitors are now available treatment options for BRCA-mutated patients with advanced OC and BC [62] and are being studied also in the early setting [63]. Recent data suggest that olaparib could reduce ovarian reserve in mice [64]; therefore, the potential detrimental effect of PARP inhibitors on women ovarian reserve should be further investigated.
Recommendation 4: The available limited evidence does not demonstrate an increased risk of chemotherapy-induced POI in BRCA carriers but the overall risk remains significant for all patients and fertility preservation should be discussed with all women diagnosed at reproductive age. No data exist on the gonadotoxicity of newer treatment options in these patients.

Question 5: Is co-administration of gonadotropin-releasing hormone agonists (GnRHa) during chemotherapy effective for ovarian function preservation in BRCA-mutated women with cancer?

Even if young cancer survivors with a BRCA pathogenic variant are counseled to undergo RRSO with subsequent iatrogenic POI, the maintenance of ovarian function up to the time of risk-reducing surgery may have a great positive impact on their quality of life, especially in women diagnosed at a very young age. To date, the use of temporary suppression of ovarian function with GnRHa during (neo-)adjuvant chemotherapy is the only recommended medical strategy to preserve ovarian function in young BC patients [65]. The three largest randomized studies on this topic (PROMISE-GIM6, POEMS-SWOG S0230, and OPTION trials) showed similar results with a significant reduction in the risk of developing treatment-induced POI in patients receiving GnRHa during chemotherapy [6668]. A recent meta-analysis reported that the use of GnRHa during chemotherapy for early BC patients was associated with an absolute 16% reduction in POI rates (from 30.9 to 14.1%; p < 0.001) and a higher number of post-treatment pregnancies (37 patients (10.3%) in the GnRHa group vs. 20 patients (5.5%) in the control group; p = 0.03) [65]. This effect was observed irrespective of hormone receptor status, patients’ age at the time of diagnosis, type, and duration of chemotherapy [65]. Nonetheless, temporary ovarian suppression with GnRHa during chemotherapy should not be considered an alternative to oocytes/embryos cryopreservation as a strategy for FP (see recommendation 5). Anyway, it may be offered as an additional option following cryopreservation options or when they are not accessible [57, 69]. In patients with cancers other than breast cancer, the use of GnRHa has shown to protect ovarian function in OC patients but not in women with hematological malignancies [70, 71].
It is important to note that there is very limited efficacy data in the specific cohort of patients with cancer and carrying germline BRCA pathogenic variants, with only one study reporting the protective effect of GnRHa use during chemotherapy in 4 carriers with BC [69]. There is no biological or clinical rationale to support a different recommendation in patients with cancer carrying or not germline BRCA pathogenic variants.
Recommendation 5: Temporary ovarian suppression obtained by administering GnRHa during (neo-)adjuvant chemotherapy should be offered to all patients with cancer who wish to preserve ovarian function, including BRCA carriers diagnosed years before the recommended age of risk-reducing surgery. Most of the available efficacy data exist in women with BC not carrying BRCA pathogenic variants. GnRHa use during chemotherapy does not replace established fertility preservation methods.

Question 6: Is oocyte/embryo cryopreservation safe among young women with a BRCA1/2 pathogenic variant?

In the setting of both BRCA-mutated healthy carriers and patients with cancer, oocyte and embryo cryopreservation should be discussed for different reasons: the first group may benefit from this technique to lengthen the fertile widow (including after the RRSO). The second one may benefit to preserve their reproductive potential before starting a gonadotoxic therapy and/or adjuvant endocrine therapy (ET, which implies a delay in childbearing) [72]. Moreover, it would give all BRCA pathogenic variant carriers the possibility to access preimplantation genetic diagnosis for monogenic diseases (PGT-M) [47, 73]. In spite of these critical issues, both the counseling and the use of this FP option are still suboptimal, essentially for safety concerns [7476].
Three major mechanisms seem to be involved in the carcinogenic effects of estrogens on the breast: stimulation of cellular proliferation through their receptor-mediated hormonal activity, direct genotoxic effects by increasing mutation rates through a cytochrome P450-mediated metabolic activation, and induction of aneuploidy. Since the BRCA1 and BRCA2 genes are involved in DNA repair, it could be postulated that the potential effect of estrogens on mammary tissue is aggravated in BRCA1/2 mutation carriers [77, 78]. To date, there is no evidence that controlled ovarian stimulation (COS) is linked to an increased risk of breast and high-grade OC in the general infertile population [79, 80]. Noteworthy, a recent nationwide cohort study showed a statistically significantly 1.8-fold higher risk of borderline ovarian tumors in women who underwent COS [81]. Perri et al. did not find an increased risk of OC in 164 healthy carriers who underwent fertility treatments versus 909 not treated for infertility [82]. Similarly, Derks-Smeets et al. reported that BC risk was not increased in 76 BRCA-pathogenic variant healthy carriers after IVF, compared to controls [77]. While these results are encouraging, they are based on small cohorts and limited follow-up time.
As for BC patients, modified protocols using letrozole in estrogen-sensitive tumors are recommended to reduce estrogen serum concentration by more than 50%, without affecting the number of mature oocytes retrieved or their fertilization capacity and cancer prognosis [57, 8386]. In a recent survey, 42% of the interviewed BC oncologists were unsure about the safety of COS in BRCA-mutated BC patients [7]. However, Kim et al. reported safety outcomes in 120 BC patients versus 217 controls, showing that the 5 years survival was not affected by FP procedures also in the specific cohort of women with a germline pathogenic variant in BRCA [86].
Recommendation 6: Despite the lack of exhaustive data, oocyte and embryo cryopreservation following COS can be considered a safe option that should be proposed both to BRCA-healthy carriers and patients with cancer interested in fertility preservation, whenever feasible. The use of letrozole to suppress supra-physiologic estrogens can be used safely without a reduction in the number or quality of oocytes.

Question 7: Is ovarian tissue cryopreservation (OTC) a safe and feasible option in young women with newly diagnosed cancer and carrying a BRCA pathogenic variant?

OTC is a widespread FP option, which is already considered standard in several countries [87]. It does not require COS, allowing FP in an urgent setting (e.g., when chemotherapy should be started as soon as possible) [88]. The ovarian cortical tissue is retrieved through laparoscopy, cryopreserved, and then re-transplanted into the ovary enabling the woman to search for a pregnancy spontaneously or through IVF [87, 89]. Literature reports live birth rates around 30–40% in experienced centers [87, 89, 90]. Two live births after OTC and re-transplantation are reported in BRCA-mutated early BC patients [52, 91].
Limited data are available about this approach in BRCA-mutated patients with cancer. OTC raises important safety concerns on transplanting a tissue in a woman at increased risk of subsequent OC. Then, OTC should not be currently offered to known BRCA-pathogenic variant carriers as a first choice for FP. Most of the patients who undergo OTC at the time of cancer diagnosis have no information available yet on their BRCA status. However, this information should be known before the transplantation of cryopreserved tissue. In this scenario, the most crucial issue is the choice of the transplantation site to ensure that all ovarian tissue is then removed at the time of RRSO [57].
The possibility of in vitro maturation (IVM) of immature oocytes retrieved during ovarian tissue processing is currently being investigated with some promising preliminary results [92], but it is not yet a clinical reality. Similarly, advancements on a fibrin matrix (the “artificial ovary”) to harvest ovarian tissue outside the body are currently at a translational research stage [93].
Recommendation 7: OTC should not be in principle recommended for known BRCA pathogenic variant carriers because of the potential OC risk associated with the transplantation of ovarian tissue. However, in selected cases and motivated patients diagnosed several years before the recommended age of RRSO, OTC may be considered with caution when other possibilities are not feasible, but special considerations also for the transplantation procedure are needed. IVM is considered a promising but still experimental strategy in this setting.

Question 8: Should PGT-M always be discussed with BRCA-pathogenic variant carriers of reproductive age?

In 2003, the Ethics Taskforce of the European Society of Human Reproduction and Embryology (ESHRE) defined PGT-M acceptable for late-onset and multifactorial diseases, including hereditary BC and OC [94]. In 2008, the first live birth following PGT-M for BRCA1 pathogenic variant carrier was reported by Jasper et al. [95]. Then, Derks-Smeets et al. published one of the largest experiences [96], consisting of 70 couples that underwent PGT for BRCA pathogenic variants. However, the reported uptake of the technique is still very low [9799]. On one hand, this may be a consequence of suboptimal knowledge and referral behavior of the involved professionals [100102]. Data on women’s uptake of the procedure, when proposed, are also conflictual. Recent surveys were focused on the attitudes about PGT-M among women with BRCA pathogenic variants. Most of the respondents declared that PGT should be offered, but less than the half would have used this option for themselves [103106]. Patients’ attitude to PGT depends on their family and/or personal history of cancer, reproductive history, or both. Several couples expressed that financial access was a major barrier to PGT. Nonetheless, even when IVF/PGT was offered at no cost to BRCA pathogenic variant carriers, its uptake was low [106]. Indeed, a previous condition of infertility seemed to be one of the most significant predictors of IVF/PGT use, suggesting that BRCA status is secondary to infertility in the decision-making process for PGT-M [107].
The ethical issues are maybe the most crucial. The American Society for Reproductive Medicine (ASRM) may justify PGT-M “when the conditions are serious and when there are no known interventions for the conditions, or the available interventions are either inadequately effective or significantly burdensome” [108]. Carrying a BRCA pathogenic variant is not a disease, but a condition that increases the risk of developing different cancers later in life. In addition, BRCA genes have an incomplete and variable penetrance and the disease is effectively curable in many cases. Despite these arguments, the anxiety and anguish involved in the need for lifetime preventive testing ethically justify PGT “as a matter of reproductive liberty” [109].
Regarding medical safety, children born after PGT seem to have comparable outcomes in terms of general health and development milestones to those born after IVF only and after natural conception in families carrying risk for a monogenic disease [110]. Recently, an increased incidence of pre-eclampsia was reported in pregnancies after trophectoderm biopsy [111]. The causative link behind the procedure and this finding is, however, still to be confirmed in larger cohorts.
All these elements deeply influence women’s choices and physician attitude to PGT-M, making it very hard to develop clear guidelines. Certainly, a better awareness of PGT-M, with both its indications and limits, has been associated with a greater PGT acceptability also among healthcare providers [112], and it will lead to less moral and ethical reservations, putting couples’ autonomy in the center of the decision-making process.
Recommendation 8: Carriers of BRCA pathogenic variants interested in avoiding the transmission of their mutation to the offspring have to be informed about the possibility to undergo PGT-M. A thorough and balanced genetic and fertility counseling should be offered to all interested carriers, underlying pros and cons of the procedure.

Conclusion

Optimal reproductive counseling in BRCA-mutation carriers still represents an unmet challenge, and several misconceptions about the topic persist among health care providers and patients.
The question of a systematic FP approach in BRCA carriers, even before the onset of cancer, remains an open issue, and several aspects should be considered in the decision-making process:
1.
Age at the time of BRCA-mutation disclosure;
 
2.
The potential negative effect of BRCA pathogenic variants on women ovarian reserve;
 
3.
The possibility to prevent the transmission of BRCA pathogenic variant to offspring with the use of PGT-M;
 
4.
The timing of RRSO, according to BRCA1 or BRCA2 pathogenic variants and family history;
 
5.
The need of gonadotoxic anticancer treatments and/or a long-lasting adjuvant ET in BRCA1/2-mutation carriers affected by cancer;
 
6.
The safety and feasibility of pregnancy after BC in patients with germline BRCA mutations.
 
During the meeting, with the participation of patient advocacy and delegates from different countries, two main criticisms emerged: (1) patients’ feeling of overload by an excess of information during fertility counseling, resulting in confusion and misunderstandings; (2) a discrepant management also related to the geographical and socioeconomic setting. A well-structured fertility counseling should be proposed to BRCA1/2 pathogenic variant carriers, both healthy and affected, pre- and post-testing, in order to early identify a personalized and suitable strategy of FP.
On the basis of the discussion, the expert panel has drafted a total of 8 recommendations (Table 1). Although there is a great interest in this field, the lack of large prospective studies on these topics highlights the need of further research efforts. Males who carry BRCA1/2 pathogenic variants should be included in the counseling process about the risk of transmission and the possible use of PGT-M. Awareness should be implemented, also among medical professionals, regarding all the mentioned reproductive health-specific issues of BRCA1/2 pathogenic variant carriers, from fertility to PGT to pregnancy after cancer, including indications to contraception and hormonal menopause therapy.
Table 1
The 8 recommendations drafted by the expert panel
Recommendations
1) In healthy BRCA mutation carriers, the impact of pregnancy and breastfeeding remains controversial. Women should be encouraged to complete childbearing at early age and discuss thorough breast follow-up during pregnancy and breastfeeding if they decide to maintain their breasts. In BRCA-mutated breast cancer patients, subsequent pregnancy following adequate treatment and follow-up does not seem to increase the risk of breast cancer recurrence and should not be discouraged.
2) A potential negative impact of BRCA pathogenic variants on women ovarian reserve and reproductive potential cannot be excluded and should be discussed; however, no strong conclusions can be drawn from existing data. Healthy carriers should be advised to not delay pregnancy beyond 35 years.
3) A potential negative impact of BRCA pathogenic variants on women ovarian reserve and reproductive potential including a diminished response to ovarian stimulation in breast cancer patients cannot be excluded and should be discussed during the oncofertility counseling; however, no strong conclusions can be drawn from existing data.
4) The available limited evidence does not demonstrate an increased risk of chemotherapy-induced POI in BRCA carriers but the overall risk remains significant for all patients and fertility preservation should be discussed with all women diagnosed at reproductive age. No data exist on the gonadotoxicity of newer treatment options in these patients.
5) Temporary ovarian suppression obtained by administering GnRHa during (neo-)adjuvant chemotherapy should be offered to all patients with cancer who wish to preserve ovarian function, including BRCA carriers diagnosed years before the recommended age of risk-reducing surgery. Most of the available efficacy data exist in women with breast cancer not carrying BRCA pathogenic variants. GnRHa use during chemotherapy does not replace established fertility preservation methods.
6) Despite the lack of exhaustive data, oocyte and embryo cryopreservation following COS can be considered a safe option that should be proposed both to BRCA-healthy carriers and patients with cancer interested in fertility preservation, whenever feasible. The use of letrozole to suppress supra-physiologic estrogens can be used safely without a reduction in the number or quality of oocytes.
7) Ovarian tissue cryopreservation should not be in principle recommended for known BRCA pathogenic variant carriers because of the potential ovarian cancer risk associated with the transplantation of ovarian tissue. However, in selected cases and motivated patients diagnosed several years before the recommended age of RRSO, ovarian tissue cryopreservation may be considered with caution when other possibilities are not feasible, but special considerations also for the transplantation procedure are needed. IVM is considered a promising but still experimental strategy in this setting.
8) Carriers of BRCA pathogenic variants interested in avoiding the transmission of their mutation to the offspring have to be informed about the possibility to undergo PGT-M. A thorough and balanced genetic and fertility counseling should be offered to all interested carriers, underlying pros and cons of the procedure.

Acknowledgements

The Authors thank the following patients’ associations for their guidance in the definition of priorities and their continuous support: aBRCAdabra Onlus and ACTO Campania Onlus.

Declarations

Not applicable
Not applicable

Competing interests

Fedro A Peccatori received honoraria from Roche Diagnostic and Ipsen outside the submitted work. Matteo Lambertini acted as a consultant for Roche, AstraZeneca, Lilly, and Novartis and received speaker honoraria from Sandoz, Roche, Takeda, Pfizer, Lilly, and Novartis outside the submitted work. Fedro A Peccatori received honoraria from Roche Diagnostic and Ipsen outside the submitted work. All the other authors declare no conflicts of interest.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
2.
Zurück zum Zitat Kuchenbaecker KB, Hopper JL, Barnes DR, Phillips KA, Mooij TM, Roos-Blom MJ, et al. Risks of breast, ovarian, and contralateral breast cancer for BRCA1 and BRCA2 mutation carriers. JAMA. 2017;317(23):2402–16.PubMedCrossRef Kuchenbaecker KB, Hopper JL, Barnes DR, Phillips KA, Mooij TM, Roos-Blom MJ, et al. Risks of breast, ovarian, and contralateral breast cancer for BRCA1 and BRCA2 mutation carriers. JAMA. 2017;317(23):2402–16.PubMedCrossRef
3.
Zurück zum Zitat Familial breast cancer: classification, care and managing breast cancer and related risks in people with a family history of breast cancer. Cardiff (UK): National Collaborating Centre for Cancer (UK); 2013. Familial breast cancer: classification, care and managing breast cancer and related risks in people with a family history of breast cancer. Cardiff (UK): National Collaborating Centre for Cancer (UK); 2013.
6.
Zurück zum Zitat Paluch-Shimon S, Cardoso F, Sessa C, Balmana J, Cardoso MJ, Gilbert F, et al. ESMO Guidelines Committee. Prevention and screening in BRCA mutation carriers and other breast/ovarian hereditary cancer syndromes: ESMO Clinical Practice Guidelines for cancer prevention and screening. Ann Oncol. 2016;27(suppl 5):v103–10.PubMedCrossRef Paluch-Shimon S, Cardoso F, Sessa C, Balmana J, Cardoso MJ, Gilbert F, et al. ESMO Guidelines Committee. Prevention and screening in BRCA mutation carriers and other breast/ovarian hereditary cancer syndromes: ESMO Clinical Practice Guidelines for cancer prevention and screening. Ann Oncol. 2016;27(suppl 5):v103–10.PubMedCrossRef
7.
Zurück zum Zitat Lambertini M, Di Maio M, Poggio F, et al. Knowledge, attitudes and practice of physicians towards fertility and pregnancy-related issues in young BRCA-mutated breast cancer patients. Reprod Biomed Online. 2019;38(5):835–44.PubMedCrossRef Lambertini M, Di Maio M, Poggio F, et al. Knowledge, attitudes and practice of physicians towards fertility and pregnancy-related issues in young BRCA-mutated breast cancer patients. Reprod Biomed Online. 2019;38(5):835–44.PubMedCrossRef
8.
Zurück zum Zitat Park B, Hopper JL, Win AK, et al. Reproductive factors as risk modifiers of breast cancer in BRCA mutation carriers and high-risk non-carriers. Oncotarget. 2017;8(60):102110–8.PubMedPubMedCentralCrossRef Park B, Hopper JL, Win AK, et al. Reproductive factors as risk modifiers of breast cancer in BRCA mutation carriers and high-risk non-carriers. Oncotarget. 2017;8(60):102110–8.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Victora CG, Bahl R, Barros AJ, França GV, Horton S, Krasevec J, et al. Breastfeeding in the 21st century: epidemiology, mechanisms, and lifelong effect. Lancet. 2016;387(10017):475–90.PubMedCrossRef Victora CG, Bahl R, Barros AJ, França GV, Horton S, Krasevec J, et al. Breastfeeding in the 21st century: epidemiology, mechanisms, and lifelong effect. Lancet. 2016;387(10017):475–90.PubMedCrossRef
17.
Zurück zum Zitat Valentini A, Lubinski J, Byrski T, et al. The impact of pregnancy on breast cancer survival in women who carry a BRCA1 or BRCA2 mutation. Breast Cancer Res Treat. 2013;142(1):177–85.PubMedPubMedCentralCrossRef Valentini A, Lubinski J, Byrski T, et al. The impact of pregnancy on breast cancer survival in women who carry a BRCA1 or BRCA2 mutation. Breast Cancer Res Treat. 2013;142(1):177–85.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Lambertini M, Blondeaux E, Bruzzone M, Perachino M, Anderson RA, de Azambuja E, Poorvu PD, Kim HJ, Villarreal-Garza C, Pistilli B, Vaz-Luis I, Saura C, Ruddy KJ, Franzoi MA, Sertoli C, Ceppi M, Azim HA Jr, Amant F, Demeestere I, Del Mastro L, Partridge AH, Pagani O, Peccatori FA. Pregnancy After Breast Cancer: A Systematic Review and Meta-Analysis. J Clin Oncol. 2021; [ahead of print] doi: https://doi.org/10.1200/JCO.21.00535. Lambertini M, Blondeaux E, Bruzzone M, Perachino M, Anderson RA, de Azambuja E, Poorvu PD, Kim HJ, Villarreal-Garza C, Pistilli B, Vaz-Luis I, Saura C, Ruddy KJ, Franzoi MA, Sertoli C, Ceppi M, Azim HA Jr, Amant F, Demeestere I, Del Mastro L, Partridge AH, Pagani O, Peccatori FA. Pregnancy After Breast Cancer: A Systematic Review and Meta-Analysis. J Clin Oncol. 2021; [ahead of print] doi: https://​doi.​org/​10.​1200/​JCO.​21.​00535.
21.
Zurück zum Zitat Pagani O, Ruggeri M, Manunta S, et al. Pregnancy after breast cancer: Are young patients willing to participate in clinical studies? Breast. 2015;24(3):201–7.PubMedCrossRef Pagani O, Ruggeri M, Manunta S, et al. Pregnancy after breast cancer: Are young patients willing to participate in clinical studies? Breast. 2015;24(3):201–7.PubMedCrossRef
23.
Zurück zum Zitat Turan V, Oktay K. BRCA-related ATM-mediated DNA double-strand break repair and ovarian aging. Hum Reprod Update. 2020;26(1):43–57.PubMedCrossRef Turan V, Oktay K. BRCA-related ATM-mediated DNA double-strand break repair and ovarian aging. Hum Reprod Update. 2020;26(1):43–57.PubMedCrossRef
24.
Zurück zum Zitat Wang ET, Pisarska MD, Bresee C, et al. BRCA1 germline mutations may be associated with reduced ovarian reserve. Fertil Steril. 2014;102(6):1723–8.PubMedPubMedCentralCrossRef Wang ET, Pisarska MD, Bresee C, et al. BRCA1 germline mutations may be associated with reduced ovarian reserve. Fertil Steril. 2014;102(6):1723–8.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Giordano S, Garrett-Mayer E, Mittal N, et al. Association of BRCA1 Mutations with Impaired Ovarian Reserve: Connection Between Infertility and Breast/Ovarian Cancer Risk. J Adolesc Young Adult Oncol. 2016;5(4):337–43.PubMedPubMedCentralCrossRef Giordano S, Garrett-Mayer E, Mittal N, et al. Association of BRCA1 Mutations with Impaired Ovarian Reserve: Connection Between Infertility and Breast/Ovarian Cancer Risk. J Adolesc Young Adult Oncol. 2016;5(4):337–43.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Phillips KA, Collins IM, Milne RL, et al. Anti-Müllerian hormone serum concentrations of women with germline BRCA1 or BRCA2 mutations. Hum Reprod. 2016;31(5):1126–32.PubMedPubMedCentralCrossRef Phillips KA, Collins IM, Milne RL, et al. Anti-Müllerian hormone serum concentrations of women with germline BRCA1 or BRCA2 mutations. Hum Reprod. 2016;31(5):1126–32.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Johnson L, Sammel MD, Domchek S, Schanne A, Prewitt M, Gracia C. Antimüllerian hormone levels are lower in BRCA2 mutation carriers. Fertil Steril. 2017;107(5):1256–1265.e6.PubMedPubMedCentralCrossRef Johnson L, Sammel MD, Domchek S, Schanne A, Prewitt M, Gracia C. Antimüllerian hormone levels are lower in BRCA2 mutation carriers. Fertil Steril. 2017;107(5):1256–1265.e6.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Ben-Aharon I, Levi M, Margel D, Yerushalmi R, Rizel S, Perry S, et al. Premature ovarian aging in BRCA carriers: a prototype of systemic precocious aging? Oncotarget. 2018;9:15931–41.PubMedPubMedCentralCrossRef Ben-Aharon I, Levi M, Margel D, Yerushalmi R, Rizel S, Perry S, et al. Premature ovarian aging in BRCA carriers: a prototype of systemic precocious aging? Oncotarget. 2018;9:15931–41.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Ponce J, Fernandez-Gonzalez S, Calvo I, et al. Assessment of ovarian reserve and reproductive outcomes in BRCA1 or BRCA2 mutation carriers. Int J Gynecol Cancer. 2020;30(1):83–8.PubMedCrossRef Ponce J, Fernandez-Gonzalez S, Calvo I, et al. Assessment of ovarian reserve and reproductive outcomes in BRCA1 or BRCA2 mutation carriers. Int J Gynecol Cancer. 2020;30(1):83–8.PubMedCrossRef
31.
Zurück zum Zitat Gal I, Sadetzki S, Gershoni-Baruch R, Oberman B, Carp H, Papa MZ, et al. Offspring gender ratio and the rate of recurrent spontaneous miscarriages in Jewish women at high risk for breast/ovarian cancer. Am J Hum Genet. 2004;74(6):1270–5.PubMedPubMedCentralCrossRef Gal I, Sadetzki S, Gershoni-Baruch R, Oberman B, Carp H, Papa MZ, et al. Offspring gender ratio and the rate of recurrent spontaneous miscarriages in Jewish women at high risk for breast/ovarian cancer. Am J Hum Genet. 2004;74(6):1270–5.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Friedman E, Kotsopoulos J, Lubinski J, Lynch HT, Ghadirian P, Neuhausen SL, et al. Spontaneous and therapeutic abortions and the risk of breast cancer among BRCA mutation carriers. Breast Cancer Res. 2006;8(2):R15.PubMedPubMedCentralCrossRef Friedman E, Kotsopoulos J, Lubinski J, Lynch HT, Ghadirian P, Neuhausen SL, et al. Spontaneous and therapeutic abortions and the risk of breast cancer among BRCA mutation carriers. Breast Cancer Res. 2006;8(2):R15.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Moslehi R, Singh R, Lessner L, Friedman JM. Impact of BRCA mutations on female fertility and offspring sex ratio. Am J Hum Biol. 2010;22(2):201–5.PubMedPubMedCentral Moslehi R, Singh R, Lessner L, Friedman JM. Impact of BRCA mutations on female fertility and offspring sex ratio. Am J Hum Biol. 2010;22(2):201–5.PubMedPubMedCentral
34.
Zurück zum Zitat Pal T, Keefe D, Sun P, Narod SA. Hereditary Breast Cancer Clinical Study Group. Fertility in women with BRCA mutations: a case-control study. Fertil Steril. 2010;93(6):1805–8.PubMedCrossRef Pal T, Keefe D, Sun P, Narod SA. Hereditary Breast Cancer Clinical Study Group. Fertility in women with BRCA mutations: a case-control study. Fertil Steril. 2010;93(6):1805–8.PubMedCrossRef
36.
Zurück zum Zitat van Tilborg TC, Derks-Smeets IAP, Bos AME, Oosterwijk JC, van Golde RJ, de Die-Smulders CE, et al. Serum AMH levels in healthy women from BRCA1/2 mutated families: are they reduced? Hum Reprod. 2016;31(11):2651–9.PubMedCrossRef van Tilborg TC, Derks-Smeets IAP, Bos AME, Oosterwijk JC, van Golde RJ, de Die-Smulders CE, et al. Serum AMH levels in healthy women from BRCA1/2 mutated families: are they reduced? Hum Reprod. 2016;31(11):2651–9.PubMedCrossRef
38.
Zurück zum Zitat Michaelson-Cohen R, Mor P, Srebnik N, Beller U, Levy-Lahad E, Eldar-Geva T. BRCA mutation carriers do not have compromised ovarian reserve. Int J Gynecol Cancer. 2014;24(2):233–7.PubMedCrossRef Michaelson-Cohen R, Mor P, Srebnik N, Beller U, Levy-Lahad E, Eldar-Geva T. BRCA mutation carriers do not have compromised ovarian reserve. Int J Gynecol Cancer. 2014;24(2):233–7.PubMedCrossRef
39.
Zurück zum Zitat Finch A, Valentini A, Greenblatt E, Lynch HT, Ghadirian P, Armel S, et al. Frequency of premature menopause in women who carry a BRCA1 or BRCA2 mutation. Fertil Steril. 2013;99(6):1724–8.PubMedCrossRef Finch A, Valentini A, Greenblatt E, Lynch HT, Ghadirian P, Armel S, et al. Frequency of premature menopause in women who carry a BRCA1 or BRCA2 mutation. Fertil Steril. 2013;99(6):1724–8.PubMedCrossRef
42.
Zurück zum Zitat Lin WT, Beattie M, Chen L-M, Oktay K, Crawford SL, Gold EB, et al. Comparison of age at natural menopause in BRCA1/2 mutation carriers with a non-clinic- based sample of women in northern California. Cancer. 2013;119(9):1652–9.PubMedCrossRef Lin WT, Beattie M, Chen L-M, Oktay K, Crawford SL, Gold EB, et al. Comparison of age at natural menopause in BRCA1/2 mutation carriers with a non-clinic- based sample of women in northern California. Cancer. 2013;119(9):1652–9.PubMedCrossRef
45.
Zurück zum Zitat Gunes S, Al-Sadaan M, Agarwal A. Spermatogenesis, DNA damage and DNA repair mechanisms in male infertility. Reprod Biomed Online. 2015;31(3):309–19.PubMedCrossRef Gunes S, Al-Sadaan M, Agarwal A. Spermatogenesis, DNA damage and DNA repair mechanisms in male infertility. Reprod Biomed Online. 2015;31(3):309–19.PubMedCrossRef
46.
Zurück zum Zitat Simhadri S, Peterson S, Patel DS, et al. Male fertility defect associated with disrupted BRCA1-PALB2 interaction in mice. J Biol Chem. 2014;289(35):24617–29.PubMedPubMedCentralCrossRef Simhadri S, Peterson S, Patel DS, et al. Male fertility defect associated with disrupted BRCA1-PALB2 interaction in mice. J Biol Chem. 2014;289(35):24617–29.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Vuković P, Peccatori FA, Massarotti C, Miralles MS, Beketić-Orešković L, Lambertini M. Preimplantation genetic testing for carriers of BRCA1/2 pathogenic variants. Crit Rev Oncol Hematol. 2021;157:103201.PubMedCrossRef Vuković P, Peccatori FA, Massarotti C, Miralles MS, Beketić-Orešković L, Lambertini M. Preimplantation genetic testing for carriers of BRCA1/2 pathogenic variants. Crit Rev Oncol Hematol. 2021;157:103201.PubMedCrossRef
48.
50.
Zurück zum Zitat Porcu E, Cillo GM, Cipriani L, et al. Impact of BRCA1 and BRCA2 mutations on ovarian reserve and fertility preservation outcomes in young women with breast cancer. J Assist Reprod Genet. 2020;37(3):709–15.PubMedCrossRef Porcu E, Cillo GM, Cipriani L, et al. Impact of BRCA1 and BRCA2 mutations on ovarian reserve and fertility preservation outcomes in young women with breast cancer. J Assist Reprod Genet. 2020;37(3):709–15.PubMedCrossRef
51.
Zurück zum Zitat Shapira M, Raanani H, Feldman B, Srebnik N, Dereck-Haim S, Manela D, et al. BRCA mutation carriers show normal ovarian response in vitro fertilization cycles. Fertil Steril. 2015;104:1162–7.PubMedCrossRef Shapira M, Raanani H, Feldman B, Srebnik N, Dereck-Haim S, Manela D, et al. BRCA mutation carriers show normal ovarian response in vitro fertilization cycles. Fertil Steril. 2015;104:1162–7.PubMedCrossRef
52.
Zurück zum Zitat Lambertini M, Goldrat O, Ferreira AR, Dechene J, Azim HA Jr, Desir J, et al. t'Kint de Roodenbeke MD, de Azambuja E, Ignatiadis M, Demeestere I. Reproductive potential and performance of fertility preservation strategies in BRCA-mutated breast cancer patients. Ann Oncol. 2018;29(1):237–43.PubMedCrossRef Lambertini M, Goldrat O, Ferreira AR, Dechene J, Azim HA Jr, Desir J, et al. t'Kint de Roodenbeke MD, de Azambuja E, Ignatiadis M, Demeestere I. Reproductive potential and performance of fertility preservation strategies in BRCA-mutated breast cancer patients. Ann Oncol. 2018;29(1):237–43.PubMedCrossRef
53.
Zurück zum Zitat Gunnala V, Fields J, Irani M, et al. BRCA carriers have similar reproductive potential at baseline to noncarriers: comparisons in cancer and cancer-free cohorts undergoing fertility preservation. Fertil Steril. 2019;111(2):363–71.PubMedCrossRef Gunnala V, Fields J, Irani M, et al. BRCA carriers have similar reproductive potential at baseline to noncarriers: comparisons in cancer and cancer-free cohorts undergoing fertility preservation. Fertil Steril. 2019;111(2):363–71.PubMedCrossRef
55.
Zurück zum Zitat Lambertini M, Olympios N, Lequesne J, Calbrix C, Fontanilles M, Loeb A, et al. Impact of Taxanes, Endocrine Therapy, and Deleterious Germline BRCA Mutations on Anti-müllerian Hormone Levels in Early Breast Cancer Patients Treated With Anthracycline- and Cyclophosphamide-Based Chemotherapy. Front Oncol. 2019;9:575.PubMedPubMedCentralCrossRef Lambertini M, Olympios N, Lequesne J, Calbrix C, Fontanilles M, Loeb A, et al. Impact of Taxanes, Endocrine Therapy, and Deleterious Germline BRCA Mutations on Anti-müllerian Hormone Levels in Early Breast Cancer Patients Treated With Anthracycline- and Cyclophosphamide-Based Chemotherapy. Front Oncol. 2019;9:575.PubMedPubMedCentralCrossRef
58.
61.
Zurück zum Zitat Tung N, Arun B, Hacker MR, Hofstatter E, Toppmeyer DL, Isakoff SJ, et al. TBCRC 031: Randomized Phase II Study of Neoadjuvant Cisplatin Versus Doxorubicin-Cyclophosphamide in Germline BRCA Carriers With HER2-Negative Breast Cancer (the INFORM trial). J Clin Oncol. 2020;38(14):1539–48.PubMedCrossRefPubMedCentral Tung N, Arun B, Hacker MR, Hofstatter E, Toppmeyer DL, Isakoff SJ, et al. TBCRC 031: Randomized Phase II Study of Neoadjuvant Cisplatin Versus Doxorubicin-Cyclophosphamide in Germline BRCA Carriers With HER2-Negative Breast Cancer (the INFORM trial). J Clin Oncol. 2020;38(14):1539–48.PubMedCrossRefPubMedCentral
65.
Zurück zum Zitat Lambertini M, Moore HCF, Leonard RCF, et al. Gonadotropin-Releasing Hormone Agonists During Chemotherapy for Preservation of Ovarian Function and Fertility in Premenopausal Patients With Early Breast Cancer: A Systematic Review and Meta-Analysis of Individual Patient-Level Data. J Clin Oncol. 2018;36(19):1981–90.PubMedPubMedCentralCrossRef Lambertini M, Moore HCF, Leonard RCF, et al. Gonadotropin-Releasing Hormone Agonists During Chemotherapy for Preservation of Ovarian Function and Fertility in Premenopausal Patients With Early Breast Cancer: A Systematic Review and Meta-Analysis of Individual Patient-Level Data. J Clin Oncol. 2018;36(19):1981–90.PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Lambertini M, Boni L, Michelotti A, Gamucci T, Scotto T, Gori S, et al. Ovarian suppression with triptorelin during adjuvant breast cancer chemotherapy and long-term ovarian function, pregnancies, and disease-free survival: a randomized clinical trial. JAMA. 2015;314(24):2632–40.PubMedCrossRef Lambertini M, Boni L, Michelotti A, Gamucci T, Scotto T, Gori S, et al. Ovarian suppression with triptorelin during adjuvant breast cancer chemotherapy and long-term ovarian function, pregnancies, and disease-free survival: a randomized clinical trial. JAMA. 2015;314(24):2632–40.PubMedCrossRef
67.
Zurück zum Zitat Moore HCF, Unger JM, Phillips KA, Boyle F, Hitre E, et al. Final Analysis of the Prevention of Early Menopause Study (POEMS)/SWOG Intergroup S0230. J Natl Cancer Inst. 2019;111(2):210–3.PubMedCrossRef Moore HCF, Unger JM, Phillips KA, Boyle F, Hitre E, et al. Final Analysis of the Prevention of Early Menopause Study (POEMS)/SWOG Intergroup S0230. J Natl Cancer Inst. 2019;111(2):210–3.PubMedCrossRef
68.
Zurück zum Zitat Leonard RCF, Adamson DJA, Bertelli G, Mansi J, Yellowlees A, Dunlop J, et al. Anderson RA; Anglo Celtic Collaborative Oncology Group and National Cancer Research Institute Trialists. GnRH agonist for protection against ovarian toxicity during chemotherapy for early breast cancer: the Anglo Celtic Group OPTION trial. Ann Oncol. 2017;28(8):1811–6.PubMedCrossRef Leonard RCF, Adamson DJA, Bertelli G, Mansi J, Yellowlees A, Dunlop J, et al. Anderson RA; Anglo Celtic Collaborative Oncology Group and National Cancer Research Institute Trialists. GnRH agonist for protection against ovarian toxicity during chemotherapy for early breast cancer: the Anglo Celtic Group OPTION trial. Ann Oncol. 2017;28(8):1811–6.PubMedCrossRef
71.
Zurück zum Zitat Demeestere I, Brice P, Peccatori FA, Kentos A, Dupuis J, Zachee P, et al. No Evidence for the Benefit of Gonadotropin-Releasing Hormone Agonist in Preserving Ovarian Function and Fertility in Lymphoma Survivors Treated With Chemotherapy: Final Long-Term Report of a Prospective Randomized Trial. J Clin Oncol. 2016;34(22):2568–74. https://doi.org/10.1200/JCO.2015.65.8864.CrossRefPubMed Demeestere I, Brice P, Peccatori FA, Kentos A, Dupuis J, Zachee P, et al. No Evidence for the Benefit of Gonadotropin-Releasing Hormone Agonist in Preserving Ovarian Function and Fertility in Lymphoma Survivors Treated With Chemotherapy: Final Long-Term Report of a Prospective Randomized Trial. J Clin Oncol. 2016;34(22):2568–74. https://​doi.​org/​10.​1200/​JCO.​2015.​65.​8864.CrossRefPubMed
76.
Zurück zum Zitat Sergentanis TN, Diamantaras AA, Perlepe C, Kanavidis P, Skalkidou A, Petridou ET. IVF and breast cancer: a systematic review and meta-analysis. Hum Reprod Update. 2014;20:106–23.PubMedCrossRef Sergentanis TN, Diamantaras AA, Perlepe C, Kanavidis P, Skalkidou A, Petridou ET. IVF and breast cancer: a systematic review and meta-analysis. Hum Reprod Update. 2014;20:106–23.PubMedCrossRef
77.
Zurück zum Zitat Derks-Smeets IAP, Schrijver LH, de Die-Smulders CEM, et al. Ovarian stimulation for IVF and risk of primary breast cancer in BRCA1/2 mutation carriers. Br J Cancer. 2018;119(3):357–63.PubMedPubMedCentralCrossRef Derks-Smeets IAP, Schrijver LH, de Die-Smulders CEM, et al. Ovarian stimulation for IVF and risk of primary breast cancer in BRCA1/2 mutation carriers. Br J Cancer. 2018;119(3):357–63.PubMedPubMedCentralCrossRef
81.
Zurück zum Zitat Spaan M, van den Belt-Dusebout AW, Lambalk CB, van Boven HH, Schats R, Kortman M, et al. Long-Term Risk of Ovarian Cancer and Borderline Tumors After Assisted Reproductive Technology. J Natl Cancer Inst. 2021;113(6):699–709. https://doi.org/10.1093/jnci/djaa163. Spaan M, van den Belt-Dusebout AW, Lambalk CB, van Boven HH, Schats R, Kortman M, et al. Long-Term Risk of Ovarian Cancer and Borderline Tumors After Assisted Reproductive Technology. J Natl Cancer Inst. 2021;113(6):699–709. https://​doi.​org/​10.​1093/​jnci/​djaa163.
82.
Zurück zum Zitat Perri T, Lifshitz D, Sadetzki S, et al. Fertility treatments and invasive epithelial ovarian cancer risk in Jewish Israeli BRCA1 or BRCA2 mutation carriers. Fertil Steril. 2015;103(5):1305–12.PubMedCrossRef Perri T, Lifshitz D, Sadetzki S, et al. Fertility treatments and invasive epithelial ovarian cancer risk in Jewish Israeli BRCA1 or BRCA2 mutation carriers. Fertil Steril. 2015;103(5):1305–12.PubMedCrossRef
83.
Zurück zum Zitat Oktay K, Harvey BE, Partridge AH, Quinn GP, Reinecke J, Taylor HS, et al. Fertility preservation in patients with cancer: ASCO clinical practice guideline update. J Clin Oncol. 2018;36:1994–2001.PubMedCrossRef Oktay K, Harvey BE, Partridge AH, Quinn GP, Reinecke J, Taylor HS, et al. Fertility preservation in patients with cancer: ASCO clinical practice guideline update. J Clin Oncol. 2018;36:1994–2001.PubMedCrossRef
84.
Zurück zum Zitat Ethics Committee of the American Society for Reproductive Medicine. Fertility preservation and reproduction in patients facing gonadotoxic therapies: an Ethics Committee opinion. Fertil Steril. 2018;110(3):380–86. Ethics Committee of the American Society for Reproductive Medicine. Fertility preservation and reproduction in patients facing gonadotoxic therapies: an Ethics Committee opinion. Fertil Steril. 2018;110(3):380–86.
86.
Zurück zum Zitat Kim J, Turan V, Oktay K. Long-Term Safety of Letrozole and Gonadotropin Stimulation for Fertility Preservation in Women With Breast Cancer. J Clin Endocrinol Metab. 2016;101(4):1364–71.PubMedPubMedCentralCrossRef Kim J, Turan V, Oktay K. Long-Term Safety of Letrozole and Gonadotropin Stimulation for Fertility Preservation in Women With Breast Cancer. J Clin Endocrinol Metab. 2016;101(4):1364–71.PubMedPubMedCentralCrossRef
91.
Zurück zum Zitat Jensen AK, Macklon KT, Fedder J, Ernst E, Humaidan P, Andersen CY. 86 successful births and 9 ongoing pregnancies worldwide in women transplanted with frozen-thawed ovarian tissue: focus on birth and perinatal outcome in 40 of these children. J Assist Reprod Genet. 2017;34(3):325–36. https://doi.org/10.1007/s10815-016-0843-9. Jensen AK, Macklon KT, Fedder J, Ernst E, Humaidan P, Andersen CY. 86 successful births and 9 ongoing pregnancies worldwide in women transplanted with frozen-thawed ovarian tissue: focus on birth and perinatal outcome in 40 of these children. J Assist Reprod Genet. 2017;34(3):325–36. https://​doi.​org/​10.​1007/​s10815-016-0843-9.
94.
Zurück zum Zitat Shenfield F, Pennings G, Devroey P, Sureau C, Tarlatzis B, Cohen J. ESHRE ethics task force. Taskforce 5: preimplantation genetic diagnosis. Hum Reprod. 2003;18:649–51.PubMedCrossRef Shenfield F, Pennings G, Devroey P, Sureau C, Tarlatzis B, Cohen J. ESHRE ethics task force. Taskforce 5: preimplantation genetic diagnosis. Hum Reprod. 2003;18:649–51.PubMedCrossRef
95.
Zurück zum Zitat Jasper MJ, Liebelt J, Hussey ND. Preimplantation genetic diagnosis for BRCA1 exon 13 duplication mutation using linked polymorphic markers resulting in a live birth. Prenat Diagn. 2008;28:292–8.PubMedCrossRef Jasper MJ, Liebelt J, Hussey ND. Preimplantation genetic diagnosis for BRCA1 exon 13 duplication mutation using linked polymorphic markers resulting in a live birth. Prenat Diagn. 2008;28:292–8.PubMedCrossRef
96.
Zurück zum Zitat Derks-Smeets IA, de Die-Smulders CE, Mackens S, van Golde R, Paulussen AD, Dreesen J, et al. Hereditary breast and ovarian cancer and reproduction: an observational study on the suitability of preimplantation genetic diagnosis for both asymptomatic carriers and breast cancer survivors. Breast Cancer Res Treat. 2014;145:673–81.PubMedCrossRef Derks-Smeets IA, de Die-Smulders CE, Mackens S, van Golde R, Paulussen AD, Dreesen J, et al. Hereditary breast and ovarian cancer and reproduction: an observational study on the suitability of preimplantation genetic diagnosis for both asymptomatic carriers and breast cancer survivors. Breast Cancer Res Treat. 2014;145:673–81.PubMedCrossRef
97.
Zurück zum Zitat Cunningham J, Goldsmith L, Skirton H. The evidence base regarding the experiences of and attitudes to preimplantation genetic diagnosis in prospective clients. Midwifery. 2015;31:288–96.PubMedCrossRef Cunningham J, Goldsmith L, Skirton H. The evidence base regarding the experiences of and attitudes to preimplantation genetic diagnosis in prospective clients. Midwifery. 2015;31:288–96.PubMedCrossRef
98.
Zurück zum Zitat Donnelly L, Watson M, Moynihan C, Bancroft E, Evans D, Eeles R, et al. Reproductive decision-making in young female carriers of a BRCA mutation. Human Reproduction. 2013;28(4):1006–12.PubMedCrossRef Donnelly L, Watson M, Moynihan C, Bancroft E, Evans D, Eeles R, et al. Reproductive decision-making in young female carriers of a BRCA mutation. Human Reproduction. 2013;28(4):1006–12.PubMedCrossRef
99.
Zurück zum Zitat Quinn GP, Pal T, Murphy D, Vadaparampil ST, Kumar A. High-risk consumers’ perceptions of preimplantation genetic diagnosis for hereditary cancers: a systematic review and meta-analysis. Genet Med. 2012;14:191–200.PubMedCrossRef Quinn GP, Pal T, Murphy D, Vadaparampil ST, Kumar A. High-risk consumers’ perceptions of preimplantation genetic diagnosis for hereditary cancers: a systematic review and meta-analysis. Genet Med. 2012;14:191–200.PubMedCrossRef
100.
Zurück zum Zitat Gietel-Habets JJG, de Die-Smulders CEM, Tjan-Heijnen VCG, et al. Professionals' knowledge, attitude and referral behaviour of preimplantation genetic diagnosis for hereditary breast and ovarian cancer. Reprod Biomed Online. 2018;36(2):137–44.PubMedCrossRef Gietel-Habets JJG, de Die-Smulders CEM, Tjan-Heijnen VCG, et al. Professionals' knowledge, attitude and referral behaviour of preimplantation genetic diagnosis for hereditary breast and ovarian cancer. Reprod Biomed Online. 2018;36(2):137–44.PubMedCrossRef
101.
Zurück zum Zitat Abbate BJ, Klitzman R, Chung WK, Ottman R, Leu CS, Appelbaum PS. Views of preimplantation genetic diagnosis (PGD) among psychiatrists and neurologists. J. Reprod. Med. 2014;59:385–92.PubMedPubMedCentral Abbate BJ, Klitzman R, Chung WK, Ottman R, Leu CS, Appelbaum PS. Views of preimplantation genetic diagnosis (PGD) among psychiatrists and neurologists. J. Reprod. Med. 2014;59:385–92.PubMedPubMedCentral
102.
Zurück zum Zitat Caldas GH, Caldas E, Araújo ED, Bonetti TCS, Leal CB, Costa AM. Opinions concerning pre-implantation genetic diagnosis and sex selection among gynecologist-obstetricians in Brazil. Eur. J. Obstet. Gynecol. Reprod. Biol. 2010;148:158–62.PubMedCrossRef Caldas GH, Caldas E, Araújo ED, Bonetti TCS, Leal CB, Costa AM. Opinions concerning pre-implantation genetic diagnosis and sex selection among gynecologist-obstetricians in Brazil. Eur. J. Obstet. Gynecol. Reprod. Biol. 2010;148:158–62.PubMedCrossRef
103.
Zurück zum Zitat Quinn GP, Vadaparampil ST, Tollin S, Miree CA, Murphy D, Bower B, et al. BRCA carriers’ thoughts on risk management in relation to preimplantation genetic diagnosis and childbearing: when too many choices are just as difficult as none. Fertil Steril. 2010;94(6):2473–5.PubMedCrossRef Quinn GP, Vadaparampil ST, Tollin S, Miree CA, Murphy D, Bower B, et al. BRCA carriers’ thoughts on risk management in relation to preimplantation genetic diagnosis and childbearing: when too many choices are just as difficult as none. Fertil Steril. 2010;94(6):2473–5.PubMedCrossRef
104.
Zurück zum Zitat Menon U, Harper J, Sharma A, Fraser L, Burnell M, ElMasry K, et al. Views of BRCA gene mutation carriers on preimplantation genetic diagnosis as a reproductive option for hereditary breast and ovarian cancer. Hum Reprod. 2007;22:1573–7.PubMedCrossRef Menon U, Harper J, Sharma A, Fraser L, Burnell M, ElMasry K, et al. Views of BRCA gene mutation carriers on preimplantation genetic diagnosis as a reproductive option for hereditary breast and ovarian cancer. Hum Reprod. 2007;22:1573–7.PubMedCrossRef
105.
Zurück zum Zitat Fortuny D, Balmana J, Grana B, Torres A, Ramon y Cajal T, Darder E, et al. Opinion about reproductive decision making among individuals undergoing BRCA1/2 genetic testing in a multicentre Spanish cohort. Hum Reprod. 2009;24:1000–6.PubMedCrossRef Fortuny D, Balmana J, Grana B, Torres A, Ramon y Cajal T, Darder E, et al. Opinion about reproductive decision making among individuals undergoing BRCA1/2 genetic testing in a multicentre Spanish cohort. Hum Reprod. 2009;24:1000–6.PubMedCrossRef
108.
Zurück zum Zitat Ethics Committee of American Society for Reproductive M. Use of preimplantation genetic diagnosis for serious adult-onset conditions: a committee opinion. Fertil Steril. 2013;100:54–7.CrossRef Ethics Committee of American Society for Reproductive M. Use of preimplantation genetic diagnosis for serious adult-onset conditions: a committee opinion. Fertil Steril. 2013;100:54–7.CrossRef
Metadaten
Titel
Reproductive issues in carriers of germline pathogenic variants in the BRCA1/2 genes: an expert meeting
verfasst von
Barbara Buonomo
Claudia Massarotti
Miriam Dellino
Paola Anserini
Alberta Ferrari
Maria Campanella
Mirosa Magnotti
Cristofaro De Stefano
Fedro Alessandro Peccatori
Matteo Lambertini
Publikationsdatum
01.12.2021
Verlag
BioMed Central
Erschienen in
BMC Medicine / Ausgabe 1/2021
Elektronische ISSN: 1741-7015
DOI
https://doi.org/10.1186/s12916-021-02081-7

Weitere Artikel der Ausgabe 1/2021

BMC Medicine 1/2021 Zur Ausgabe

Leitlinien kompakt für die Allgemeinmedizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Facharzt-Training Allgemeinmedizin

Die ideale Vorbereitung zur anstehenden Prüfung mit den ersten 24 von 100 klinischen Fallbeispielen verschiedener Themenfelder

Mehr erfahren

Reizdarmsyndrom: Diäten wirksamer als Medikamente

29.04.2024 Reizdarmsyndrom Nachrichten

Bei Reizdarmsyndrom scheinen Diäten, wie etwa die FODMAP-arme oder die kohlenhydratreduzierte Ernährung, effektiver als eine medikamentöse Therapie zu sein. Das hat eine Studie aus Schweden ergeben, die die drei Therapieoptionen im direkten Vergleich analysierte.

Akuter Schwindel: Wann lohnt sich eine MRT?

28.04.2024 Schwindel Nachrichten

Akuter Schwindel stellt oft eine diagnostische Herausforderung dar. Wie nützlich dabei eine MRT ist, hat eine Studie aus Finnland untersucht. Immerhin einer von sechs Patienten wurde mit akutem ischämischem Schlaganfall diagnostiziert.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Therapiestart mit Blutdrucksenkern erhöht Frakturrisiko

25.04.2024 Hypertonie Nachrichten

Beginnen ältere Männer im Pflegeheim eine Antihypertensiva-Therapie, dann ist die Frakturrate in den folgenden 30 Tagen mehr als verdoppelt. Besonders häufig stürzen Demenzkranke und Männer, die erstmals Blutdrucksenker nehmen. Dafür spricht eine Analyse unter US-Veteranen.

Update Allgemeinmedizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.