1
|
Loubersac S, Chaillot M, Reignier A, Lefebvre T, Dezellus A, Colombel A, Barriere P, Masson D, Freour T. Serum androgen dynamics in young women aged 18-40 treated with chemotherapy for breast cancer: an observational, multicentric, prospective study in France. HUM FERTIL 2024; 27:2350758. [PMID: 38957151 DOI: 10.1080/14647273.2024.2350758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 04/24/2024] [Indexed: 07/04/2024]
Abstract
Although the deleterious impact of chemotherapy regimen used to treat women of reproductive age with breast cancer on ovarian reserve has been extensively studied, hardly anything has been reported on the effect of these protocols on theca cell function and ovarian androgen secretion. The aim of this prospective multicentric cohort study was to describe serum levels of total testosterone and androstenedione during chemotherapy and 24-month follow-up in 250 patients <40 years treated for breast cancer. Mean basal levels of androstenedione and total testosterone at diagnosis were 1.68 ng/mL and 0.20 ng/mL respectively. No correlation with age was found. Serum levels of androstenedione and total testosterone rapidly decreased after chemotherapy completion, before slowly increasing and almost returning to basal levels in all patients during 2-year follow-up. In conclusion our study demonstrates a chemotherapy-induced alteration of ovarian thecal function, resulting in a significant decrease in serum androgen levels. This alteration of theca cell function adds to the well-known alteration of granulosa cell function, resulting in a global, but partly transient, ovarian failure in young women treated for breast cancer. These data bring new insight into ovarian physiology and emphasize the need for pre and post-treatment ovarian follow-up. Trial registration: ClinicalTrial.gov identifier NCT01114464.
Collapse
Affiliation(s)
- S Loubersac
- Service de médecine et biologie du développement et de la reproduction, CHU de Nantes, Bd Jean Monnet, Nantes, France
| | - M Chaillot
- Service de médecine et biologie du développement et de la reproduction, CHU de Nantes, Bd Jean Monnet, Nantes, France
- Faculté de médecine, Université de Nantes, Nantes, France
| | - A Reignier
- Service de médecine et biologie du développement et de la reproduction, CHU de Nantes, Bd Jean Monnet, Nantes, France
- Faculté de médecine, Université de Nantes, Nantes, France
- Centre de Recherche en Transplantation et Immunologie (ou CRTI), Inserm, Université de Nantes, Nantes, France
| | - T Lefebvre
- Service de médecine et biologie du développement et de la reproduction, CHU de Nantes, Bd Jean Monnet, Nantes, France
- Faculté de médecine, Université de Nantes, Nantes, France
| | - A Dezellus
- Service d'oncologie, Institut de Cancérologie de l'Ouest, Saint Herblain, France
| | - A Colombel
- Service de médecine et biologie du développement et de la reproduction, CHU de Nantes, Bd Jean Monnet, Nantes, France
| | - P Barriere
- Service de médecine et biologie du développement et de la reproduction, CHU de Nantes, Bd Jean Monnet, Nantes, France
- Faculté de médecine, Université de Nantes, Nantes, France
- Centre de Recherche en Transplantation et Immunologie (ou CRTI), Inserm, Université de Nantes, Nantes, France
| | - D Masson
- Faculté de médecine, Université de Nantes, Nantes, France
- Laboratoire de biochimie, CHU de Nantes, place Alexis-Ricordeau, Nantes, France
- INSERM UMR 913, rue Gaston Veil, Nantes, France
| | - T Freour
- Service de médecine et biologie du développement et de la reproduction, CHU de Nantes, Bd Jean Monnet, Nantes, France
- Faculté de médecine, Université de Nantes, Nantes, France
- Centre de Recherche en Transplantation et Immunologie (ou CRTI), Inserm, Université de Nantes, Nantes, France
| |
Collapse
|
2
|
Arecco L, Borea R, Magaton IM, Janković K, Mariamizde E, Stana M, Scavone G, Ottonello S, Spinaci S, Genova C, de Azambuja E, Lambertini M. Current practices in oncofertility counseling: updated evidence on fertility preservation and post-treatment pregnancies in young women affected by early breast cancer. Expert Rev Anticancer Ther 2024; 24:803-817. [PMID: 38913581 DOI: 10.1080/14737140.2024.2372337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 06/21/2024] [Indexed: 06/26/2024]
Abstract
INTRODUCTION Anticancer treatments have significantly contributed to increasing cure rates of breast cancer in the last years; however, they can also lead to short- and long-term side effects, including gonadotoxicity, and compromised fertility in young women. Oncofertility is a crucial issue for young patients who have not yet completed their family planning at the time of cancer diagnosis. AREAS COVERED This review aims to cover all the latest available evidence in the field of oncofertility, including the gonadotoxicity of currently adopted anticancer therapies in the curative breast cancer setting, the available strategies for fertility preservation and the feasibility of achieving a pregnancy following anticancer treatment completion. EXPERT OPINION Over the past years, a significant progress has been made in oncofertility care for young women with breast cancer. In the context of the currently available evidence, every young woman with newly diagnosed breast cancer should receive a proper and complete oncofertility counseling before starting any anticancer treatment to increase her chances of future pregnancies.
Collapse
Affiliation(s)
- Luca Arecco
- Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genova, Genova, Italy
- Academic Trials Promoting Team, Institut Jules Bordet, Université Libre de Bruxelles (U.L.B.), Hôpital Universitaire de Bruxelles (HUB), Brussels, Belgium
| | - Roberto Borea
- Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Isotta Martha Magaton
- Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genova, Genova, Italy
- Division of Gynaecological Endocrinology and Reproductive Medicine, University Women's Hospital, Bern, Switzerland
| | | | - Elene Mariamizde
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Oncology and Hematology, Todua Clinic, Tbilisi, Georgia
| | - Mihaela Stana
- Department of Medical Oncology, Elysee Hospital, Alba Iulia, Romania
| | - Graziana Scavone
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Silvia Ottonello
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy
| | - Stefano Spinaci
- ASL3 Breast Unit Department, Division of Breast Surgery, Ospedale Villa Scassi, Genova, Italy
| | - Carlo Genova
- Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Evandro de Azambuja
- Academic Trials Promoting Team, Institut Jules Bordet, Université Libre de Bruxelles (U.L.B.), Hôpital Universitaire de Bruxelles (HUB), Brussels, Belgium
| | - Matteo Lambertini
- Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
3
|
Peigné M, Mur P, Laup L, Hamy AS, Sifer C, Mayeur A, Eustache F, Sarandi S, Vinolas C, Rakrouki S, Benoit A, Grynberg M, Sonigo C. Fertility outcomes several years after urgent fertility preservation for patients with breast cancer. Fertil Steril 2024; 122:504-513. [PMID: 38679360 DOI: 10.1016/j.fertnstert.2024.04.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 04/22/2024] [Accepted: 04/22/2024] [Indexed: 05/01/2024]
Abstract
OBJECTIVE To study the fertility outcomes of women who tried to conceive after breast cancer (BC) treatment and fertility preservation. DESIGN Retrospective observational, bicentric cohort study. SETTING University hospital. PATIENTS Patients with BC. INTERVENTION All patients who had undergone fertility preservation before BC treatment between January 2013 and July 2019 were included (n = 844). The endpoint date was March 1, 2022. Patients with missing data on pregnancy attempts after a cancer diagnosis (n = 195) were excluded from the pregnancy analysis. MAIN OUTCOME MEASURES Cumulative incidences of pregnancy and live birth (LB) were calculated. For women who became pregnant, the time to conception was calculated between the first fertility preservation consultation and the estimated day of conception. For those who did not conceive, we considered the time between the first fertility preservation consultation and the endpoint date, or the date of patient death. A Cox regression model was used to study the predictive factors for pregnancy and LB. RESULTS Among the 649 patients with available data on pregnancy attempts after BC diagnosis, 255 (39.3% [35.5-43.2]) tried to conceive (median follow-up of 6.5 years). Overall, 135 (52.9% [46.6-59.2]) of these patients achieved a pregnancy, mainly through unassisted conception (79.3% [72.8-84.8]), and 99 reported an LB (representing 38.8% of patients who attempted conception). In our cohort, 48 months after the first fertility preservation consultation, the cumulative incidence of pregnancy was 33.1% ([27.6-37.9]). After adjustment for age, parity, type of chemotherapy administration, and endocrine therapy, only multiparity at diagnosis and absence of chemotherapy were positive predictive factors of pregnancy after cancer. Of the 793 patients who had vitrified oocytes and embryos, 68 used them (27% [21.3-32.5] of the patients who tried to conceive), resulting in 8 LBs (11.8% [5.2-21.9]). Women who used their cryopreserved oocytes and embryos were older at the first consultation of fertility preservation (hazard ratio 1.71 [1.42-2.21]), and chose more often to vitrify embryos (hazard ratio 1.76 [1.28-2.23]). CONCLUSION Although pregnancy rates after fertility preservation for patients with BC are low, most conceptions are achieved without medical assistance. Our findings provide useful information to advise women on the different techniques of fertility preservation, their efficacy, and safety, as well as the relatively high chances of unassisted conception.
Collapse
Affiliation(s)
- Maëliss Peigné
- Department of Reproductive Medicine and Fertility Preservation, Jean Verdier Hospital, AP-HP-Université Sorbonne Paris Nord, Bondy, France.
| | - Pauline Mur
- Department of Reproductive Medicine and Fertility Preservation, Antoine Beclère Hospital, AP-HP-Université Paris-Saclay, Clamart, France
| | - Laëtitia Laup
- Department of Reproductive Medicine and Fertility Preservation, Jean Verdier Hospital, AP-HP-Université Sorbonne Paris Nord, Bondy, France
| | - Anne-Sophie Hamy
- Department of Medical Oncology, Institut Curie, Université Paris Cité, Paris, France
| | - Christophe Sifer
- Embryology Unit, Jean Verdier Hospital, AP-HP-Université Sorbonne Paris Nord, Bondy, France
| | - Anne Mayeur
- Histology-Embryology-Cytogenetic Laboratory, Antoine Beclère Hospital, AP-HP-Université Paris- Saclay, Clamart, France
| | - Florence Eustache
- Le Centre d'Études et de Conservation des Œufs et du Sperme (CECOS), Jean Verdier Hospital, AP-HP-Université Sorbonne Paris Nord, Bondy, France
| | - Solmaz Sarandi
- Embryology Unit, Jean Verdier Hospital, AP-HP-Université Sorbonne Paris Nord, Bondy, France
| | - Claire Vinolas
- Department of Reproductive Medicine and Fertility Preservation, Jean Verdier Hospital, AP-HP-Université Sorbonne Paris Nord, Bondy, France
| | - Sophia Rakrouki
- Department of Reproductive Medicine and Fertility Preservation, Jean Verdier Hospital, AP-HP-Université Sorbonne Paris Nord, Bondy, France
| | - Alexandra Benoit
- Department of Reproductive Medicine and Fertility Preservation, Antoine Beclère Hospital, AP-HP-Université Paris-Saclay, Clamart, France
| | - Michaël Grynberg
- Department of Reproductive Medicine and Fertility Preservation, Jean Verdier Hospital, AP-HP-Université Sorbonne Paris Nord, Bondy, France; Department of Reproductive Medicine and Fertility Preservation, Antoine Beclère Hospital, AP-HP-Université Paris-Saclay, Clamart, France
| | - Charlotte Sonigo
- Department of Reproductive Medicine and Fertility Preservation, Antoine Beclère Hospital, AP-HP-Université Paris-Saclay, Clamart, France; Physiologie et Physiopathologie Endocrinienne, Inserm, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| |
Collapse
|
4
|
Holtzman S, McCarthy L, Estevez SL, Lee JA, Baird MF, Gounko D, Copperman AB, Blank SV. Walking the tightrope: Fertility preservation among hereditary breast and ovarian Cancer syndrome Previvors. Gynecol Oncol 2024; 186:176-181. [PMID: 38696905 DOI: 10.1016/j.ygyno.2024.03.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 03/21/2024] [Accepted: 03/26/2024] [Indexed: 05/04/2024]
Abstract
INTRODUCTION Fertility-related concerns cause significant anxiety among patients with Hereditary Breast and Ovarian Cancer Syndrome (HBOC). The Society of Gynecologic Oncology and the American Society for Reproductive Medicine recommend patients diagnosed with HBOC receive early referral to a reproductive endocrinologist. However, evidence about fertility trends in this patient population are limited and guidelines are scarce. The aim of this study is to compare fertility preservation among patients with HBOC to control patients undergoing fertility treatment without a diagnosis of infertility. METHODS This retrospective study included patients who presented to a single academic institution for fertility preservation in the setting of diagnosis of HBOC. In this study, HBOC patients are referred to as those who had tested positive for pathogenic mutations in BRCA1, BRCA2 or were at high-risk for HBOC based on a strong family history (defined as >3 family members diagnosed with HBOC) without a genetic mutation. HBOC patients were matched in a 1:1 fashion to a control group undergoing fertility preservation without a diagnosis of infertility or HBOC. All analysis was done using SPSS version 9.4 (SAS Institute, Cary, NC). RESULTS Between August 1st, 2016 and August 1st, 2022, 81 patients presented to the study center for consultation in the setting of HBOC. Of those who presented, 48 (59.2%) ultimately underwent oocyte cryopreservation and 33 (40.7%) underwent embryo cryopreservation. Patients who underwent oocyte cryopreservation due to BRCA1 status were more likely to present for fertility consultation at a younger age compared to control patients (32.6 vs. 34.7 years, p = 0.03) and were more likely to undergo oocyte cryopreservation at a younger age (32.1 vs. 34.6 years, p = 0.007). There was no difference in age at initial consultation or age at procedure for patients with BRCA2 or patients with a strong family history compared to control patients (p > 0.05). There was no difference in the mean age of patients with HBOC at presentation for consultation for embryo cryopreservation or the mean age the patient with HBOC underwent embryo cryopreservation compared to control patients (p > 0.05). Patients with BRCA1 or BRCA2 did not have expedited time from consultation to first cycle start (p > 0.05). After adjusting for factors including anti-Müllerian hormone (AMH) level and age, patients considered in the HBOC group due to family history had less time between consultation and oocyte cryopreservation cycle compared to control patients. (179 vs. 317 days, p = 0.045). There was no difference in time from consultation to starting cycle for embryo cryopreservation for patients with HBOC compared to controls (p > 0.05). CONCLUSION Patients with HBOC did not undergo expedited fertility treatment compared to control patients undergoing oocyte and embryo cryopreservation for non-infertility reasons. Patients diagnosed with BRCA1 had more oocytes retrieved compared to the control population which is possibly due to earlier age of presentation in the setting of recommended age of risk reducing surgery being age 35-40. When age matched, cycle outcomes did not differ between HBOC and control patients. Given the known cancer prevention benefit and recommendations for risk-reducing surgery, future studies should focus on guidelines for fertility preservation for patients with HBOC.
Collapse
Affiliation(s)
- Sharonne Holtzman
- Icahn School of Medicine at the Mount Sinai Department of Obstetrics, Gynecology, and Reproductive Science, New York, NY, USA.
| | - Lily McCarthy
- Icahn School of Medicine at the Mount Sinai Department of Obstetrics, Gynecology, and Reproductive Science, New York, NY, USA
| | - Samantha L Estevez
- Icahn School of Medicine at the Mount Sinai Department of Obstetrics, Gynecology, and Reproductive Science, New York, NY, USA; Reproductive Medicine Associates of New York, New York, NY, USA
| | - Joseph A Lee
- Reproductive Medicine Associates of New York, New York, NY, USA
| | - Morgan F Baird
- Reproductive Medicine Associates of New York, New York, NY, USA
| | - Dmitry Gounko
- Reproductive Medicine Associates of New York, New York, NY, USA
| | - Alan B Copperman
- Icahn School of Medicine at the Mount Sinai Department of Obstetrics, Gynecology, and Reproductive Science, New York, NY, USA; Reproductive Medicine Associates of New York, New York, NY, USA
| | - Stephanie V Blank
- Icahn School of Medicine at the Mount Sinai Department of Obstetrics, Gynecology, and Reproductive Science, New York, NY, USA
| |
Collapse
|
5
|
Magaton IM, Arecco L, Mariamidze E, Jankovic K, Stana M, Buzzatti G, Trevisan L, Scavone G, Ottonello S, Fregatti P, Massarotti C, von Wolff M, Lambertini M. Fertility and Pregnancy-Related Issues in Young BRCA Carriers With Breast Cancer. Breast Cancer (Auckl) 2024; 18:11782234241261429. [PMID: 38882447 PMCID: PMC11179469 DOI: 10.1177/11782234241261429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 05/06/2024] [Indexed: 06/18/2024] Open
Abstract
Approximately 10% to 15% of breast cancer cases in young women are diagnosed in patients harbouring germline (g) pathogenic or likely pathogenic variants (PVs) in the BReast CAncer 1 (BRCA1) or BReast CAncer 2 (BRCA2) genes. Preclinical and clinical studies showed a potential negative effect of germline BRCA1/2 (gBRCA1/2) PVs on ovarian reserve and reproductive potential, even before starting anticancer therapies. The aim of this article is to summarize the current literature on the fertility potential of young gBRCA1/2 PVs carriers with breast cancer and the risk of gonadotoxicity associated with anticancer treatments. Moreover, we describe the available evidence on the efficacy of fertility preservation techniques in young gBRCA1/2 PVs carriers and the safety data on having a pregnancy after breast cancer treatment.
Collapse
Affiliation(s)
- Isotta Martha Magaton
- Division of Gynaecological Endocrinology and Reproductive Medicine, University Women's Hospital, Inselspital, Bern, Switzerland
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Luca Arecco
- Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genoa, Genoa, Italy
| | - Elene Mariamidze
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Medical Oncology & Hematology, Todua Clinic, Tbilisi, Georgien
| | - Kristina Jankovic
- Department of Medical Oncology, University Clinic Center Nis, Nis, Serbia
| | - Mihaela Stana
- Department of Medical Oncology, Elysee Hospital, Alba Iulia, Romania
| | - Giulia Buzzatti
- Department of Medical Oncology, Unit of Hereditary Cancer, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Lucia Trevisan
- Department of Medical Oncology, Unit of Hereditary Cancer, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Graziana Scavone
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Silvia Ottonello
- Departent of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Piero Fregatti
- Department of Surgery, U.O.C. Clinica di Chirurgia Senologica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Surgical Sciences and Integrated Diagnostic (DISC), School of Medicine, University of Genoa, Genoa, Italy
| | - Claudia Massarotti
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics and Maternal and Child Health (DINOGMI Department), University of Genoa, Genoa, Italy
- Academic Unit of Obstetrics and Gynecology, Maternal-Child Department, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Michael von Wolff
- Division of Gynaecological Endocrinology and Reproductive Medicine, University Women's Hospital, Inselspital, Bern, Switzerland
| | - Matteo Lambertini
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genoa, Genoa, Italy
| |
Collapse
|
6
|
Stenta T, Assis M, Ayers K, Tucker EJ, Halman A, Gook D, Sinclair AH, Elliott DA, Jayasinghe Y, Conyers R. Pharmacogenomic studies of fertility outcomes in pediatric cancer survivors - A systematic review. Clin Transl Sci 2024; 17:e13827. [PMID: 38924306 PMCID: PMC11199333 DOI: 10.1111/cts.13827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 04/18/2024] [Accepted: 04/25/2024] [Indexed: 06/28/2024] Open
Abstract
For the same age, sex, and dosage, there can be significant variation in fertility outcomes in childhood cancer survivors. Genetics may explain this variation. This study aims to: (i) review the genetic contributions to infertility, (ii) search for pharmacogenomic studies looking at interactions of cancer treatment, genetic predisposition and fertility-related outcomes. Systematic searches in MEDLINE Ovid, Embase Classic+Embase, and PubMed were conducted using the following selection criteria: (i) pediatric, adolescent, and young adult cancer survivors, below 25 years old at the time of diagnosis, (ii) fertility outcome measures after cancer therapy, (iii) genetic considerations. Studies were excluded if they were (i) conducted in animal models, (ii) were not published in English, (iii) editorial letters, (iv) theses. Articles were screened in Covidence by at least two independent reviewers, followed by data extraction and a risk of bias assessment using the Quality in Prognostic Studies tool. Eight articles were reviewed with a total of 29 genes. Outcome measures included sperm concentration, azoospermia, AMH levels, assessment of premature menopause, ever being pregnant or siring a pregnancy. Three studies included replication cohorts, which attempted replication of SNP findings for NPY2R, BRSK1, FANCI, CYP2C19, CYP3A4, and CYP2B6. Six studies were rated with a high risk of bias. Differing methods may explain a lack of replication, and small cohorts may have contributed to few significant findings. Larger, prospective longitudinal studies with an unbiased genome-wide focus will be important to replicate significant results, which can be applied clinically.
Collapse
Affiliation(s)
- Tayla Stenta
- Cancer Therapies, Stem Cell MedicineMurdoch Children's Research InstituteParkvilleVictoriaAustralia
| | - Michael Assis
- Cancer Therapies, Stem Cell MedicineMurdoch Children's Research InstituteParkvilleVictoriaAustralia
- Department of Obstetrics, Gynaecology and Newborn HealthRoyal Women's Hospital, University of MelbourneParkvilleVictoriaAustralia
| | - Katie Ayers
- Department of PaediatricsUniversity of MelbourneParkvilleVictoriaAustralia
- Reproductive DevelopmentMurdoch Children's Research InstituteParkvilleVictoriaAustralia
| | - Elena J. Tucker
- Department of PaediatricsUniversity of MelbourneParkvilleVictoriaAustralia
- Reproductive DevelopmentMurdoch Children's Research InstituteParkvilleVictoriaAustralia
| | - Andreas Halman
- Cancer Therapies, Stem Cell MedicineMurdoch Children's Research InstituteParkvilleVictoriaAustralia
- Victorian Clinical Genetics ServicesMurdoch Children's Research InstituteMelbourneVictoriaAustralia
| | - Debra Gook
- Department of Obstetrics, Gynaecology and Newborn HealthRoyal Women's Hospital, University of MelbourneParkvilleVictoriaAustralia
- Gynaecology, Royal Children‘s HospitalParkvilleVictoriaAustralia
- Reproductive Services, The Royal Women's HospitalParkvilleVictoriaAustralia
| | - Andrew H. Sinclair
- Department of PaediatricsUniversity of MelbourneParkvilleVictoriaAustralia
- Reproductive DevelopmentMurdoch Children's Research InstituteParkvilleVictoriaAustralia
| | - David A. Elliott
- Cancer Therapies, Stem Cell MedicineMurdoch Children's Research InstituteParkvilleVictoriaAustralia
- Department of PaediatricsUniversity of MelbourneParkvilleVictoriaAustralia
| | - Yasmin Jayasinghe
- Department of Obstetrics, Gynaecology and Newborn HealthRoyal Women's Hospital, University of MelbourneParkvilleVictoriaAustralia
- Gynaecology, Royal Children‘s HospitalParkvilleVictoriaAustralia
| | - Rachel Conyers
- Cancer Therapies, Stem Cell MedicineMurdoch Children's Research InstituteParkvilleVictoriaAustralia
- Department of PaediatricsUniversity of MelbourneParkvilleVictoriaAustralia
- Children's Cancer Centre, The Royal Children's HospitalParkvilleVictoriaAustralia
| |
Collapse
|
7
|
Silvestris E, Cormio G, Loizzi V, Corrado G, Arezzo F, Petracca EA. Fertility Preservation in BRCA1/2 Germline Mutation Carriers: An Overview. Life (Basel) 2024; 14:615. [PMID: 38792636 PMCID: PMC11122448 DOI: 10.3390/life14050615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 04/28/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
BRCA1 and BRCA2 mutations are responsible for a higher incidence of breast and ovarian cancer (from 55% up to 70% vs. 12% in the general population). If their functions have been widely investigated in the onset of these malignancies, still little is known about their role in fertility impairment. Cancer patients treated with antineoplastic drugs can be susceptible to their gonadotoxicity and, in women, some of them can induce apoptotic program in premature ovarian follicles, progressive depletion of ovarian reserve and, consequently, cancer treatment-related infertility (CTRI). BRCA variants seem to be associated with early infertility, thus accelerating treatment impairment of ovaries and making women face the concrete possibility of an early pregnancy. In this regard, fertility preservation (FP) procedures should be discussed in oncofertility counseling-from the first line of prevention with risk-reducing salpingo-oophorectomy (RRSO) to the new experimental ovarian stem cells (OSCs) model as a new way to obtain in vitro-differentiated oocytes, several techniques may represent a valid option to BRCA-mutated patients. In this review, we revisit knowledge about BRCA involvement in lower fertility, pregnancy feasibility, and the fertility preservation (FP) options available.
Collapse
Affiliation(s)
- Erica Silvestris
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (G.C.); (V.L.); (F.A.); (E.A.P.)
| | - Gennaro Cormio
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (G.C.); (V.L.); (F.A.); (E.A.P.)
- Department of Interdisciplinary Medicine (DIM), University of Bari “Aldo Moro”, 70121 Bari, Italy
| | - Vera Loizzi
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (G.C.); (V.L.); (F.A.); (E.A.P.)
- Department of Interdisciplinary Medicine (DIM), University of Bari “Aldo Moro”, 70121 Bari, Italy
| | - Giacomo Corrado
- Department of Woman, Child Health and Public Health, Gynecologic Oncology Unit, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00136 Rome, Italy;
| | - Francesca Arezzo
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (G.C.); (V.L.); (F.A.); (E.A.P.)
| | - Easter Anna Petracca
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (G.C.); (V.L.); (F.A.); (E.A.P.)
| |
Collapse
|
8
|
Hong YH, Park C, Paik H, Lee KH, Lee JR, Han W, Park S, Chung S, Kim HJ. Fertility Preservation in Young Women With Breast Cancer: A Review. J Breast Cancer 2023; 26:221-242. [PMID: 37387349 DOI: 10.4048/jbc.2023.26.e28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/11/2023] [Accepted: 05/12/2023] [Indexed: 07/01/2023] Open
Abstract
Fertility preservation is a major concern in young patients diagnosed with breast cancer and planning to receive multimodality treatment, including gonadotoxic chemotherapy with or without age-related decline through long-term endocrine therapy. Most breast cancer patients undergo multimodality treatments; many short-term and long-term side effects arise during these therapies. One of the most detrimental side effects is reduced fertility due to gonadotoxic treatments with resultant psychosocial stress. Cryopreservation of oocytes, embryos, and ovarian tissue are currently available fertility preservation methods for these patients. As an adjunct to these methods, in vitro maturation or gonadotropin-releasing hormone agonist could also be considered. It is also essential to communicate well with patients in the decision-making process on fertility preservation. It is essential to refer patients diagnosed with breast cancer on time to fertility specialists for individualized treatment, which may lead to desirable outcomes. To do so, a multimodal team-based approach and in-depth discussion on the treatment of breast cancer and fertility preservation is crucial. This review aims to summarize infertility risk related to currently available breast cancer treatment, options for fertility preservation and its details, barriers to oncofertility counseling, and psychosocial issues.
Collapse
Affiliation(s)
- Yeon Hee Hong
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Changhee Park
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Haerin Paik
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Kyung-Hun Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Cancer Research Institute, Seoul National University, Seoul, Korea
| | - Jung Ryeol Lee
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Wonshik Han
- Cancer Research Institute, Seoul National University, Seoul, Korea
- Department of Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Seho Park
- Division of Breast Surgery, Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Seockhoon Chung
- Department of Psychiatry, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| | - Hee Jeong Kim
- Division of Breast Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| |
Collapse
|
9
|
Lewinsohn R, Zheng Y, Rosenberg SM, Ruddy KJ, Tamimi RM, Schapira L, Peppercorn J, Borges VF, Come S, Snow C, Ginsburg ES, Partridge AH. Fertility Preferences and Practices Among Young Women With Breast Cancer: Germline Genetic Carriers Versus Noncarriers. Clin Breast Cancer 2023; 23:317-323. [PMID: 36628811 DOI: 10.1016/j.clbc.2022.12.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Young women with breast cancer who carry germline genetic pathogenic variants may face distinct fertility concerns, yet limited data exist comparing fertility preferences and practices between carriers and noncarriers. PATIENTS AND METHODS Participants in the Young Women's Breast Cancer Study (NCT01468246), a prospective cohort of women diagnosed with breast cancer at ≤40 years, who completed a modified Fertility Issues Survey were included in this analysis. RESULTS Of 1052 eligible participants, 118 (11%) tested positive for a pathogenic variant. Similar proportions (P = .23) of carriers (46%, [54/118]) and noncarriers (37%, [346/934]) desired more biologic children prediagnosis, and desire decreased similarly postdiagnosis (carriers, 30% [35/118] vs. noncarriers, 26% [244/934], P = .35). Among those desiring children postdiagnosis (n = 279), concern about cancer risk heritability was more common among carriers (74% [26/35] vs. noncarriers, 36% [88/244], P < .01). Carriers were more likely to report that concern about cancer risk heritability contributed to a lack of certainty or interest in future pregnancies (20% [16/81] vs. noncarriers, 7% [49/674], P = .001). Similar proportions (P = .65) of carriers (36% [43/118]) and noncarriers (38% [351/934]) were somewhat or very concerned about infertility post-treatment; utilization of fertility preservation strategies was also similar (carriers, 14% [17/118] vs. noncarriers, 12% [113/934], P = .78). CONCLUSION Carriers were similarly concerned about future fertility and as likely to pursue fertility preservation as noncarriers. Concern about cancer risk heritability was more frequent among carriers and impacted decisions not to pursue future pregnancies for some, underscoring the importance of counseling regarding strategies to prevent transmission to offspring, including preimplantation genetic testing.
Collapse
Affiliation(s)
- Rebecca Lewinsohn
- Harvard Medical School, Boston, MA; Medical Oncology, Dana-Farber Cancer Institute, Boston, MA; Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
| | - Yue Zheng
- Data Sciences, Dana-Farber Cancer Institute, Boston, MA
| | | | | | | | | | - Jeffrey Peppercorn
- Harvard Medical School, Boston, MA; Massachusetts General Hospital, Boston, MA
| | | | - Steven Come
- Harvard Medical School, Boston, MA; Beth Israel Deaconess Medical Center, Boston, MA
| | - Craig Snow
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA; Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
| | | | - Ann H Partridge
- Harvard Medical School, Boston, MA; Medical Oncology, Dana-Farber Cancer Institute, Boston, MA; Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA.
| |
Collapse
|
10
|
Prokurotaite E, Condorelli M, Dechene J, Bouziotis J, Lambertini M, Demeestere I. Impact of Breast Cancer and Germline BRCA Pathogenic Variants on Fertility Preservation in Young Women. Life (Basel) 2023; 13:930. [PMID: 37109459 PMCID: PMC10146760 DOI: 10.3390/life13040930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/24/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
BACKGROUND Several studies have suggested that breast cancer (BC) and germline BRCA pathogenic variants (gBRCA PVs) could have a deleterious impact on ovarian reserve. Nevertheless, data are limited and mixed. Our objective was to evaluate the performance of fertility preservation (FP) in terms of the number of collected mature oocytes after ovarian stimulation (OS) in young women carrying a gBRCA PV, associated or not with BC. METHODS We conducted a retrospective monocentric study at HUB-Hôpital Erasme in Brussels. All women aged between 18 and 41 years diagnosed with invasive non-metastatic BC and/or gBRCA PV carriers who underwent OS for FP or preimplantation genetic testing for monogenic disorder (PGT-M) between November 2012 and October 2021 were included. Three groups were compared: BC patients without a gBRCA PV, BC patients with a gBRCA PV, and healthy gBRCA PV carriers. Ovarian reserve was evaluated based on the efficacy of OS and AMH levels. RESULTS A total of 85 patients underwent 100 cycles. The mean age (32.2 ± 3.9 years; p = 0.61) and median AMH level (1.9 [0.2-13] μg/L; p = 0.22) were similar between groups. Correlations between the number of mature oocytes and AMH level (p < 0.001) and between AMH and age (p < 0.001) were observed. No differences in the number of retrieved mature oocytes were observed between groups (p = 0.41), or for other OS parameters. CONCLUSION Neither BC nor a gBRCA PV significantly affects ovarian reserve and FP efficacy in terms of the number of mature oocytes retrieved.
Collapse
Affiliation(s)
- Elze Prokurotaite
- Fertility Clinic, Department of Obstetrics and Gynecology, H.U.B—Hôpital Erasme, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Margherita Condorelli
- Fertility Clinic, Department of Obstetrics and Gynecology, H.U.B—Hôpital Erasme, Université Libre de Bruxelles, 1070 Brussels, Belgium
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Julie Dechene
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Jason Bouziotis
- Department of Biomedical Research, H.U.B—Hôpital Erasme, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Matteo Lambertini
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, 16132 Genova, Italy
- Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Isabelle Demeestere
- Fertility Clinic, Department of Obstetrics and Gynecology, H.U.B—Hôpital Erasme, Université Libre de Bruxelles, 1070 Brussels, Belgium
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles, 1070 Brussels, Belgium
| |
Collapse
|
11
|
Razeti MG, Soldato D, Arecco L, Levaggi A, Puglisi S, Solinas C, Agostinetto E, Spinaci S, Lapuchesky L, Genova C, Massarotti C, Lambertini M. Approaches to Fertility Preservation for Young Women With Breast Cancer. Clin Breast Cancer 2023; 23:241-248. [PMID: 36710145 DOI: 10.1016/j.clbc.2023.01.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 12/23/2022] [Accepted: 01/10/2023] [Indexed: 01/15/2023]
Abstract
In patients with early breast cancer, the combination of different systemic treatment strategies, including chemotherapy, endocrine therapy, targeted therapy, and more recently also immunotherapy has demonstrated to significantly improve their survival outcomes. However, this gain is often obtained at the cost of higher toxicity calling for the need of increased attention toward survivorship-related issues, including fertility preservation in young women. According to available guidelines, health care providers should offer oncofertility counseling to all patients with cancer diagnosed at reproductive age. Counselling should focus on the risk of gonadotoxicity of anticancer treatments and on the access to fertility preservation techniques. However, several surveys have demonstrated suboptimal implementation of these recommendations. This review aims at summarizing the available evidence on oncofertility to guide health care providers involved in the management of young women with breast cancer. Available and effective options for fertility preservation include oocyte/embryo cryopreservation or ovarian tissue cryopreservation. Patient, disease, and treatment characteristics should be carefully considered when offering these strategies. Ovarian function preservation with gonadotrophin-releasing hormone agonists during chemotherapy should be discussed and offered to every premenopausal woman concerned about developing premature ovarian insufficiency and independently of her wish to preserve fertility. Current available data confirm that pregnancy occurring after proper treatment for breast cancer is safe, both in terms of long-term clinical outcomes and for the babies. Fertility preservation and pregnancy desire should be pivotal components of the multimodal management of breast cancer in young women, and require a multidisciplinary approach based on close collaborations between oncologists and fertility specialists.
Collapse
Affiliation(s)
- Maria G Razeti
- Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - Davide Soldato
- Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - Luca Arecco
- Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - Alessia Levaggi
- Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Silvia Puglisi
- Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - Cinzia Solinas
- Medical Oncology, AOU Cagliari, Policlinico Duilio Casula, Monserrato, Cagliari, Italy
| | - Elisa Agostinetto
- Academic Trials Promoting Team, Institut Jules Bordet, Université Libre de Bruxelles (U.L.B.), Brussels, Belgium
| | | | - Laura Lapuchesky
- Medical Oncology, Instituto Alexander Fleming, Ciudad Autónoma De Buenos Aires, Argentina
| | - Carlo Genova
- Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - Claudia Massarotti
- Physiopatology of Human Reproduction Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics and Maternal-Child Health (DiNOGMI Department), School of Medicine, University of Genova, Genova, Italy
| | - Matteo Lambertini
- Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy.
| |
Collapse
|
12
|
Baltacı E, Kazancı F, Şahin Fİ. BRCA, infertility, and fertility preservation: a review for counseling. J Assist Reprod Genet 2023; 40:465-472. [PMID: 36695945 PMCID: PMC10033813 DOI: 10.1007/s10815-023-02725-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 01/10/2023] [Indexed: 01/26/2023] Open
Abstract
BRCA mutations as a triggering factor in breast cancer have been reported to result in fertility problems and oocyte aging in young patients with cancer diagnosis. These patients are concerned about fertility problems and family planning before undergoing treatment modalities that may result in infertility. In this review, we conducted analysis of the literature on the association between BRCA mutations and infertility, possible fertility preservation options, and their safety and tried to gather results from different disciplines and points of view on the matter. Our aim is to provide a general summary of recent studies to provide further insight on the matter for counseling BRCA mutation carriers on fertility preservation methods and their implications.
Collapse
Affiliation(s)
- Ege Baltacı
- Department of Medical Genetics, Başkent University Hospital, Ankara, Turkey
| | - Ferah Kazancı
- Department of Gynecology and Obstetrics, Nezip Fazıl City Hospital, Kahramanmaraş, Turkey
| | - Feride İffet Şahin
- Department of Medical Genetics, Başkent University Hospital, Ankara, Turkey
| |
Collapse
|
13
|
Sonigo C, Amsellem N, Mayeur A, Laup L, Pistilli B, Delaloge S, Eustache F, Sifer C, Rakrouki S, Benoit A, Peigné M, Grynberg M. Disease-free survival does not differ according to fertility preservation technique for young women with breast cancer. Fertil Steril 2023; 119:465-473. [PMID: 36473609 DOI: 10.1016/j.fertnstert.2022.11.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 11/24/2022] [Accepted: 11/29/2022] [Indexed: 12/07/2022]
Abstract
OBJECTIVE To study whether fertility preservation strategies using ovarian stimulation or without using it impact long-term disease-free survival of patients with breast cancer. DESIGN Retrospective bicentric cohort study. SETTING Two university hospitals. PATIENT(S) In this study, 740 women with breast cancer, aged 18-43 years, who received primary fertility preservation between 2013 and 2019 after a diagnosis of localized breast cancer were included. INTERVENTION(S) Overall, 328 patients underwent at least 1 ovarian stimulation cycle (STIM group) and 412 had a technique without hormonal administration (no STIM group). MAIN OUTCOME MEASURE(S) Disease-free survival and overall survival up to May 2021 were compared between the 2 groups by log-rank test. Cox proportional-hazard regression model was used for multivariable analyses. RESULT(S) Out of the 740 women who underwent fertility preservation, follow-up data were available for 269 women in the STIM group (82%) and 330 (80%) in the no STIM group. Kaplan-Meier estimates of disease-free survival at 4 years were 87.9% (82.8%-92.2%) and 83.1% (78.4%-87.3%) in the STIM and no STIM groups, respectively. After adjustment on prognostic parameters, no significant difference in breast cancer recurrence rate was observed between the STIM and no STIM groups (hazard ratios, 0.83 [0.64-1.08]). Kaplan-Meier estimate of overall survival at 4 years was 97.6% (95.3%-99.2%) and 93.6% (90.9%-95.9%) in the STIM and no STIM groups, respectively. Overall survival was higher in the STIM group than no STIM group (log-rank test). After adjustment on prognostic parameters, the risk of death remained significantly lower in the STIM group (Hazard Ratio, 0.55 [0.35-0.85]). CONCLUSION(S) In our cohort, STIM for fertility preservation in breast cancer did not significantly impact disease-free survival but was associated with higher overall survival. The disease-free survival and overall survival of young patients with breast cancer were not impacted by fertility preservation techniques irrespective of the timing of chemotherapy (neoadjuvant or adjuvant) and the use of ovarian stimulation. Nevertheless, because death and recurrence were rare events, these results should be taken with caution.
Collapse
Affiliation(s)
- Charlotte Sonigo
- Department of Reproductive Medicine and Fertility Preservation, Universite Paris-Saclay, Assistance Publique Hopitaux de Paris, Antoine Beclere Hospital, Clamart, France; Universite Paris-Saclay, Inserm, Physiologie et physiopathologie endocrinienne, Le Kremlin-Bicetre, France.
| | - Noémi Amsellem
- Department of Reproductive Medicine and Fertility Preservation, Universite Paris-Saclay, Assistance Publique Hopitaux de Paris, Antoine Beclere Hospital, Clamart, France
| | - Anne Mayeur
- Histology-Embryology-Cytogenetic Laboratory, Universite Paris-Saclay, Assistance Publique Hopitaux de Paris, Antoine Beclere Hospital, Clamart, France
| | - Laetitia Laup
- Department of Reproductive Medicine and Fertility Preservation, Universite Sorbonne Paris Nord, Assistance Publique - Hopitaux de Paris, Jean Verdier Hospital, Bondy, France
| | - Barbara Pistilli
- Department of Medical Oncology, Institut Gustave Roussy, Villejuif, France
| | - Suzette Delaloge
- Department of Medical Oncology, Institut Gustave Roussy, Villejuif, France
| | - Florence Eustache
- Department of Biology of Reproduction and CECOS, Universite Sorbonne Paris Nord, Assistance Publique - Hopitaux de Paris, Jean Verdier Hospital, Bondy, France
| | - Christophe Sifer
- Department of Biology of Reproduction and CECOS, Universite Sorbonne Paris Nord, Assistance Publique - Hopitaux de Paris, Jean Verdier Hospital, Bondy, France
| | - Sophia Rakrouki
- Department of Reproductive Medicine and Fertility Preservation, Universite Sorbonne Paris Nord, Assistance Publique - Hopitaux de Paris, Jean Verdier Hospital, Bondy, France
| | - Alexandra Benoit
- Department of Reproductive Medicine and Fertility Preservation, Universite Paris-Saclay, Assistance Publique Hopitaux de Paris, Antoine Beclere Hospital, Clamart, France
| | - Maeliss Peigné
- Department of Reproductive Medicine and Fertility Preservation, Universite Sorbonne Paris Nord, Assistance Publique - Hopitaux de Paris, Jean Verdier Hospital, Bondy, France
| | - Michael Grynberg
- Department of Reproductive Medicine and Fertility Preservation, Universite Paris-Saclay, Assistance Publique Hopitaux de Paris, Antoine Beclere Hospital, Clamart, France; Department of Reproductive Medicine and Fertility Preservation, Universite Sorbonne Paris Nord, Assistance Publique - Hopitaux de Paris, Jean Verdier Hospital, Bondy, France
| |
Collapse
|
14
|
El Moujahed L, Philis R, Grynberg M, Laot L, Mur P, Amsellem N, Mayeur A, Benoit A, Rakrouki S, Sifer C, Peigné M, Sonigo C. Response to Ovarian Stimulation for Urgent Fertility Preservation before Gonadotoxic Treatment in BRCA-Pathogenic-Variant-Positive Breast Cancer Patients. Cancers (Basel) 2023; 15:cancers15030895. [PMID: 36765851 PMCID: PMC9913552 DOI: 10.3390/cancers15030895] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/19/2023] [Accepted: 01/24/2023] [Indexed: 02/05/2023] Open
Abstract
BRCA 1/2 pathogenic variants increase the risk of developing early and aggressive breast cancers (BC). For these patients, fertility potential can be directly affected by oncologic treatments. In addition, evidence indicates that BRCA-mutated women had a significant reduction in their ovarian reserve. In order to improve their chances of conception after the completion of cancer treatments, fertility preservation should be proposed before the administration of gonadotoxic drugs, ideally by oocyte vitrification after controlled ovarian hyperstimulation (COH). The present investigation aims to assess the ovarian response to COH in BRCA 1/2-pathogenic-variant carriers diagnosed with BC. Patient characteristics and COH outcomes were compared between BRCA-positive (n = 54) and BRCA-negative (n = 254) patients. The number of oocytes recovered did not differ between the two groups. However, the oocyte maturation rate and the number of mature oocytes obtained (7 (4.5-11.5) vs. 9 (5-14) oocytes, p = 0.05) were significantly lower in the BRCA-mutated patients. Although individualized COH protocols should be discussed, BRCA-mutated patients would benefit from FP before BC occurs, in order to cope with the potential accelerated decline of their ovarian reserve, optimize the success rate of FP by repeating COH cycles, and to preserve the feasibility of PGT-M by collecting a large amount of eggs.
Collapse
Affiliation(s)
- Lina El Moujahed
- Department of Reproductive Medicine and Fertility Preservation, Université Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Antoine Beclere Hospital, 92140 Clamart, France
| | - Robin Philis
- Department of Reproductive Medicine and Fertility Preservation, Université Sorbonne Paris Nord, Assistance Publique-Hôpitaux de Paris, Jean Verdier Hospital, 93143 Bondy, France
| | - Michael Grynberg
- Department of Reproductive Medicine and Fertility Preservation, Université Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Antoine Beclere Hospital, 92140 Clamart, France
- Department of Reproductive Medicine and Fertility Preservation, Université Sorbonne Paris Nord, Assistance Publique-Hôpitaux de Paris, Jean Verdier Hospital, 93143 Bondy, France
| | - Lucie Laot
- Department of Reproductive Medicine and Fertility Preservation, Université Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Antoine Beclere Hospital, 92140 Clamart, France
| | - Pauline Mur
- Department of Reproductive Medicine and Fertility Preservation, Université Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Antoine Beclere Hospital, 92140 Clamart, France
| | - Noemi Amsellem
- Department of Reproductive Medicine and Fertility Preservation, Université Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Antoine Beclere Hospital, 92140 Clamart, France
| | - Anne Mayeur
- Service de Biologie de la Reproduction—CECOS, Université Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Antoine Beclere Hospital, 92140 Clamart, France
| | - Alexandra Benoit
- Department of Reproductive Medicine and Fertility Preservation, Université Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Antoine Beclere Hospital, 92140 Clamart, France
| | - Sophia Rakrouki
- Department of Reproductive Medicine and Fertility Preservation, Université Sorbonne Paris Nord, Assistance Publique-Hôpitaux de Paris, Jean Verdier Hospital, 93143 Bondy, France
| | - Christophe Sifer
- Department of Biology of Reproduction and CECOS, Université Sorbonne Paris Nord, Assistance Publique-Hôpitaux de Paris, Jean Verdier Hospital, 93143 Bondy, France
| | - Maeliss Peigné
- Department of Reproductive Medicine and Fertility Preservation, Université Sorbonne Paris Nord, Assistance Publique-Hôpitaux de Paris, Jean Verdier Hospital, 93143 Bondy, France
| | - Charlotte Sonigo
- Department of Reproductive Medicine and Fertility Preservation, Université Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Antoine Beclere Hospital, 92140 Clamart, France
- Inserm, Physiologie et Physiopathologie Endocrinienne, Université Paris-Saclay, 94276 Le Kremlin-Bicêtre, France
- Department of Reproductive Medicine, Hôpital Antoine Béclère, 157 Avenue de la Porte Trivaux, 92140 Clamart, France
- Correspondence: ; Tel.: +33-1-45-374-053; Fax: +33-8-97-500-086
| |
Collapse
|
15
|
Sparidaens EM, Beerendonk CCM, Fleischer K, Nelen WLDM, Braat DDM, Hermens RPMG. Exploration of fertility and early menopause related information needs and development of online information for young breast cancer survivors. BMC Womens Health 2022; 22:329. [PMID: 35922784 PMCID: PMC9351061 DOI: 10.1186/s12905-022-01901-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 07/24/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Approximately half of premenopausal women diagnosed with breast cancer desire to conceive after they finish treatment. Counseling about the risk of infertility prior to cancer treatment has been proven to improve quality of life after cancer treatment. As a result of this, guidelines focus on informing women on this topic prior to treatment. However, it is equally important to provide fertility related information after primary treatment has been completed, when the wish to conceive might become actual. Therefore, the aim of this study was to identify the fertility and early menopause related information needs of young breast cancer survivors and to design, develop and implement online information material with input of stakeholders. METHODS A phenomenological qualitative study consisting of four phases was performed: identification of information needs through semi-structured interviews from a professional perspective (1) and a patient perspective (2). Exploration of stakeholders perspective regarding development and implementation of online information material (3) and development and implementation of the information material (4). RESULTS Professionals indicated that there are no guidelines regarding the provision of fertility related information during cancer survivorship. Survivors reported unmet information needs. Women identified the following as most important information needs (a) fertility preservation options, (b) the risk of menopause or infertility, and (c) long term consequences of early menopause. A wide range of stakeholders involved in breast cancer care were interviewed. Based on their proposed design the information material was implemented on a nationwide website aiming at informing and supporting breast cancer patients. CONCLUSIONS Fertility and early menopause related information needs of young breast cancer survivors and their professionals were identified. Information material has been designed, developed and nationally implemented. This way, professionals in breast cancer care are provided with an information tool that helps them meet the information needs and preferences of their patients.
Collapse
Affiliation(s)
- Ellen Marie Sparidaens
- Department of Obstetrics and Gynaecology, Radboud University Medical Center, P.O. Box 9101, 6500HB, Nijmegen, The Netherlands.
| | - Catharina C M Beerendonk
- Department of Obstetrics and Gynaecology, Radboud University Medical Center, P.O. Box 9101, 6500HB, Nijmegen, The Netherlands
| | - Kathrin Fleischer
- Department of Obstetrics and Gynaecology, Radboud University Medical Center, P.O. Box 9101, 6500HB, Nijmegen, The Netherlands
| | - Willianne L D M Nelen
- Department of Obstetrics and Gynaecology, Radboud University Medical Center, P.O. Box 9101, 6500HB, Nijmegen, The Netherlands
| | - Didi D M Braat
- Department of Obstetrics and Gynaecology, Radboud University Medical Center, P.O. Box 9101, 6500HB, Nijmegen, The Netherlands
| | - Rosella P M G Hermens
- Scientific Institute for Quality of Healthcare (IQ Healthcare), Radboud University Medical Center, P.O. Box 9101, 6500HB, Nijmegen, The Netherlands
| |
Collapse
|
16
|
Kufel-Grabowska J, Podolak A, Maliszewski D, Bartoszkiewicz M, Ramlau R, Lukaszuk K. Fertility Counseling in BRCA1/2-Mutated Women with Breast Cancer and Healthy Individuals. J Clin Med 2022; 11:jcm11143996. [PMID: 35887761 PMCID: PMC9321124 DOI: 10.3390/jcm11143996] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 06/27/2022] [Accepted: 07/03/2022] [Indexed: 11/16/2022] Open
Abstract
Breast cancer is the most commonly diagnosed cancer worldwide and the fifth leading cause of cancer death. In 2020, there were 2.3 million new cases, and 685,000 women died from it. Breast cancer among young women under 40 years of age accounts for 5% to 10% of all cases of this cancer. The greater availability of multi-gene sequence analysis by next-generation sequencing has improved diagnosis and, consequently, the possibility of using appropriate therapeutic approaches in BRCA1/2 gene mutation carriers. Treatment of young breast cancer patients affects their reproductive potential by reducing ovarian reserve. It can lead to reversible or permanent premature menopause, decreased libido, and other symptoms of sex hormone deficiency. This requires that, in addition to oncological treatment, patients are offered genetic counseling, oncofertility, psychological assistance, and sexological counseling. Given the number of BRCA1/2 gene mutation carriers among young breast cancer patients, but also thanks to growing public awareness, among their healthy family members planning offspring, the possibility of benefiting from preimplantation testing and performing cancer-risk-reduction procedures: RRM (risk-reducing mastectomy) and RRSO (risk-reducing salpingo-oophorectomy) significantly increase the chance of a genetically burdened person living a healthy life and giving birth to a child not burdened by the parent's germline mutation. The goal of this paper is to show methods and examples of fertility counselling for BRCA1/2 gene mutation carriers, including both patients already affected by cancer and healthy individuals.
Collapse
Affiliation(s)
- Joanna Kufel-Grabowska
- Department of Oncology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (J.K.-G.); (R.R.)
| | - Amira Podolak
- Department of Obstetrics and Gynecological Nursing, Faculty of Health Sciences, Medical University of Gdansk, 80-210 Gdansk, Poland; (A.P.); (K.L.)
| | - Daniel Maliszewski
- Department of General and Oncological Surgery, Wojewódzki Szpital Specjalistyczny im. Janusza Korczaka w Słupsku Sp. z o.o., 76-200 Słupsk, Poland;
- Department of General and Oncological Surgery at Specialist Hospital in Koscierzyn, Sp.z.o.o., 83-400 Kościerzyna, Poland
- Swissmed Health Center, 80-210 Gdansk, Poland
| | - Mikołaj Bartoszkiewicz
- Department of Immunobiology, Poznan University of Medical Sciences, 60-806 Poznan, Poland
- Correspondence: ; Tel.: +48-61-854-76-53
| | - Rodryg Ramlau
- Department of Oncology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (J.K.-G.); (R.R.)
| | - Krzysztof Lukaszuk
- Department of Obstetrics and Gynecological Nursing, Faculty of Health Sciences, Medical University of Gdansk, 80-210 Gdansk, Poland; (A.P.); (K.L.)
- Invicta Research and Development Center, 81-740 Sopot, Poland
| |
Collapse
|
17
|
Kaidar-Person O, Yoeli-Ullman R, Pillar N, Paluch-Shimon S, Poortmans P, Lawrence YR. Obstetric complications at time of delivery amongst breast cancer survivors: A population-based cohort study. Breast 2022; 62:170-178. [PMID: 35219114 PMCID: PMC8873951 DOI: 10.1016/j.breast.2022.02.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 02/14/2022] [Accepted: 02/14/2022] [Indexed: 12/14/2022] Open
Abstract
PURPOSE Our aim was to determine whether breast cancer survivors are at increased risk of obstetric and maternal complications at time of delivery. METHODS The USA 'National Inpatient Sample' database was queried for hospitalizations associated with deliveries, between 2015 and 2018. The incidence of maternal and fetal complications was compared between women with, and without, a personal history of breast cancer. RESULTS Of the 2,103,216 birth related admissions, 617 (0.03%) of the women were breast cancer survivors, with the proportion increasing over time (from 0.02% in 2015 to 0.04% in 2018). Breast cancer survivors had a higher socioeconomic status (p < 0.001) and were significantly older compared to other mothers (34 vs. 28 years, p < 0.001). Additionally, they were more likely to suffer from preexisting chronic diseases including cardiopulmonary disease and diabetes mellitus, and had a higher incidence of multiple gestation (4.4% vs. 1.6%) [OR 2.7, 95% CI 1.9-4.0, p < 0.001]. The incidence of acute adverse events at time of delivery including fetal distress, preterm labor, cesarean section and maternal infection was higher amongst the breast cancer survivors. On multivariate analysis age, ethnic group, comorbidities, multiple gestations, and a previous breast cancer diagnosis, but not cancer treatment, were associated with an increased risk of an obstetric adverse event. CONCLUSION Breast cancer survivors have more comorbidities and are at increased risk of acute obstetrical complications at time of delivery. Further studies are required to validate these findings, and evaluate the ability of interventions to improve obstetrical outcomes amongst breast cancer survivors.
Collapse
Affiliation(s)
- Orit Kaidar-Person
- Department of Radiation Oncology, Sheba Medical Center, Tel HaShomer, 5265601, Israel; Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| | - Rakefet Yoeli-Ullman
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Department of Obstetrics and Gynecology, Sheba Medical Center, Tel-Hashomer, Israel
| | - Nir Pillar
- Department of Pathology, Hadassah-Hebrew-University-Medical-Center, Jerusalem, 91120, Israel
| | - Shani Paluch-Shimon
- Sharett Institute of Oncology, Hadassah University Hospital & Faculty of Medicine Hebrew University, Jerusalem, Israel
| | - Philip Poortmans
- Department of Radiation Oncology, Iridium Netwerk, Antwerp, 2610, Belgium; University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Yaacov R Lawrence
- Department of Radiation Oncology, Sheba Medical Center, Tel HaShomer, 5265601, Israel; Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Department Radiation Oncology, Sidney Kimmel Medical College at Thomas Jefferson University, USA
| |
Collapse
|
18
|
Kim SW, Kim TH, Han JY, Kim SK, Lee JR, Jee UC, Suh CS, Kim SH. Impact of BRCA mutations and hormone receptor status on reproductive potential in breast cancer patients undergoing fertility preservation. Gynecol Endocrinol 2022; 38:227-230. [PMID: 34775902 DOI: 10.1080/09513590.2021.2002294] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
OBJECTIVE To identify whether the BRCA mutations and hormone receptor status affect the ovarian reserve and ovarian stimulation outcomes in breast cancer patients undergoing fertility preservation. METHODS A total of 117 women diagnosed with breast cancer who were referred to the fertility preservation clinics at Seoul National University Bundang Hospital and Seoul National University Hospital between September 2012 and July 2019 undergone ovarian stimulation for oocyte retrieval. Basal characteristics including age, BMI, basal AMH, basal FSH, and fertility preservation outcomes such as the number of retrieved oocytes and mature oocytes were compared retrospectively. RESULTS BRCA1 mutation was noted in 25 women, and BRCA2 mutation was observed in 35 women. Positive estrogen receptor status was noted in 87 women, and positive progesterone receptor status was noted in 69 women. HER2 was positive in 55 women, and 19 women were diagnosed with triple-negative breast cancers. The number of total oocytes retrieved was lower in patients with BRCA mutation (8.3 ± 5.4 vs. 15.3 ± 8.7, p = .002). The number of mature oocytes retrieved was also lower in BRCA carriers (4.7 ± 4.2 vs. 8.7 ± 7.9, p = .025). Triple-negative breast cancer (TNBC) patients were younger than non-TNBC patients (30.3 ± 4.8 vs. 33.9 ± 5.0, p = .007). The rate of mature oocyte rate was higher in TNBC patients (68.6%±20.6 vs. 52.5%±29.7, p = .037). CONCLUSIONS Our study demonstrated that BRCA carriers with breast cancer had comparable ovarian reserve to non-carriers but the response to ovarian stimulation was lower. We also observed that oocyte maturity was higher in TNBC patients, however age might be a confounding factor of this result.
Collapse
Affiliation(s)
- Sung Woo Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine,, Seoul, Korea
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Tae Hee Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine,, Seoul, Korea
| | - Ji Yeon Han
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine,, Seoul, Korea
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Seul Ki Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine,, Seoul, Korea
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Gyeonggi-do, Republic of Korea
| | - Jung Ryeol Lee
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine,, Seoul, Korea
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Gyeonggi-do, Republic of Korea
| | - Ung Chul Jee
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine,, Seoul, Korea
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Gyeonggi-do, Republic of Korea
| | - Chang Suk Suh
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine,, Seoul, Korea
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Seok Hyun Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine,, Seoul, Korea
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, Republic of Korea
| |
Collapse
|
19
|
Salama M, Lambertini M, Christianson MS, Jayasinghe Y, Anazodo A, De Vos M, Amant F, Stern C, Appiah L, Woodard TL, Anderson RA, Westphal LM, Leach RE, Rodriguez-Wallberg KA, Patrizio P, Woodruff TK. Installing oncofertility programs for breast cancer in limited versus optimum resource settings: Empirical data from 39 surveyed centers in Repro-Can-OPEN Study Part I & II. J Assist Reprod Genet 2022; 39:505-516. [PMID: 35032286 PMCID: PMC8760079 DOI: 10.1007/s10815-022-02394-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 01/05/2022] [Indexed: 01/09/2023] Open
Abstract
PURPOSE As a further step to elucidate the actual diverse spectrum of oncofertility practices for breast cancer around the globe, we present and discuss the comparisons of oncofertility practices for breast cancer in limited versus optimum resource settings based on data collected in the Repro-Can-OPEN Study Part I & II. METHODS We surveyed 39 oncofertility centers including 14 in limited resource settings from Africa, Asia & Latin America (Repro-Can-OPEN Study Part I), and 25 in optimum resource settings from the United States, Europe, Australia and Japan (Repro-Can-OPEN Study Part II). Survey questions covered the availability of fertility preservation and restoration options offered to young female patients with breast cancer as well as the degree of utilization. RESULTS In the Repro-Can-OPEN Study Part I & II, responses for breast cancer and calculated oncofertility scores showed the following characteristics: (1) higher oncofertility scores in optimum resource settings than in limited resource settings especially for established options, (2) frequent utilization of egg freezing, embryo freezing, ovarian tissue freezing, GnRH analogs, and fractionation of chemo- and radiotherapy, (3) promising utilization of oocyte in vitro maturation (IVM), (4) rare utilization of neoadjuvant cytoprotective pharmacotherapy, artificial ovary, and stem cells reproductive technology as they are still in preclinical or early clinical research settings, (5) recognition that technical and ethical concerns should be considered when offering advanced and innovative oncofertility options. CONCLUSIONS We presented a plausible oncofertility best practice model to guide oncofertility teams in optimizing care for breast cancer patients in various resource settings.
Collapse
Affiliation(s)
- Mahmoud Salama
- Oncofertility Consortium, Michigan State University, 965 Wilson Road, Room A626B, East Lansing, MI 48824-1316 USA
| | - M. Lambertini
- Department of Medical Oncology, UOC Clinica Di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - MS Christianson
- Division of Reproductive Endocrinology and Infertility, Johns Hopkins Fertility Center, Johns Hopkins University School of Medicine, 10751 Falls Road, Suite 280, Lutherville, MD 21093 USA
| | - Y. Jayasinghe
- Royal Children’s Hospital, Flemington Rd, Parkville, Melbourne, Vic 3054 Australia
- Department of Obstetrics & Gynecology, Royal Womens Hospital Melbourne, Parkville, Australia
| | - A. Anazodo
- Fertility Research Centre, Royal Hospital for Women, Barker Street, Sydney, Australia
| | - M. De Vos
- Centre for Reproductive Medicine of UZ Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium
| | - F. Amant
- Department of Oncology, KU Leuven, Leuven, Belgium
- Department of Surgery, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - C. Stern
- Fertility Preservation Service, Reproductive Services Unit, Royal Women’s Hospital, Parkville, 3051 Australia
- Fertility Preservation Service, Melbourne IVF, East Melbourne, 3002 Australia
| | - L. Appiah
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO USA
| | - T. L. Woodard
- Department of Gynecologic Oncology and Reproductive Medicine, MD Anderson Oncofertility Program, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - R. A. Anderson
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - L. M. Westphal
- Stanford University Hospital, 300 Pasteur Drive, Stanford, CA USA
| | - R. E. Leach
- Oncofertility Consortium, Michigan State University, 965 Wilson Road, Room A626B, East Lansing, MI 48824-1316 USA
| | - K. A. Rodriguez-Wallberg
- Department of Oncology-Pathology, Karolinska University Hospital, 14186 Stockholm, Sweden
- Department of Reproductive Medicine, Division of Gynecology and Reproduction, Karolinska University Hospital, 14186 Stockholm, Sweden
| | - P. Patrizio
- Yale Fertility Center and Yale Fertility Preservation Program, 200 West Campus Dr, Orange, CT 06477 USA
| | - Teresa K. Woodruff
- Oncofertility Consortium, Michigan State University, 965 Wilson Road, Room A626B, East Lansing, MI 48824-1316 USA
| |
Collapse
|
20
|
Ntemou E, Vidal PD, Alexandri C, Van den Steen G, Lambertini M, Demeestere I. Ovarian toxicity of carboplatin and paclitaxel in mouse carriers of mutation in BRIP1 tumor suppressor gene. Sci Rep 2022; 12:1658. [PMID: 35105904 PMCID: PMC8807594 DOI: 10.1038/s41598-022-05357-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 01/11/2022] [Indexed: 11/19/2022] Open
Abstract
More than 10% of women diagnosed with breast cancer during reproductive age carry hereditary germline pathogenic variants in high-penetrance BRCA genes or in others genes involved in DNA repair mechanisms such as PALB2, BRIP or ATM. Anticancer treatments may have an additional negative impact on the ovarian reserve and subsequently on the fertility of young patients carrying such mutations. Recently, the combination of carboplatin and paclitaxel is being recommended to these BRCA-mutated patients as neoadjuvant therapy. However, the impact on the ovary is unknown. Here, we investigated their effect of on the ovarian reserve using mice carriers of BRCA1-interacting protein C-terminal helicase-1 (BRIP1) mutation that plays an important role in BRCA1-dependent DNA repair. Results revealed that the administration of carboplatin or paclitaxel did not affect the ovarian reserve although increased DNA double-strand breaks were observed with carboplatin alone. Co-administration of carboplatin and paclitaxel resulted in a significant reduction of the ovarian reserve leading to a lower IVF performance, and an activation of the PI3K-Pten pathway, irrespective of the genetic background. This study suggests that co-administration of carboplatin and paclitaxel induces cumulative ovarian damage and infertility but a heterozygote genetic predisposition for DNA damage related to BRCA1 gene function does not increase this risk.
Collapse
Affiliation(s)
- E Ntemou
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles, Brussels, Belgium
| | - P Diaz Vidal
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles, Brussels, Belgium
| | - C Alexandri
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles, Brussels, Belgium
| | - G Van den Steen
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles, Brussels, Belgium
| | - M Lambertini
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genoa, Italy
- Department of Medical Oncology, UOC Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - I Demeestere
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles, Brussels, Belgium.
- Obstetrics and Gynaecology Department, Erasme Hospital, Brussels, Belgium.
| |
Collapse
|
21
|
Vanni VS, Campo G, Cioffi R, Papaleo E, Salonia A, Viganò P, Lambertini M, Candiani M, Meirow D, Orvieto R. The neglected members of the family: non-BRCA mutations in the Fanconi anemia/BRCA pathway and reproduction. Hum Reprod Update 2022; 28:296-311. [PMID: 35043201 DOI: 10.1093/humupd/dmab045] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 10/27/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND BReast CAncer (BRCA) genes are extensively studied in the context of fertility and reproductive aging. BRCA proteins are part of the DNA repair Fanconi anemia (FA)/BRCA pathway, in which more than 20 proteins are implicated. According to which gene is mutated and which interactions are lost owing to the mutation, carriers and patients with monoallelic or biallelic FA/BRCA mutations exhibit very different phenotypes, from overt FA to cancer predisposition or no pathological implications. The effect of the so far neglected non-BRCA FA mutations on fertility also deserves consideration. OBJECTIVE AND RATIONALE As improved treatments allow a longer life expectancy in patients with biallelic FA mutations and overt FA, infertility is emerging as a predominant feature. We thus reviewed the mechanisms for such a manifestation, as well as whether they also occur in monoallelic carriers of FA non-BRCA mutations. SEARCH METHODS Electronic databases PUBMED, EMBASE and CENTRAL were searched using the following term: 'fanconi' OR 'FANC' OR 'AND' 'fertility' OR 'pregnancy' OR 'ovarian reserve' OR 'spermatogenesis' OR 'hypogonadism'. All pertinent reports in the English-language literature were retrieved until May 2021 and the reference lists were systematically searched in order to identify any potential additional studies. OUTCOMES Biallelic FA mutations causing overt FA disease are associated with premature ovarian insufficiency (POI) occurring in the fourth decade in women and with primary non-obstructive azoospermia (NOA) in men. Hypogonadism in FA patients seems mainly associated with a defect in primordial germ cell proliferation in fetal life. In recent small, exploratory whole-exome sequencing studies, biallelic clinically occult mutations in the FA complementation group A (Fanca) and M (Fancm) genes were found in otherwise healthy patients with isolated NOA or POI, and also monoallelic carrier status for a loss-of-function mutation in Fanca has been implicated as a possible cause for POI. In those patients with known monoallelic FA mutations undergoing pre-implantation genetic testing, poor assisted reproduction outcomes are reported. However, the mechanisms underlying the repeated failures and the high miscarriage rates observed are not fully known. WIDER IMPLICATIONS The so far 'neglected' members of the FA/BRCA family will likely emerge as a relevant focus of investigation in the genetics of reproduction. Several (rather than a single) non-BRCA genes might be implicated. State-of-the-art methods, such as whole-genome/exome sequencing, and further exploratory studies are required to understand the prevalence and mechanisms for occult FA mutations in infertility and recurrent miscarriage.
Collapse
Affiliation(s)
- Valeria Stella Vanni
- Università Vita-Salute San Raffaele, Milan, Italy.,Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Raffaella Cioffi
- Università Vita-Salute San Raffaele, Milan, Italy.,Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Enrico Papaleo
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Salonia
- Università Vita-Salute San Raffaele, Milan, Italy.,Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Paola Viganò
- Reproductive Sciences Laboratory, Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Matteo Lambertini
- Department of Medical Oncology, U.O.C Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy.,Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - Massimo Candiani
- Università Vita-Salute San Raffaele, Milan, Italy.,Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Dror Meirow
- Department of Obstetrics and Gynecology, Chaim Sheba Medical Center, Tel-Hashomer, Ramat Gan, Israel
| | - Raoul Orvieto
- Department of Obstetrics and Gynecology, Chaim Sheba Medical Center, Tel-Hashomer, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv-Yafo, Israel
| |
Collapse
|
22
|
Hassel JC, Livingstone E, Allam JP, Behre HM, Bojunga J, Klein HH, Landsberg J, Nawroth F, Schüring A, Susok L, Thoms KM, Kiesel L, Berking C. Fertility preservation and management of pregnancy in melanoma patients requiring systemic therapy. ESMO Open 2021; 6:100248. [PMID: 34438241 PMCID: PMC8390524 DOI: 10.1016/j.esmoop.2021.100248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/21/2021] [Accepted: 07/27/2021] [Indexed: 01/09/2023] Open
Abstract
Melanoma is one of the most common cancers in adolescents and adults at fertile age, especially in women. With novel and more effective systemic therapies that began to profoundly change the dismal outcome of melanoma by prolonging overall survival, the wish for fertility preservation or even parenthood has to be considered for a growing portion of melanoma patients-from the patients' as well as from the physicians' perspective. The dual blockade of the mitogen-activated protein kinase pathway by B-Raf proto-oncogene serine/threonine kinase and mitogen-activated protein kinase inhibitors and the immune checkpoint inhibition by anti-programmed cell death protein 1 and anti-cytotoxic T-lymphocyte-associated protein-4 monoclonal antibodies constitute the current standard systemic approaches to combat locally advanced or metastatic melanoma. Here, the preclinical data and clinical evidence of these systemic therapies are reviewed in terms of their potential gonadotoxicity, teratogenicity, embryotoxicity and fetotoxicity. Recommendations for routine fertility and contraception counseling of melanoma patients at fertile age are provided in line with interdisciplinary recommendations for the diagnostic work-up of these patients and for fertility-protective measures. Differentiated recommendations for the systemic therapy in both the adjuvant and the advanced, metastatic treatment situation are given. In addition, the challenges of pregnancy during systemic melanoma therapy are discussed.
Collapse
Affiliation(s)
- J C Hassel
- Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany
| | - E Livingstone
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | - J P Allam
- Department of Andrology, University Hospital Bonn, Bonn, Germany
| | - H M Behre
- Center for Reproductive Medicine and Andrology, University Hospital Halle (Saale), Halle (Saale), Germany
| | - J Bojunga
- Department of Endocrinology, Diabetology and Nutrition Medicine, University Hospital Frankfurt, Frankfurt, Germany
| | - H H Klein
- Department of Internal Medicine, Endocrinology & Diabetology & Gastroenterology and Hepatology, BG University Hospital Bergmannsheil Bochum, Bochum, Germany
| | - J Landsberg
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
| | - F Nawroth
- Center for Infertility, Prenatal Medicine, Endocrinology and Osteology, Amedes Medical Center MVZ Hamburg, Hamburg, Germany
| | - A Schüring
- Fertility Center MVZ KITZ Regensburg, Regensburg, Germany
| | - L Susok
- Department of Dermatology, University Hospital Bochum, Bochum, Germany
| | - K M Thoms
- Department of Dermatology, Venereology and Allergy, University Medical Center Goettingen, Goettingen, Germany
| | - L Kiesel
- Department of Gynecology and Obstetrics, University Hospital Münster, Münster, Germany
| | - C Berking
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Comprehensive Cancer Center Erlangen EMN, Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany.
| |
Collapse
|
23
|
Denis-Laroque L, Drouet Y, Plotton I, Chopin N, Bonadona V, Lornage J, Salle B, Lasset C, Rousset-Jablonski C. Anti-müllerian hormone levels and antral follicle count in women with a BRCA1 or BRCA2 germline pathogenic variant: A retrospective cohort study. Breast 2021; 59:239-247. [PMID: 34304065 PMCID: PMC8326804 DOI: 10.1016/j.breast.2021.07.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/05/2021] [Accepted: 07/08/2021] [Indexed: 12/22/2022] Open
Abstract
Background Some studies suggested a decreased ovarian reserve among BRCA1/2 pathogenic variant carriers, with conflicting results. Methods We conducted a retrospective single-center observational study of ovarian reserve and spontaneous fertility comparing BRCA1/2 pathogenic variant carriers to controls (women who attended consultations to discuss fertility preservation before gonadotoxic treatment). Measures of associations between plasma AMH concentration, AFC and BRCA1/2 status were modelled by nonlinear generalized additive regression models and logistic regressions adjusted for age at plasma storage, oral contraceptive use, body mass index, cigarette smoking, and the AMH assay technique. Results The whole population comprised 119 BRCA1/2 pathogenic variant carriers and 92 controls. A total of 110 women (42 carriers, among whom 30 were cancer-free, and 68 controls) underwent an ovarian reserve evaluation. Spontaneous fertility analysis included all women who previously attempted to become pregnant (134 women). We observed a tendency towards a premature decrease in ovarian reserve in BRCA1/2 pathogenic variant carriers, but no difference in mean AMH or AFC levels was found between BRCA1/2 pathogenic variant carriers and controls. An analysis of the extreme levels of AMH (≤5 pmol/l) and AFC (≤7 follicles) by logistic regression suggested a higher risk of low ovarian reserve among BRCA1/2 pathogenic variant carriers (adjusted odds ratio (OR) = 3.57, 95% CI = 1.00–12.8, p = 0.05; and adjusted OR = 4.99, 95% CI = 1.10–22.62, p = 0.04, respectively). Discussion Attention should be paid to BRCA1/2 pathogenic variant carriers’ ovarian reserve, considering this potential risk of premature alteration. A tendency towards a premature decrease in the ovarian reserve in BRCA1/2 pathogenic variant carriers was found. BRCA1/2 pathogenic variant carriers presented a higher risk of a low ovarian reserve. Information and education regarding reproduction is mandatory among BRCA1/2 pathogenic variant carriers.
Collapse
Affiliation(s)
- Laurie Denis-Laroque
- Centre Léon Bérard, Department of Surgical Oncology, 28 rue Laënnec, 69008, Lyon, France
| | - Youenn Drouet
- Centre Léon Bérard, Département Prévention et Santé Publique, 28, Rue Laënnec, Lyon, 69008, France; CNRS UMR 5558, Laboratoire de Biométrie et Biologie évolutive, 16, rue Raphael Dubois, Villeurbanne Cedex, 69622, France
| | - Ingrid Plotton
- Hormonology and Molecular Endocrinology, Biology and Est Pathology Center, Hospices Civils de Lyon, 69500, U1208, Université Claude Bernard Lyon1, Bron, France
| | - Nicolas Chopin
- Centre Léon Bérard, Department of Surgical Oncology, 28 rue Laënnec, 69008, Lyon, France
| | - Valérie Bonadona
- Centre Léon Bérard, Département Prévention et Santé Publique, 28, Rue Laënnec, Lyon, 69008, France; CNRS UMR 5558, Laboratoire de Biométrie et Biologie évolutive, 16, rue Raphael Dubois, Villeurbanne Cedex, 69622, France
| | - Jacqueline Lornage
- Hospices Civils de Lyon, Groupement Hospitalier Est, Service de Médecine de La Reproduction, Bron, France
| | - Bruno Salle
- Hospices Civils de Lyon, Groupement Hospitalier Est, Service de Médecine de La Reproduction, Bron, France
| | - Christine Lasset
- Centre Léon Bérard, Département Prévention et Santé Publique, 28, Rue Laënnec, Lyon, 69008, France; CNRS UMR 5558, Laboratoire de Biométrie et Biologie évolutive, 16, rue Raphael Dubois, Villeurbanne Cedex, 69622, France
| | - Christine Rousset-Jablonski
- Centre Léon Bérard, Department of Surgical Oncology, 28 rue Laënnec, 69008, Lyon, France; Hospices Civils de Lyon, Lyon Sud University Hospital, Department of Obstetrics and Gynecology, 165 Chemin Du Grand Revoyet, 69310, Pierre-Bénite, France; Research on Healthcare Performance RESHAPE, INSERM U1290, Université Claude Bernard Lyon 1, France.
| |
Collapse
|
24
|
Turan V, Lambertini M, Lee DY, Wang E, Clatot F, Karlan BY, Demeestere I, Bang H, Oktay K. Association of Germline BRCA Pathogenic Variants With Diminished Ovarian Reserve: A Meta-Analysis of Individual Patient-Level Data. J Clin Oncol 2021; 39:2016-2024. [PMID: 33891474 PMCID: PMC8260903 DOI: 10.1200/jco.20.02880] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 01/27/2021] [Accepted: 03/09/2021] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To determine whether germline BRCA (gBRCA) pathogenic variants are associated with decreased ovarian reserve. MATERIALS AND METHODS An individual patient-level data meta-analysis was performed using five data sets on 828 evaluable women who were tested for gBRCA. Of those, 250 carried gBRCA, whereas 578 had tested negative and served as controls. Of the women with gBRCA, four centers studied those affected with breast cancer (n = 161) and one studied unaffected individuals (n = 89). The data were adjusted for the center, age, body mass index, smoking, and oral contraceptive pill use before the final analysis. Anti-Müllerian hormone (AMH) levels in affected women were drawn before presystemic therapy. RESULTS The mean age of women with versus without gBRCA1/2 (34.1 ± 4.9 v 34.3 ± 4.8 years; P = .48) and with gBRCA1 versus gBRCA2 (33.7 ± 4.9 v 34.6 ± 4.8 years; P = .16) was similar. After the adjustments, women with gBRCA1/2 had significantly lower AMH levels compared with controls (23% lower; 95% CI, 4 to 38; P = .02). When the adjusted analysis was limited to affected women (157 with gBRCA v 524 without, after exclusions), the difference persisted (25% lower; 95% CI, 9 to 38; P = .003). The serum AMH levels were lower in women with gBRCA1 (33% lower; 95% CI, 12 to 49; P = .004) but not gBRCA2 compared with controls (7% lower; 95% CI, 31% lower to 26% higher; P = .64). CONCLUSION Young women with gBRCA pathogenic variants, particularly those affected and with gBRCA1, have lower serum AMH levels compared with controls. They may need to be preferentially counseled about the possibility of shortened reproductive lifespan because of diminished ovarian reserve.
Collapse
Affiliation(s)
- Volkan Turan
- Department of Obstetrics and Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT
- Department of Obstetrics and Gynecology, Health and Technology University School of Medicine, Istanbul, Turkey
| | - Matteo Lambertini
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, UOC Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Dong-Yun Lee
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Erica Wang
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Florian Clatot
- Department of Medical Oncology, Henri Becquerel Centre, Rouen, France
| | - Beth Y. Karlan
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA
| | - Isabelle Demeestere
- Fertility Clinic, Research Laboratory on Human Reproduction, CUB-Erasme, and Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Heejung Bang
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, CA
| | - Kutluk Oktay
- Department of Obstetrics and Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
25
|
Martelli V, Latocca MM, Ruelle T, Perachino M, Arecco L, Beshiri K, Razeti MG, Tagliamento M, Cosso M, Fregatti P, Lambertini M. Comparing the Gonadotoxicity of Multiple Breast Cancer Regimens: Important Understanding for Managing Breast Cancer in Pre-Menopausal Women. BREAST CANCER (DOVE MEDICAL PRESS) 2021; 13:341-351. [PMID: 34079366 PMCID: PMC8164347 DOI: 10.2147/bctt.s274283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/14/2021] [Indexed: 11/23/2022]
Abstract
Over the last several decades, improvements in breast cancer treatment have contributed to increased cure rates for women diagnosed with this malignancy. Consequently, great importance should be paid to the long-term side effects of systemic therapies. For young women (defined as per guideline ≤40 years at diagnosis) who undergo chemotherapy, one of the most impactful side effects on their quality of life is premature ovarian insufficiency (POI) leading to fertility-related problems and the side effects of early menopause. Regimens, type, and doses of chemotherapy, as well as the age of patients and their ovarian reserve at the time of treatment are major risk factors for treatment-induced POI. For these reasons, childbearing desire and preservation of ovarian function and/or fertility should be discussed with all premenopausal patients before planning the treatments. This manuscript summarizes the available fertility preservation techniques in breast cancer patients, the risk of treatment-induced POI with different anticancer treatments, and the possible procedures to prevent it. A special focus is paid to the role of oncofertility counseling, as a central part of the visit in this setting, during which the patient should receive all the information about the potential consequences of the disease and of the proposed treatment on her future life.
Collapse
Affiliation(s)
- Valentino Martelli
- Department of Medical Oncology, U.O.C Clinica Di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - Maria Maddalena Latocca
- Department of Medical Oncology, U.O.C Clinica Di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - Tommaso Ruelle
- Department of Medical Oncology, U.O.C Clinica Di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - Marta Perachino
- Department of Medical Oncology, U.O.C Clinica Di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - Luca Arecco
- Department of Medical Oncology, U.O.C Clinica Di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - Kristi Beshiri
- Department of Medical Oncology, U.O.C Clinica Di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - Maria Grazia Razeti
- Department of Medical Oncology, U.O.C Clinica Di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - Marco Tagliamento
- Department of Medical Oncology, U.O.C Clinica Di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - Maurizio Cosso
- Department of Radiology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Piero Fregatti
- U.O.C. Clinica Di Chirurgia Senologica, Department of Surgery, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Integrated Diagnostic Surgical Sciences, School of Medicine, University of Genova, Genova, Italy
| | - Matteo Lambertini
- Department of Medical Oncology, U.O.C Clinica Di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
| |
Collapse
|
26
|
La Marca A, Mastellari E. Fertility preservation for genetic diseases leading to premature ovarian insufficiency (POI). J Assist Reprod Genet 2021; 38:759-777. [PMID: 33495935 DOI: 10.1007/s10815-021-02067-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 01/07/2021] [Indexed: 12/13/2022] Open
Abstract
PURPOSE The current review aims to summarize the data available concerning the applicability of fertility preservation techniques to genetic conditions at risk of premature ovarian insufficiency (POI). METHODS A literature review through the PubMed Database was carried out. RESULTS About 10% of cases of POI is related to genetic diseases. The most frequent conditions associated with POI are Turner syndrome and fragile X pre-mutation; mutation of BRCA 1-2 genes and several other mutations and genetic syndromes have recently been highlighted, although they rarely occur. If a diagnosis is issued before POI onset, counseling on currently available fertility preservation techniques is advisable. In case of spontaneous menarche (this can occur variably depending on the mutation) established techniques like embryo or oocyte cryopreservation can be proposed, even if, in some cases, their effectiveness may be reduced by ovarian alterations connected to the mutation. Ovarian tissue cryopreservation has recently been defined as an established medical procedure for fertility preservation in young cancer patients and may be an option for prepubertal patients. However, it is still experimental in special populations with genetic diseases causing POI. New innovative experimental techniques, like in vitro maturation of immature oocytes (IVM) and vitro activation (IVA) of immature follicles on ovarian tissue, have shown limited but encouraging data and they will be probably available in the near future. For a correct risk-benefit evaluation, the following aspects should be considered: actual knowledge about the pathology-specific efficacy of the various techniques, the average age of onset of POI, the possible risks associated with the procedure in relation to the underlying pathology, the probability of spontaneous conception, as well as the health implications of a possible future pregnancy.. CONCLUSIONS Fertility preservation techniques represent a crucial opportunity for patients with genetic risk of POI. Early diagnosis increases the chances to apply these techniques. No specific recommendations concerning fertility preservation for each genetic pathology are available, and clinicians should first counsel the patient and her relatives about known risks and benefits of the available techniques, both those established and those considered as experimental.
Collapse
Affiliation(s)
- Antonio La Marca
- Department of Medical and Surgical Sciences for Mother, Child and Adult, University of Modena and Reggio Emilia, Modena, Italy. .,Clinica Eugin Modena, Modena, Italy.
| | - Elisa Mastellari
- Department of Medical and Surgical Sciences for Mother, Child and Adult, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
27
|
Shulman Y, Almog B, Kalma Y, Fouks Y, Azem F, Cohen Y. Effects of letrozole or tamoxifen coadministered with a standard stimulation protocol on fertility preservation among breast cancer patients. J Assist Reprod Genet 2021; 38:743-750. [PMID: 33409757 DOI: 10.1007/s10815-020-02030-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 12/02/2020] [Indexed: 12/21/2022] Open
Abstract
PURPOSE To assess the effects of letrozole or tamoxifen coadministration on fertility preservation treatment outcomes. METHODS Retrospective cohort study of 118 breast cancer patients undergoing fertility preservation treatment between 2008 and 2018. Patients who received letrozole (n = 36) or tamoxifen (n = 30) were compared to controls (n = 52) who underwent standard ovarian stimulation protocols. The primary outcome measures included the number of retrieved oocytes, mature oocytes (MII), fertilization, and top-quality embryo rates. The secondary outcome measures included duration of stimulation, gonadotropin dose and peak estradiol level. RESULTS The number of oocytes retrieved, MII oocytes, fertilization rate, duration of stimulation, or gonadotropin dose were similar in the letrozole and tamoxifen groups, compared to controls. Top-quality embryo rate was lower in the tamoxifen group compared to controls (25% vs 39.4%, respectively, P = 0.034). The abnormal fertilization rate was higher in the letrozole group compared to controls (7.8% vs 3.60%, respectively, P = 0.015). A stepwise logistic regression analysis revealed that letrozole and peak estradiol were significantly associated with abnormal fertilization (OR 11.94; 95% CI 2.35-60.4, P = 0.003 for letrozole and OR 1.075; 95% CI 1.024-1.12, P = 0.004 per 100 unit change in estradiol). CONCLUSIONS There may be a negative effect of letrozole or tamoxifen on fertilization and embryo quality, in fertility preservation cycles. Further studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Yael Shulman
- IVF Unit, Lis Maternity Hospital, Tel-Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, 6 Weizmann Street, 6423906, Tel Aviv, Israel.
| | - Benny Almog
- IVF Unit, Lis Maternity Hospital, Tel-Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, 6 Weizmann Street, 6423906, Tel Aviv, Israel
| | - Yael Kalma
- IVF Unit, Lis Maternity Hospital, Tel-Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, 6 Weizmann Street, 6423906, Tel Aviv, Israel
| | - Yuval Fouks
- IVF Unit, Lis Maternity Hospital, Tel-Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, 6 Weizmann Street, 6423906, Tel Aviv, Israel
| | - Foad Azem
- IVF Unit, Lis Maternity Hospital, Tel-Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, 6 Weizmann Street, 6423906, Tel Aviv, Israel
| | - Yoni Cohen
- IVF Unit, Lis Maternity Hospital, Tel-Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, 6 Weizmann Street, 6423906, Tel Aviv, Israel
| |
Collapse
|
28
|
Installing oncofertility programs for common cancers in optimum resource settings (Repro-Can-OPEN Study Part II): a committee opinion. J Assist Reprod Genet 2021; 38:163-176. [PMID: 33452592 PMCID: PMC7810602 DOI: 10.1007/s10815-020-02012-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 11/09/2020] [Indexed: 01/03/2023] Open
Abstract
PURPOSE The main objective of Repro-Can-OPEN Study Part 2 is to learn more about oncofertility practices in optimum resource settings to provide a roadmap to establish oncofertility best practice models. METHODS As an extrapolation for oncofertility best practice models in optimum resource settings, we surveyed 25 leading and well-resourced oncofertility centers and institutions from the USA, Europe, Australia, and Japan. The survey included questions on the availability and degree of utilization of fertility preservation options in case of childhood cancer, breast cancer, and blood cancer. RESULTS All surveyed centers responded to all questions. Responses and their calculated oncofertility scores showed three major characteristics of oncofertility practice in optimum resource settings: (1) strong utilization of sperm freezing, egg freezing, embryo freezing, ovarian tissue freezing, gonadal shielding, and fractionation of chemo- and radiotherapy; (2) promising utilization of GnRH analogs, oophoropexy, testicular tissue freezing, and oocyte in vitro maturation (IVM); and (3) rare utilization of neoadjuvant cytoprotective pharmacotherapy, artificial ovary, in vitro spermatogenesis, and stem cell reproductive technology as they are still in preclinical or early clinical research settings. Proper technical and ethical concerns should be considered when offering advanced and experimental oncofertility options to patients. CONCLUSIONS Our Repro-Can-OPEN Study Part 2 proposed installing specific oncofertility programs for common cancers in optimum resource settings as an extrapolation for best practice models. This will provide efficient oncofertility edification and modeling to oncofertility teams and related healthcare providers around the globe and help them offer the best care possible to their patients.
Collapse
|
29
|
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 2020; 157:103201. [PMID: 33333149 DOI: 10.1016/j.critrevonc.2020.103201] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 11/29/2020] [Accepted: 11/30/2020] [Indexed: 12/12/2022] Open
Abstract
The detection of germline BRCA1/2 pathogenic variant has relevant implications for the patients and their family members. Family planning, prophylactic surgery and the possibility of preimplantation genetic testing for monogenic disorders (PGT-M) to avoid transmittance of pathogenic variants to the offspring are relevant topics in this setting. PGT-M is valuable option for BRCA carriers, but it remains a controversial and underdiscussed topic. Although the advances in PGT technologies have improved pregnancy rate, there are still several important challenges associated with its use. The purpose of this review is to report the current evidence on PGT-M for BRCA1/2 carriers, ethical concerns and controversy associated with its use, reproductive implications of BRCA pathogenic variants, underlying areas in which an educational effort would be beneficial as well as possibilities for future research efforts in the field.
Collapse
Affiliation(s)
- Petra Vuković
- Division of Radiotherapy and Medical Oncology, University Hospital for Tumors, University Hospital Center Sestre Milosrdnice, Zagreb, 10000, Croatia.
| | - Fedro Alessandro Peccatori
- Fertility and Procreation Unit, Gynecologic Oncology Program, IEO European Institute of Oncology IRCCS, Milan, 20125, Italy.
| | - Claudia Massarotti
- Physiopathology of Human Reproduction Unit, IRCCS Ospedale Policlinico San Martino, Genova, 16132, Italy.
| | | | - Lidija Beketić-Orešković
- Division of Radiotherapy and Medical Oncology, University Hospital for Tumors, University Hospital Center Sestre Milosrdnice, Zagreb, 10000, Croatia; Department of Clinical Oncology, School of Medicine, University of Zagreb, Zagreb, 10000, Croatia.
| | - Matteo Lambertini
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, 16132, Italy; Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, 16126, Italy.
| |
Collapse
|
30
|
Poorvu PD, Gelber SI, Zheng Y, Ruddy KJ, Tamimi RM, Peppercorn J, Schapira L, Borges VF, Come SE, Lambertini M, Rosenberg SM, Partridge AH. Pregnancy after breast cancer: Results from a prospective cohort of young women with breast cancer. Cancer 2020; 127:1021-1028. [PMID: 33259061 DOI: 10.1002/cncr.33342] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/12/2020] [Accepted: 11/02/2020] [Indexed: 01/04/2023]
Abstract
BACKGROUND Many young women with newly diagnosed breast cancer are interested in future pregnancies. Prospective data regarding fertility interest and reproductive patterns after diagnosis are needed to counsel patients. METHODS The Young Women's Breast Cancer Study is a multicenter, prospective cohort of women who were diagnosed with breast cancer at age ≤40 years between 2006 and 2016. Women complete surveys at baseline, every 6 months for 3 years, then annually. Here, the authors describe fertility interest and pregnancies within 5 years of diagnosis for women with stage 0 through III breast cancer. RESULTS Of 1026 eligible participants, 368 (36%) reported interest in future biologic children at least once within 5 years after diagnosis, including 16% at 5 years after diagnosis. Among 130 women who attempted to become pregnant, 90 (69.2%) conceived; and, among 896 women who did not attempt to conceive, 18 (2.0%) became pregnant, with a total of 152 pregnancies resulting in 91 live births. Factors associated with pregnancy included younger versus older age at diagnosis (aged ≤30 vs 36-40 years: odds ratio [OR], 6.63; 95% CI, 3.18-13.83; P < .0001; aged 31-35 vs 36-40 years: OR, 5.86; 95% CI, 3.37-10.17; P < .0001) and being nulliparous versus parous (OR, 2.66; 95% CI, 1.56-4.53; P = .001). The receipt of endocrine therapy versus no endocrine therapy (OR, 0.35; 95% CI, 0.20-0.59; P = .001) was inversely associated with pregnancy. CONCLUSIONS Many women remain interested in future fertility in the 4 years after a breast cancer diagnosis, indicating that longitudinal fertility discussions are needed. Although a minority of those interested in having children attempted to become pregnant in the first 5 years, most who attempted to conceive did so and had live births.
Collapse
Affiliation(s)
- Philip D Poorvu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Shari I Gelber
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Yue Zheng
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kathryn J Ruddy
- Department of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - Rulla M Tamimi
- Department of Population Health Sciences, Weill Cornell Medicine, New York, New York
| | - Jeffrey Peppercorn
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | | | - Virginia F Borges
- Department of Medical Oncology, University of Colorado Cancer Center, Aurora, Colorado
| | - Steven E Come
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Matteo Lambertini
- Department of Internal Medicine and Medical Specialties, University of Genoa, Genoa, Italy.,Medical Oncology Clinic, IRCCS San Maritino Polyclinic Hospital, Genoa, Italy
| | - Shoshana M Rosenberg
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Social and Behavioral Sciences, Harvard School of Public Health, Boston, Massachusetts
| | - Ann H Partridge
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
31
|
Malacarne E, Devesa M, Martinez F, Rodriguez I, Coroleu B. COH outcomes in breast cancer patients for fertility preservation: a comparison with the expected response by age. J Assist Reprod Genet 2020; 37:3069-3076. [PMID: 32945994 PMCID: PMC7714818 DOI: 10.1007/s10815-020-01944-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 09/08/2020] [Indexed: 12/23/2022] Open
Abstract
PURPOSE Breast cancer is the most common cancer diagnosed during childbearing age, and fertility preservation is becoming increasingly more essential. However, recent studies indicate a possible poorer response to controlled ovarian hyperstimulation (COH) in cancer patients than in non-cancer controls and a negative impact of BRCA mutations on female fertility. This study aims to evaluate ovarian response and the number of mature oocytes (MII) vitrified in women with breast cancer, with or without BRCA mutation, comparing them to the expected response according to an age-related nomogram. METHODS This is a retrospective observational study involving sixty-one breast cancer patients who underwent COH for oocyte cryopreservation. The age-specific nomogram was built using 3871 patients who underwent COH due to oocyte donation, fertility preservation for non-medical reasons, or FIVET for male factor exclusively. RESULTS The mean number of oocytes retrieved was 13.03, whereas the mean number of MII oocytes was 10.00. After the application of the z-score, no statistically significant differences were found compared with the expected response in the general population, neither by dividing patients according to the presence or absence of BRCA mutation nor according to the phase in which they initiated stimulation. CONCLUSION The results obtained do not support the notion of a negative impact of the BRCA mutation on the ovarian response of women with breast cancer. Women with breast cancer undergoing COH for fertility preservation can expect the ovarian response predicted for their age.
Collapse
Affiliation(s)
- Elisa Malacarne
- Department of Clinical and Experimental Medicine, Division of Obstetrics and Gynecology, University of Pisa, Via Roma, 67, 56126, Pisa, Italy.
| | - Marta Devesa
- Human Reproduction Service, Department of Obstetrics, Gynaecology and Reproduction, University Hospital Dexeus, Barcelona, Spain
| | - Francisca Martinez
- Human Reproduction Service, Department of Obstetrics, Gynaecology and Reproduction, University Hospital Dexeus, Barcelona, Spain
| | - Ignacio Rodriguez
- Human Reproduction Service, Department of Obstetrics, Gynaecology and Reproduction, University Hospital Dexeus, Barcelona, Spain
| | - Buenaventura Coroleu
- Human Reproduction Service, Department of Obstetrics, Gynaecology and Reproduction, University Hospital Dexeus, Barcelona, Spain
| |
Collapse
|
32
|
Abstract
Fertility represents a biological and psychological requirement for women. Some genetic diseases represent a rare cause of infertility, being responsible for 10% of cases of premature ovarian insufficiency. Among these, the most frequent and also those most studied by researchers are Turner Syndrome - due to a karyotype abnormality of the X chromosome pair - and the presence of fragile X premutation (FMR1). To exclude these conditions the diagnostic workup for non-iatrogenic premature ovarian insufficiency (POI) involves the performance of a karyotype analysis and the search for the FMR1 gene mutation, as well as the search for the presence of Y-chromosomal material. However, several other mutations and genetic syndromes associated with POI development have recently been highlighted, although they occur rarely, such as the GALT gene mutation in galactosemia or the FOXL2 gene mutation in BPES and many others, and further autosomal genetic testing are indicated if clinical suspicion is present. Mutations of BRCA 1 and 2 genes, make patients at genetically determined high risk of developing early ovarian or breast cancer and of getting POIs for the treatments they must undergo to prevent it (prophylactic bilateral oophorectomy) or treat it (chemotherapy). The management of impaired fertility is not less important than that of other syndromic manifestations for the quality of life of patients. Few data are available regarding the efficiency of cryopreservation of reproductive material (oocytes, embryos or ovarian tissue) in order to preserve fertility in this particular subgroup of patients, but certainly it represents a promising chance and a hope for the future.
Collapse
Affiliation(s)
- Elisa Mastellari
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Antonio La Marca
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy -
- Clinica Eugin Modena, Modena, Italy
| |
Collapse
|
33
|
Landry DA, Vaishnav HT, Vanderhyden BC. The significance of ovarian fibrosis. Oncotarget 2020; 11:4366-4370. [PMID: 33315987 PMCID: PMC7720769 DOI: 10.18632/oncotarget.27822] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 11/03/2020] [Indexed: 02/06/2023] Open
Abstract
Ovarian aging is associated with significant changes in the structural organization of collagen, resulting in ovarian fibrosis. In many other tissues, fibrosis increases risks associated with tumorigenesis and metastasis. Thus, it is possible that ovarian fibrosis increases the risk of ovarian cancer by creating a microenvironment more permissive to tumor growth. In this research perspective, we review the impact of female reproduction on the development of ovarian fibrosis and the contributions of genetic and hormonal disruptions such as BRCA mutation, polycystic ovarian syndrome, and infertility to structural changes in the ovary and their relative risk of ovarian cancer. We also explore new fundamental questions in the field of ovarian fibrosis and possible prevention strategies such as metformin.
Collapse
Affiliation(s)
- David A Landry
- Cancer Therapeutics Program, Ottawa Hospital Research Institute and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | - Het T Vaishnav
- Cancer Therapeutics Program, Ottawa Hospital Research Institute and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | - Barbara C Vanderhyden
- Cancer Therapeutics Program, Ottawa Hospital Research Institute and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| |
Collapse
|
34
|
Butler E, Collier S, Hevey D. The factors associated with distress a minimum of six months after BRCA1/2 confirmation: A systematic review. J Psychosoc Oncol 2020; 39:646-672. [PMID: 33089755 DOI: 10.1080/07347332.2020.1836109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PROBLEM IDENTIFICATION Many BRCA1/2 carriers experience an increase in distress after diagnosis; however, there is a need to review the longer term psychological implications of genetic confirmation and the factors associated with persistent distress. LITERATURE SEARCH This article systematically reviewed the literature in line with PRISMA guidelines on distress a minimum of six months after BRCA1/2 confirmation focusing on prevalence rates and factors associated with distress. DATA EVALUATION AND SYNTHESIS Fifteen studies were identified for inclusion and a narrative synthesis was conducted. Distress was associated with a range of demographic, clinical and psychological factors. A consistent finding was that although most carriers experience a reduction in distress 6-12 months after BRCA1/2 confirmation, those who experience persistent distress are more likely to have had higher distress levels at time of genetic testing. Risk reducing surgery may also play a role in reducing distress. CONCLUSION The review highlights the importance of psychological assessment and the use of specific distress measures. Given the considerable challenges in synthesizing the data there is a need for further prospective studies of high methodological quality.
Collapse
Affiliation(s)
- Ellen Butler
- School of Psychology, Trinity College Dublin, Dublin, Ireland
| | - Sonya Collier
- Department of Psychological Medicine, St James's Hospital, Dublin, Ireland
| | - David Hevey
- School of Psychology, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
35
|
Bonardi B, Massarotti C, Bruzzone M, Goldrat O, Mangili G, Anserini P, Spinaci S, Arecco L, Del Mastro L, Ceppi M, Demeestere I, Lambertini M. Efficacy and Safety of Controlled Ovarian Stimulation With or Without Letrozole Co-administration for Fertility Preservation: A Systematic Review and Meta-Analysis. Front Oncol 2020; 10:574669. [PMID: 33117711 PMCID: PMC7575927 DOI: 10.3389/fonc.2020.574669] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 09/01/2020] [Indexed: 01/01/2023] Open
Abstract
Background: The co-administration of letrozole during controlled ovarian stimulation (COS) with gonadotropins is used to limit the potentially harmful effects of a supra-physiological rise in estrogen levels on hormone-sensitive cancers. However, the efficacy and safety of adding letrozole to COS remain debated. Methods: This is a systematic review and meta-analysis of published studies that compared the efficacy and safety of COS with co-administration of letrozole vs. COS without letrozole in all patient populations. A secondary analysis was done including only the studies in breast cancer patients. The primary efficacy endpoint was the number of retrieved mature Metaphase II (MII) oocytes. Secondary efficacy and safety endpoints were total number of oocytes, maturation rate, fertilization rate, number of cryopreserved embryos, peak estradiol levels, progesterone levels, and total gonadotropin dose. Data for each endpoint were reported and analyzed thorough mean ratio (MR) with 95% confidence interval (CI). Results: A total of 11 records were selected including 2,121 patients (990 patients underwent COS with letrozole and 1,131 COS without letrozole). The addition of letrozole to COS did not have any negative effect on the number of mature oocytes collected (MR = 1.00, 95% CI = 0.87–1.16; P = 0.967) and the other efficacy endpoints. COS with letrozole was associated with significantly decreased peak estradiol levels (MR = 0.28, 95% CI = 0.24–0.32; P < 0.001). Similar results were observed in the secondary analysis including only breast cancer patients. Conclusions: These findings are reassuring on the efficacy and safety of COS with gonadotropins and letrozole and are particularly important for fertility preservation in women with hormone-sensitive cancers.
Collapse
Affiliation(s)
- Benedetta Bonardi
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles (ULB), Brussels, Belgium.,Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Claudia Massarotti
- Physiopathology of Human Reproduction Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Marco Bruzzone
- Clinical Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | | | - Giorgia Mangili
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paola Anserini
- Physiopathology of Human Reproduction Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Stefano Spinaci
- Division of Breast Surgery, Ospedale Villa Scassi, Genova, Italy
| | - Luca Arecco
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy.,Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Lucia Del Mastro
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy.,Breast Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Marcello Ceppi
- Clinical Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Isabelle Demeestere
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles (ULB), Brussels, Belgium.,Fertility Clinic, CUB-Hôpital Erasme, Brussels, Belgium
| | - Matteo Lambertini
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy.,Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
36
|
Perachino M, Massarotti C, Razeti MG, Parisi F, Arecco L, Damassi A, Fregatti P, Solinas C, Lambertini M. Gender-specific aspects related to type of fertility preservation strategies and access to fertility care. ESMO Open 2020; 5:e000771. [PMID: 33115753 PMCID: PMC7594356 DOI: 10.1136/esmoopen-2020-000771] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/17/2020] [Accepted: 08/14/2020] [Indexed: 12/11/2022] Open
Abstract
Survivorship is an area of paramount importance to be addressed as early as possible after cancer diagnosis by all health care providers. On this regard, cancer care in young patients often poses several age-related considerations among which fertility and pregnancy-related issues have a crucial role. According to the available guidelines on the topic, all patients with cancer diagnosed during their reproductive years should be provided a proper oncofertility counselling before starting anticancer treatments. This is an important step in order to inform patients about the potential treatment-induced gonadotoxicity and the available strategies for fertility preservation so that they can be referred as early as possible to fertility specialists if potentially interested in these options.In this manuscript, we aim to provide an up to date overview on the available efficacy and safety data with the main strategies for fertility preservation in male and female cancer patients in order to help optimising the oncofertility counselling performed by healthcare providers involved in cancer care and dealing with young patients. In male patients with cancer, sperm cryopreservation is the standard technique for fertility preservation. Oocyte/embryo cryopreservation, ovarian tissue cryopreservation and temporary ovarian suppression with luteinising hormone-releasing hormone agonists during chemotherapy are the main options in female patients with cancer.A multidisciplinary management building a strong network between fertility and oncology/haematology units is crucial to properly address fertility care in all young patients with cancer, at both diagnosis and during oncologic follow-up. Discussing fertility and pregnancy-related issues with young patients with cancer has to be considered mandatory nowadays keeping in mind that returning to a normal life (including the possibility to have a family and to live with as few side effects as possible) should be considered an important ambition in cancer care in the 21st century .
Collapse
Affiliation(s)
- Marta Perachino
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Liguria, Italy; Department of Medical Oncology, U.O.C Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Claudia Massarotti
- Physiopatology of Human Reproduction Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Maria Grazia Razeti
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Liguria, Italy; Department of Medical Oncology, U.O.C Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Francesca Parisi
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Liguria, Italy; Department of Medical Oncology, U.O.C Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Luca Arecco
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Liguria, Italy; Department of Medical Oncology, U.O.C Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Alessandra Damassi
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Liguria, Italy; Department of Medical Oncology, U.O.C Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Piero Fregatti
- Department of Surgery, U.O.C. Clinica di Chirurgia Senologica, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Integrated Diagnostic Surgical Sciences, School of Medicine, University of Genova, Genova, Italy
| | - Cinzia Solinas
- Medical Oncology, Azienda Tutela della Salute Sardegna, Hospital A. Segni Ozieri, Sassari, Italy
| | - Matteo Lambertini
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Liguria, Italy; Department of Medical Oncology, U.O.C Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| |
Collapse
|
37
|
Loubersac S, Dezellus A, Lefebvre T, Reignier A, Barriere P, Masson D, Freour T. Evolution of serum Anti-Müllerian Hormone (AMH) level in young women treated with chemotherapy for breast cancer according to basal AMH level. Eur J Obstet Gynecol Reprod Biol 2020; 254:132-137. [PMID: 32971432 DOI: 10.1016/j.ejogrb.2020.09.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 09/09/2020] [Accepted: 09/11/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Serum AMH level has been shown to decrease in women treated for breast cancer in several studies. However, whether basal AMH status affects AMH dynamics during chemotherapy remains to be clarified. The objective of this study was to compare serum AMH dynamics in young women with either low, normal or high basal serum AMH level at diagnosis, during and after treatment with chemotherapy for breast cancer. STUDY DESIGN In this secondary analysis of a prospective cohort study, serum AMH was measured during and after chemotherapy in 239 women of reproductive age diagnosed with breast cancer and treated with chemotherapy. The association between AMH dynamics throughout chemotherapy and during follow-up and basal AMH status, i.e. low AMH (<1 μg/l, <7 pmol/l), normal AMH (1-4.9 μg/l, 7-36 pmol/l) and high AMH (≥5 μg/l, >36 pmol/l), was evaluated. Menses occurrence was also recorded. RESULTS A total of 21 women had low, 154 had normal and 64 had high basal AMH level. Serum AMH rapidly decreased during chemotherapy in all groups, and its variation during chemotherapy and follow-up was not significantly different between the 3 groups. CONCLUSION No association was found between AMH variation during chemotherapy and follow-up, and basal AMH level at diagnosis. However, women with high basal AMH levels have significantly higher AMH levels throughout chemotherapy and follow-up than women with normal or low basal AMH levels at diagnosis.
Collapse
Affiliation(s)
- S Loubersac
- Service de médecine et biologie du développement et de la Reproduction, CHU de Nantes, Bd Jean Monnet, 44000, Nantes, France
| | - A Dezellus
- Service de médecine et biologie du développement et de la Reproduction, CHU de Nantes, Bd Jean Monnet, 44000, Nantes, France; Service d'oncologie, Institut de Cancérologie de l'Ouest, Bd Jacques Monod, 44805, Saint Herblain, France
| | - T Lefebvre
- Service de médecine et biologie du développement et de la Reproduction, CHU de Nantes, Bd Jean Monnet, 44000, Nantes, France; Faculté de médecine, Université de Nantes, rue Gaston Veil, 44035, Nantes, France
| | - A Reignier
- Service de médecine et biologie du développement et de la Reproduction, CHU de Nantes, Bd Jean Monnet, 44000, Nantes, France; Faculté de médecine, Université de Nantes, rue Gaston Veil, 44035, Nantes, France; Inserm, Université de Nantes, Centre de Recherche en Transplantation et Immunologie (ou CRTI), Nantes, France
| | - P Barriere
- Service de médecine et biologie du développement et de la Reproduction, CHU de Nantes, Bd Jean Monnet, 44000, Nantes, France; Faculté de médecine, Université de Nantes, rue Gaston Veil, 44035, Nantes, France; Inserm, Université de Nantes, Centre de Recherche en Transplantation et Immunologie (ou CRTI), Nantes, France
| | - D Masson
- Faculté de médecine, Université de Nantes, rue Gaston Veil, 44035, Nantes, France; Laboratoire de biochimie, CHU de Nantes, place Alexis-Ricordeau, 44000, Nantes, France; INSERM UMR 913, rue Gaston Veil, 44035, Nantes, France
| | - T Freour
- Service de médecine et biologie du développement et de la Reproduction, CHU de Nantes, Bd Jean Monnet, 44000, Nantes, France; Faculté de médecine, Université de Nantes, rue Gaston Veil, 44035, Nantes, France; Inserm, Université de Nantes, Centre de Recherche en Transplantation et Immunologie (ou CRTI), Nantes, France.
| | | |
Collapse
|
38
|
Arecco L, Perachino M, Damassi A, Latocca MM, Soldato D, Vallome G, Parisi F, Razeti MG, Solinas C, Tagliamento M, Spinaci S, Massarotti C, Lambertini M. Burning Questions in the Oncofertility Counseling of Young Breast Cancer Patients. Breast Cancer (Auckl) 2020; 14:1178223420954179. [PMID: 32952399 PMCID: PMC7476336 DOI: 10.1177/1178223420954179] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 08/10/2020] [Indexed: 12/28/2022] Open
Abstract
The improved prognosis of breast cancer patients makes survivorship issues an area of crucial importance. In this regard, an increased attention is needed toward the development of potential anticancer treatment-related long-term side-effects, including gonadal failure and infertility in young women. Therefore, fertility preservation and family planning are crucial issues to be addressed in all young women of reproductive age with newly diagnosed cancer. Despite a growing availability of data on the efficacy and safety of fertility preservation options and the fact that conceiving after prior history of breast cancer has become more accepted over time, there are still several gray zones in this field so that many physicians remain uncomfortable to deal with these topics. The purpose of this review is to answer some of the most controversial questions frequently asked by patients during their oncofertility counseling, in order to provide a detailed and up-to-date overview on the evidence available in this field to physicians involved in the care of young women with breast cancer.
Collapse
Affiliation(s)
- Luca Arecco
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, University of Genova, Genova, Italy
| | - Marta Perachino
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, University of Genova, Genova, Italy
| | - Alessandra Damassi
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, University of Genova, Genova, Italy
| | - Maria Maddalena Latocca
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, University of Genova, Genova, Italy
| | - Davide Soldato
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, University of Genova, Genova, Italy
| | - Giacomo Vallome
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, University of Genova, Genova, Italy
| | - Francesca Parisi
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, University of Genova, Genova, Italy
| | - Maria Grazia Razeti
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, University of Genova, Genova, Italy
| | - Cinzia Solinas
- Department of Medical Oncology, Azienda Tutela della Salute Sardegna, Hospital A.Segni Ozieri, Sassari, Italy
| | - Marco Tagliamento
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, University of Genova, Genova, Italy
| | - Stefano Spinaci
- Division of Breast Surgery, Ospedale Villa Scassi e ASL3, Genova, Italy
| | - Claudia Massarotti
- Physiopathology of Human Reproduction Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Matteo Lambertini
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, University of Genova, Genova, Italy
| |
Collapse
|
39
|
Merlino L, Chiné A, Galli C, Piccioni MG. BRCA1/2 genes mutations, ovarian reserve and female reproductive outcomes: a systematic review of the literature. ACTA ACUST UNITED AC 2020; 72:339-348. [PMID: 32744451 DOI: 10.23736/s0026-4784.20.04624-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION BRCA1 and BRCA2 genes mutations seems to impact female fertility, in addition to increasing the risk of ovarian and breast cancer. Several studies had investigated this issue but data available are still controversial. In order to clarify the role of BRCA1 and BRCA2 mutations in female fertility and ovarian function we carried out a systematic review of the literature with the aim to establish a possible management's strategy of these patients. EVIDENCE ACQUISITION A review of current literature regarding BRCA mutation (BRCAm) and fertility was conducted using the PubMed tool to select remarkable articles with the keywords "BRCA1/2 gene," "BRCA1/2 mutation," "anti-Müllerian hormone," "female fertility," "ovarian reserve" and "premature ovarian failure." EVIDENCE SYNTHESIS In current literature there are controversial findings about the relation between BRCA genes mutations and lifespan of female reproductive age. Several studies showed an higher risk of premature ovarian insufficiency of BRCAs mutations carriers, according to lower serum AMH level, primordial follicle count, or fewer oocyte yield after ovarian stimulation; on the other hand more recent studies reported not significant differences in serum AMH level or in reproductive outcomes between mutated and non-mutated BRCA patients. For this reason, currently there is not a strict recommendation for routine evaluation of fertility in female carriers of BRCA mutations. Nevertheless, the strong advice to complete childbearing by age 40 and then to undergo a risk-reducing salpingo-oophorectomy and the increased risk of infertility as a result of anticancer treatment in breast cancer BRCAm patients, make the issue of fertility and pregnancy planning in these women worthy of consideration. CONCLUSIONS A dedicated counseling to discuss these issues, eventually associated with a personalized assessment of serum AMH or antral follicle count in order to have a panoramic view of ovarian reserve, may be useful in the management of these patients.
Collapse
Affiliation(s)
- Lucia Merlino
- Department of Maternal and Child Health and Urological Sciences, Umberto I Polyclinic Hospital, Sapienza University, Rome, Italy -
| | - Alessandra Chiné
- Department of Maternal and Child Health and Urological Sciences, Umberto I Polyclinic Hospital, Sapienza University, Rome, Italy
| | - Cecilia Galli
- Department of Maternal and Child Health and Urological Sciences, Umberto I Polyclinic Hospital, Sapienza University, Rome, Italy
| | - Maria G Piccioni
- Department of Maternal and Child Health and Urological Sciences, Umberto I Polyclinic Hospital, Sapienza University, Rome, Italy
| |
Collapse
|
40
|
Lambertini M, Ameye L, Hamy AS, Zingarello A, Poorvu PD, Carrasco E, Grinshpun A, Han S, Rousset-Jablonski C, Ferrari A, Paluch-Shimon S, Cortesi L, Senechal C, Miolo G, Pogoda K, Pérez-Fidalgo JA, De Marchis L, Ponzone R, Livraghi L, Estevez-Diz MDP, Villarreal-Garza C, Dieci MV, Clatot F, Berlière M, Graffeo R, Teixeira L, Córdoba O, Sonnenblick A, Luna Pais H, Ignatiadis M, Paesmans M, Partridge AH, Caron O, Saule C, Del Mastro L, Peccatori FA, Azim HA. Pregnancy After Breast Cancer in Patients With Germline BRCA Mutations. J Clin Oncol 2020; 38:3012-3023. [PMID: 32673153 DOI: 10.1200/jco.19.02399] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
PURPOSE Young women with germline BRCA mutations have unique reproductive challenges. Pregnancy after breast cancer does not increase the risk of recurrence; however, very limited data are available in patients with BRCA mutations. This study investigated the impact of pregnancy on breast cancer outcomes in patients with germline BRCA mutations. PATIENTS AND METHODS This is an international, multicenter, hospital-based, retrospective cohort study. Eligible patients were diagnosed between January 2000 and December 2012 with invasive early breast cancer at age ≤ 40 years and harbored deleterious germline BRCA mutations. Primary end points were pregnancy rate, and disease-free survival (DFS) between patients with and without a pregnancy after breast cancer. Pregnancy outcomes and overall survival (OS) were secondary end points. Survival analyses were adjusted for guarantee-time bias controlling for known prognostic factors. RESULTS Of 1,252 patients with germline BRCA mutations (BRCA1, 811 patients; BRCA2, 430 patients; BRCA1/2, 11 patients) included, 195 had at least 1 pregnancy after breast cancer (pregnancy rate at 10 years, 19%; 95% CI, 17% to 22%). Induced abortions and miscarriages occurred in 16 (8.2%) and 20 (10.3%) patients, respectively. Among the 150 patients who gave birth (76.9%; 170 babies), pregnancy complications and congenital anomalies occurred in 13 (11.6%) and 2 (1.8%) cases, respectively. Median follow-up from breast cancer diagnosis was 8.3 years. No differences in DFS (adjusted hazard ratio [HR], 0.87; 95% CI, 0.61 to 1.23; P = .41) or OS (adjusted HR, 0.88; 95% CI, 0.50 to 1.56; P = .66) were observed between the pregnancy and nonpregnancy cohorts. CONCLUSION Pregnancy after breast cancer in patients with germline BRCA mutations is safe without apparent worsening of maternal prognosis and is associated with favorable fetal outcomes. These results provide reassurance to patients with BRCA-mutated breast cancer interested in future fertility.
Collapse
Affiliation(s)
- Matteo Lambertini
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genoa, Genoa, Italy.,Department of Medical Oncology, UOC Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Lieveke Ameye
- Data Centre, Institut Jules Bordet and Université Libre de Bruxelles, Brussels, Belgium
| | | | - Anna Zingarello
- Département Médecine Oncologique, Institut Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Philip D Poorvu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Estela Carrasco
- Hereditary Cancer Genetics Group, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Albert Grinshpun
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Sileny Han
- Multidisciplinary Breast Center, Department of Gynaecology, University Hospitals Leuven, Department of Oncology, KU Leuven, Leuven, Belgium
| | | | - Alberta Ferrari
- Department of Surgical Sciences, General Surgery III-Breast Surgery, Fondazione IRCCS Policlinico San Matteo, and Department of Clinical Surgical Sciences, University of Pavia, Pavia, Italy
| | | | - Laura Cortesi
- Department of Oncology and Haematology, Azienda Ospedaliero-Universitaria Policlinico di Modena, Modena, Italy
| | | | - Gianmaria Miolo
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano, IRCCS, Aviano, Italy
| | - Katarzyna Pogoda
- Department of Breast Cancer and Reconstructive Surgery, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | | | - Laura De Marchis
- Division of Medical Oncology, Department of Radiological, Oncological and Pathological Sciences, "La Sapienza" University of Rome, Rome, Italy
| | - Riccardo Ponzone
- Gynecological Oncology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
| | - Luca Livraghi
- Medical Oncology Unit, ASST Papa Giovanni XXIII, Bergamo, Italy.,University of Siena, Siena, Italy
| | - Maria Del Pilar Estevez-Diz
- Department of Oncology, Instituto do Cancer do Estado de São Paulo-Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Cynthia Villarreal-Garza
- Departamento de Investigacion y de Tumores Mamarios, Instituto Nacional de Cancerologia, Mexico City, Mexico.,Tecnologico de Monterrey, Centro de Cancer de Mama del Hospital Zambrano Hellion, Nuevo Leon, Mexico
| | - Maria Vittoria Dieci
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy.,Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
| | - Florian Clatot
- Department of Medical Oncology, Centre Henri Becquerel, Rouen, France
| | - Martine Berlière
- Department of Oncology, Breast Clinic, Cliniques Universitaires Saint-Luc UCL, Brussels, Belgium
| | - Rossella Graffeo
- Breast Unit of Southern Switzerland, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Luis Teixeira
- Breast Disease Unit Saint-Louis Hospital, APHP, Université de Paris, Inserm, U976 HIPI Unit, F-75010, Paris, France
| | - Octavi Córdoba
- Obstetrics and Gynecology Department, Hospital Universitari Son Espases, Palma, Spain
| | - Amir Sonnenblick
- Oncology Division, Tel Aviv Sourasky Medical Center, and Sackler Faculty of Medicine, Tel Aviv, Israel
| | - Helena Luna Pais
- Department of Medical Oncology, Centro Hospitalar Universitário Lisboa Norte-Hospital de Santa Maria, Lisbon, Portugal
| | - Michail Ignatiadis
- Department of Medical Oncology, Institut Jules Bordet and Université Libre de, Brussels, Belgium
| | - Marianne Paesmans
- Data Centre, Institut Jules Bordet and Université Libre de Bruxelles, Brussels, Belgium
| | - Ann H Partridge
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Olivier Caron
- Département Médecine Oncologique, Institut Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Claire Saule
- Department of Genetics, Institut Curie, Paris, France
| | - Lucia Del Mastro
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genoa, Genoa, Italy.,Breast Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Fedro A Peccatori
- Gynecologic Oncology Department, European Institute of Oncology IRCCS, Milan, Italy
| | - Hatem A Azim
- Tecnologico de Monterrey, Centro de Cancer de Mama del Hospital Zambrano Hellion, Nuevo Leon, Mexico
| |
Collapse
|
41
|
Lorzadeh N, Kazemirad N, Kazemirad Y. Human immunodeficiency: Extragonadal comorbidities of infertility in women. IMMUNITY INFLAMMATION AND DISEASE 2020; 8:447-457. [PMID: 32621331 PMCID: PMC7416027 DOI: 10.1002/iid3.327] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 06/21/2020] [Indexed: 12/12/2022]
Abstract
Introduction Infertility is mediated by several changes system‐wide. These changes are likely to cause other systems‐related pathologies, such as changes in systemic immune response, particularly inflammatory response can lead to cardiovascular diseases and breast cancer. Methods These morbidities can exist immediately or years after the diagnosis of infertility. Therefore, understanding the mechanism is important to move toward therapeutic interventions. Results Several extragonadal pathologies are reported due to infertility, as well as, how these might also contribute to reproductive disabilities. Detailed evidence are still not present that can give stronger result. Conclusion This review highlights some of the most frequent comorbidities that are seen in infertile women, hence requiring a need for complete clinical screening and care, as well as diagnosis and treatment in early stages.
Collapse
Affiliation(s)
- Nahid Lorzadeh
- Department of Obstetrics and Gynecology, Faculty of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Nastaran Kazemirad
- Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
42
|
Call for assessing treatment-induced gonadotoxicity of platinum-based chemotherapy in early breast cancer. Breast Cancer Res Treat 2020; 183:239-240. [PMID: 32596767 DOI: 10.1007/s10549-020-05767-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 06/20/2020] [Indexed: 12/31/2022]
|
43
|
Grynberg M, Dagher Hayeck B, Papanikolaou EG, Sifer C, Sermondade N, Sonigo C. BRCA1/2 gene mutations do not affect the capacity of oocytes from breast cancer candidates for fertility preservation to mature in vitro. Hum Reprod 2020; 34:374-379. [PMID: 30561604 DOI: 10.1093/humrep/dey358] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 12/07/2018] [Indexed: 12/14/2022] Open
Abstract
STUDY QUESTION Are the maturation rates of oocytes recovered from small antral follicles different between breast cancer patients presenting with or without a BRCA 1/2 gene mutation? SUMMARY ANSWER BRCA 1/2 gene mutations do not affect the capacity of oocytes from breast cancer candidates for fertility preservation to mature in vitro. WHAT IS KNOWN ALREADY Mutations in the BRCA1 and BRCA2 genes are associated with an increased risk for developing breast and ovarian cancer. Controversy exists about fertility and ovarian reserve in BRCA mutation carriers. Studies suggest that these patients may have low ovarian reserve and poor response to ovarian stimulation. The impaired ability of the mutated BRCA gene to repair double-strand breaks in DNA may prompt oocyte aging, apoptosis and meiotic errors. IVM of oocytes retrieved at germinal vesicle stage, followed by vitrification of metaphase II (MII) oocytes has recently emerged as an option for young women seeking fertility preservation, when ovarian stimulation is unfeasible. STUDY DESIGN, SIZE, DURATION Retrospective cohort study involving 329 breast cancer candidates for fertility preservation using IVM between January 2014 and December 2017. PARTICIPANTS/MATERIALS, SETTING, METHODS Inclusion criteria were: age 18-40 years; two ovaries present; no history of chemotherapy; test for BRCA 1/2 mutations performed. Before immature oocyte retrieval, all follicles measuring 2-9 mm in diameter were precisely counted on both ovaries and serum anti-Müllerian hormone (AMH) was measured irrespective of the phase of the cycle. Number of cumulus oocyte complexes (COC) retrieved, maturation rate and number of MII oocytes cryopreserved were compared according to BRCA mutation status. MAIN RESULTS AND THE ROLE OF CHANCE Overall, BRCA-mutated women (n = 52) and BRCA-negative women (n = 277) were comparable in terms of age (31.7 ± 3.9 versus 32.3 ± 3.8 years, respectively, P = 0.3), BMI (23.4 ± 4.7 versus 22.6 ± 3.7 kg/m2, respectively, P = 0.3) and ovarian reserve tests (antral follicle count: 20.5 ± 11.4 versus 21.7 ± 12.1 follicles, P = 0.5; serum AMH levels: 3.6 ± 2.9 versus 4.1 ± 3.6 ng/ml, P = 0.3, respectively). The number of COCs retrieved did not differ significantly between both groups (8.9 ± 6.9 versus 9.9 ± 8.1 oocytes, P = 0.5). After similar IVM rates (67 ± 24 versus 62 ± 23%, P = 0.2), the number of MII oocytes cryopreserved was similar in patients presenting BRCA mutation or not (5.1 ± 3.8 versus 6.1 ± 5.1, P = 0.1, respectively). LIMITATIONS, REASONS FOR CAUTION Given the low incidence of the mutation, these preliminary findings should be confirmed by further multi-center studies. WIDER IMPLICATIONS OF THE FINDINGS Although BRCA mutations are known to alter DNA repair mechanism, it does not seem to impair oocyte capacity to mature in vitro. STUDY FUNDING/COMPETING INTEREST(s) None.
Collapse
Affiliation(s)
- Michaël Grynberg
- Department of Reproductive Medicine and Fertility Preservation, Hôpital Antoine Béclère, Hôpitaux Universitaires Paris Sud, Assistance Publique - Hôpitaux de Paris, Clamart, France.,Department of Reproductive Medicine and Fertility Preservation, Hôpital Jean Verdier, Hôpitaux Universitaires Paris-Seine-Saint-Denis, Assistance Publique- Hôpitaux de Paris, Bondy, France.,Université Paris-Sud, Université Paris Saclay, Le Kremlin Bicêtre, France.,Inserm U1133 Université Paris Diderot, Paris, France
| | - Bénédicte Dagher Hayeck
- Department of Reproductive Medicine and Fertility Preservation, Hôpital Jean Verdier, Hôpitaux Universitaires Paris-Seine-Saint-Denis, Assistance Publique- Hôpitaux de Paris, Bondy, France
| | | | - Christophe Sifer
- Department of Cytogenetic and Reproductive Biology, Hôpital Jean Verdier, Hôpitaux Universitaires Paris-Seine-Saint-Denis, Assistance Publique - Hôpitaux de Paris, Bondy, France
| | - Nathalie Sermondade
- Department of Cytogenetic and Reproductive Biology, Hôpital Jean Verdier, Hôpitaux Universitaires Paris-Seine-Saint-Denis, Assistance Publique - Hôpitaux de Paris, Bondy, France
| | - Charlotte Sonigo
- Department of Reproductive Medicine and Fertility Preservation, Hôpital Antoine Béclère, Hôpitaux Universitaires Paris Sud, Assistance Publique - Hôpitaux de Paris, Clamart, France.,Inserm U1185 Université Paris-Sud, Université Paris Saclay, Le Kremlin Bicêtre, France
| |
Collapse
|
44
|
Hu KL, Wang S, Ye X, Zhang D. Effects of BRCA gene mutation on female reproductive potential: A systematic review. Maturitas 2020; 137:11-17. [PMID: 32498931 DOI: 10.1016/j.maturitas.2020.04.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/21/2020] [Accepted: 04/15/2020] [Indexed: 11/18/2022]
Abstract
Mutation of BRCA genes significantly increases the lifetime risk of breast, ovarian, fallopian tube and primary peritoneal cancers. In addition to the increased risk of these multiple malignancies, the recent literature suggests that mutations in BRCA genes may lead to decreased reproductive potential. In this systematic review, we focus on the effect of BRCA gene mutation on reproductive potential. The main outcomes included the rate of nulliparity, ovarian reserve, ovarian response, and the age at natural menopause. A total of 23 observational studies were included for quality analysis. The certainty of evidence was low to moderate: the main limitations were imprecision and statistically significant heterogeneity. Meta-analysis suggested that the rate of nulliparity, serum anti-müllerian hormone levels, antral follicle counts and ovarian response were not significantly affected in BRCA gene mutation carriers (P > 0.05). BRCA gene mutation carriers tended to have a lower number of primordial follicles (P = 0.0002) and lower age at natural menopause than non-carriers. In conclusion, there is no compelling evidence indicating that the rate of nulliparity, serum AMH, antral follicle counts and ovarian response are affected in BRCA mutation carriers.
Collapse
Affiliation(s)
- Kai-Lun Hu
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China.
| | - Siwen Wang
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China.
| | - Xiaohang Ye
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China.
| | - Dan Zhang
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China.
| |
Collapse
|
45
|
Sella T, Partridge AH. Fertility Counseling and Preservation in Breast Cancer. CURRENT BREAST CANCER REPORTS 2020. [DOI: 10.1007/s12609-019-00348-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
46
|
Hampe ME, Rhoton-Vlasak AS. Fertility preservation in breast cancer with case-based examples for guidance. J Assist Reprod Genet 2020; 37:717-729. [PMID: 32008180 PMCID: PMC7125269 DOI: 10.1007/s10815-019-01665-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 12/12/2019] [Indexed: 12/12/2022] Open
Abstract
With more young breast cancer survivors, a trend toward having children later in life, and improvements in assisted reproductive technology (ART), fertility preserving techniques are of growing importance prior to initiation of gonadotoxic treatments. The American Society for Clinical Oncology (ASCO) updated their Fertility Preservation in Patients with Cancer guidelines in April of 2018. ASCO continues to recognize oocyte and embryo cryopreservation as standard practice for women interested in preserving fertility and sperm cryopreservation as standard practice for men. ASCO has clarified their statement on ovarian suppression during chemotherapy as an option when standard methods are unavailable but should not be used as the sole method of fertility preservation (FP) due to conflicting evidence. ASCO also updated their statement on ovarian tissue cryopreservation, which is still labeled experimental but ASCO acknowledges that it can restore global ovarian function and could be of use in specific patients. The NCCN's Version 1.2018 Clinical Practice Guidelines® for treatment of breast cancer include fertility counseling as part of their work-up in all types of breast cancer for premenopausal women.The purpose of this review is to explain the indications and evidence for the different methods of FP for young breast cancer patients in accordance with ASCO and NCCN guidelines. The guidance will then be applied to three theoretical clinical cases in order to highlight actual use in clinical practice.
Collapse
Affiliation(s)
- Mary E Hampe
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Alice S Rhoton-Vlasak
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Florida College of Medicine, Gainesville, FL, USA.
| |
Collapse
|
47
|
Zhang X, Niu J, Che T, Zhu Y, Zhang H, Qu J. Fertility preservation in BRCA mutation carriers-efficacy and safety issues: a review. Reprod Biol Endocrinol 2020; 18:11. [PMID: 32070378 PMCID: PMC7027288 DOI: 10.1186/s12958-019-0561-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 12/27/2019] [Indexed: 02/08/2023] Open
Abstract
BRCA mutation carriers face various situations that influence their fertility potential. There is still a lack of guideline or expert consensus on Fertility Preservation (FP) in BRCA mutation carriers and the necessity and safety of FP in BRCA mutation carriers is still in dispute. This review aims to focus on the population of BRCA mutation carriers by analyzing the existing FP strategies, comprehensively comparing the pros and cons of each strategy and its applicability.FP is a suggestion for BRCA mutation carriers with birth planning. Different FP strategies have different characteristics. Considering the particularity of BRCA mutation carriers, multiple factors need to be carefully considered. This review focuses on the applicability of each FP method for carriers under various circumstances. Available FP strategies including oocyte cryopreservation, ovarian tissue cryopreservation, preimplantation genetic diagnosis, and egg/embryo donation are analyzed by comparing existing methods comprehensively. In the attempt to provide an up-to-date decision-making guidance. Conditions taking into consideration were the carrier's age, the risk of breast and ovarian metastasis, plans for oncotherapy, FP outcome, time available for FP intervention and accessibility.Overall, FP is necessary and safe for BRCA mutation carriers. Among all available FP methods, oocyte cryopreservation is the most reliable procedure; ovarian tissue cryopreservation is the only way for preserving both fertility and endocrine function, recommended for pre-pubertal carriers and when time is limited for oocyte stimulation. A clear framework provides frontline clinical practitioners a new thought and eventually benefit thousands of BRCA mutation carriers.
Collapse
Affiliation(s)
- Xiaofu Zhang
- Department of Clinical Medicine, Medical College of Soochow University, Ren Ai Road 199, Suzhou Industrial Park, Suzhou, 215123, China
| | - Jingxin Niu
- Department of Clinical Medicine, Medical College of Soochow University, Ren Ai Road 199, Suzhou Industrial Park, Suzhou, 215123, China
| | - Tuanjie Che
- Laboratory of Precision Medicine and Translational Medicine, Suzhou Hospital Affiliated to Nanjing Medical University, Suzhou Science and Technology Town Hospital, Suzhou, 215153, China
| | - Yibei Zhu
- Department of Immunology, Medical College of Soochow University, Ren Ai Road 199, Suzhou Industrial Park, Suzhou, 215123, China
| | - Hongtao Zhang
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
| | - Jing Qu
- Department of Cell Biology, Medical College of Soochow University, Ren Ai Road 199, Suzhou Industrial Park, Suzhou, 215123, China.
| |
Collapse
|
48
|
Lambertini M, Olympios N, Lequesne J, Calbrix C, Fontanilles M, Loeb A, Leheurteur M, Demeestere I, Di Fiore F, Perdrix A, Clatot F. 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. [PMID: 31355134 PMCID: PMC6640206 DOI: 10.3389/fonc.2019.00575] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 06/13/2019] [Indexed: 11/30/2022] Open
Abstract
Background: Limited evidence exists on the impact of adding a taxane, using endocrine therapy and carrying a deleterious germline BRCA mutation on ovarian reserve measured by anti-müllerian hormone (AMH) levels of young breast cancer patients receiving (neo)adjuvant cyclophosphamide- and anthracycline-based chemotherapy. Methods: This is a biomarker analysis including young (≤ 40 years) early breast cancer patients with known germline BRCA mutational status and available prospectively collected frozen plasma samples before and after chemotherapy. Chemotherapy consisted of either six cycles of FEC (5 fluorouracil 500 mg/m2, epirubicin 100 mg/m2, cyclophosphamide 500 mg/m2) or three cycles of FEC followed by three cycles of docetaxel (D, 100 mg/m2). Endocrine therapy consisted of tamoxifen (±GnRH agonists). AMH levels at baseline, 1 and 3 years after diagnosis were compared according to type of chemotherapy (FEC only vs. FEC-D), use of endocrine therapy (yes vs. no) and deleterious germline BRCA mutations (mutated vs. negative). Results: Out of 148 included patients, 127 (86%) received D following FEC chemotherapy, 90 (61%) underwent endocrine therapy, and 35 (24%) had deleterious germline BRCA mutations. In the whole cohort, AMH levels drastically dropped 1 year after diagnosis (p < 0.0001) with a slight but significant recovery at 3 years (p < 0.0001). One year after diagnosis, patients treated with FEC only had higher median AMH levels than those who received FEC-D (0.22 vs. 0.04 μg/L, p = 0.0006); no difference was observed at 3 years (0.06 and 0.18 μg/L, p = 0.47). Patients under endocrine therapy had significantly higher AMH levels than those who did not receive this treatment 1 year after diagnosis (0.12 vs. 0.02 μg/L; p = 0.008), with no difference at 3 years (0.11 and 0.20 μg/L, p = 0.22). AMH levels were similar between BRCA-mutated and BRCA-negative patients at baseline (1.94 vs. 1.66 μg/L, p = 0.53), 1 year (0.09 vs. 0.06 μg/L, p = 0.39) and 3 years (0.25 vs. 0.16 μg/L; p = 0.43) after diagnosis. Conclusions: In breast cancer patients receiving FEC chemotherapy, adding D appeared to negatively impact on their ovarian reserve in the short-term; no further detrimental effect was observed for endocrine therapy use and presence of a deleterious germline BRCA mutation.
Collapse
Affiliation(s)
- Matteo Lambertini
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy.,Department of Internal Medicine and Medical Specialties, School of Medicine, University of Genova, Genova, Italy
| | - Nathalie Olympios
- Department of Medical Oncology, Centre Henri Becquerel, Rouen, France
| | - Justine Lequesne
- Department of Clinical Research and Biostatistics, Centre Henri Becquerel, Rouen, France
| | - Céline Calbrix
- Department of Bio-Pathology, Centre Henri Becquerel, Rouen, France
| | - Maxime Fontanilles
- Department of Medical Oncology, Centre Henri Becquerel, Rouen, France.,IRON Group, UNIROUEN, Inserm U1245, Normandy Centre for Genomic and Personalized Medicine, Rouen University Hospital, Normandie University, Rouen, France
| | - Agnès Loeb
- Department of Medical Information, Henri Becquerel Centre, Rouen, France
| | | | - Isabelle Demeestere
- Research Laboratory on Human Reproduction, Fertility Clinic, CUB-Hôpital Erasme and Université Libre de Bruxelles, Brussels, Belgium
| | - Frédéric Di Fiore
- Department of Medical Oncology, Centre Henri Becquerel, Rouen, France.,IRON Group, UNIROUEN, Inserm U1245, Normandy Centre for Genomic and Personalized Medicine, Rouen University Hospital, Normandie University, Rouen, France
| | - Anne Perdrix
- Department of Bio-Pathology, Centre Henri Becquerel, Rouen, France.,IRON Group, UNIROUEN, Inserm U1245, Normandy Centre for Genomic and Personalized Medicine, Rouen University Hospital, Normandie University, Rouen, France
| | - Florian Clatot
- Department of Medical Oncology, Centre Henri Becquerel, Rouen, France.,IRON Group, UNIROUEN, Inserm U1245, Normandy Centre for Genomic and Personalized Medicine, Rouen University Hospital, Normandie University, Rouen, France
| |
Collapse
|
49
|
|
50
|
Lambertini M, Goldrat O, Ferreira AR, Dechene J, Azim HA, Desir J, Delbaere A, 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 2019; 29:237-243. [PMID: 29045555 DOI: 10.1093/annonc/mdx639] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Preclinical evidence suggests a possible negative impact of deleterious BRCA mutations on female fertility. However, limited and rather conflicting clinical data are available. This study assessed the reproductive potential and performance of fertility preservation strategies in BRCA-mutated breast cancer patients. Patients and methods This was a retrospective analysis of two prospective studies investigating oocyte cryopreservation and ovarian tissue cryopreservation in newly diagnosed early breast cancer patients. In the current analysis, baseline anti-Mullerian hormone (AMH) and performance of cryopreservation strategies were compared between patients with or without germline deleterious BRCA mutations. Results Out of 156 patients included, 101 had known BRCA status of whom 29 (18.6%) were BRCA-mutated and 72 (46.1%) had no mutation. Median age in the entire cohort was 31 years [interquartile range (IQR) 28-33). Median AMH levels were 1.8 μg/l (IQR 1.0-2.7) and 2.6 µg/l (IQR 1.5-4.1) in the BRCA-positive and BRCA-negative cohorts, respectively (P = 0.109). Among patients who underwent oocyte cryopreservation (N = 29), women in the BRCA-positive cohort tended to retrieve (6.5 versus 9; P = 0.145) and to cryopreserve (3.5 versus 6; P = 0.121) less oocytes than those in the BRCA-negative cohort. Poor response rate (i.e. retrieval of ≤4 oocytes) was 40.0% and 11.1% in the BRCA-positive and BRCA-negative cohorts, respectively (P = 0.147). Among patients who underwent ovarian tissue cryopreservation (N = 72), women in the BRCA-positive cohort tended to have a numerically lower number of oocytes per fragment (0.08 versus 0.14; P = 0.193) and per square millimeter (0.33 versus 0.78; P = 0.153) than those in the BRCA-negative cohort. Two BRCA-mutated patients were transplanted after chemotherapy and one delivered at term a healthy baby. No difference between BRCA1- and BRCA2-mutated patients was observed in any of the above-mentioned outcomes. Conclusion A consistent trend for reduced reproductive potential and performance of cryopreservation strategies was observed in BRCA-mutated breast cancer patients. Independent validation of these results is needed.
Collapse
Affiliation(s)
- M Lambertini
- Department of Medicine, Institut Jules Bordet and Université Libre de Bruxelles (U.L.B.), Brussels, Belgium.,Breast Cancer Translational Research Laboratory, Institut Jules Bordet and Université Libre de Bruxelles (U.L.B.), Brussels, Belgium
| | - O Goldrat
- Fertility Clinic, CUB-Hôpital Erasme and Research Laboratory on Human Reproduction, Université Libre de Bruxelles (U.L.B.), Brussels, Belgium
| | - A R Ferreira
- Hospital de Santa Maria and Instituto de Medicina Molecular, Faculdade de Medicina, Universiade de Lisboa, Lisbon, Portugal
| | - J Dechene
- Fertility Clinic, CUB-Hôpital Erasme and Research Laboratory on Human Reproduction, Université Libre de Bruxelles (U.L.B.), Brussels, Belgium
| | - H A Azim
- Department of Internal Medicine, American University of Beirut (AUB), Beirut, Lebanon
| | - J Desir
- Medical Genetics Department, CUB-Hôpital Erasme, Université Libre de Bruxelles (U.L.B.), Brussels, Belgium
| | - A Delbaere
- Fertility Clinic, CUB-Hôpital Erasme and Research Laboratory on Human Reproduction, Université Libre de Bruxelles (U.L.B.), Brussels, Belgium
| | - M-D t'Kint de Roodenbeke
- Department of Medicine, Institut Jules Bordet and Université Libre de Bruxelles (U.L.B.), Brussels, Belgium
| | - E de Azambuja
- Department of Medicine, Institut Jules Bordet and Université Libre de Bruxelles (U.L.B.), Brussels, Belgium
| | - M Ignatiadis
- Department of Medicine, Institut Jules Bordet and Université Libre de Bruxelles (U.L.B.), Brussels, Belgium
| | - I Demeestere
- Fertility Clinic, CUB-Hôpital Erasme and Research Laboratory on Human Reproduction, Université Libre de Bruxelles (U.L.B.), Brussels, Belgium
| |
Collapse
|