145
Views
0
CrossRef citations to date
0
Altmetric
Review

Current practices in oncofertility counseling: updated evidence on fertility preservation and post-treatment pregnancies in young women affected by early breast cancer

ORCID Icon, ORCID Icon, , , , , , , ORCID Icon, ORCID Icon, ORCID Icon & ORCID Icon show all
Received 04 Mar 2024, Accepted 21 Jun 2024, Published online: 09 Jul 2024

References

  • Siegel RL, Giaquinto AN, Jemal A. Cancer statistics, 2024. CA Cancer J Clin. 2024;74(1):12–49. doi: 10.3322/caac.21820
  • Vaz-Luis I, Masiero M, Cavaletti G, et al. ESMO expert consensus statements on cancer survivorship: promoting high-quality survivorship care and research in Europe. Ann Oncol Off J Eur Soc Med Oncol. 2022;33(11):1119–1133. doi: 10.1016/j.annonc.2022.07.1941
  • Soldato D, Arecco L, Agostinetto E, et al. The future of breast cancer research in the survivorship field. Oncol Ther. 2023;11(2):199–229. doi: 10.1007/s40487-023-00225-8
  • Loibl S, André F, Bachelot T, et al. Early breast cancer: ESMO clinical practice guideline for diagnosis, treatment and follow-up†. Ann Oncol Off J Eur Soc Med Oncol. 2023 Dec 8:S0923–7534(23)05104–9. doi: 10.1016/j.annonc.2023.11.016
  • Denduluri N, Somerfield MR, Chavez-MacGregor M, et al. Selection of optimal adjuvant chemotherapy and targeted therapy for early breast cancer: ASCO guideline update. J Clin Oncol Off J Am Soc Clin Oncol. 2021;39(6):685–693. doi: 10.1200/JCO.20.02510
  • Anderson RA, Amant F, Braat D, et al.; ESHRE Guideline Group on Female Fertility Preservation ESHRE guideline: female fertility preservation†. Human Reproduction Open. 2020;2020(4):hoaa052. doi: 10.1093/hropen/hoaa052
  • Lambertini M, Arecco L, Woodard TL, et al. Advances in the management of menopausal symptoms, fertility preservation, and bone health for women with breast cancer on endocrine therapy. Am Soc Clin Oncol Educ Book Am Soc Clin Oncol Annu Meet. 2023;43(43):e390442. doi: 10.1200/EDBK_390442
  • Ruddy KJ, Gelber SI, Tamimi RM, et al. Prospective study of fertility concerns and preservation strategies in young women with breast cancer. J Clin Oncol. 2014;32(11):1151–1156. doi: 10.1200/JCO.2013.52.8877
  • Blondeaux E, Massarotti C, Fontana V, et al. The PREgnancy and FERtility (PREFER) study investigating the need for ovarian function and/or fertility preservation strategies in premenopausal women with early breast cancer. Front Oncol. 2021;11:690320. doi: 10.3389/fonc.2021.690320
  • Zaami S, Melcarne R, Patrone R, et al. Oncofertility and reproductive counseling in patients with breast cancer: a retrospective study. J Clin Med. 2022;11(5):1311. doi: 10.3390/jcm11051311
  • Khan SZ, Arecco L, Villarreal-Garza C, et al. Knowledge, practice, and attitudes of physicians in low- and middle-income countries on fertility and pregnancy-related issues in young women with breast cancer. JCO Glob Oncol. 2022;8(8):e2100153. doi: 10.1200/GO.21.00153
  • Lambertini M, Blondeaux E, Bruzzone M, et al. Pregnancy after breast cancer: a systematic review and meta-analysis. J Clin Oncol Off J Am Soc Clin Oncol. 2021;39(29):3293–3305. doi: 10.1200/JCO.21.00535
  • Lambertini M, Peccatori FA, Demeestere I, et al. Fertility preservation and post-treatment pregnancies in post-pubertal cancer patients: ESMO clinical practice guidelines†. Ann Oncol. 2020;31(12):1664–1678. doi: 10.1016/j.annonc.2020.09.006
  • Paluch-Shimon S, Cardoso F, Partridge AH, et al. ESO-ESMO fifth international consensus guidelines for breast cancer in young women (BCY5). Ann Oncol Off J Eur Soc Med Oncol. 2022;33(11):1097–1118. doi: 10.1016/j.annonc.2022.07.007
  • Vaz-Luis I, Francis PA, Di Meglio A, et al. Challenges in adjuvant therapy for premenopausal women diagnosed with luminal breast cancers. Am Soc Clin Oncol Educ Book. 2021;41:e47–e61. doi: 10.1200/EDBK_320595
  • Chon SJ, Umair Z, Yoon MS. Premature ovarian insufficiency: past, present, and future. Front Cell Dev Biol. 2021;9:672890. doi: 10.3389/fcell.2021.672890
  • Anderson RA, Cameron D, Clatot F, et al. Anti-Müllerian hormone as a marker of ovarian reserve and premature ovarian insufficiency in children and women with cancer: a systematic review. Hum Reprod Update. 2022;28(3):417–434. doi: 10.1093/humupd/dmac004
  • Codacci-Pisanelli G, Del Pup L, Del Grande M, et al. Mechanisms of chemotherapy-induced ovarian damage in breast cancer patients. Crit Rev Oncol Hematol. 2017;113:90–96. doi: 10.1016/j.critrevonc.2017.03.009
  • Razeti MG, Spinaci S, Spagnolo F, et al. How I perform fertility preservation in breast cancer patients. ESMO Open. 2021;6(3):100112. doi: 10.1016/j.esmoop.2021.100112
  • Zhao J, Liu J, Chen K, et al. What lies behind chemotherapy-induced amenorrhea for breast cancer patients: a meta-analysis. Breast Cancer Res Treat. 2014;145(1):113–128. doi: 10.1007/s10549-014-2914-x
  • Silva C, Caramelo O, Almeida-Santos T, et al. Factors associated with ovarian function recovery after chemotherapy for breast cancer: a systematic review and meta-analysis. Hum Reprod. 2016;31(12):2737–2749. doi: 10.1093/humrep/dew224
  • Lambertini M, Olympios N, Lequesne J, et al. Impact of taxanes, endocrine therapy, and deleterious germline BRCA mutations on anti-müllerian hormone levels in early breast cancer patients treated with anthracycline- and cyclophosphamide-based chemotherapy. Front Oncol. 2019;9:575. doi: 10.3389/fonc.2019.00575
  • Poggio F, Bruzzone M, Ceppi M, et al. Platinum-based neoadjuvant chemotherapy in triple-negative breast cancer: a systematic review and meta-analysis. Ann Oncol. 2018;29(7):1497–1508. doi: 10.1093/annonc/mdy127
  • Poggio F, Tagliamento M, Ceppi M, et al. Adding a platinum agent to neoadjuvant chemotherapy for triple-negative breast cancer: the end of the debate. Ann Oncol Off J Eur Soc Med Oncol. 2022;33(3):347–349. doi: 10.1016/j.annonc.2021.11.016
  • Martelli V, Latocca MM, Ruelle T, et al. Comparing the gonadotoxicity of multiple breast cancer regimens: important understanding for managing breast cancer in pre-menopausal women. Breast Cancer Targets Ther. 2021;13:341–351. doi: 10.2147/BCTT.S274283
  • Kabirian R, Franzoi MA, Havas J, et al. Chemotherapy-related amenorrhea and quality of life among premenopausal women with breast cancer. JAMA Netw Open. 2023;6(11):e2343910. doi: 10.1001/jamanetworkopen.2023.43910
  • Swain SM, Shastry M, Hamilton E. Targeting HER2-positive breast cancer: advances and future directions. Nat Rev Drug Discov. 2023;22(2):101–126. doi: 10.1038/s41573-022-00579-0
  • Rastogi P, O’Shaughnessy J, Martin M, et al. Adjuvant abemaciclib plus endocrine therapy for hormone receptor–positive, human epidermal growth factor receptor 2–negative, high-risk early breast cancer: results from a preplanned monarche overall survival interim analysis, including 5-year efficacy outcomes. J Clin Oncol. 2024 Jan 9:JCO.23.01994. doi: 10.1200/JCO.23.01994
  • Slamon D, Lipatov O, Nowecki Z, et al. Ribociclib plus Endocrine Therapy in Early Breast Cancer. N Engl J Med. 2024 Mar 21;390(12):1080–1091. doi: 10.1056/NEJMoa2305488
  • Schmid P, Cortes J, Dent R, et al. Event-free survival with pembrolizumab in Early Triple-negative breast cancer. N Engl J Med. 2022;386(6):556–567. doi: 10.1056/NEJMoa2112651
  • Geyer CE, Garber JE, Gelber RD, et al. Overall survival in the OlympiA phase III trial of adjuvant olaparib in patients with germline pathogenic variants in BRCA1/2 and high-risk, early breast cancer. Ann Oncol. 2022;33(12):1250–1268. doi: 10.1016/j.annonc.2022.09.159
  • Lambertini M, Marrocco C, Spinaci S, et al. Risk of gonadotoxicity with immunotherapy and targeted agents remains an unsolved but crucial issue. Eur J Clin Invest. 2022;52(7):e13779. doi: 10.1111/eci.13779
  • Lambertini M, Campbell C, Bines J, et al. Adjuvant anti-HER2 therapy, treatment-related amenorrhea, and survival in premenopausal HER2-positive early breast cancer patients. JNCI J Natl Cancer Inst. 2019;111(1):86–94. doi: 10.1093/jnci/djy094
  • Ruddy KJ, Zheng Y, Tayob N, et al. Chemotherapy-related amenorrhea (CRA) after adjuvant ado-trastuzumab emtansine (T-DM1) compared to paclitaxel in combination with trastuzumab (TH) (TBCRC033: ATEMPT Trial). Breast Cancer Res Treat. 2021 Jun 12;189(1):103–110. doi: 10.1007/s10549-021-06267-8
  • Lambertini M, Ceppi M, Anderson RA, et al. Impact of Anti-HER2 therapy alone and with weekly paclitaxel on the ovarian reserve of young women with HER2-positive breast cancer. J Natl Compr Cancer Netw JNCCN. 2023;21(1):33–41.e16. doi: 10.6004/jnccn.2022.7065
  • von Minckwitz G, Huang CS, Mano MS, et al. Trastuzumab emtansine for residual invasive HER2-positive breast cancer. N Engl J Med. 2019;380(7):617–628. doi: 10.1056/NEJMoa1814017
  • Hurvitz SA, Hegg R, Chung WP, et al. Trastuzumab deruxtecan versus trastuzumab emtansine in patients with HER2-positive metastatic breast cancer: updated results from DESTINY-Breast03, a randomised, open-label, phase 3 trial. Lancet. 2023;401(10371):105–117. doi: 10.1016/S0140-6736(22)02420-5
  • Modi S, Jacot W, Yamashita T, et al. Trastuzumab deruxtecan in Previously treated HER2-low advanced breast cancer. N Engl J Med. 2022;387(1):9–20. doi: 10.1056/NEJMoa2203690
  • Mayer EL, Dueck AC, Martin M, et al. Palbociclib with adjuvant endocrine therapy in early breast cancer (PALLAS): interim analysis of a multicentre, open-label, randomised, phase 3 study. Lancet Oncol. 2021;22(2):212–222. doi: 10.1016/S1470-2045(20)30642-2
  • Loibl S, Marmé F, Martin M, et al. Palbociclib for residual high-risk invasive HR-Positive and HER2-negative early breast cancer-the penelope-B Trial. J Clin Oncol Off J Am Soc Clin Oncol. 2021;39(14):1518–1530. doi: 10.1200/JCO.20.03639
  • Research C for DE and. FDA D.I.S.C.O.; Burst Edition: FDA approval of Verzenio (abemaciclib) with endocrine therapy for patients with HR-positive, HER2-negative, node-positive, early breast cancer. FDA. [UpdatedUpdated 2023 Mar 24; cited 2023 Mar 24]. Available from: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-disco-burst-edition-fda-approval-verzenio-abemaciclib-endocrine-therapy-patients-hr-positive
  • Gao X, Leone GW, Wang H. Cyclin D-CDK4/6 functions in cancer. Adv Cancer Res. 2020;148:147–169. doi: 10.1016/bs.acr.2020.02.002
  • Scavone G, Ottonello S, Blondeaux E, et al. The Role of cyclin-dependent kinases (CDK) 4/6 in the ovarian tissue and the possible effects of their exogenous inhibition. Cancers (Basel). 2023;15(20):4923. doi: 10.3390/cancers15204923
  • Sheffield KM, Peachey JR, Method M, et al. A real-world US study of recurrence risks using combined clinicopathological features in HR-positive, HER2-negative early breast cancer. Future Oncol Lond Engl. 2022;18(21):2667–2682. doi: 10.2217/fon-2022-0310
  • Dannehl D, Volmer LL, Weiss M, et al. Feasibility of adjuvant treatment with abemaciclib-real-world data from a large German breast center. J Pers Med. 2022;12(3):382. doi: 10.3390/jpm12030382
  • Schäffler H, Mergel F, Pfister K, et al. The clinical relevance of the NATALEE study: application of the NATALEE criteria to a real-world cohort from two large German breast cancer centers. Int J Mol Sci. 2023;24(22):16366. doi: 10.3390/ijms242216366
  • Catlin NR, Bowman CJ, Engel SM, et al. Reproductive and developmental toxicity assessment of palbociclib, a CDK4/6 inhibitor, in Sprague-Dawley rats and New Zealand White rabbits. Reprod Toxicol Elmsford N. 2019;88:76–84. doi: 10.1016/j.reprotox.2019.07.016
  • Furlanetto J, Marmé F, Thode C, et al. 60MO Ovarian function in young patients (pts) treated with postneoadjuvant palbociclib (PAL) and endocrine therapy (ET) for hormone receptor (HR)-positive, HER2-negative early breast cancer (BC): Explorative analysis in Penelope-B. Ann Oncol. 2022;33:S149–S150. doi: 10.1016/j.annonc.2022.03.076
  • Robson ME, Tung N, Conte P, et al. OlympiAD final overall survival and tolerability results: olaparib versus chemotherapy treatment of physician’s choice in patients with a germline BRCA mutation and HER2-negative metastatic breast cancer. Ann Oncol. 2019;30(4):558–566. doi: 10.1093/annonc/mdz012
  • Litton JK, Hurvitz SA, Mina LA, et al. Talazoparib versus chemotherapy in patients with germline BRCA1/2-mutated HER2-negative advanced breast cancer: final overall survival results from the EMBRACA trial. Ann Oncol Off J Eur Soc Med Oncol. 2020;31(11):1526–1535. doi: 10.1016/j.annonc.2020.08.2098
  • Winship AL, Griffiths M, Lliberos Requesens C, et al. The PARP inhibitor, olaparib, depletes the ovarian reserve in mice: implications for fertility preservation. Hum Reprod. 2020;35(8):1864–1874. doi: 10.1093/humrep/deaa128
  • Mittendorf EA, Zhang H, Barrios CH, et al. Neoadjuvant atezolizumab in combination with sequential nab-paclitaxel and anthracycline-based chemotherapy versus placebo and chemotherapy in patients with early-stage triple-negative breast cancer (IMpassion031): a randomised, double-blind, phase 3 trial. Lancet. 2020;396(10257):1090–1100. doi: 10.1016/S0140-6736(20)31953-X
  • Cardoso F, McArthur HL, Schmid P, et al. LBA21 KEYNOTE-756: phase III study of neoadjuvant pembrolizumab (pembro) or placebo (pbo) + chemotherapy (chemo), followed by adjuvant pembro or pbo + endocrine therapy (ET) for early-stage high-risk ER+/HER2– breast cancer. Ann Oncol. 2023;34:S1260–S1261. doi: 10.1016/j.annonc.2023.10.011
  • Loi S, McArthur HL, Harbeck N, et al. A phase III trial of nivolumab with neoadjuvant chemotherapy and adjuvant endocrine therapy in ER+/HER2- primary breast cancer: checkMate 7FL. J Clin Oncol. 2020;38(15_suppl):TPS604–TPS604. doi: 10.1200/JCO.2020.38.15_suppl.TPS604
  • Winship AL, Alesi LR, Sant S, et al. Checkpoint inhibitor immunotherapy diminishes oocyte number and quality in mice. Nat Cancer. 2022;3(8):1–13. doi: 10.1038/s43018-022-00413-x
  • Garutti M, Lambertini M, Puglisi F. Checkpoint inhibitors, fertility, pregnancy, and sexual life: a systematic review. ESMO Open. 2021;6(5):100276. doi: 10.1016/j.esmoop.2021.100276
  • Boots CE, Meister M, Cooper AR, et al. Ovarian stimulation in the luteal phase: systematic review and meta-analysis. J Assist Reprod Genet. 2016;33(8):971–980. doi: 10.1007/s10815-016-0721-5
  • Orvieto R. Triggering final follicular maturation- hCG, GnRH-agonist or both, when and to whom? J Ovarian Res. 2015;8(1):60. doi: 10.1186/s13048-015-0187-6
  • Cobo A, García-Velasco JA, Coello A, et al. Oocyte vitrification as an efficient option for elective fertility preservation. Fertil Steril. 2016;105(3):755–764.e8. doi: 10.1016/j.fertnstert.2015.11.027
  • Sessa C, Balmaña J, Bober SL, et al. Risk reduction and screening of cancer in hereditary breast-ovarian cancer syndromes: ESMO clinical practice guideline. Ann Oncol Off J Eur Soc Med Oncol. 2023;34(1):33–47. doi: 10.1016/j.annonc.2022.10.004
  • Azim AA, Costantini-Ferrando M, Oktay K. Safety of fertility preservation by ovarian stimulation with letrozole and gonadotropins in patients with breast cancer: a prospective controlled study. J Clin Oncol. 2008;26(16):2630–2635. doi: 10.1200/JCO.2007.14.8700
  • Bonardi B, Massarotti C, Bruzzone M, et al. 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. doi: 10.3389/fonc.2020.574669
  • Meirow D, Raanani H, Maman E, et al. Tamoxifen co-administration during controlled ovarian hyperstimulation for in vitro fertilization in breast cancer patients increases the safety of fertility-preservation treatment strategies. Fertil Steril. 2014;102(2):488–495.e3. doi: 10.1016/j.fertnstert.2014.05.017
  • Yoshida T, Takahashi O, Suzuki Y, et al. The effectiveness of controlled ovarian stimulation with tamoxifen for patients with estrogen‐sensitive breast cancer: a systematic review and meta‐analysis. Reprod Med Biol. 2023;22(1):e12543. doi: 10.1002/rmb2.12543
  • Arecco L, Blondeaux E, Bruzzone M, et al. Safety of fertility preservation techniques before and after anticancer treatments in young women with breast cancer: a systematic review and meta-analysis. Hum Reprod Oxf Eng. 2022;37(5):954–968. doi: 10.1093/humrep/deac035
  • Benvenuti C, Laot L, Grinda T, et al. Is controlled ovarian stimulation safe in patients with hormone receptor-positive breast cancer receiving neoadjuvant chemotherapy? ESMO Open. 2024;9(2):102228. doi: 10.1016/j.esmoop.2023.102228
  • İsrafilova G, Şükür YE, Özkavukcu S, et al. Comparison of oocyte and embryo quality between random start and controlled ovarian stimulation cycles in cancer patients undergoing fertility preservation. Reprod Sci Thousand Oaks Calif. 2021;28(8):2200–2207. doi: 10.1007/s43032-020-00412-2
  • Cakmak H, Katz A, Cedars MI, et al. Effective method for emergency fertility preservation: random-start controlled ovarian stimulation. Fertil Steril. 2013;100(6):1673–1680. doi: 10.1016/j.fertnstert.2013.07.1992
  • Cobo A, García-Velasco J, Domingo J, et al. Elective and Onco-fertility preservation: factors related to IVF outcomes. Hum Reprod Oxf Eng. 2018;33(12):2222–2231. doi: 10.1093/humrep/dey321
  • Cardozo ER, Thomson AP, Karmon AE, et al. Ovarian stimulation and in-vitro fertilization outcomes of cancer patients undergoing fertility preservation compared to age matched controls: a 17-year experience. J Assist Reprod Genet. 2015;32(4):587–596. doi: 10.1007/s10815-015-0428-z
  • Khattak H, Malhas R, Craciunas L, et al. Fresh and cryopreserved ovarian tissue transplantation for preserving reproductive and endocrine function: a systematic review and individual patient data meta-analysis. Hum Reprod Update. 2022;28(3):400–416. doi: 10.1093/humupd/dmac003
  • Dueholm Hjorth IM, Kristensen SG, Dueholm M, et al. Reproductive outcomes after in vitro fertilization treatment in a cohort of Danish women transplanted with cryopreserved ovarian tissue. Fertil Steril. 2020;114(2):379–387. doi: 10.1016/j.fertnstert.2020.03.035
  • Fleury A, Pirrello O, Maugard C, et al. Breast cancer and ovarian tissue cryopreservation: review of the literature. J Gynecol Obstet Hum Reprod. 2018;47(8):351–357. doi: 10.1016/j.jogoh.2018.05.008
  • Buonomo B, Massarotti C, Dellino M, et al. Reproductive issues in carriers of germline pathogenic variants in the BRCA1/2 genes: an expert meeting. BMC Med. 2021;19(1):205. doi: 10.1186/s12916-021-02081-7
  • Arecco L, Ruelle T, Martelli V, et al. How to Protect Ovarian Function before and during chemotherapy? J Clin Med. 2021;10(18):4192. doi: 10.3390/jcm10184192
  • Conn PM, Crowley WF. Gonadotropin-releasing hormone and its analogues. N Engl J Med. 1991;324(2):93–103. doi: 10.1056/NEJM199101103240205
  • Blumenfeld Z. Fertility preservation using gnrh agonists: rationale, possible mechanisms, and explanation of controversy. Clin Med Insights Reprod Health. 2019;13:117955811987016. doi: 10.1177/1179558119870163
  • Scaruffi P, Stigliani S, Cardinali C, et al. Gonadotropin releasing hormone agonists have an anti-apoptotic effect on cumulus cells. Int J Mol Sci. 2019;20(23):6045. doi: 10.3390/ijms20236045
  • Del Mastro L, Boni L, Michelotti A, et al. Effect of the gonadotropin-releasing hormone analogue triptorelin on the occurrence of chemotherapy-induced early menopause in premenopausal women with breast cancer: a randomized trial. JAMA. 2011;306(3):269–276. doi: 10.1001/jama.2011.991
  • Gerber B, von Minckwitz G, Stehle H, et al. Effect of luteinizing hormone-releasing hormone agonist on ovarian function after modern adjuvant breast cancer chemotherapy: the GBG 37 ZORO study. J Clin Oncol Off J Am Soc Clin Oncol. 2011;29(17):2334–2341. doi: 10.1200/JCO.2010.32.5704
  • Munster PN, Moore AP, Ismail-Khan R, et al. Randomized trial using gonadotropin-releasing hormone agonist triptorelin for the preservation of ovarian function during (Neo)adjuvant chemotherapy for breast cancer. J Clin Oncol. 2012;30(5):533–538. doi: 10.1200/JCO.2011.34.6890
  • Moore HCF, Unger JM, Phillips KA, et al. Goserelin for ovarian protection during breast-cancer adjuvant chemotherapy. N Engl J Med. 2015;372(10):923–932. doi: 10.1056/NEJMoa1413204
  • Leonard RCF, Adamson DJA, Bertelli G, et al. GnRH agonist for protection against ovarian toxicity during chemotherapy for early breast cancer: the anglo celtic group OPTION trial. Ann Oncol. 2017;28(8):1811–1816. doi: 10.1093/annonc/mdx184
  • Lambertini M, Ceppi M, Poggio F, et al. Ovarian suppression using luteinizing hormone-releasing hormone agonists during chemotherapy to preserve ovarian function and fertility of breast cancer patients: a meta-analysis of randomized studies. Ann Oncol. 2015;26(12):2408–2419. doi: 10.1093/annonc/mdv374
  • Lambertini M, Moore HCF, Leonard RCF, et al. Gonadotropin-releasing hormone agonists during chemotherapy for preservation of ovarian function and fertility in premenopausal patients with early breast cancer: a systematic review and meta-analysis of individual patient–level data. J Clin Oncol. 2018;36(19):1981–1990. doi: 10.1200/JCO.2018.78.0858
  • Oktay K, Harvey BE, Partridge AH, et al. Fertility preservation in patients with cancer: ASCO clinical practice guideline update. J Clin Oncol. 2018;36(19):1994–2001. doi: 10.1200/JCO.2018.78.1914
  • Lambertini M, Boni L, Michelotti A, et al. Long-term outcomes with pharmacological ovarian suppression during chemotherapy in premenopausal early breast cancer patients. J Natl Cancer Inst. 2022;114(3):400–408. doi: 10.1093/jnci/djab213
  • Regan MM, Walley BA, Francis PA, et al. Concurrent and sequential initiation of ovarian function suppression with chemotherapy in premenopausal women with endocrine-responsive early breast cancer: an exploratory analysis of TEXT and SOFT. Ann Oncol. 2017;28(9):2225–2232. doi: 10.1093/annonc/mdx285
  • Zong X, Yu Y, Yang H, et al. Effects of Gonadotropin-releasing hormone analogs on ovarian function against chemotherapy-induced gonadotoxic effects in premenopausal women with breast cancer in china: a randomized clinical trial. JAMA Oncol. 2022;8(2):252–258. doi: 10.1001/jamaoncol.2021.6214
  • Lambertini M, Goldrat O, Toss A, et al. Fertility and pregnancy issues in BRCA -mutated breast cancer patients. Cancer Treat Rev. 2017;59:61–70. doi: 10.1016/j.ctrv.2017.07.001
  • Turan V, Oktay K. BRCA-related ATM-mediated DNA double-strand break repair and ovarian aging. Hum Reprod Update. 2020;26(1):43–57. doi: 10.1093/humupd/dmz043
  • Oktay K, Turan V, Titus S, et al. BRCA Mutations, DNA repair deficiency, and ovarian aging. Biol Reprod. 2015;93(3):67. doi: 10.1095/biolreprod.115.132290
  • Son KA, Lee DY, Choi D. Association of BRCA mutations and anti-müllerian hormone level in young breast cancer patients. Front Endocrinol. 2019;10:235. doi: 10.3389/fendo.2019.00235
  • Oktay KH, Bedoschi G, Goldfarb SB, et al. Increased chemotherapy-induced ovarian reserve loss in women with germline BRCA mutations due to oocyte deoxyribonucleic acid double strand break repair deficiency. Fertil Steril. 2020;113(6):1251–1260.e1. doi: 10.1016/j.fertnstert.2020.01.033
  • Porcu E, Cillo GM, Cipriani L, et al. Impact of BRCA1 and BRCA2 mutations on ovarian reserve and fertility preservation outcomes in young women with breast cancer. J Assist Reprod Genet. 2020;37(3):709–715. doi: 10.1007/s10815-019-01658-9
  • Daum H, Peretz T, Laufer N. BRCA mutations and reproduction. Fertil Steril. 2018;109(1):33–38. doi: 10.1016/j.fertnstert.2017.12.004
  • Oktay KH, Turan V, Bedoschi G, et al. A prospective longitudinal analysis of the predictors of amenorrhea after breast cancer chemotherapy: impact of BRCA pathogenic variants. Cancer Med. 2023;12(18):19225–19233. doi: 10.1002/cam4.6527
  • Kuchenbaecker KB, Hopper JL, Barnes DR, et al. Risks of breast, ovarian, and contralateral breast cancer for BRCA1 and BRCA2 Mutation Carriers. JAMA. 2017;317(23):2402–2416. doi: 10.1001/jama.2017.7112
  • Kim J, Turan V, Oktay K. Long-term safety of letrozole and gonadotropin stimulation for fertility preservation in women with breast cancer. J Clin Endocrinol Metab. 2016;101(4):1364–1371. doi: 10.1210/jc.2015-3878
  • Greer AC, Lanes A, Poorvu PD, et al. The impact of fertility preservation on the timing of breast cancer treatment, recurrence, and survival. Cancer. 2021 Jun 23;cncr.33601. doi: 10.1002/cncr.33601
  • Kim SW, Kim TH, Han JY, et al. Impact of BRCA mutations and hormone receptor status on reproductive potential in breast cancer patients undergoing fertility preservation. Gynecol Endocrinol Off J Int Soc Gynecol Endocrinol. 2022;38(3):227–230. doi: 10.1080/09513590.2021.2002294
  • Lambertini M, Goldrat O, Ferreira AR, et al. Reproductive potential and performance of fertility preservation strategies in BRCA-mutated breast cancer patients. Ann Oncol. 2018;29(1):237–243. doi: 10.1093/annonc/mdx639
  • Turan V, Bedoschi G, Emirdar V, et al. Ovarian stimulation in patients with cancer: impact of letrozole and BRCA mutations on fertility preservation cycle outcomes. Reprod Sci Thousand Oaks Calif. 2018;25(1):26–32. doi: 10.1177/1933719117728800
  • El Moujahed L, Philis R, Grynberg M, et al. Response to ovarian stimulation for urgent fertility preservation before gonadotoxic treatment in BRCA-pathogenic-variant-positive breast cancer patients. Cancers (Basel). 2023;15(3):895. doi: 10.3390/cancers15030895
  • Shapira M, Raanani H, Feldman B, et al. BRCA mutation carriers show normal ovarian response in in vitro fertilization cycles. Fertil Steril. 2015;104(5):1162–1167. doi: 10.1016/j.fertnstert.2015.07.1162
  • Gunnala V, Fields J, Irani M, et al. BRCA carriers have similar reproductive potential at baseline to noncarriers: comparisons in cancer and cancer-free cohorts undergoing fertility preservation. Fertil Steril. 2019;111(2):363–371. doi: 10.1016/j.fertnstert.2018.10.014
  • Grynberg M, Dagher Hayeck B, Papanikolaou EG, et al. BRCA1/2 gene mutations do not affect the capacity of oocytes from breast cancer candidates for fertility preservation to mature in vitro. Hum Reprod Oxf Eng. 2019;34(2):374–379. doi: 10.1093/humrep/dey358
  • Vuković P, Peccatori FA, Massarotti C, et al. Preimplantation genetic testing for carriers of BRCA1/2 pathogenic variants. Crit Rev Oncol Hematol. 2021;157:103201. doi: 10.1016/j.critrevonc.2020.103201
  • Shenfield F, Pennings G, Devroey P, et al. Taskforce 5: preimplantation genetic diagnosis. Hum Reprod Oxf Eng. 2003;18(3):649–651. doi: 10.1093/humrep/deg110
  • Jensen AK, Macklon KT, Fedder J, et al. 86 successful births and 9 ongoing pregnancies worldwide in women transplanted with frozen-thawed ovarian tissue: focus on birth and perinatal outcome in 40 of these children. J Assist Reprod Genet. 2017;34(3):325–336. doi: 10.1007/s10815-016-0843-9
  • Lambertini M, Blondeaux E, Bruzzone M, et al. Pregnancy after breast cancer: a systematic review and meta-analysis. J Clin Oncol. 2021 Jul 1:JCO.21.00535. doi: 10.1200/JCO.21.00535
  • Azim HA, Metzger-Filho O, de Azambuja E, et al. Pregnancy occurring during or following adjuvant trastuzumab in patients enrolled in the HERA trial (BIG 01-01). Breast Cancer Res Treat. 2012;133(1):387–391. doi: 10.1007/s10549-012-1996-6
  • Lambertini M, Martel S, Campbell C, et al. Pregnancies during and after trastuzumab and/or lapatinib in patients with human epidermal growth factor receptor 2-positive early breast cancer: Analysis from the NeoALTTO (BIG 1-06) and ALTTO (BIG 2-06) trials: pregnancies in women with HER2+ Breast Cancer. Cancer. 2019;125(2):307–316. doi: 10.1002/cncr.31784
  • Arecco L, Blondeaux E, Bruzzone M, et al. Safety of pregnancy after breast cancer in young women with hormone receptor-positive disease: a systematic review and meta-analysis. ESMO Open. 2023;8(6):102031. doi: 10.1016/j.esmoop.2023.102031
  • Partridge AH, Niman SM, Ruggeri M, et al. Interrupting endocrine therapy to attempt pregnancy after breast cancer. N Engl J Med. 2023;388(18):1645–1656. doi: 10.1056/NEJMoa2212856
  • Arecco L, Lambertini M. Safety of interrupting adjuvant endocrine therapy to conceive: early data are POSITIVE. Nat Rev Clin Oncol. 2023 Jul 6;20(10):662–663. doi: 10.1038/s41571-023-00797-4
  • Valentini A, Lubinski J, Byrski T, et al. The impact of pregnancy on breast cancer survival in women who carry a BRCA1 or BRCA2 mutation. Breast Cancer Res Treat. 2013;142(1):177–185. doi: 10.1007/s10549-013-2729-1
  • Lambertini M, Ameye L, Hamy AS, et al. Pregnancy after breast cancer in patients with germline BRCA mutations. J Clin Oncol Off J Am Soc Clin Oncol. 2020;38(26):3012–3023. doi: 10.1200/JCO.19.02399
  • Lambertini M, Blondeaux E, Agostinetto E, et al. Pregnancy after breast cancer in young BRCA carriers: an international hospital-based cohort study. JAMA. 2024;331(1):49–59. doi: 10.1001/jama.2023.25463
  • Razeti MG, Soldato D, Arecco L, et al. Approaches to fertility preservation for young women with breast cancer. Clin Breast Cancer. 2023;23(3):241–248. doi: 10.1016/j.clbc.2023.01.006
  • Zaami S, Montanari Vergallo G, Moscatelli M, et al. Oncofertility: the importance of counseling for fertility preservation in cancer patients. Eur Rev Med Pharmacol Sci. 2021;25(22):6874–6880. doi: 10.26355/eurrev_202111_27235
  • Jones G, Hughes J, Mahmoodi N, et al. What factors hinder the decision-making process for women with cancer and contemplating fertility preservation treatment? Hum Reprod Update. 2017;23(4):433–457. doi: 10.1093/humupd/dmx009
  • Zaami S, Stark M, Signore F, et al. Fertility preservation in female cancer sufferers: (only) a moral obligation? Eur J Contracept Reprod Health Care Off J Eur Soc Contracept. 2022;27(4):335–340. doi: 10.1080/13625187.2022.2045936
  • Lambertini M, Goldrat O, Clatot F, et al. Controversies about fertility and pregnancy issues in young breast cancer patients: current state of the art. Curr Opin Oncol. 2017;29(4):243–252. doi: 10.1097/CCO.0000000000000380
  • Massarotti C, Scaruffi P, Lambertini M, et al. Beyond fertility preservation: role of the oncofertility unit in the reproductive and gynecological follow-up of young cancer patients. Hum Reprod. 2019;34(8):1462–1469. doi: 10.1093/humrep/dez108
  • Logan S, Anazodo A. The psychological importance of fertility preservation counseling and support for cancer patients. Acta Obstet Gynecol Scand. 2019;98(5):583–597. doi: 10.1111/aogs.13562
  • Lambertini M, Massarotti C, Havas J, et al. Contraceptive use in premenopausal women with early breast cancer. JAMA Netw Open. 2022;5(9):e2233137. doi: 10.1001/jamanetworkopen.2022.33137
  • Bosch E, Broer S, Griesinger G, et al.; Ovarian Stimulation TEGGO. ESHRE guideline: ovarian stimulation for IVF/ICSI†. Hum Reprod Open. 2020;2020(2):hoaa009. doi: 10.1093/hropen/hoaa009
  • van der Kooi ALLF, Kelsey TW, van den Heuvel-Eibrink MM, et al. Perinatal complications in female survivors of cancer: a systematic review and meta-analysis. Eur J Cancer Oxf Engl 1990. 2019;111:126–137. doi: 10.1016/j.ejca.2019.01.104
  • Hartnett KP, Mertens AC, Kramer MR, et al. Pregnancy after cancer: Does timing of conception affect infant health? pregnancy timing after cancer. Cancer. 2018;124(22):4401–4407. doi: 10.1002/cncr.31732
  • Buonomo B, Brunello A, Noli S, et al. Tamoxifen Exposure during pregnancy: a systematic review and three more cases. Breast Care. 2019 Jul 25:1–9. doi: 10.1159/000501473

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.