1
|
Turan V, Oktay K. Developments in pharmacotherapy for the preservation of ovarian function during cancer treatment. Expert Opin Pharmacother 2025; 26:897-907. [PMID: 40271805 DOI: 10.1080/14656566.2025.2495090] [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: 02/19/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/25/2025]
Abstract
INTRODUCTION Cancer is one of the major causes of human death, and anti-cancer therapy often results in premature ovarian failure and infertility, depending on factors such as age, initial ovarian reserve, and chemotherapy type and dose. Fertility preservation procedures, such as oocyte, embryo, and ovarian cortex cryopreservation, can help women achieve pregnancy after cancer treatment. However, the development of pharmacological therapies to protect ovarian function during chemotherapy would represent a significant advancement. AREAS COVERED We searched the published articles in PubMed up to December 2024, containing key words '"chemotherapy",' 'cancer,' '"ovarian protection",' '"pharmacological therapy",' '"ovarian reserve"' and '"fertility".' Chemotherapeutic agents act via various mechanisms in the human ovary, including direct DNA damage leading to oocyte apoptosis, as well as damage to ovarian stroma and microvascular architecture. In recent years, numerous protective agents have emerged, showing promise in protecting ovaries from chemotherapy-induced damage. However, most studies have relied on animal models, and only a limited number have directly tested these agents in human ovarian tissue. At present, no pharmacological treatment has been conclusively proven effective for preserving fertility. EXPERT OPINION A comprehensive understanding of the mechanisms underlying chemotherapy-induced ovarian damage is critical for the development of efficient and targeted pharmacological therapies.
Collapse
Affiliation(s)
- Volkan Turan
- Department of Obstetrics, Health and Technology University School of Medicine, Istanbul, Turkey
- Innovation Institute for Fertility Preservation, Newyork, CT, USA
| | - Kutluk Oktay
- Innovation Institute for Fertility Preservation, Newyork, CT, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
2
|
Su HI, Lacchetti C, Letourneau J, Partridge AH, Qamar R, Quinn GP, Reinecke J, Smith JF, Tesch M, Wallace WH, Wang ET, Loren AW. Fertility Preservation in People With Cancer: ASCO Guideline Update. J Clin Oncol 2025; 43:1488-1515. [PMID: 40106739 DOI: 10.1200/jco-24-02782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 01/09/2025] [Indexed: 03/22/2025] Open
Abstract
PURPOSE To provide updated fertility preservation (FP) recommendations for people with cancer. METHODS A multidisciplinary Expert Panel convened and updated the systematic review. RESULTS One hundred sixty-six studies comprise the evidence base. RECOMMENDATIONS People with cancer should be evaluated for and counseled about reproductive risks at diagnosis and during survivorship. Patients interested in or uncertain about FP should be referred to reproductive specialists. FP approaches should be discussed before cancer-directed therapy. Sperm cryopreservation should be offered to males before cancer-directed treatment, with testicular sperm extraction if unable to provide semen samples. Testicular tissue cryopreservation in prepubertal males is experimental and should be offered only in a clinical trial. Males should be advised of potentially higher genetic damage risks in sperm collected soon after cancer-directed therapy initiation and completion. For females, established FP methods should be offered, including embryo, oocyte, and ovarian tissue cryopreservation (OTC), ovarian transposition, and conservative gynecologic surgery. In vitro maturation of oocytes may be offered as an emerging method. Post-treatment FP may be offered to people who did not undergo pretreatment FP or cryopreserve enough oocytes or embryos. Gonadotropin-releasing hormone agonist (GnRHa) should not be used in place of established FP methods but may be offered as an adjunct to females with breast cancer. For patients with oncologic emergencies requiring urgent oncologic therapy, GnRHa may be offered for menstrual suppression. Established FP methods in children who have begun puberty should be offered with patient assent and parent/guardian consent. The only established method for prepubertal females is OTC. Oncology teams should ensure prompt access to a multidisciplinary FP team. Clinicians should advocate for comprehensive FP services coverage and help patients access benefits.Additional information is available at www.asco.org/survivorship-guidelines.
Collapse
Affiliation(s)
- H Irene Su
- University of California, San Diego, San Diego, CA
| | | | | | | | | | | | | | - James F Smith
- University of California, San Francisco, San Francisco, CA
| | | | - W Hamish Wallace
- Royal Hospital for Children & Young People & University of Edinburgh, Edinburgh, United Kingdom
| | | | - Alison W Loren
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
3
|
Maiorano MFP, Cormio G, Loizzi V, Maiorano BA, D’Oronzo S, Silvestris E. Tamoxifen and Fertility in Women with Breast Cancer: A Systematic Review on Reproductive Outcomes and Oncological Safety of Treatment Interruption. Int J Mol Sci 2025; 26:3787. [PMID: 40332441 PMCID: PMC12028241 DOI: 10.3390/ijms26083787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 05/08/2025] Open
Abstract
Breast cancer (BC) is the most prevalent malignancy among women worldwide, with a rising incidence in young, premenopausal patients. For those diagnosed with hormone receptor-positive (HR+) BC, tamoxifen is a cornerstone of adjuvant endocrine therapy, significantly reducing recurrence risk and improving long-term survival. However, its prolonged use poses challenges for women desiring pregnancy, prompting interest in temporary treatment interruption as a strategy to achieve reproductive goals while maintaining oncological safety. This systematic review evaluates the impact of tamoxifen on fertility, the feasibility of treatment interruption, and associated reproductive and oncological outcomes. Following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, we conducted a comprehensive search across major databases, identifying three relevant studies, including one randomized controlled trial (RCT) and two observational cohort studies. The findings suggest that temporary tamoxifen interruption allows for successful pregnancies without significantly increasing short-term recurrence rates. Notably, the POSITIVE trial demonstrated a pregnancy achievement rate of 74% and a live birth rate of 63.8%, with comparable three-year disease-free survival between patients who interrupted tamoxifen and those who continued therapy. However, concerns remain regarding tamoxifen's teratogenic risks, emphasizing the need for strict contraceptive measures and preconception counseling. Despite emerging evidence supporting this approach, long-term safety data are limited. Further research is warranted to refine clinical recommendations and optimize reproductive counseling for young BC survivors.
Collapse
Affiliation(s)
| | - Gennaro Cormio
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (G.C.); (V.L.); (E.S.)
| | - Vera Loizzi
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (G.C.); (V.L.); (E.S.)
- Department of Translational Biomedicine and Neuroscience (DiBraiN), University of Bari “Aldo Moro”, Policlinico of Bari, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Brigida Anna Maiorano
- Department of Medical Oncology, IRCCS San Raffaele Hospital, Via Olgettina 60, 20132 Milan, Italy;
| | - Stella D’Oronzo
- Medicine and Surgery Department, LUM University, Casamassima, 70010 Bari, Italy
- Oncology and Oncohematology Division, “F. Miulli” General Regional Hospital, Acquaviva delle Fonti, 70021 Bari, Italy
| | - Erica Silvestris
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (G.C.); (V.L.); (E.S.)
| |
Collapse
|
4
|
Nguyen TTA, Condorelli M, Demeestere I. Can we really protect the ovary from chemotherapy damage? Best Pract Res Clin Obstet Gynaecol 2025; 99:102603. [PMID: 40120392 DOI: 10.1016/j.bpobgyn.2025.102603] [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: 10/31/2024] [Revised: 02/04/2025] [Accepted: 02/24/2025] [Indexed: 03/25/2025]
Abstract
Future alternatives to current fertility preservation methods such as pharmacological strategies to prevent chemotherapy-induced ovarian damage in female cancer patients are of growing interest. Chemotherapeutic agents, especially alkylating agents, cause DNA damage and apoptosis in ovarian follicles, significantly reducing ovarian reserve. To mitigate this gonadotoxicity, various emerging strategies are being explored, including kinase inhibitors, PI3K/Akt/mTOR pathway inhibitors, antioxidants, miRNAs and GnRH agonists. These treatments work by preventing follicular apoptosis or excessive activation of primordial follicles. Although promising results have been observed in vitro and in vivo in rodent models, further investigations to bypass their limitations are needed to confirm their efficacy and safety. These challenges include the non-interference with anti-tumoral effect of chemotherapy and the specificity of fertoprotective agents to ovaries.
Collapse
Affiliation(s)
- Thuy Truong An Nguyen
- Research Laboratory on Human Reproduction, Faculty of Medicine, Université Libre de Bruxelles (ULB), 1070, Brussels, Belgium
| | - Margherita Condorelli
- Fertility Clinic, Department of Obstetrics and Gynecology, HUB Erasme, Brussels, Belgium
| | - Isabelle Demeestere
- Research Laboratory on Human Reproduction, Faculty of Medicine, Université Libre de Bruxelles (ULB), 1070, Brussels, Belgium; Fertility Clinic, Department of Obstetrics and Gynecology, HUB Erasme, Brussels, Belgium.
| |
Collapse
|
5
|
Tanaka Y, Amano T, Nakamura A, Takahashi A, Takebayashi A, Hanada T, Tsuji S, Murakami T. Balancing Fertility Preservation and Treatment Efficacy in (Neo)adjuvant Therapy for Adolescent and Young Adult Breast Cancer Patients: a Narrative Review. Curr Oncol Rep 2024; 26:1563-1574. [PMID: 39499484 DOI: 10.1007/s11912-024-01615-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2024] [Indexed: 11/07/2024]
Abstract
PURPOSE OF REVIEW Adolescent and young adult (AYA) breast cancer survivors face a significant risk of infertility due to the gonadotoxic effects of (neo)adjuvant therapy, which complicates their ability to conceive post-treatment. While (neo)adjuvant therapy primarily aims to improve recurrence-free and overall survival, fertility preservation strategies should also be considered for young patients. This narrative review explores recent advancements in fertility preservation techniques, such as oocyte, embryo, and ovarian tissue cryopreservation, and evaluates the feasibility of modifying breast cancer (neo)adjuvant therapy to preserve fertility without compromising survival outcomes. RECENT FINDINGS Our review highlights that clinical trials with co-primary endpoints of oncological safety and fertility preservation are limited, and substituting standard treatment regimens solely for fertility preservation is currently not recommended. Nevertheless, new clinical studies have emerged that either exclude highly ovarian-toxic agents, such as cyclophosphamide, or omit adjuvant therapy altogether, even if fertility preservation is not their primary endpoint. Unfortunately, many of these trials have not evaluated ovarian toxicity. Notably, since 2020, major oncology organizations, including the American Society of Clinical Oncology (ASCO), the European Society of Medical Oncology (ESMO) have advocated for the routine assessment of ovarian toxicity in all clinical trials. The review underscores the importance of incorporating ovarian toxicity as a standard endpoint in future trials involving premenopausal breast cancer patients to identify treatment regimens that can effectively balance fertility preservation with treatment efficacy.
Collapse
Affiliation(s)
- Yuji Tanaka
- Department of Obstetrics and Gynaecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-Cho, Otsu, Shiga, Japan.
| | - Tsukuru Amano
- Department of Obstetrics and Gynaecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-Cho, Otsu, Shiga, Japan
| | - Akiko Nakamura
- Department of Obstetrics and Gynaecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-Cho, Otsu, Shiga, Japan
| | - Akimasa Takahashi
- Department of Obstetrics and Gynaecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-Cho, Otsu, Shiga, Japan
| | - Akie Takebayashi
- Department of Obstetrics and Gynaecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-Cho, Otsu, Shiga, Japan
| | - Tetsuro Hanada
- Department of Obstetrics and Gynaecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-Cho, Otsu, Shiga, Japan
| | - Shunichiro Tsuji
- Department of Obstetrics and Gynaecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-Cho, Otsu, Shiga, Japan
| | - Takashi Murakami
- Department of Obstetrics and Gynaecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-Cho, Otsu, Shiga, Japan
| |
Collapse
|
6
|
Markowska A, Antoszczak M, Markowska J, Huczyński A. Gynotoxic Effects of Chemotherapy and Potential Protective Mechanisms. Cancers (Basel) 2024; 16:2288. [PMID: 38927992 PMCID: PMC11202309 DOI: 10.3390/cancers16122288] [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: 05/29/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
Chemotherapy is one of the leading cancer treatments. Unfortunately, its use can contribute to several side effects, including gynotoxic effects in women. Ovarian reserve suppression and estrogen deficiency result in reduced quality of life for cancer patients and are frequently the cause of infertility and early menopause. Classic alkylating cytostatics are among the most toxic chemotherapeutics in this regard. They cause DNA damage in ovarian follicles and the cells they contain, and they can also induce oxidative stress or affect numerous signaling pathways. In vitro tests, animal models, and a few studies among women have investigated the effects of various agents on the protection of the ovarian reserve during classic chemotherapy. In this review article, we focused on the possible beneficial effects of selected hormones (anti-Müllerian hormone, ghrelin, luteinizing hormone, melatonin), agents affecting the activity of apoptotic pathways and modulating gene expression (C1P, S1P, microRNA), and several natural (quercetin, rapamycin, resveratrol) and synthetic compounds (bortezomib, dexrazoxane, goserelin, gonadoliberin analogs, imatinib, metformin, tamoxifen) in preventing gynotoxic effects induced by commonly used cytostatics. The presented line of research appears to provide a promising strategy for protecting and/or improving the ovarian reserve in the studied group of cancer patients. However, well-designed clinical trials are needed to unequivocally assess the effects of these agents on improving hormonal function and fertility in women treated with ovotoxic anticancer drugs.
Collapse
Affiliation(s)
- Anna Markowska
- Department of Perinatology and Women’s Health, Poznań University of Medical Sciences, 60-535 Poznań, Poland
| | - Michał Antoszczak
- Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, 61-614 Poznań, Poland
| | - Janina Markowska
- Gynecological Oncology Center, Poznańska 58A, 60-850 Poznań, Poland;
| | - Adam Huczyński
- Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, 61-614 Poznań, Poland
| |
Collapse
|
7
|
Bedoschi G, Ingold C, Navarro PA. Fertility Preservation and Ovarian Hyperstimulation Syndrome Management in Cancer Care: A Pathophysiological Perspective on Gonadotropin-Releasing Hormone Agonists and Antagonists. PATHOPHYSIOLOGY 2024; 31:288-297. [PMID: 38921726 PMCID: PMC11206524 DOI: 10.3390/pathophysiology31020021] [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: 12/31/2023] [Revised: 03/02/2024] [Accepted: 06/04/2024] [Indexed: 06/27/2024] Open
Abstract
This narrative review delves into the evolving landscape of fertility preservation techniques, with a particular focus on their use in patients undergoing oncology treatment that carries a risk of ovarian insufficiency. Advances in established methods such as cryopreservation of oocytes and embryos are highlighted, and the increasing use of gonadotropin-releasing hormone (GnRH) agonists is discussed. The review also addresses the complexities and controversies associated with these approaches, such as the 'flare-up' effect associated with GnRH agonists and the potential of GnRH antagonists to reduce the risk of ovarian hyperstimulation syndrome. Despite advances in fertility preservation, the report highlights the challenges we face, including the need for personalized treatment protocols and the management of associated risks. It calls for continued research and collaboration between healthcare professionals to refine these techniques and ultimately improve reproductive outcomes for patients facing the prospect of fertility-impairing treatment.
Collapse
Affiliation(s)
- Giuliano Bedoschi
- Department of Gynecology and Obstetrics, Reproductive Medicine Division, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14048-900, Brazil;
| | - Caroline Ingold
- Department of Collective Health, Sexual, Reproductive Health and Population Genetics, Faculdade de Medicina do ABC, Santo André 09060-870, Brazil;
| | - Paula Andrea Navarro
- Department of Gynecology and Obstetrics, Reproductive Medicine Division, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14048-900, Brazil;
| |
Collapse
|
8
|
Rodriguez-Wallberg KA, Nilsson HP, Bergh J, Malmros J, Ljungman P, Foukakis T, Stragliotto CL, Friman EI, Linderholm B, Valachis A, Andersson A, Harrysson S, Vennström L, Frisk P, Mörse H, Eloranta S. ProFertil study protocol for the investigation of gonadotropin-releasing hormone agonists (GnRHa) during chemotherapy aiming at fertility protection of young women and teenagers with cancer in Sweden-a phase III randomised double-blinded placebo-controlled study. BMJ Open 2023; 13:e078023. [PMID: 38070906 PMCID: PMC10728964 DOI: 10.1136/bmjopen-2023-078023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 10/31/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Gonadotropin-releasing hormone agonists (GnRHa) cotreatment used to transiently suppress ovarian function during chemotherapy to prevent ovarian damage and preserve female fertility is used globally but efficacy is debated. Most clinical studies investigating a beneficial effect of GnRHa cotreatment on ovarian function have been small, retrospective and uncontrolled. Unblinded randomised studies on women with breast cancer have suggested a beneficial effect, but results are mixed with lack of evidence of improvement in markers of ovarian reserve. Unblinded randomised studies of women with lymphoma have not shown any benefit regarding fertility markers after long-term follow-up and no placebo-controlled study has been conducted so far. The aim of this study is to investigate if administration of GnRHa during cancer treatment can preserve fertility in young female cancer patients in a double-blind, placebo-controlled clinical trial. METHODS AND ANALYSIS A prospective, randomised, double-blinded, placebo-controlled, phase III study including 300 subjects with breast cancer. In addition, 200 subjects with lymphoma, acute leukemias and sarcomas will be recruited. Women aged 14-42 will be randomised 1:1 to treatment with GnRHa (triptorelin) or placebo for the duration of their gonadotoxic chemotherapy. Follow-up until 5 years from end of treatment (EoT). The primary endpoint will be change in anti-Müllerian hormone (AMH) recovery at follow-up 12 months after EoT, relative to AMH levels at EoT, comparing the GnRHa group and the placebo group in women with breast cancer. ETHICS AND DISSEMINATION This study is designed in accordance with the principles of Good Clinical Practice (ICH-GCP E6 (R2)), local regulations (ie, European Directive 2001/20/EC) and the ethical principles of the Declaration of Helsinki. Within 6 months of study completion, the results will be analysed and the study results shall be reported in the EudraCT database. STUDY REGISTRATION The National Institutional review board in Sweden dnr:2021-03379, approval date 12 October 2021 (approved amendments 12 June 2022, dnr:2022-02924-02 and 13 December 2022, dnr:2022-05565-02). The Swedish Medical Product Agency 19 January 2022, Dnr:5.1-2021-98927 (approved amendment 4 February 2022). Manufacturing authorisation for authorised medicinal products approved 6 December 2021, Dnr:6.2.1-2020-079580. Stockholm Medical Biobank approved 22 June 2022, RBC dnr:202 253. TRIAL REGISTRATION NUMBER NCT05328258; EudraCT number:2020-004780-71.
Collapse
Affiliation(s)
- Kenny A Rodriguez-Wallberg
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
- Department of Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
| | | | - Jonas Bergh
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
- Theme cancer, Karolinska Comprehensive Cancer Center and University Hospital, Stockholm, Sweden
| | - Johan Malmros
- Pediatric Theme Astrid Lindgren's Pediatric Hospital, Stockholm, Sweden
| | - Per Ljungman
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska University Hospital, Stockholm, Sweden
- Division of Hematology, Department of Medicine Huddinge, Karolinska Institute, Huddinge, Sweden
| | - Theodoros Foukakis
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
- Theme cancer, Karolinska Comprehensive Cancer Center and University Hospital, Stockholm, Sweden
| | | | | | - Barbro Linderholm
- Department of Oncology, Sahlgrenska University Hospital, Goteborg, Sweden
| | - Antonis Valachis
- Oncology, Örebro universitet Fakulteten för medicin och hälsa, Orebro, Sweden
| | - Anne Andersson
- Department of Oncology, Norrlands University Hospital, Umeå, Sweden
| | - Sara Harrysson
- Department of Hematology, Cancer Theme, Karolinska University Hospital, Stockholm, Sweden
| | - Lovisa Vennström
- Department of Hematology and Coagulation, Sahlgrenska University Hospital, Goteborg, Sweden
| | - Per Frisk
- Akademiska Hospital, Uppsala, Sweden
| | - Helena Mörse
- Center for Pediatric Oncology, Skåne University Hospital, Lund, Sweden
| | - Sandra Eloranta
- Department of Medicine, Karolinska Institute, Solna, Sweden
- Division of Clinical Epidemiology, Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
9
|
Gayete-Lafuente S, Turan V, Oktay KH. Oocyte cryopreservation with in vitro maturation for fertility preservation in girls at risk for ovarian insufficiency. J Assist Reprod Genet 2023; 40:2777-2785. [PMID: 37715873 PMCID: PMC10656385 DOI: 10.1007/s10815-023-02932-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 09/06/2023] [Indexed: 09/18/2023] Open
Abstract
PURPOSE To assess the feasibility and outcomes of oocyte cryopreservation with in vitro maturation (IVM) in post-pubertal girls undergoing fertility preservation (FP) for primary ovarian insufficiency (POI) risk. METHODS Ovarian stimulation was performed with an antagonist protocol or progesterone priming. Ultrasound monitoring was performed transabdominally. Oocytes were retrieved transvaginally under IV sedation. Immature oocytes were subjected to IVM for up to 36 h. All MII oocytes were vitrified. The main outcome measure was the total number of mature oocytes cryopreserved. The secondary outcome was the increase in the mature oocyte yield after IVM. RESULTS Indications for FP included mosaic Turner syndrome (mTS; n = 10), malignancy (n = 3), and POI risk (n = 2). The mean ± SD age, antral follicle count (AFC), and AMH levels were 14.2 ± 1.4 years, 8 ± 5.2 and 1.3 ± 1.3 ng/mL. In girls with mTS, the ovarian reserve was low for age (AFC 7.4 ± 4.7 and AMH 1.4 ± 1.6 ng/mL). Oocyte cryopreservation was possible in all girls with a range of 1-27 mature oocytes obtained, even in those who were previously exposed to chemotherapy or with low ovarian reserve, and no surgical complications were encountered. After IVM, the median mature oocyte yield increased significantly from 7.5 to 10.5 (p = 0.001). CONCLUSIONS Oocyte cryopreservation appears to be feasible and safe in girls as young as 12 years of age at risk for POI The utility of IVM increases the yield of cryopreserved mature oocytes. Prior exposure to chemotherapy or low ovarian reserve should not be an automatic reason to exclude these girls from FP consideration.
Collapse
Affiliation(s)
- Sonia Gayete-Lafuente
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, CT, 310 Cedar Street, FMB Room 224. New Haven, CT, USA
| | - Volkan Turan
- Innovation Institute for Fertility Preservation, NY and CT, USA
- Istanbul Health and Technology University School of Medicine, Istanbul, Turkey
| | - Kutluk H Oktay
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, CT, 310 Cedar Street, FMB Room 224. New Haven, CT, USA.
- Innovation Institute for Fertility Preservation, NY and CT, USA.
| |
Collapse
|
10
|
Oktay KH, Turan V, Bedoschi G, Abdo N, Bang H, Goldfarb S. A prospective longitudinal analysis of the predictors of amenorrhea after breast cancer chemotherapy: Impact of BRCA pathogenic variants. Cancer Med 2023; 12:19225-19233. [PMID: 37698031 PMCID: PMC10557848 DOI: 10.1002/cam4.6527] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 08/18/2023] [Accepted: 08/31/2023] [Indexed: 09/13/2023] Open
Abstract
BACKGROUND Better tools for post-chemotherapy amenorrhea risk assessment are needed for fertility preservation decision-making. Our aim was to determine the predictors of amenorrhea risk at 12 and 18 months post-chemotherapy in women with breast cancer. METHODS 142 women with breast cancer were longitudinally followed for their menstrual changes at 6, 12, and 18 months after the completion of adjuvant chemotherapy with an Anthracycline-Cyclophosphamide-based (AC-based) or Cyclophosphamide-Methotrexate +5-Fluorouracil regimen. Pre- and/or post-chemo AMH levels, age, BMI, tamoxifen use, regimen type, and germline BRCA pathogenic variant (gBRCApv) status were evaluated for the prediction of amenorrhea at 6-18 months. RESULTS In multivariable-adjusted logistic regression, age (p = 0.03) and AMH (p = 0.03) at 12 months, and gBRCApv status (p = 0.03) at 18 months were significant predictors of amenorrhea (areas under the ROC curve of 0.77 and 0.76, for 12 and 18 months, respectively) among 102 evaluable subjects. An undetectable AMH immediately post-chemotherapy was predictive of amenorrhea with <18 month follow-up. In longitudinal analysis estimating time trends, baseline AMH and gBRCApv status was associated with the risk of amenorrhea over 6-18 months; the AMH >2.0 ng/mL group showed attenuated time-trend risk of amenorrhea versus AMH ≤2.0 group (ratio of ORs = 0.91, 95% CI = 0.86-0.97, p = 0.002), while the gBRCApv + showed a steeper time trend, versus the controls (ratio of ORs = 1.12, 95% CI = 1.04-1.20, p = 0.003). CONCLUSIONS In addition to the pre- and post-treatment AMH levels, gBRCApv status is a novel potential predictor of amenorrhea at 12 and 18 months after chemotherapy. The higher likelihood of amenorrhea in women gBRCApv suggests that they are more prone to losing their fertility post-chemotherapy.
Collapse
Affiliation(s)
- Kutluk H. Oktay
- Department of Obstetrics and Gynecology and Reproductive SciencesYale University School of MedicineNew HavenConnecticutUSA
| | - Volkan Turan
- Department of Obstetrics and GynecologyIstanbul Health and Technology University School of MedicineIstanbulTurkey
| | - Giuliano Bedoschi
- Department of Obstetrics and Gynecology, Ribeirao Preto School of MedicineUniversity of Sao PauloRibeirao PretoBrazil
| | - Nadia Abdo
- Memorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Heejung Bang
- Division of Biostatistics, Department of Public Health SciencesUniversity of CaliforniaDavisCaliforniaUSA
| | - Shari Goldfarb
- Memorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
- Weill Cornell Medical CenterNew YorkNew YorkUSA
| |
Collapse
|
11
|
Trujillo M, Odle AK, Aykin-Burns N, Allen AR. Chemotherapy induced oxidative stress in the ovary: drug-dependent mechanisms and potential interventions†. Biol Reprod 2023; 108:522-537. [PMID: 36539327 PMCID: PMC10106837 DOI: 10.1093/biolre/ioac222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/07/2022] [Accepted: 12/14/2022] [Indexed: 01/19/2023] Open
Abstract
Cancer incidence and relative survival are expected to increase over the next few decades. With the majority of patients receiving combinatorial chemotherapy, an increasing proportion of patients experience long-term side effects from treatment-including reproductive disorders and infertility. A limited number of studies have examined mechanisms of single-agent chemotherapy-induced gonadotoxicity, with chemotherapy-induced oxidative stress being implicated in the loss of reproductive functions. Current methods of female fertility preservation are costly, invasive, only moderately successful, and seldom presented to cancer patients. The potential of antioxidants to alleviate chemotherapy has been overlooked at a time when it is becoming increasingly important to develop strategies to protect reproductive functions during chemotherapy. This review will summarize the importance of reactive oxygen species homeostasis in reproduction, chemotherapy-induced mitochondrial dysfunction in oocytes, chemotherapy-induced oxidative stress, and several promising natural adjuvants.
Collapse
Affiliation(s)
- Madison Trujillo
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Angela K Odle
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Nukhet Aykin-Burns
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Antiño R Allen
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
12
|
Yildiz S, Bildik G, Benlioglu C, Turan V, Dilege E, Ozel M, Kim S, Oktem O. Breast cancer treatment and ovarian function. Reprod Biomed Online 2023; 46:313-331. [PMID: 36400663 DOI: 10.1016/j.rbmo.2022.09.014] [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: 06/13/2022] [Revised: 09/14/2022] [Accepted: 09/14/2022] [Indexed: 02/07/2023]
Abstract
The aim of this study was to provide an update on ovarian function and the mechanisms of gonadal damage after exposure to chemotherapy in breast cancer survivors. The alkylating agents are toxic to both primordial and growing follicles. However, anti-metabolite drugs are more likely to destroy preantral and antral follicles. Younger patients are more likely to have a higher ovarian reserve, and therefore, more likely to retain some residual ovarian function after exposure to gonadotoxic regimens. However, there can be significant variability in ovarian reserve among patients of the same age. Furthermore, patients with critically diminished ovarian reserve may continue to menstruate regularly. Therefore age and menstrual status are not reliable indicators of good ovarian reserve and might give a false sense of security and result in an adverse outcome if the patient is consulted without considering more reliable quantitative markers of ovarian reserve (antral follicle count and anti-Müllerian hormone) and fertility preservation is not pursued. In contrast to well-documented ovarian toxicity of older chemotherapy regimens, data for newer taxane-containing protocols have only accumulated in the last decade and data are still very limited regarding the impact of targeted therapies on ovarian function.
Collapse
Affiliation(s)
- Sule Yildiz
- The Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Koç University Hospital, Koç University School of Medicine, Istanbul, Turkey
| | - Gamze Bildik
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston TX 77054, USA
| | - Can Benlioglu
- Department of Obstetrics and Gynecology, Koç University Hospital, Istanbul, Turkey
| | - Volkan Turan
- Istanbul Tema Hospital, Assisted Reproduction Unit, Istanbul
| | - Ece Dilege
- Department of General Surgery, Koç University Hospital, Koç University School of Medicine, Istanbul, Turkey
| | - Melis Ozel
- Department of Gynecology and Obstetrics Klinikum Ingolstadt, Bavaria, Germany
| | - Samuel Kim
- Eden Centers for Advanced Fertility, Fullerton CA 92835, USA
| | - Ozgur Oktem
- The Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Koç University Hospital, Koç University School of Medicine, Istanbul, Turkey.
| |
Collapse
|
13
|
Investigation of the female infertility risk associated with anti-cancer therapy. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023:10.1007/s12094-023-03087-8. [PMID: 36689055 DOI: 10.1007/s12094-023-03087-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/11/2023] [Indexed: 01/24/2023]
Abstract
Female infertility is a significant health issue worldwide with a rising incidence. Anti-cancer therapy is one of the most important reasons for increasing infertility. Although anti-cancer treatment increases the rate of survival, it decreases the quality of life through its side effects. The most substantial side effects are sexual dysfunction and infertility. Breast cancer is the most common cancer. The first-line treatment of breast cancer is chemotherapy by alkylating agents like cyclophosphamide, which leads to infertility. For instance, persistent chemotherapy-induced amenorrhea among breast cancer patients could affect almost half of the patients that undergo such therapy. However, some agents or therapeutic methods can ameliorate these intoxicating effects. Chemotherapy plus gonadotropin-releasing hormone agonist, in breast cancer patients, can not only improve overall survival but also reduce ovarian toxicity. Age plays an essential role in chemotherapy-induced amenorrhea. Chemotherapy at a younger age can reduce the risk of infertility. Gynecological cancers including uterine and ovarian cancer, which have high mortality rates, are the most related cancers to infertility. Surgery is the primary treatment of gynecological cancers. Studies demonstrated that fertility-sparing surgery is a better option than radical surgery. In addition, neoadjuvant chemotherapy is mostly a better option than primary cytoreductive surgery in terms of survival and fertility. Immune checkpoint inhibitors (ICIs) have recently played a major role in treating various cancer types. However, ICIs are associated with hypophysitis, which affects ovaries and can lead to infertility. There are some options for ovarian preservation such as embryo cryopreservation, oocyte cryopreservation, ovarian transposition, ovarian tissue cryopreservation, and ovarian suppression by GnRH agonists. Anti-müllerian hormone level can be utilized to monitor the ovarian reserve. Moreover, to avoid fertility loss, approaches such as using transplantation of human placenta mesenchymal stem cells, administrating anti-inflammatory agents and hormone therapy are under investigation.
Collapse
|
14
|
Di Nardo P, Lisanti C, Garutti M, Buriolla S, Alberti M, Mazzeo R, Puglisi F. Chemotherapy in patients with early breast cancer: clinical overview and management of long-term side effects. Expert Opin Drug Saf 2022; 21:1341-1355. [DOI: 10.1080/14740338.2022.2151584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Paola Di Nardo
- Department of Medical Oncology, CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
| | - Camilla Lisanti
- Department of Medical Oncology, CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
| | - Mattia Garutti
- Department of Medical Oncology, CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
| | - Silvia Buriolla
- Department of Medicine (DAME), University of Udine, Udine, Italy
| | - Martina Alberti
- Department of Medicine (DAME), University of Udine, Udine, Italy
| | - Roberta Mazzeo
- Department of Medicine (DAME), University of Udine, Udine, Italy
| | - Fabio Puglisi
- Department of Medical Oncology, CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
- Department of Medicine (DAME), University of Udine, Udine, Italy
| |
Collapse
|
15
|
Yan F, Zhao Q, Li Y, Zheng Z, Kong X, Shu C, Liu Y, Shi Y. The role of oxidative stress in ovarian aging: a review. J Ovarian Res 2022; 15:100. [PMID: 36050696 PMCID: PMC9434839 DOI: 10.1186/s13048-022-01032-x] [Citation(s) in RCA: 107] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 08/21/2022] [Indexed: 11/29/2022] Open
Abstract
Ovarian aging refers to the process by which ovarian function declines until eventual failure. The pathogenesis of ovarian aging is complex and diverse; oxidative stress (OS) is considered to be a key factor. This review focuses on the fact that OS status accelerates the ovarian aging process by promoting apoptosis, inflammation, mitochondrial damage, telomere shortening and biomacromolecular damage. Current evidence suggests that aging, smoking, high-sugar diets, pressure, superovulation, chemotherapeutic agents and industrial pollutants can be factors that accelerate ovarian aging by exacerbating OS status. In addition, we review the role of nuclear factor E2-related factor 2 (Nrf2), Sirtuin (Sirt), mitogen-activated protein kinase (MAPK), protein kinase B (AKT), Forkhead box O (FoxO) and Klotho signaling pathways during the process of ovarian aging. We also explore the role of antioxidant therapies such as melatonin, vitamins, stem cell therapies, antioxidant monomers and Traditional Chinese Medicine (TCM), and investigate the roles of these supplements with respect to the reduction of OS and the improvement of ovarian function. This review provides a rationale for antioxidant therapy to improve ovarian aging.
Collapse
Affiliation(s)
- Fei Yan
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Qi Zhao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Ying Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Zhibo Zheng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Xinliang Kong
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Chang Shu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Yanfeng Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, People's Republic of China.
| | - Yun Shi
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, People's Republic of China.
| |
Collapse
|
16
|
Zhou B, Kwan B, Desai MJ, Nalawade V, Ruddy KJ, Nathan PC, Henk HJ, Murphy JD, Whitcomb BW, Su HI. Long-term antimüllerian hormone patterns differ by cancer treatment exposures in young breast cancer survivors. Fertil Steril 2022; 117:1047-1056. [PMID: 35216831 PMCID: PMC9081208 DOI: 10.1016/j.fertnstert.2022.01.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 01/13/2022] [Accepted: 01/14/2022] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To compare antimüllerian hormone (AMH) patterns by cancer status and treatment exposures across 6 years after incident breast cancer using administrative data. DESIGN In a cross-sectional design, AMH levels in patients who developed incident breast cancer between ages 15-39 years during 2005-2019 were matched 1:10 to levels in females without cancer in the OptumLabs Data Warehouse. Modeled AMH patterns were compared among cyclophosphamide-based chemotherapy, non-cyclophosphamide-based chemotherapy, no chemotherapy, and no breast cancer groups. SETTING Commercially insured females in the United States. PATIENT(S) Females with and without breast cancer. EXPOSURE(S) Breast cancer, cyclophosphamide- and non-cyclophosphamide-based chemotherapy. MAIN OUTCOME MEASURE(S) AMH levels. RESULT(S) A total of 233 patients with breast cancer (mean age, 34 years; standard deviation, 3.7 years) contributed 278 AMH levels over a median of 2 years (range, 0-6.7 years) after diagnosis; 52% received cyclophosphamide-based chemotherapy, 17% received non-cyclophosphamide-based chemotherapy (80% platinum-based), and 31% received no chemotherapy. A total of 2,777 matched females without cancer contributed 2,780 AMH levels. The pattern of AMH levels differed among the 4 groups. Among females without cancer and breast cancer survivors who did not undergo chemotherapy, AMH declined linearly over time. In contrast, among those who received cyclophosphamide-based and noncyclophosphamide-based chemotherapy, a nonlinear pattern of AMH level of initial fall during chemotherapy, followed by an increase over 2-4 years, and then by a plateau over 1-2 years before a decline was observed. CONCLUSION(S) In breast cancer survivors, AMH levels from administrative data supported ovarian toxicity of non-cyclophosphamide-based chemotherapy in breast cancer and efficiently depicted the timing and duration of changes in ovarian reserve to reflect the residual reproductive lifespan.
Collapse
Affiliation(s)
- Beth Zhou
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Brian Kwan
- Division of Biostatistics and Bioinformatics, Herbert Wertheim School of Public Health, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
- Moores Cancer Center, University of California, San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093, USA
| | - Milli J. Desai
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Vinit Nalawade
- Department of Radiation Medicine and Applied Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Kathryn J. Ruddy
- Department of Oncology, Mayo Clinic, Rochester, 200 1st St SW, Rochester, MN 55905, USA
| | - Paul C. Nathan
- The Hospital for Sick Children, 555 University Avenue Toronto ON M5G 1X8, CA
| | - Henry J. Henk
- OptumLabs, 11000 Optum Cir, Eden Prarie, MN 55344, USA
| | - James D. Murphy
- Department of Radiation Medicine and Applied Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Brian W. Whitcomb
- Department of Biostatistics & Epidemiology, School of Public Health & Health Sciences, University of Massachusetts, 433 Arnold House, Amherst, MA, 01003, USA
| | - H. Irene Su
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
- Moores Cancer Center, University of California, San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093, USA
| |
Collapse
|
17
|
Anderson RA, Cameron D, Clatot F, Demeestere I, Lambertini M, Nelson SM, Peccatori F. 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; 28:417-434. [PMID: 35199161 PMCID: PMC9071067 DOI: 10.1093/humupd/dmac004] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 11/18/2021] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Female patients undergoing anticancer treatment are at elevated risk of adverse ovarian outcomes including infertility and premature ovarian insufficiency (POI), which is associated with short- and long-term health risks. Anti-Müllerian hormone (AMH) is a key biomarker of ovarian reserve, but its role prior to and after cancer treatment is less well understood. OBJECTIVE AND RATIONALE To conduct a systematic review evaluating AMH as a biomarker of ovarian reserve and POI before and after anticancer treatment, which has become a pressing clinical issue in reproductive medicine. There are a large number of observational studies, but differences in patient groups, cancer diagnoses and study design make this a confusing field that will benefit from a thorough and robust review. SEARCH METHODS A systematic literature search for AMH in women with cancer was conducted in PubMed, Embase and Cochrane Central Register of Controlled Trials up to 1 April 2021. Bias review was conducted using the Risk of Bias In Non-randomized Studies of Interventions (ROBINS-I) protocol along with qualitative assessment of quality. Exploratory subgroups were established based on age, cancer type and length of follow-up. OUTCOMES Ninety-two publications (N = 9183 patients) were included in this analysis after quality and bias review. Reduced/undetectable AMH was consistently identified in 69/75 studies (92%) following chemotherapy or radiotherapy, with reductions ranging from 42% to concentrations below the limit of detection, and many reporting mean or median declines of ≥90%. Where longitudinal data were analysed (42 studies), a majority (33/42 (79%)) of studies reported at least partial recovery of AMH at follow-up, however, effect estimates were highly variable, reflecting that AMH levels were strongly impacted by anticancer treatment (i.e. the chemotherapy regimen used and the number of treatment cycles need), with recovery and its degree determined by treatment regimen, age and pre-treatment AMH level. In 16/31 (52%) publications, oligo/amenorrhoea was associated with lower post-treatment AMH consistent with impending POI, although menstruation and/or pregnancy were reported in patients with low or undetectable AMH. Long-term (>5 years) follow-up of paediatric patients following cancer treatment also found significantly lower AMH compared with control groups in 14/20 (70%) of studies, with very variable effect sizes from complete loss of AMH to full recovery depending on treatment exposure, as in adult patients. WIDER IMPLICATIONS AMH can be used to identify the damaging effect of cancer treatments on ovarian function. This can be applied to individual women, including pre-pubertal and adolescent girls, as well as comparing different treatment regimens, ages and pre-treatment AMH levels in populations of women. While there was evidence for its value in the diagnosis of POI after cancer treatment, further studies across a range of diagnoses/treatment regimens and patient ages are required to clarify this, and to quantify its predictive value. A major limitation for the use of AMH clinically is the very limited data relating post-treatment AMH levels to fertility, duration of reproductive lifespan or time to POI; analysis of these clinically relevant outcomes will be important in further research.
Collapse
Affiliation(s)
- Richard A Anderson
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK,Correspondence address. MRC Centre for Reproductive Health, The Queen’s Medical Research Institute, Edinburgh BioQuarter, 47 Little France Crescent, Edinburgh EH16 4TJ, UK. Tel: +44-(0)-131-242-6386; E-mail:https://orcid.org/0000-0002-7495-518X
| | - David Cameron
- Edinburgh University Cancer Centre, IGMM, Edinburgh, UK
| | | | - Isabelle Demeestere
- Fertility clinic, CUB-Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - 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 Sciences (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - Scott M Nelson
- School of Medicine, University of Glasgow, Glasgow, UK,NIHR Bristol Biomedical Research Centre, Bristol, UK,The Fertility Partnership, Oxford, UK
| | | |
Collapse
|
18
|
Oktay KH, Marin L, Titus S. Impact of chemotherapy on the ovarian reserve: Are all primordial follicles created equal? Fertil Steril 2022; 117:396-398. [PMID: 34998576 DOI: 10.1016/j.fertnstert.2021.12.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 11/04/2022]
Affiliation(s)
- Kutluk H Oktay
- Laboratory of Molecular Reproduction and Fertility Preservation, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut; Innovation Institute for Fertility Preservation, New York, New York
| | - Loris Marin
- Laboratory of Molecular Reproduction and Fertility Preservation, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut; Department of Women's and Children's Health, University of Padua, Padua, Italy
| | - Shiny Titus
- Laboratory of Molecular Reproduction and Fertility Preservation, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
19
|
Anderson RA, Kelsey TW, Morrison DS, Wallace WHB. Family size and duration of fertility in female cancer survivors: a population-based analysis. Fertil Steril 2022; 117:387-395. [PMID: 34933761 PMCID: PMC8865032 DOI: 10.1016/j.fertnstert.2021.11.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/01/2021] [Accepted: 11/04/2021] [Indexed: 01/04/2023]
Abstract
OBJECTIVE To assess family size and timescale for achieving pregnancy in women who remain fertile after cancer. DESIGN Population-based analysis. SETTING National databases. PATIENT(S) All women diagnosed with cancer before the age of 40 years in Scotland, 1981-2012 (n = 10,267) with no previous pregnancy; each was matched with 3 population controls. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) The number and timing of pregnancy and live birth after cancer diagnosis, to 2018. RESULT(S) In 10,267 cancer survivors, the hazard ratio for a subsequent live birth was 0.56 (95% confidence interval, 0.53-0.58) overall. In women who achieved a subsequent pregnancy, age at live birth increased (mean ± SD, 31.2 ± 5.5 vs. 29.7 ± 6.1 in controls), and the family size was lower (2.0 ± 0.8 vs. 2.3 ± 1.1 live births). These findings were consistent across several diagnoses. The interval from diagnosis to last pregnancy was similar to that of controls (10.7 ± 6.4 vs. 10.9 ± 7.3 years) or significantly increased, for example, after breast cancer (6.2 ± 2.8 vs. 5.3 ± 3.3 years) and Hodgkin lymphoma (11.1 ± 5.1 vs. 10.1 ± 5.8 years). CONCLUSION(S) These data quantify the reduced chance of live birth after cancer. Women who subsequently conceived achieved a smaller family size than matched controls, but the period of time after cancer diagnosis across which pregnancies occurred was similar or, indeed, increased. Thus, we did not find evidence that women who were able to achieve a pregnancy after cancer had a shorter timescale over which they have pregnancies.
Collapse
Affiliation(s)
- Richard A Anderson
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom.
| | - Tom W Kelsey
- School of Computer Science, University of St Andrews, St Andrews, United Kingdom
| | | | - W Hamish B Wallace
- Department of Hematology and Oncology, Royal Hospital for Children and Young People, Edinburgh, United Kingdom
| |
Collapse
|
20
|
Mailliez A, Pigny P, Bogart E, Keller L, D'orazio E, Vanseymortier M, le Deley MC, Decanter C. Is ovarian recovery after chemotherapy in young patients with early breast cancer influenced by controlled ovarian hyperstimulation for fertility preservation or tumor characteristics? Results of a prospective study in 126 patients. Int J Cancer 2022; 150:1850-1860. [PMID: 35038360 DOI: 10.1002/ijc.33933] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/29/2021] [Accepted: 12/01/2021] [Indexed: 11/06/2022]
Abstract
Young individuals, aged <40 years, represent 7% of all patients with early breast cancer (EBC), most of whom receive chemotherapy. Preserving future fertility in these patients has become a major concern. This prospective study assessed ovarian function during and after chemotherapy according to patient and tumor characteristics and evaluated the outcome of controlled ovarian hyperstimulation (COH). Ovarian reserve was evaluated in terms of amenorrhea duration and by longitudinal serum anti-Müllerian hormone (AMH) level variations measured at study entry, during treatment, and until 24 months thereafter. COH has been proposed for patients receiving adjuvant chemotherapy. We studied the association between clinical factors and ovarian function using Cox models and logistic regression. In this young population (age <38 years, median=32), 85 of 90 evaluable patients (94%) experienced chemo-induced amenorrhea, including six persistent amenorrhea and one chemotherapy-induced definitive ovarian failure. Overall, 33% of patients still had undetectable AMH values 12 months after the end of chemotherapy, although most had recovered spontaneous and regular menstrual function. No specific factor was associated with clinical or biological late ovarian dysfunction, except for age and baseline AMH value. Overall, 58 patients underwent COH. The mean number of total retrieved oocytes and metaphase II oocytes were of 11.7 and 6.9, respectively. Thus, our study confirms the importance of fertility preservation in young patients with EBC. Our findings indicates that sequential chemotherapy is associated with a higher risk of persistent amenorrhea. There was no significant association between tumor characteristics, fertility preservation, or recovery of ovarian reserve. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Audrey Mailliez
- Breast Cancer Unit, Medical Oncology Department, Oscar Lambret Center, Lille, France
| | - Pascal Pigny
- Laboratoire de Biochimie « Hormonologie, Métabolisme-Nutrition & Oncologie » Centre de Biologie Pathologie, Centre Hospitalier Régional Universitaire, Lille, France.,INSERM UMR-S1277 CANTHER, Université de Lille, Lille, France
| | - Emilie Bogart
- Clinical Research and Innovation Department, Centre Oscar Lambret, Lille, France
| | - Laura Keller
- Institut de Biologie de la Reproduction-Spermiologie-CECOS, hôpital Jeanne de Flandre, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Emmanuelle D'orazio
- Centre d'Assistance médicale à la Procréation et de Préservation de la Fertilité, Hôpital Jeanne de Flandre, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Marie Vanseymortier
- Clinical Research and Innovation Department, Centre Oscar Lambret, Lille, France
| | | | - Christine Decanter
- Centre d'Assistance médicale à la Procréation et de Préservation de la Fertilité, Hôpital Jeanne de Flandre, Centre Hospitalier Universitaire de Lille, Lille, France.,EA 4308 Gamétogénèse et qualité du gamète, Centre Hospitalier Universitaire de Lille, Lille, France
| |
Collapse
|
21
|
Lei YY, Yeo W. The risk of menopausal symptoms in premenopausal breast cancer patients and current pharmacological prevention strategies. Expert Opin Drug Saf 2021; 20:1163-1175. [PMID: 33951990 DOI: 10.1080/14740338.2021.1926980] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 05/04/2021] [Indexed: 10/21/2022]
Abstract
Introduction: For young premenopausal breast cancer (BC) patients, adjuvant chemotherapy and other anti-cancer treatments can increase the risk of menopausal symptoms and may cause chemotherapy-related amenorrhea (CRA), infertility and premature ovarian insufficiency (POI).Areas covered: In this report, menopausal symptoms related to anti-cancer treatment are described. Menstrual disturbances associated with the use of adjuvant chemotherapy, endocrine therapy, and targeted therapy against human epidermal growth factor receptor 2 (HER2) in premenopausal women withBC are discussed. To prevent menopausal symptoms, CRA and POI, data on the efficacy of temporary ovarian suppression with gonadotropin-releasing hormone analogues (GnRHa) during chemotherapy are highlighted. Pooled analyses have confirmed that concurrent administration of GnRHa during chemotherapy could significantly reduce the risk of developing chemotherapy-induced POI in premenopausal women with early-stageBC. In addition, reports have suggested that embryo/oocyte cryopreservation may increase the chance of pregnancy after the diagnosis ofBC, although such data remain limited.Expert opinion: Commonly experienced by pre-menopausal women withBC, anti-cancer treatment could cause severe menopausal symptoms. Temporary ovarian suppression with GnRHa during chemotherapy provided asafe and efficient strategy to reduce the likelihood of chemotherapy-induced POI in premenopausal patients with early-stageBC undergoing (neo)-adjuvant chemotherapy.
Collapse
Affiliation(s)
- Yuan-Yuan Lei
- Department of Clinical Oncology, Prince of Wales Hospital, the Chinese University of Hong Kong, New Territories, Hong Kong SAR, China
| | - Winnie Yeo
- Department of Clinical Oncology, Prince of Wales Hospital, the Chinese University of Hong Kong, New Territories, Hong Kong SAR, China
- Hong Kong Cancer Institute, State Key Laboratory in Oncology in South China, Faculty of Medicine, The Chinese University of Hong Kong, New Territories, Hong Kong SAR, China
| |
Collapse
|
22
|
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
|