1
|
Murphy HG, Isaacson E, Moravek MB, Ellman E, Compton SD, Rosen MW. Disparities in access to fertility preservation among adolescents undergoing gonadotoxic therapies. J Assist Reprod Genet 2024:10.1007/s10815-024-03253-z. [PMID: 39325343 DOI: 10.1007/s10815-024-03253-z] [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: 04/01/2024] [Accepted: 09/05/2024] [Indexed: 09/27/2024] Open
Abstract
PURPOSE Fertility preservation (FP) for adolescents prior to potentially gonadotoxic therapies is not accessible for all patients. Current literature acknowledges multiple barriers to FP, but research surrounding disparities for accessing these services is limited. We aimed to identify inequities in receiving FP services among adolescents undergoing gonadotoxic therapy. METHODS A retrospective chart review was performed at a single academic medical center for patients aged 0-21 referred for FP counseling prior to gonadotoxic therapy. Exclusions included referral after treatment, prior to gender-affirming therapy, or for fertility discussion due to a genetic condition. Minority patients were defined as non-White race and/or Hispanic ethnicity. Non-minority patients were defined as White, non-Hispanic. Analyses to assess differences in receiving FP based on minority identity and insurance status were performed via logistic regression, with receiving desired care as the outcome variable. RESULTS Our cohort included 136 patients-38 minority and 98 non-minority. Forty-six (33.8%) patients had Medicaid, which did not differ between minority and non-minority (42.1% vs. 38.8%, P = .73). Most patients (83.1%) had a cancer diagnosis. Similar proportions of minority and non-minority patients had gonadotoxic treatment starting urgently (52.6% vs. 55.1%, P = .80), while more minority than non-minority patients desired FP (89.5% vs .77.5%, P = .10). When controlling for insurance type and age, minority participants were 12.8% less likely to receive desired FP (marginal effect = - .128, P = .05). CONCLUSIONS This study identified significant inequities for minority populations in accessing FP. Further research is needed to determine how to make FP services more accessible to all patients, regardless of minority status.
Collapse
Affiliation(s)
- Hana G Murphy
- Department of Obstetrics and Gynecology, University of Michigan, 1500 E. Medical Center Dr., Ann Arbor, MI, 48109-5276, USA
| | - Erin Isaacson
- Department of Obstetrics and Gynecology, University of Michigan, 1500 E. Medical Center Dr., Ann Arbor, MI, 48109-5276, USA
| | - Molly B Moravek
- Division of Reproductive Endocrinology and Infertility, University of Michigan, Ann Arbor, MI, USA
| | - Erin Ellman
- Division of Reproductive Endocrinology and Infertility, University of Michigan, Ann Arbor, MI, USA
| | - Sarah D Compton
- Department of Obstetrics and Gynecology, University of Michigan, 1500 E. Medical Center Dr., Ann Arbor, MI, 48109-5276, USA
| | - Monica W Rosen
- Department of Obstetrics and Gynecology, University of Michigan, 1500 E. Medical Center Dr., Ann Arbor, MI, 48109-5276, USA.
| |
Collapse
|
2
|
Arecco L, Borea R, Magaton IM, Janković K, Mariamizde E, Stana M, Scavone G, Ottonello S, Spinaci S, Genova C, de Azambuja E, Lambertini M. Current practices in oncofertility counseling: updated evidence on fertility preservation and post-treatment pregnancies in young women affected by early breast cancer. Expert Rev Anticancer Ther 2024; 24:803-817. [PMID: 38913581 DOI: 10.1080/14737140.2024.2372337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 06/21/2024] [Indexed: 06/26/2024]
Abstract
INTRODUCTION Anticancer treatments have significantly contributed to increasing cure rates of breast cancer in the last years; however, they can also lead to short- and long-term side effects, including gonadotoxicity, and compromised fertility in young women. Oncofertility is a crucial issue for young patients who have not yet completed their family planning at the time of cancer diagnosis. AREAS COVERED This review aims to cover all the latest available evidence in the field of oncofertility, including the gonadotoxicity of currently adopted anticancer therapies in the curative breast cancer setting, the available strategies for fertility preservation and the feasibility of achieving a pregnancy following anticancer treatment completion. EXPERT OPINION Over the past years, a significant progress has been made in oncofertility care for young women with breast cancer. In the context of the currently available evidence, every young woman with newly diagnosed breast cancer should receive a proper and complete oncofertility counseling before starting any anticancer treatment to increase her chances of future pregnancies.
Collapse
Affiliation(s)
- Luca Arecco
- Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genova, Genova, Italy
- Academic Trials Promoting Team, Institut Jules Bordet, Université Libre de Bruxelles (U.L.B.), Hôpital Universitaire de Bruxelles (HUB), Brussels, Belgium
| | - Roberto Borea
- Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Isotta Martha Magaton
- Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genova, Genova, Italy
- Division of Gynaecological Endocrinology and Reproductive Medicine, University Women's Hospital, Bern, Switzerland
| | | | - Elene Mariamizde
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Oncology and Hematology, Todua Clinic, Tbilisi, Georgia
| | - Mihaela Stana
- Department of Medical Oncology, Elysee Hospital, Alba Iulia, Romania
| | - Graziana Scavone
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Silvia Ottonello
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy
| | - Stefano Spinaci
- ASL3 Breast Unit Department, Division of Breast Surgery, Ospedale Villa Scassi, Genova, Italy
| | - Carlo Genova
- Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Evandro de Azambuja
- Academic Trials Promoting Team, Institut Jules Bordet, Université Libre de Bruxelles (U.L.B.), Hôpital Universitaire de Bruxelles (HUB), Brussels, Belgium
| | - Matteo Lambertini
- Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
3
|
CVETANOVIC ANAS, LAMBERTINI MATTEO, PUNIE KEVIN, MATOVINA BRKO GORANAG, ZIVKOVIC NIKOLAD, POPOVIC MAJAJ, MILOVIC KOVACEVIC MARIJANAM, POPOVIC LAZARS. Pharmacological methods for ovarian function and fertility preservation in women with cancer: A literature review. Oncol Res 2024; 32:1309-1322. [PMID: 39055889 PMCID: PMC11267035 DOI: 10.32604/or.2024.049743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 05/06/2024] [Indexed: 07/28/2024] Open
Abstract
Oncofertility is an extremely significant topic that is increasingly being discussed owing to increased evidence indicating that fertility preservation does not affect the treatment outcomes of patients with cancer but significantly contributes to preserving life quality. The effect of chemotherapy can range from minimal effects to complete ovarian atrophy. Limited data are available on the effects of monoclonal antibodies and targeted therapies on the ovaries and fertility. Temporary ovarian suppression by administering a gonadotropin-releasing hormone agonist (GnRHa) during chemotherapy decreases the gonadotoxic effect of chemotherapy, thereby diminishing the chance of developing premature ovarian insufficiency (POI). At present, the concomitant administration of GnRH analogs during chemotherapy is the only accepted pharmacological method for preserving ovarian function. Notably, most randomized studies on the effectiveness of luteinizing hormone-releasing hormone agonists during chemotherapy in preventing POI have been conducted in women with breast cancer, with a considerably small number of studies on patients with hematological malignancies. Furthermore, most randomized controlled trials on breast cancer have revealed a decrease in treatment-induced POI risk, regardless of the hormone receptor status. In addition, studies on hematological malignancies have yielded negative results; nevertheless, the findings must be interpreted with caution owing to numerous limitations. Current guidelines from the American Society of Clinical Oncology and ESMO Clinical Practice Guidelines recommend sperm, oocyte, and embryo cryopreservation as a standard practice and only offering GnRHa to patients when proven fertility preservation methods are not feasible. In this manuscript, we present a comprehensive literature overview on the application of ovarian suppression with GnRHa during chemotherapy in patients with cancer by addressing preclinical and clinical data, as well as future perspectives in this field that upcoming research should focus on.
Collapse
Affiliation(s)
- ANA S. CVETANOVIC
- Department for Oncology, Medical Faculty Nis, University of Nis, Nis, 18000, Serbia
- Clinic of Oncology, University Clinical Centre Nis, Nis, 18000, Serbia
| | - MATTEO LAMBERTINI
- Department of Internal Medicine and Medical Sciences (DiMI), School of Medicine, University of Genova, Genova, 16100, Italy
- Department of Medical Oncology, U.O.C Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, 16100, Italy
| | - KEVIN PUNIE
- Department of General Medical Oncology and Multidisciplinary Breast Centre, Leuven Cancer Institute, University Hospitals Leuven, Leuven, 3001, Belgium
| | - GORANA G. MATOVINA BRKO
- Department of Medical Oncology, Oncology Institute of Vojvodina, Sremska Kamenica, 21208, Serbia
| | - NIKOLA D. ZIVKOVIC
- Department for Pathology, Medical Faculty Nis, University of Nis, Nis, 18000, Serbia
- Center for Pathology, University Clinical Centre Nis, Nis, 18000, Serbia
| | - MAJA J. POPOVIC
- Department of Medical Oncology, Oncology Institute of Vojvodina, Sremska Kamenica, 21208, Serbia
- Faculty of Medicine, University of Novi Sad, Novi Sad, 21000, Serbia
| | | | - LAZAR S. POPOVIC
- Department of Medical Oncology, Oncology Institute of Vojvodina, Sremska Kamenica, 21208, Serbia
- Faculty of Medicine, University of Novi Sad, Novi Sad, 21000, Serbia
| |
Collapse
|
4
|
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
|
5
|
Tanaka Y, Amano T, Nakamura A, Yoshino F, Takebayashi A, Takahashi A, Yamanaka H, Inatomi A, Hanada T, Yoneoka Y, Tsuji S, Murakami T. Rapamycin prevents cyclophosphamide-induced ovarian follicular loss and potentially inhibits tumour proliferation in a breast cancer xenograft mouse model. Hum Reprod 2024:deae085. [PMID: 38734930 DOI: 10.1093/humrep/deae085] [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: 09/09/2023] [Revised: 03/26/2024] [Indexed: 05/13/2024] Open
Abstract
STUDY QUESTION To what extent and via what mechanism does the concomitant administration of rapamycin (a follicle activation pathway inhibitor and antitumour agent) and cyclophosphamide (a highly toxic ovarian anticancer agent) prevent cyclophosphamide-induced ovarian reserve loss and inhibit tumour proliferation in a breast cancer xenograft mouse model? SUMMARY ANSWER Daily concomitant administration of rapamycin and a cyclic regimen of cyclophosphamide, which has sufficient antitumour effects as a single agent, suppressed cyclophosphamide-induced primordial follicle loss by inhibiting primordial follicle activation in a breast cancer xenograft mouse model, suggesting the potential of an additive inhibitory effect against tumour proliferation. WHAT IS KNOWN ALREADY Cyclophosphamide stimulates primordial follicles by activating the mammalian target of the rapamycin (mTOR) pathway, resulting in the accumulation of primary follicles, most of which undergo apoptosis. Rapamycin, an mTOR inhibitor, regulates primordial follicle activation and exhibits potential inhibitory effects against breast cancer cell proliferation. STUDY DESIGN, SIZE, DURATION To assess ovarian follicular apoptosis, 3 weeks after administering breast cancer cells, 8-week-old mice were randomized into three treatment groups: control, cyclophosphamide, and cyclophosphamide + rapamycin (Cy + Rap) (n = 5 or 6 mice/group). Mice were treated with rapamycin or vehicle control for 1 week, followed by a single dose of cyclophosphamide or vehicle control. Subsequently, the ovaries were resected 24 h after cyclophosphamide administration (short-term treatment groups). To evaluate follicle abundance and the mTOR pathway in ovaries, as well as the antitumour effects and impact on the mTOR pathway in tumours, 8-week-old xenograft breast cancer transplanted mice were randomized into three treatment groups: vehicle control, Cy, and Cy + Rap (n = 6 or 7 mice/group). Rapamycin (5 mg/kg) or the vehicle was administered daily for 29 days. Cyclophosphamide (120 mg/kg) or the vehicle was administered thrice weekly (long-term treatment groups). The tumour diameter was measured weekly. Seven days after the last cyclophosphamide treatment, the ovaries were harvested, fixed, and sectioned (for follicle counting) or frozen (for further analysis). Similarly, the tumours were resected and fixed or frozen. PARTICIPANTS/MATERIALS, SETTING, METHODS Terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL) was performed to examine ovarian follicular apoptosis in the short-term treatment groups. All subsequent experiments were conducted in the long-term treatment groups. Tumour growth was evaluated using the tumour volume index. The tumour volume index indicates the relative volume, compared to the volume 3 weeks after tumour cell injection (at treatment initiation) set to 100%. Tumour cell proliferation was evaluated by Ki-67 immunostaining. Activation of the mTOR pathway in tumours was assessed using the protein extracts from tumours and analysed by western blotting. Haematoxylin and eosin staining of ovaries was used to perform differential follicle counts for primordial, primary, secondary, antral, and atretic follicles. Activation of the mTOR pathway in ovaries was assessed using protein extracts from whole ovaries and analysed by western blotting. Localization of mTOR pathway activation within ovaries was assessed by performing anti-phospho-S6 kinase (downstream of mTOR pathway) immunohistochemistry. MAIN RESULTS AND THE ROLE OF CHANCE Ovaries of the short-term treatment groups were resected 24 h after cyclophosphamide administration and subjected to TUNEL staining of apoptotic cells. No TUNEL-positive primordial follicles were detected in the control, Cy, and Cy + Rap groups. Conversely, many granulosa cells of growing follicles were TUNEL positive in the Cy group but negative in the control and Cy + Rap groups. All subsequent experimental results were obtained from the long-term treatment groups. The tumour volume index stabilized at a mean of 160-200% in the Cy group and 130% in the Cy + Rap group throughout the treatment period. In contrast, tumours in the vehicle control group grew continuously with a mean tumour volume index of 600%, significantly greater than that of the two treatment groups. Based on the western blot analysis of tumours, the mTOR pathway was activated in the vehicle control group and downregulated in the Cy + Rap group when compared with the control and Cy groups. Ki-67 immunostaining of tumours showed significant inhibition of cell proliferation in the Cy + Rap group when compared with that in the control and Cy groups. The ovarian follicle count revealed that the Cy group had significantly fewer primordial follicles (P < 0.001) than the control group, whereas the Cy + Rap group had significantly higher number of primordial follicles (P < 0.001, 2.5 times) than the Cy group. The ratio of primary to primordial follicles was twice as high in the Cy group than in the control group; however, no significant difference was observed between the control group and the Cy + Rap group. Western blot analysis of ovaries revealed that the mTOR pathway was activated by cyclophosphamide and inhibited by rapamycin. The phospho-S6 kinase (pS6K)-positive primordial follicle rate was 2.7 times higher in the Cy group than in the control group. However, this effect was suppressed to a level similar to the control group in the Cy + Rap group. LARGE SCALE DATA None. LIMITATIONS, REASONS FOR CAUTION The combinatorial treatment of breast cancer tumours with rapamycin and cyclophosphamide elicited inhibitory effects on cell proliferative potential compared to cyclophosphamide monotherapy. However, no statistically significant additive effect was observed on tumour volume. Thus, the beneficial antitumour effect afforded by rapamycin administration on breast cancer could not be definitively proven. Although rapamycin has ovarian-protective effects, it does not fully counteract the ovarian toxicity of cyclophosphamide. Nevertheless, rapamycin is advantageous as an ovarian protective agent as it can be used in combination with other ovarian protective agents, such as hormonal therapy. Hence, in combination with other agents, mTOR inhibitors may be sufficiently ovario-protective against high-dose and cyclic cyclophosphamide regimens. WIDER IMPLICATIONS OF THE FINDINGS Compared with a cyclic cyclophosphamide regimen that replicates human clinical practice under breast cancer-bearing conditions, the combination with rapamycin mitigates the ovarian follicle loss of cyclophosphamide without interfering with the anticipated antitumour effects. Hence, rapamycin may represent a new non-invasive treatment option for cyclophosphamide-induced ovarian dysfunction in breast cancer patients. STUDY FUNDING/COMPETING INTEREST(S) This work was not financially supported. The authors declare that they have no conflict of interest.
Collapse
Affiliation(s)
- Yuji Tanaka
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Tsukuru Amano
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Akiko Nakamura
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Fumi Yoshino
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Akie Takebayashi
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Akimasa Takahashi
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Hiroyuki Yamanaka
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Ayako Inatomi
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Tetsuro Hanada
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Yutaka Yoneoka
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Shunichiro Tsuji
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Takashi Murakami
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| |
Collapse
|
6
|
Benor A, Decherney A. Gonadotropin-Releasing Hormone (GnRH) Agonists Do Not Protect Ovarian Function in Patients Undergoing Stem Cell Transplants. Cureus 2024; 16:e58387. [PMID: 38756303 PMCID: PMC11097921 DOI: 10.7759/cureus.58387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2024] [Indexed: 05/18/2024] Open
Abstract
Introduction Medical indications for fertility preservation include any malignancy, chronic illness, or disease that would require gonadotoxic chemotherapy or radiation (conditioning regimens), which would impede a woman's ability to conceive in the future. Thus, any patient who plans to undergo a gonadotoxic regimen is advised to cryopreserve oocytes or embryos, which can be used in the future at the patient's convenience. Attempts have been made to suppress ovarian function with gonadotropin-releasing hormone agonists (GnRH-a) to induce ovarian quiescence and, thereby, theoretically limit the gonadotoxic impact on the follicular pool. We explored the use of leuprolide (a type of GnRH-a) in preventing primary ovarian insufficiency (POI) in a cohort study of patients who underwent hematopoietic stem cell transplants (HSCT) at the National Institutes of Health (NIH); since the conditioning regimens for HSCT include cyclophosphamide and other gonadotoxic therapies, we hypothesized that GnRH-a would be ineffective in preventing POI. Methods We assessed patients who underwent fertility preservation prior to their stem cell transplant, as their follicular-stimulating hormone (FSH) levels were evaluated prior to and post-chemotherapy. Twenty-nine patients who underwent hormonal evaluation prior to and post-chemotherapy were included. The control group did not receive GnRH-a prior to chemotherapy, while the treatment group did receive GnRH-a pre-chemotherapy. Results Our data revealed that 80% of the control group had menopausal levels post-chemotherapy, while 91% of the treatment group still had menopausal levels post-chemotherapy (p=0.33). Conclusions Thus, our hypothesis that GnRH-a is ineffective in reducing the risk of POI in a cohort of patients who receive conditioning regimens for HSCT was confirmed.
Collapse
Affiliation(s)
- Ariel Benor
- Reproductive Endocrinology and Infertility, National Institutes of Health, Bethesda, USA
| | - Alan Decherney
- Reproductive Endocrinology and Infertility, National Institutes of Health, Bethesda, USA
| |
Collapse
|
7
|
Molinelli C, Jacobs F, Nader-Marta G, Borea R, Scavone G, Ottonello S, Fregatti P, Villarreal-Garza C, Bajpai J, Kim HJ, Puglisi S, de Azambuja E, Lambertini M. Ovarian Suppression: Early Menopause and Late Effects. Curr Treat Options Oncol 2024; 25:523-542. [PMID: 38478329 PMCID: PMC10997548 DOI: 10.1007/s11864-024-01190-8] [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: 02/07/2024] [Indexed: 04/06/2024]
Abstract
OPINION STATEMENT Around 90% of breast tumours are diagnosed in the early stage, with approximately 70% being hormone receptor-positive. The cornerstone of adjuvant therapy for early-stage hormone receptor-positive breast cancer is endocrine therapy, tailored according to disease stage, biological characteristics of the tumour, patient's comorbidities, preferences and age. In premenopausal patients with hormone receptor-positive breast cancer, ovarian function suppression is a key component of the adjuvant endocrine treatment in combination with an aromatase inhibitor or tamoxifen. Moreover, it can be used during chemotherapy as a standard strategy for ovarian function preservation in all breast cancer subtypes. In the metastatic setting, ovarian function suppression should be used in all premenopausal patients with hormone receptor-positive breast cancer to achieve a post-menopausal status. Despite its efficacy, ovarian function suppression may lead to several side effects that can have a major negative impact on patients' quality of life if not properly managed (e.g. hot flashes, depression, cognitive impairment, osteoporosis, sexual dysfunction, weight gain). A deep knowledge of the side effects of ovarian function suppression is necessary for clinicians. A correct counselling in this regard and proactive management should be considered a fundamental part of survivorship care to improve treatment adherence and patients' quality of life.
Collapse
Affiliation(s)
- Chiara Molinelli
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genoa, Italy
- Department of Medical Oncology, U.O. Clinical Di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Flavia Jacobs
- Humanitas Clinical and Research Center - IRCCS, Humanitas Cancer Center, Via Manzoni 56, 20089, Rozzano, Milan, Italy
| | - Guilherme Nader-Marta
- Academic Trials Promoting Team, Institut Jules Bordet and l'Université Libre de Bruxelles (U.L.B), 90, Rue Meylemeersch, 1070, Anderlecht, Brussels, Belgium
| | - Roberto Borea
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genoa, Italy
- Department of Medical Oncology, U.O. Clinical Di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Graziana Scavone
- Department of Medical Oncology, U.O. Clinical Di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Silvia Ottonello
- Department of Medical Oncology, U.O. Clinical Di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Piero Fregatti
- Department of Surgery, U.O. Senologia Chirurgica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Surgical Sciences and Integrated Diagnostic (DISC), School of Medicine, University of Genoa, 16132, Genoa, Italy
| | - Cynthia Villarreal-Garza
- Breast Cancer Center, Hospital Zambrano Hellion - TecSalud, Tecnologico de Monterrey, Monterrey, Mexico
| | - Jyoti Bajpai
- Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Ernest Borges Rd, Parel East, Parel, Mumbai, Maharashtra, 400012, India
| | - Hee Jeong Kim
- Division of Breast Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, South Korea
| | - Silvia Puglisi
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Evandro de Azambuja
- Academic Trials Promoting Team, Institut Jules Bordet and l'Université Libre de Bruxelles (U.L.B), 90, Rue Meylemeersch, 1070, Anderlecht, Brussels, Belgium
| | - Matteo Lambertini
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genoa, Italy.
- Department of Medical Oncology, U.O. Clinical Di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132, Genoa, Italy.
| |
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: 2.0] [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
|
Boutas I, Kontogeorgi A, Koufopoulos N, Dimas DT, Sitara K, Kalantaridou SN, Dimitrakakis C. Breast Cancer and Fertility Preservation in Young Female Patients: A Systematic Review of the Literature. Clin Pract 2023; 13:1413-1426. [PMID: 37987428 PMCID: PMC10660549 DOI: 10.3390/clinpract13060127] [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: 09/05/2023] [Revised: 10/20/2023] [Accepted: 11/08/2023] [Indexed: 11/22/2023] Open
Abstract
INTRODUCTION Breast cancer affects almost 1.5 million women worldwide below the age of 45 years each year. Many of these women will be advised to undergo adjuvant chemotherapy to minimize the risk of death or recurrence of the tumor. For these patients, chemotherapy is a known cause of infertility, as it can damage primordial follicles, which can lead to early menopause or premature ovarian insufficiency. This systematic review aims to synthesize the current evidence of the most suitable treatments for fertility preservation. METHODOLOGY This review was performed following the PRISMA guidelines. The authors conducted an extensive search from the last 15 years. Relevant studies were pursued in PubMed, Embase, and the Cochrane Library up until 31 July 2023. A total of seven eligible studies were identified. RESULTS From the reviewed literature, ovarian suppression with gonadotropin-releasing hormone agonists showed promising results in preserving fertility for breast cancer patients undergoing chemotherapy. Additionally, oocyte and embryo cryopreservation demonstrated successful outcomes, with embryo cryopreservation being the most effective option. Notably, the slow-freezing and vitrification methods were both effective in preserving embryos, with vitrification showing superior results in clinical-assisted reproductive technologies. Ovarian tissue cryopreservation emerged as a viable option for prepubertal girls and those unable to undergo conventional ovarian stimulation. The potential of in vitro maturation (IVM) as an alternative method presents a promising avenue for future fertility preservation research. DISCUSSION The most suitable treatments for fertility preservation in young patients is the temporary suppression with luteinizing hormone-releasing analogs, while the patient undergoes chemotherapy and cryopreservation. For cryopreservation, the physicians might deem it necessary to either cryopreserve ovarian tissue taken from the patient before any treatment or cryopreserve embryos/oocytes. Cryopreservation of oocytes and/or embryos is the most effective solution for fertility preservation in women of reproductive age, who have a sufficient ovarian reserve and are diagnosed with breast cancer, regardless of the histological type of the tumor. Because approximately 50% of young breast cancer patients are interested in becoming pregnant right after completion of therapy, the evolution and development of fertility preservation techniques promise to be very exciting.
Collapse
Affiliation(s)
- Ioannis Boutas
- Breast Unit, Rea Maternity Hospital, P. Faliro, 17564 Athens, Greece
| | - Adamantia Kontogeorgi
- Third Department of Obstetrics and Gynecology, Attikon University Hospital, National and Kapodistrian University of Athens, Rimini 1, 12462 Chaidari, Greece; (A.K.); (S.N.K.)
| | - Nektarios Koufopoulos
- Second Pathology Department, Attikon University Hospital, National and Kapodistrian University of Athens, Rimini 1, 12462 Chaidari, Greece;
| | - Dionysios T. Dimas
- Breast Unit, Athens Medical Center, Psychiko Clinic, 11525 Athens, Greece;
| | - Kyparissia Sitara
- Department of Internal Medicine, “Elpis” General Hospital, 11522 Athens, Greece;
| | - Sophia N. Kalantaridou
- Third Department of Obstetrics and Gynecology, Attikon University Hospital, National and Kapodistrian University of Athens, Rimini 1, 12462 Chaidari, Greece; (A.K.); (S.N.K.)
| | - Constantine Dimitrakakis
- First Department of Obstetrics and Gynecology, Alexandra University Hospital, National and Kapodistrian University of Athens, Lourou 4-2, 11528 Athens, Greece;
| |
Collapse
|
10
|
Zhao P, Guo C, Du H, Xiao Y, Su J, Wang X, Yeung WSB, Li G, Wang T. Chemotherapy-induced ovarian damage and protective strategies. HUM FERTIL 2023; 26:887-900. [PMID: 38054300 DOI: 10.1080/14647273.2023.2275764] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 10/14/2023] [Indexed: 12/07/2023]
Abstract
More than 9.2 million women worldwide suffer from cancer, and about 5% of them are at reproductive age. Chemotherapy-induced impairment of fertility affects the quality of life of these women. Several chemotherapeutic agents have been proven to cause apoptosis and autophagy by inducing DNA damage and cellular stress. Injuries to the ovarian stroma and micro-vessel network are also considered as pivotal factors resulting in ovarian dysfunction induced by chemotherapeutic agents. Primordial follicle pool over-activation may also be the mechanism inducing damage to the ovarian reserve. Although many studies have explored the mechanisms involved in chemotherapy-induced reproductive toxicity, the exact molecular mechanisms have not been elucidated. It is essential to understand the mechanisms involved in ovarian damage, in order to develop potential protective treatments to preserve fertility. In this article, we reviewed the current knowledge on the mechanism of chemotherapy-induced ovarian damage and possible protective strategies that prevent the ovary from such damages.
Collapse
Affiliation(s)
- Peikun Zhao
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, PR China
| | - Chenxi Guo
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, PR China
| | - Huijia Du
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, PR China
| | - Yuan Xiao
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, PR China
| | - Jiaping Su
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, PR China
| | - Xiaohui Wang
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, PR China
| | - Willian S B Yeung
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, PR China
| | - Guangxin Li
- Department of Breast and Thyroid Surgery, Peking University Shenzhen Hospital, Shenzhen, PR China
| | - Tianren Wang
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, PR China
| |
Collapse
|
11
|
Yuan Y, Zhang C, Lei X, Ren T, Chen H, Zhao Q. Gonadotropin-Releasing Hormone Agonists during Gonadal Chemotherapy for the Effect on Pregnancy Outcome and Ovarian Function in Premenopausal Patients with Breast Cancer: A Systematic Review and Meta-Analysis. Breast Care (Basel) 2023; 18:270-278. [PMID: 37900550 PMCID: PMC10601703 DOI: 10.1159/000528028] [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: 06/08/2022] [Accepted: 11/07/2022] [Indexed: 10/31/2023] Open
Abstract
Objectives The aim of this study was to evaluate the effects of gonadotropin-releasing hormone agonists (GnRHas) on pregnancy outcomes, premature ovarian failure (POF), menstrual recovery, disease-free survival (DFS), and adverse events in premenopausal breast cancer patients during gonadal chemotherapy. Methods We systematically searched PubMed, Cochrane Library, and Embase databases. The trials were eligible if they included premenopausal breast cancer patients treated with chemotherapy alone or with concurrent GnRHa and reported ovarian function recovery data. Heterogeneity for the eligible data was assessed, and a pooled risk ratio (RR) with 95% confidence interval (CI) was calculated. A meta-analysis was conducted using a fixed-effect model. Results Fifteen randomized controlled trials were included in this analysis. The results indicated that GnRHa combined with chemotherapy significantly increased pregnancy rates compared with chemotherapy alone (RR = 1.76; 95% CI: 1.16-2.67) and decreased rates of POF (RR = 0.42; 95% CI: 0.35-0.51). For secondary endpoints, the GnRHa group improved menstrual recovery rates (RR = 1.20; 95% CI: 1.11-1.30) and decreased the rate of amenorrhea 1-2 years after chemotherapy (RR = 0.50; 95% CI: 0.40-0.63). Furthermore, the 5-year DFS and overall survival (OS) rates were significantly improved in the GnRHa group. Conclusion For premenopausal breast cancer patients receiving gonadal toxic chemotherapy, adjuvant chemotherapy with GnRHa can better protect the ovarian function of patients, reduce the rate of POF and amenorrhea, and improve the pregnancy rate, menstrual recovery rate, DFS rate, and OS rate of patients.
Collapse
Affiliation(s)
- Yuan Yuan
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, China
| | - Chu Zhang
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Xueli Lei
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Tianshu Ren
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, China
| | - Han Chen
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, China
| | - Qingchun Zhao
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, China
| |
Collapse
|
12
|
White R, Wilson A, Bechman N, Keay SD, McAvan L, Quenby S, Odendaal J. Fertility preservation, its effectiveness and its impact on disease status in pre-menopausal women with breast cancer: A systematic review and meta-analysis. Eur J Obstet Gynecol Reprod Biol 2023; 287:8-19. [PMID: 37269752 DOI: 10.1016/j.ejogrb.2023.05.030] [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: 06/16/2022] [Revised: 05/14/2023] [Accepted: 05/21/2023] [Indexed: 06/05/2023]
Abstract
INTRODUCTION Preservation of reproductive function is a key concern for many premenopausal women with breast cancer, given the known gonadotoxic effects of treatments. The present systematic review aimed to investigate the effectiveness and safety of fertility preservation strategies in pre-menopausal women with breast cancer. METHODS Primary research assessing fertility preservation strategies of any type was identified. Markers of preservation of fertility including return of menstrual function, clinical pregnancy rates and live birth rates were selected as main outcome measures. An additional analysis of safety data was also performed. RESULTS Fertility preservation interventions were overall associated with higher fertility outcomes: with a pooled odds ratio 4.14 (95% CI 3.59-4.77) for any kind of fertility preservation intervention. This was seen both for return of menstruation and for clinical pregnancy rate, but not for live birth rates. Fertility preservation was associated with a reduced rate of disease recurrence (OR 0.63 (95% CI 0.49-0.81)), while there was no significant difference in disease free survival (OR 0.88 (95% CI 0.74-1.05)) or in overall survival (OR 0.9 (95% CI 0.74-1.10)) between the fertility preservation group and those who had not undergone fertility preservation. CONCLUSION Fertility preservation is both effective in preserving reproductive function, and safe with regard to disease recurrence, disease free survival and overall survival in premenopausal women with breast cancer.
Collapse
Affiliation(s)
- Rhiannon White
- Division of Biomedical Sciences, Clinical Sciences Research Laboratories, Warwick Medical School, University of Warwick, Coventry CV2 2DX, United Kingdom
| | - Anna Wilson
- Division of Biomedical Sciences, Clinical Sciences Research Laboratories, Warwick Medical School, University of Warwick, Coventry CV2 2DX, United Kingdom
| | - Natasha Bechman
- Division of Biomedical Sciences, Clinical Sciences Research Laboratories, Warwick Medical School, University of Warwick, Coventry CV2 2DX, United Kingdom
| | - Stephen D Keay
- University Hospitals Coventry & Warwickshire, Coventry CV2 2DX, United Kingdom
| | - Lucy McAvan
- University Hospitals Coventry & Warwickshire, Coventry CV2 2DX, United Kingdom
| | - Siobhan Quenby
- Division of Biomedical Sciences, Clinical Sciences Research Laboratories, Warwick Medical School, University of Warwick, Coventry CV2 2DX, United Kingdom; University Hospitals Coventry & Warwickshire, Coventry CV2 2DX, United Kingdom
| | - Joshua Odendaal
- Division of Biomedical Sciences, Clinical Sciences Research Laboratories, Warwick Medical School, University of Warwick, Coventry CV2 2DX, United Kingdom; University Hospitals Coventry & Warwickshire, Coventry CV2 2DX, United Kingdom.
| |
Collapse
|
13
|
Cerić T, Sokolović E, Hasanbegović B, Pašić A, Gojković Z, Mašić JV, Dukić N, Marijanović I, Abazović AM, Šišić I, Koprić D, Hammami M, Bajramović S, Delić T, Bešlija S. The Oncology Association of Bosnia and Herzegovina's recommendations for fertility preservation in oncologic patients. Bosn J Basic Med Sci 2022; 22:646-650. [PMID: 35348448 PMCID: PMC9519169 DOI: 10.17305/bjbms.2021.6977] [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: 01/23/2022] [Accepted: 02/27/2022] [Indexed: 11/24/2022] Open
Abstract
Malignancy is one of the major public health problems in Bosnia and Herzegovina. Along with breakthroughs in specific oncological therapy, improving the quality of life of cancer patients and management of therapy-induced side effects need to be recognized as a priority in the comprehensive cancer patient care. Fertility loss after cancer treatment is a field requiring special attention due to its various consequences on patients themselves. Although oncofertility is well-recognized area of oncology, low- to middle-income countries are facing issues with its implementation in everyday practice. Increased awareness about fertility preservation is of high priority for all specialists who participate in the medical care of cancer patients. The absence of a systemic solution and lack of expertise led to the founding of Fertility Preservation Working Group of the Oncology Association of Bosnia and Herzegovina. We have made recommendationsas an expert consensus with the ultimate goal of making the first step towards enhancement of oncofertility implementation in Bosnia and Herzegovina.
Collapse
Affiliation(s)
- Timur Cerić
- Clinic of Oncology, Clinical Center University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Emir Sokolović
- Clinic of Oncology, Clinical Center University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Berisa Hasanbegović
- Clinic of Oncology, Clinical Center University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Anes Pašić
- Clinic of Oncology, Clinical Center University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Zdenka Gojković
- Oncology Clinic, Clinical Center of Republic of Srpska, Banja Luka, Bosnia and Herzegovina
| | | | - Nikolina Dukić
- Department of Oncology, University Hospital Foča, Foča, Bosnia and Herzegovina
| | - Inga Marijanović
- Oncology Clinic, University Clinical Hospital Mostar, Mostar, Bosnia and Herzegovina
| | - Alma Mekić Abazović
- Department of Oncology, Cantonal Hospital Zenica, Zenica, Bosnia and Herzegovina
| | - Ibrahim Šišić
- Department of Oncology, Cantonal Hospital Zenica, Zenica, Bosnia and Herzegovina
| | - Dijana Koprić
- Department of Oncology, University Clinical Center Tuzla, Tuzla, Bosnia and Herzegovina
| | - Mustafa Hammami
- Department of Oncology, General hospital “Dr. Irfan Ljubijankić” Bihać, Bihać, Bosnia and Herzegovina
| | - Senad Bajramović
- Clinic of Urology, Clinical Center University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Taib Delić
- Polyclinic Sunce, Sarajevo, Bosnia and Herzegovina
| | - Semir Bešlija
- Clinic of Oncology, Clinical Center University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| |
Collapse
|
14
|
Li ZY, Dong YL, Cao XZ, Ren SS, Zhang Z. Gonadotropin-releasing hormone agonists for ovarian protection during breast cancer chemotherapy: a systematic review and meta-analysis. Menopause 2022; 29:1093-1100. [PMID: 35917530 PMCID: PMC9422762 DOI: 10.1097/gme.0000000000002019] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/17/2022] [Indexed: 11/25/2022]
Abstract
IMPORTANCE The increasing trend of delaying childbirth means that more women are being diagnosed with breast cancer before having given birth to their desired number of children. Although chemotherapy can significantly improve the prognosis of this population, it also causes ovarian damage, including premature ovarian insufficiency and infertility. Gonadotropin-releasing hormone agonists (GnRHa) have shown promising fertility protective activity in premenopausal women, but their clinical usage remains controversial. OBJECTIVE Here, we conducted a meta-analysis to assess the efficacy of GnRHa when administered concurrently with chemotherapy that included cyclophosphamide in the prevention of chemotherapy-induced ovarian damage in premenopausal women. EVIDENCE REVIEW An extensive literature search was performed using the PubMed, Embase, and Cochrane databases. Pooled odds ratios (ORs) with 95% confidence intervals (CIs) were determined. FINDINGS Eleven randomized controlled trials with a total of 1,219 participants were included in the analyses. A significantly higher number of women treated with GnRHa experienced the resumption of ovarian function after chemotherapy than those who did not receive this treatment (OR, 3.04; 95% CI, 1.87-4.94; P < 0.001). Regarding spontaneous pregnancy, a statistically significant difference was observed only in hormone receptor-negative participants (OR, 2.06; 95% CI, 1.03-4.11; P = 0.04). CONCLUSIONS AND RELEVANCE When treating premenopausal women with breast cancer, the administration of GnRHa concurrently with chemotherapy appeared to improve the resumption rate of ovarian function; however, the spontaneous pregnancy rate only improved in hormone receptor-negative patients. Thus, the use of GnRHa during chemotherapy may represent a feasible strategy for preserving ovarian function in women with breast cancer.
Collapse
Affiliation(s)
- Zhen-Yu Li
- From the Department of Breast Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China; and the
| | - Ying-Li Dong
- Department of Stomatology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Xiao-Zhong Cao
- From the Department of Breast Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China; and the
| | - Sha-Sha Ren
- From the Department of Breast Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China; and the
| | - Zhen Zhang
- From the Department of Breast Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China; and the
| |
Collapse
|
15
|
Torrisi R, Basilico V, Giordano L, Caruso M, Musolino A, Monari MN, Carnaghi C, Santoro A. Is Anti-Müllerian Hormone a Marker of Ovarian Reserve in Young Breast Cancer Patients Receiving a GnRH Analog during Chemotherapy? Breast Care (Basel) 2022; 17:10-15. [PMID: 35355699 PMCID: PMC8914273 DOI: 10.1159/000514445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 01/14/2021] [Indexed: 02/03/2023] Open
Abstract
Introduction Anti-Müllerian hormone (AMH) is the most reliable biomarker of ovarian reserve; however, its role in predicting ovarian recovery after chemotherapy is unclear. Administration of a GnRH analog (GnRHa) during chemotherapy significantly reduces the ovarian failure rate and increases the pregnancy rate. The available data on the behavior of AMH during concurrent administration of chemotherapy and GnRHa are inconsistent. We investigated whether concurrent administration of triptorelin and adjuvant chemotherapy might reduce the expected drop of AMH. Methods Eligible patients were premenopausal women aged <40 years, with a diagnosis of early breast cancer, and candidates to 4-8 cycles of adjuvant chemotherapy. Triptorelin (3.75 mg i.m.) was started before chemotherapy and administered every 4 weeks thereafter. The principal endpoint was the proportion of patients with an AMH percent change ≤50% between 12 months after chemotherapy and basal levels. The secondary endpoint was the proportion of patients achieving postchemotherapy AMH levels above the threshold of 0.2 ng/mL. Results Fifty patients were enrolled, 31 of whom had blood samples available at baseline and 1 year after the end of chemotherapy. AMH decreased to nearly undetectable levels after chemotherapy and recovered after 12 months, but they did not exceed 1 tenth of the pretreatment levels. As for the secondary endpoint, 15 of the 31 patients recovered AMH levels above the threshold. Conclusions This study did not reach its principal endpoint; however, the rate of 48% of patients who recovered AMH above threshold levels favorably compared with those in studies without concurrent GnRHa, supporting a better recovery of AMH with triptorelin.
Collapse
Affiliation(s)
- Rosalba Torrisi
- IRCCS Humanitas Research Hospital Medical Oncology Unit, Rozzano, Italy,*Rosalba Torrisi, IRCCS Humanitas Research Hospital Medical Oncology Unit, Department of Medical Oncology and Hematology, Via Manzoni 56, IT–20089 Rozzano (Italy),
| | - Vera Basilico
- Medical Oncology Unit, Istituto Clinico Mater Domini Humanitas, Castellanza, Italy
| | - Laura Giordano
- IRCCS Humanitas Research Hospital Medical Oncology Unit, Rozzano, Italy
| | - Michele Caruso
- Medical Oncology Humanitas Centro Catanese di Oncologia, Catania, Italy
| | | | - Marta Noemi Monari
- Laboratory of Clinical Analysis, Humanitas Clinical and Research Center, IRCCS, Rozzano, Italy
| | - Carlo Carnaghi
- Medical Oncology Unit, Istituto Clinico Mater Domini Humanitas, Castellanza, Italy,Medical Oncology Humanitas Centro Catanese di Oncologia, Catania, Italy
| | - Armando Santoro
- IRCCS Humanitas Research Hospital Medical Oncology Unit, Rozzano, Italy,Department of Biomedical Sciences, Humanitas University, Rozzano, Italy
| |
Collapse
|
16
|
McClam M, Xiao S. Preserving Oocytes in Oncofertility†. Biol Reprod 2022; 106:328-337. [PMID: 35040934 PMCID: PMC8862718 DOI: 10.1093/biolre/ioac008] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/09/2021] [Accepted: 01/13/2022] [Indexed: 01/19/2023] Open
Abstract
The prodigious rise of cancer survival rates enables many cancer survivors to live long lives. Therefore, the side effects of cancer treatments as well as the long-term quality of life after cancer have become more relevant. Ovarian toxicity is a major off-target effect of anticancer agents for childhood and young adult female cancer patients. Both chemotherapy and irradiation have been demonstrated to damage the ovary and increase the risks of premature ovarian failure (POF), early menopause, ovarian endocrine disorders, and sub- or infertility. Oncofertility is an emerging and multidisciplinary research and medical field that focuses on providing cancer patients with fertility preservation options. Oocyte quality and quantity are one of the most important factors to determine women's fertility success; therefore, preserving oocytes is paramount for maintaining the ability of young female cancer patients' reproduction after their recovery. This review summarizes peer-reviewed literature on current oocyte preservation options in oncofertility. We describe in-depth oocyte and embryo cryopreservation, ovarian suppression, ovarian tissue cryopreservation, in vitro maturation, ovarian transposition, and adjuvant therapy. Further, we discuss current guidelines and practices of female fertility preservation that cover preserving oocytes.
Collapse
Affiliation(s)
- Maria McClam
- Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Shuo Xiao
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Environmental and Occupational Health Sciences Institute (EOHSI), Rutgers University, Piscataway, NJ, USA,Correspondence: 170 Frelinghuysen Rd, Piscataway, NJ 08854, USA. Tel: + 1-848-445-3729; E-mail:
| |
Collapse
|
17
|
Graft-versus-host disease in the female genital tract: a prospective cohort study. Arch Gynecol Obstet 2021; 305:1551-1558. [DOI: 10.1007/s00404-021-06330-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 11/05/2021] [Indexed: 10/19/2022]
|
18
|
Borges VF. The Expanded Role of Ovarian Suppression for Young Women's Breast Cancer: An Era of Patient-Tailored Decision Making. J Natl Cancer Inst 2021; 114:342-344. [PMID: 34850044 DOI: 10.1093/jnci/djab214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 11/16/2021] [Indexed: 11/12/2022] Open
Affiliation(s)
- Virginia F Borges
- Division of Medical Oncology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
19
|
Chung EH, Acharya CR, Harris BS, Acharya KS. Development of a fertility risk calculator to predict individualized chance of ovarian failure after chemotherapy. J Assist Reprod Genet 2021; 38:3047-3055. [PMID: 34495476 PMCID: PMC8609057 DOI: 10.1007/s10815-021-02311-0] [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: 06/06/2021] [Accepted: 09/01/2021] [Indexed: 10/20/2022] Open
Abstract
PURPOSE To develop an innovative machine learning (ML) model that predicts personalized risk of primary ovarian insufficiency (POI) after chemotherapy for reproductive-aged women. Currently, individualized prediction of a patient's risk of POI is challenging. METHODS Authors of published studies examining POI after gonadotoxic therapy were contacted, and six authors shared their de-identified data (N = 435). A composite outcome for POI was determined for each patient and validated by 3 authors. The primary dataset was partitioned into training and test sets; random forest binary classifiers were trained, and mean prediction scores were computed. Institutional data collected from a cross-sectional survey of cancer survivors (N = 117) was used as another independent validation set. RESULTS Our model predicted individualized risk of POI with an accuracy of 88% (area under the ROC 0.87, 95% CI: 0.77-0.96; p < 0.001). Mean prediction scores for patients who developed POI and who did not were 0.60 and 0.38 (t-test p < 0.001), respectively. Highly weighted variables included age, chemotherapy dose, prior treatment, smoking, and baseline diminished ovarian reserve. CONCLUSION We developed an ML-based model to estimate personalized risk of POI after chemotherapy. Our web-based calculator will be a user-friendly decision aid for individualizing risk prediction in oncofertility consultations.
Collapse
Affiliation(s)
- Esther H Chung
- Division of Reproductive Endocrinology & Infertility, Department of Obstetrics and Gynecology, Duke University, 200 Trent Drive (Baker House 236), Durham, NC, 27710, USA.
| | - Chaitanya R Acharya
- Duke Center for Applied Therapeutics, Department of Surgery, Durham, NC, 27710, USA
| | - Benjamin S Harris
- Division of Reproductive Endocrinology & Infertility, Department of Obstetrics and Gynecology, Duke University, 200 Trent Drive (Baker House 236), Durham, NC, 27710, USA
| | - Kelly S Acharya
- Division of Reproductive Endocrinology & Infertility, Department of Obstetrics and Gynecology, Duke University, 200 Trent Drive (Baker House 236), Durham, NC, 27710, USA
| |
Collapse
|
20
|
Odeh OM, Awwad J, Khalife D, Ghunaim S. The use of GnRH analogs in preserving ovarian function during chemotherapy. MIDDLE EAST FERTILITY SOCIETY JOURNAL 2021. [DOI: 10.1186/s43043-021-00088-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
The literature has always been controversial on the use of gonadotropin-releasing hormone agonists in preserving fertility in women of childbearing age after chemotherapy; thereby, in this article, we will be discussing its use in preserving fertility.
Main body of abstract
When it comes to preserving fertility, it is crucial to consider all available options in this topic due to its very sensitive nature, thereby we have found that while a lot of trials favor the use of gonadotropin-releasing hormone agonists, the lack of proper follow-up and long-term trials renders its use highly debatable, and since the longest follow-up trial showed non-significant results, it also opens the floor for debate on whether this short-term benefit is worth adding another drug to the regimen or not.
Short conclusion
As described in this review, while the use of gonadotropin-releasing hormone agonists is beneficial in a lot of studies, the lack of long-term reports still makes its use debatable, thereby more trials should be done.
Collapse
|
21
|
Arecco L, Ruelle T, Martelli V, Boutros A, Latocca MM, Spinaci S, Marrocco C, Massarotti C, Lambertini M. How to Protect Ovarian Function before and during Chemotherapy? J Clin Med 2021; 10:jcm10184192. [PMID: 34575299 PMCID: PMC8467797 DOI: 10.3390/jcm10184192] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 08/29/2021] [Accepted: 09/03/2021] [Indexed: 12/22/2022] Open
Abstract
A significant number of women receive a cancer diagnosis before their age of natural menopause. Among these patients, the most frequent neoplasms are breast cancer, gynecological, and hematological malignancies. Premature ovarian insufficiency and infertility are among the most feared short- to long-term consequences of anticancer treatments in premenopausal patients. Both patient- and treatment-related characteristics are key factors in influencing the risk of gonadotoxicity with the use of chemotherapy. The cryopreservation of oocytes/embryos is a standard strategy for fertility preservations offered to young women interested in future family planning, but it does not allow gonadal function protection during chemotherapy. Ovarian suppression with gonadotropin-releasing hormone agonist (GnRHa) during chemotherapy is now recommended as an option to reduce the risk of gonadotoxicity in order to avoid the negative consequences of premature ovarian insufficiency in premenopausal women receiving cytotoxic therapy, including those not interested in fertility preservation. This review summarizes the risk of treatment-induced gonadotoxicity in premenopausal patients and the evidence available on the protective role of administering GnRHa during chemotherapy to preserve ovarian function.
Collapse
Affiliation(s)
- Luca Arecco
- U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (L.A.); (M.M.L.); (C.M.)
- Department of Internal Medicine and Medical Sciences (DiMI), School of Medicine, University of Genova, 16132 Genova, Italy; (T.R.); (V.M.); (A.B.)
| | - Tommaso Ruelle
- Department of Internal Medicine and Medical Sciences (DiMI), School of Medicine, University of Genova, 16132 Genova, Italy; (T.R.); (V.M.); (A.B.)
- U.O. Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Valentino Martelli
- Department of Internal Medicine and Medical Sciences (DiMI), School of Medicine, University of Genova, 16132 Genova, Italy; (T.R.); (V.M.); (A.B.)
- U.O. Oncologia Medica 1, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Andrea Boutros
- Department of Internal Medicine and Medical Sciences (DiMI), School of Medicine, University of Genova, 16132 Genova, Italy; (T.R.); (V.M.); (A.B.)
- U.O. Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Maria Maddalena Latocca
- U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (L.A.); (M.M.L.); (C.M.)
- Department of Internal Medicine and Medical Sciences (DiMI), School of Medicine, University of Genova, 16132 Genova, Italy; (T.R.); (V.M.); (A.B.)
| | - Stefano Spinaci
- Division of Breast Surgery, Ospedale Villa Scassi ASL3, 16149 Genova, Italy;
| | - Camilla Marrocco
- U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (L.A.); (M.M.L.); (C.M.)
| | - Claudia Massarotti
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), School of Medicine, University of Genova, 16132 Genova, Italy;
- Academic Unit of Obstetrics and Gynaecology, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Matteo Lambertini
- U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (L.A.); (M.M.L.); (C.M.)
- Department of Internal Medicine and Medical Sciences (DiMI), School of Medicine, University of Genova, 16132 Genova, Italy; (T.R.); (V.M.); (A.B.)
- Correspondence: ; Tel.: +39-010-555-4254; Fax: +39-010-555-6536
| |
Collapse
|
22
|
Wang S, Pei L, Hu T, Jia M, Wang S. Protective effect of goserelin on ovarian reserve during (neo)adjuvant chemotherapy in young breast cancer patients: a prospective cohort study in China. Hum Reprod 2021; 36:976-986. [PMID: 33411897 DOI: 10.1093/humrep/deaa349] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 11/15/2020] [Indexed: 12/15/2022] Open
Abstract
STUDY QUESTION Does goserelin, a GnRH agonist, have a protective effect in young breast cancer patients in terms of ovarian reserve markers anti-Müllerian hormone (AMH) and antral follicle count (AFC) during chemotherapy? SUMMARY ANSWER Compared with chemotherapy alone, concurrent goserelin is associated with a higher probability of ovarian reserve recovery at 1 year after chemotherapy. WHAT IS KNOWN ALREADY Previous studies on the administration of goserelin to protect ovarian function during chemotherapy have produced conflicting results because of the endpoint used, namely, chemotherapy-induced amenorrhoea. Reproductive medicine specialists consider AMH and AFC as the most sensitive ovarian reserve markers; however, they have never been used as biomarkers to assess the potential protective effects on ovarian reserve of goserelin during chemotherapy. STUDY DESIGN, SIZE, DURATION This was a prospective cohort study in which patients were assigned to receive (neo)adjuvant chemotherapy with goserelin (the goserelin group) or without goserelin (the control group) according to each patient's preference. Of 242 breast cancer patients enrolled between December 2015 and November 2019, 76 in control group and 73 in goserelin group were able to be assessed at 1 year after chemotherapy. PARTICIPANTS/MATERIALS, SETTING, METHODS Premenopausal patients with a regular menstrual cycle and aged 18-45 years were eligible for enrolment if they were newly diagnosed with stages I-III breast cancer for which treatment with adjuvant or neoadjuvant chemotherapy was planned. Each patient in the goserelin group was given a subcutaneous dose of 3.6 mg at least 1 week before the first cycle of chemotherapy and then every 4 weeks for the duration of chemotherapy. Ovarian reserve markers and menstrual status were evaluated before and after chemotherapy in the two treatment groups. The primary endpoint was the AMH recovery rate, the secondary endpoints were the recovery rates of AFC, estradiol (E2), follicle-stimulating hormone (FSH) and menstruation. MAIN RESULTS AND THE ROLE OF CHANCE Among 149 patients (76 in the control group and 73 in the goserelin group) with complete data at 1 year after chemotherapy, the adjusted recovery rate of AMH was 46.5% and 21.8% in the goserelin group and control group, respectively (odds ratio: 3.08; P = 0.002). The trends in AFC and FSH recovery rates were consistent with that in AMH recovery rate. Notably, AMH levels remained low in 41.3% of patients whose menstrual activity had resumed. LIMITATIONS, REASONS FOR CAUTION Randomisation was not performed because of ethical considerations, so selection bias was inevitable, although propensity score weighting was done. The study was also underpowered because 21.5% (52/242) of enrolled patients received GnRH agonist-containing endocrine therapy and could not be analysed at 1 and 2 years after chemotherapy. WIDER IMPLICATIONS OF THE FINDINGS Our results indicate that co-administration of goserelin with chemotherapy provides obvious ovarian reserve protection in these young breast cancer patients. We expect that these results will be applicable in clinical practice for young breast cancer patients. STUDY FUNDING/COMPETING INTEREST(S) This study was funded by the National Key R&D Program of China No. 2016YFC0901302, by the Research and Development Fund of Peking University People's Hospital No. RD2014-13, RDY2017-19 and by AstraZeneca. The authors have no disclosures. TRIAL REGISTRATION NUMBER NCT02430103.
Collapse
Affiliation(s)
- Siyuan Wang
- Department of Breast Surgery, Peking University People's Hospital, Beijing, China
| | - Lin Pei
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Taobo Hu
- Department of Breast Surgery, Peking University People's Hospital, Beijing, China
| | - Mei Jia
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Shu Wang
- Department of Breast Surgery, Peking University People's Hospital, Beijing, China
| |
Collapse
|
23
|
Dolmans MM, Taylor HS, Rodriguez-Wallberg KA, Blumenfeld Z, Lambertini M, von Wolff M, Donnez J. Utility of gonadotropin-releasing hormone agonists for fertility preservation in women receiving chemotherapy: pros and cons. Fertil Steril 2021; 114:725-738. [PMID: 33040981 DOI: 10.1016/j.fertnstert.2020.08.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 08/06/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Marie-Madeleine Dolmans
- Pôle de Gynécologie, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium; Gynecology Department, Cliniques Universitaires Saint Luc, Brussels, Belgium
| | - Hugh S Taylor
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut
| | - Kenny A Rodriguez-Wallberg
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Reproductive Medicine, Division of Gynecology and Reproduction, Karolinska University Hospital, Stockholm, Sweden
| | - Zeev Blumenfeld
- Department of Reproductive Endocrinology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Matteo Lambertini
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
| | | | - Jacques Donnez
- Société de Recherche pour l'Infertilité, Brussels, Belgium; Catholic University of Louvain, Brussels, Belgium.
| |
Collapse
|
24
|
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: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [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
|
25
|
Lee JH, Choi YS. The role of gonadotropin-releasing hormone agonists in female fertility preservation. Clin Exp Reprod Med 2021; 48:11-26. [PMID: 33648041 PMCID: PMC7943347 DOI: 10.5653/cerm.2020.04049] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 10/13/2020] [Indexed: 11/19/2022] Open
Abstract
Advances in anticancer treatments have resulted in increasing survival rates among cancer patients. Accordingly, the quality of life after treatment, particularly the preservation of fertility, has gradually emerged as an essential consideration. Cryopreservation of embryos or unfertilized oocytes has been considered as the standard method of fertility preservation among young women facing gonadotoxic chemotherapy. Other methods, including ovarian suppression and ovarian tissue cryopreservation, have been considered experimental. Recent large-scale randomized controlled trials have demonstrated that temporary ovarian suppression using gonadotropin-releasing hormone agonists (GnRHa) during chemotherapy is beneficial for preventing chemotherapy-induced premature ovarian insufficiency in breast cancer patients. It should also be emphasized that GnRHa use during chemotherapy does not replace established fertility preservation methods. All young women facing gonadotoxic chemotherapy should be counseled about and offered various options for fertility preservation, including both GnRHa use and cryopreservation of embryos, oocytes, and/or ovarian tissue.
Collapse
Affiliation(s)
- Jae Hoon Lee
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.,Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Young Sik Choi
- Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Korea.,Department of Obstetrics and Gynecology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
26
|
Chelariu-Raicu A, Cobb LP, Gershenson DM. Fertility preservation in rare ovarian tumors. Int J Gynecol Cancer 2021; 31:432-441. [PMID: 33649010 DOI: 10.1136/ijgc-2020-001775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 09/30/2020] [Indexed: 12/22/2022] Open
Abstract
Although gynecologic cancers usually affect older women, a significant proportion of patients with rare ovarian tumors are of reproductive age. In a young patient who presents with a pelvic mass, a primary consideration should be the probability of a malignancy. If there is any suspicion of a cancer diagnosis, the patient should be referred to a gynecologic oncologist. Key factors in clinical management include assessment of preoperative studies (physical examination, tumor markers, and imaging) to determine the likelihood of a malignancy, appropriate preoperative counseling (including discussion of fertility preservation), choice of surgical approach (minimally invasive vs open), frozen section examination by a gynecologic pathologist, and intraoperative decision making. Fortunately, the clinical features of several rare ovarian tumors are compatible with fertility preservation. These characteristics include a high proportion of stage I disease and unilateral ovarian involvement for most rare histotypes. Once a final diagnosis of a rare ovarian tumor is determined, further clinical management may include the need for further studies, possible referral to a fertility expert, consideration of further surgery (if the initial surgery was incomplete), and recommendations for postoperative therapy. This article reviews the literature on fertility preservation in the context of the treatment of several rare ovarian tumor subtypes, including malignant germ cell tumors, sex cord-stromal tumors, borderline tumors, low grade serous carcinoma, clear cell carcinoma, mucinous carcinoma, and small cell carcinoma of the hypercalcemic type.
Collapse
Affiliation(s)
- Anca Chelariu-Raicu
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany
| | - Lauren P Cobb
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - David M Gershenson
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
27
|
Qu J, Li Y, Liao S, Yan J. The Effects of Negative Elements in Environment and Cancer on Female Reproductive System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1300:283-313. [PMID: 33523439 DOI: 10.1007/978-981-33-4187-6_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
With the development of human society, factors that contribute to the impairment of female fertility is accumulating. Lifestyle-related risk factors, occupational risk factors, and iatrogenic factors, including cancer and anti-cancer treatments, have been recognized with their negative effects on the function of female reproductive system. However, the exact influences and their possible mechanism have not been elucidated yet. It is impossible to accurately estimate the indexes of female fertility, but many researchers have put forward that the general fertility has inclined through the past decades. Thus the demand for fertility preservation has increased more and more dramatically. Here we described some of the factors which may influence female reproductive system and methods for fertility preservation in response to female infertility.
Collapse
Affiliation(s)
- Jiangxue Qu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Yuehan Li
- Department of Gynaecology and Obstetrics, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shujie Liao
- Department of Gynaecology and Obstetrics, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Jie Yan
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
28
|
Yamazaki R, Ono M, Sugie T, Inokuchi M, Ishikawa S, Iizuka T, Masumoto S, Myojo S, Uchida S, Horie A, Matsuzaki T. Nationwide survey of Japanese breast oncology and reproductive endocrinology departments about the impact of breast cancer treatment on fertility. J Obstet Gynaecol Res 2020; 46:2488-2496. [PMID: 33063425 DOI: 10.1111/jog.14469] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 08/26/2020] [Indexed: 01/19/2023]
Abstract
The purpose of this study is to assess the impact of breast cancer treatment on the reproductive potential. We conducted a nationwide survey of breast oncology and reproductive endocrinology and infertility (REI) departments using a questionnaire designed to assess the impact of breast cancer treatment on fertility. We received responses from 312 breast oncology departments (response rate, 31.9%) and 541 REI departments (response rate, 50.9%). The most common method of achieving pregnancy reported by breast oncology departments was natural insemination (69.6%), followed by assisted reproductive technology ( 15.6%) and intrauterine insemination (IUI; 14.8%). The most common method of achieving pregnancy reported by REI departments was conventional in vitro fertilization and/or intracytoplasmic sperm injection (51.0%), followed by natural insemination with or without ovulation induction (40.0%) and IUI (8.0%). The overall pregnancy rate for patients who underwent treatment for infertility at REI departments after breast cancer treatment was 39.0%. Vast patients who experienced breast cancer treatments conceived mainly by natural insemination based on the data from breast oncology departments. On the other hand, 61.0% of the patients who visited REI departments presumably due to infertility by natural insemination did not conceive even by infertility treatments with exclusive knowledge in REI departments.
Collapse
Affiliation(s)
- Rena Yamazaki
- Department of Obstetrics and Gynecology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Masanori Ono
- Department of Obstetrics and Gynecology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Tomoharu Sugie
- Department of Breast Surgery, Kansai Medical University Hospital, Kansai, Japan
| | - Masafumi Inokuchi
- Department of Breast and Endocrine Surgery, Kanazawa Medical University, Kanazawa, Japan
| | - Satoko Ishikawa
- Department of Breast Oncology, Division of Cancer Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Takashi Iizuka
- Department of Obstetrics and Gynecology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Sakiko Masumoto
- Department of Obstetrics and Gynecology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Subaru Myojo
- Department of Obstetrics and Gynecology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Soko Uchida
- Department of Gynecology, National Hospital Organization Fukuokahigashi Medical Center, Koga, Japan
| | - Akihito Horie
- Department of Obstetrics and Gynecology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Toshiya Matsuzaki
- Department of Obstetrics and Gynecology, Yoshinogawa Medical Center, Yoshinogawa, Japan
| |
Collapse
|
29
|
Arecco L, Perachino M, Damassi A, Latocca MM, Soldato D, Vallome G, Parisi F, Razeti MG, Solinas C, Tagliamento M, Spinaci S, Massarotti C, Lambertini M. Burning Questions in the Oncofertility Counseling of Young Breast Cancer Patients. Breast Cancer (Auckl) 2020; 14:1178223420954179. [PMID: 32952399 PMCID: PMC7476336 DOI: 10.1177/1178223420954179] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 08/10/2020] [Indexed: 12/28/2022] Open
Abstract
The improved prognosis of breast cancer patients makes survivorship issues an area of crucial importance. In this regard, an increased attention is needed toward the development of potential anticancer treatment-related long-term side-effects, including gonadal failure and infertility in young women. Therefore, fertility preservation and family planning are crucial issues to be addressed in all young women of reproductive age with newly diagnosed cancer. Despite a growing availability of data on the efficacy and safety of fertility preservation options and the fact that conceiving after prior history of breast cancer has become more accepted over time, there are still several gray zones in this field so that many physicians remain uncomfortable to deal with these topics. The purpose of this review is to answer some of the most controversial questions frequently asked by patients during their oncofertility counseling, in order to provide a detailed and up-to-date overview on the evidence available in this field to physicians involved in the care of young women with breast cancer.
Collapse
Affiliation(s)
- Luca Arecco
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, University of Genova, Genova, Italy
| | - Marta Perachino
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, University of Genova, Genova, Italy
| | - Alessandra Damassi
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, University of Genova, Genova, Italy
| | - Maria Maddalena Latocca
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, University of Genova, Genova, Italy
| | - Davide Soldato
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, University of Genova, Genova, Italy
| | - Giacomo Vallome
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, University of Genova, Genova, Italy
| | - Francesca Parisi
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, University of Genova, Genova, Italy
| | - Maria Grazia Razeti
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, University of Genova, Genova, Italy
| | - Cinzia Solinas
- Department of Medical Oncology, Azienda Tutela della Salute Sardegna, Hospital A.Segni Ozieri, Sassari, Italy
| | - Marco Tagliamento
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, University of Genova, Genova, Italy
| | - Stefano Spinaci
- Division of Breast Surgery, Ospedale Villa Scassi e ASL3, Genova, Italy
| | - Claudia Massarotti
- Physiopathology of Human Reproduction Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Matteo Lambertini
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, University of Genova, Genova, Italy
| |
Collapse
|
30
|
Lee DY, Kim JY, Yu J, Kim SW. Prediction of Successful Ovarian Protection Using Gonadotropin-Releasing Hormone Agonists During Chemotherapy in Young Estrogen Receptor-Negative Breast Cancer Patients. Front Oncol 2020; 10:863. [PMID: 32656076 PMCID: PMC7326007 DOI: 10.3389/fonc.2020.00863] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 05/01/2020] [Indexed: 01/13/2023] Open
Abstract
Background: It is important to identify factors predicting successful ovarian protection using gonadotropin-releasing hormone (GnRH) agonists during chemotherapy. However, only a few studies have prospectively assessed this issue in young breast cancer patients. Objective: This study evaluated the predictive factors for successful ovarian protection with GnRH agonists during chemotherapy in young estrogen receptor-negative breast cancer patients. Materials and Methods: This prospective study analyzed 67 estrogen receptor-negative breast cancer patients ≤40 years of age who were longitudinally assessed after receiving GnRH agonists during cyclophosphamide-based chemotherapy for ovarian protection. Associations between clinical characteristics or pretreatment hormones and successful ovarian protection [resumption of menstruation and anti-Müllerian hormone (AMH) ≥1 ng/ml]. Results: The mean age and pretreatment serum level of AMH were 33.2 years and 4.57 ng/ml, respectively. At 12 months after the completion of chemotherapy, most women (97%) experienced the resumption of menstruation. However, the proportion of patients with AMH ≥1 ng/ml at 12 months was 70.1%. In multivariate analyses, only the pretreatment serum AMH level (P < 0.001) was predictive for AMH ≥1 ng/ml at 12 months. Receiver operating characteristic curve analyses of pretreatment AMH exhibited an area under the curve of 0.866 (95% CI = 0.777–0.955) for AMH ≥1 ng/ml at 12 months. The cutoff value for the prediction of AMH concentration ≥1 ng/ml at 12 months was 2.87 ng/ml of pretreatment AMH with a sensitivity of 0.87 and a specificity of 0.75. Conclusions: Pretreatment AMH (2.87 ng/ml) is a useful predictor for AMH ≥1 ng/ml at 12 months after receiving GnRH agonists in young estrogen receptor-negative breast cancer patients. This finding can help improve decision-making regarding fertility preservation.
Collapse
Affiliation(s)
- Dong-Yun Lee
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Ji-Yeon Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jonghan Yu
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Seok Won Kim
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| |
Collapse
|
31
|
Yu B, Davidson NE. Gonadotropin-Releasing Hormone (GnRH) Agonists for Fertility Preservation: Is POEMS the Final Verse? J Natl Cancer Inst 2020; 111:107-108. [PMID: 30371813 DOI: 10.1093/jnci/djy188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 09/11/2018] [Indexed: 12/28/2022] Open
Affiliation(s)
- Bo Yu
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA
| | - Nancy E Davidson
- Seattle Cancer Care Alliance, Fred Hutchinson Cancer Research Center, Department of Medicine, University of Washington, Seattle, WA
| |
Collapse
|
32
|
Klipstein S, Fallat ME, Savelli S. Fertility Preservation for Pediatric and Adolescent Patients With Cancer: Medical and Ethical Considerations. Pediatrics 2020; 145:peds.2019-3994. [PMID: 32071259 DOI: 10.1542/peds.2019-3994] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Many cancers presenting in children and adolescents are curable with surgery, chemotherapy, and/or radiotherapy. Potential adverse consequences of treatment include sterility, infertility, or subfertility as a result of gonad removal, damage to germ cells as a result of adjuvant therapy, or damage to the pituitary and hypothalamus or uterus as a result of irradiation. In recent years, treatment of solid tumors and hematologic malignancies has been modified in an attempt to reduce damage to the gonadal axis. Simultaneously, advances in assisted reproductive technology have led to new possibilities for the prevention and treatment of infertility. This clinical report reviews the medical aspects and ethical considerations that arise when considering fertility preservation in pediatric and adolescent patients with cancer.
Collapse
Affiliation(s)
- Sigal Klipstein
- Department of Obstetrics and Gynecology, Pritzker School of Medicine, University of Chicago, Chicago, Illinois; .,InVia Fertility Specialists, Chicago, Illinois
| | - Mary E Fallat
- Division of Pediatric Surgery, Hiram C. Polk Jr MD Department of Surgery, University of Louisville, Louisville, Kentucky; and
| | - Stephanie Savelli
- Division of Pediatric Hematology/Oncology, Akron Children's Hospital, Akron, Ohio
| | | | | | | |
Collapse
|
33
|
Durrani S, Heena H. Controversies Regarding Ovarian Suppression and Infertility in Early Stage Breast Cancer. Cancer Manag Res 2020; 12:813-817. [PMID: 32104064 PMCID: PMC7008199 DOI: 10.2147/cmar.s231524] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 01/15/2020] [Indexed: 01/24/2023] Open
Abstract
A common side effect of chemotherapy in breast cancer is early menopause in premenopausal patients, which is mainly a result of an indirect form of ovarian ablation, and is associated with substantial impairment of quality of life. Suppressing the production of ovarian estrogen has been shown to reduce the recurrence of hormone receptor-positive early breast cancer in premenopausal women, but whether it has an added advantage over tamoxifen is being discussed. Types of permanent ablation of the ovarian function include surgical oophorectomy and radiation-induced ovarian failure. Both are associated with similar response rates in hormone receptor-positive metastatic breast cancer. Medical castration with luteinizing hormone-releasing hormone analogs (LHRHa) has the benefit of being a reversible approach. Another advantage that premenopausal patients who wish to reduce the risk of developing premature ovarian insufficiency induced by chemotherapy may be offered LHRHa irrespective of whether they desire pregnancy and their age at diagnosis. This also helps reduce the risk of menopausal signs and symptoms as well as the loss of bone density in the long-term, which are primary concerns for women. This is of utmost importance to premenopausal women who do not want to conceive after treatment or are not candidates for fertility preservation strategies because of age. It should be emphasized that for women who are interested in fertility preservation, gamete cryopreservation remains the first option, and LHRHa is not an alternative. During chemotherapy, however, temporary ovarian suppression with LHRHa may be given to women who either have no access to a fertility clinic or who have declined chemotherapy or have contraindications.
Collapse
Affiliation(s)
- Sajid Durrani
- Comprehensive Cancer Center, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Humariya Heena
- Research Center, King Fahad Medical City, Riyadh, Saudi Arabia
| |
Collapse
|
34
|
Soussoko M, Salleron J, Desandes E, Lesur A. [Breast cancer management before 40 years: what change in one decade? A retrospective study at the "Institut de cancérologie de Lorraine": 2002-2012]. Bull Cancer 2020; 106:S60-S74. [PMID: 32008740 DOI: 10.1016/s0007-4551(20)30049-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
INTRODUCTION Breast cancer of young women has medico-psychological issues, particularly in the perspective of "after cancer" period. The objective of the study was to analyze the changes in management, in a decade including the publication of Cancer Plans I and II. METHODS This retrospective unicentric study involved two populations of women aged 40 and treated at the "Institut de cancérologie de Lorraine" in 2002-2004 (P1) and 2012-2014 (P2). Epidemiological-demographic, histological, diagnostic and therapeutic sequences were analyzed. RESULTS 216 patients were included. The mean age was 35.7 years (range 22-40), with no significant epidemiological difference, except for an increased rate of obesity in P2. Preoperative biopsies became almost systematic and the pre-therapeutic care plan rate increased over the decade from 3.8% to 8% (p < 0.001). The rate of breast-conserving treatment remained stable. The sentinel lymph node rate increased from 4.4% in P1 to 31.9% in P2 (p < 0.001), while the rate of lymph node dissection decreased (p < 0.001) between the two periods. Resort to breast nurses, psycho-oncologists, fertility experts and oncogenetic consultations has also increased. CONCLUSION There is an improvement in the care and management of young women under 40 because of the first Cancer Plans. A study about the impact on the prognosis and a reflection to improve the experience of illness for these women are to lead.
Collapse
Affiliation(s)
- Mariam Soussoko
- Institut de cancérologie de Lorraine Alexis-Vautrin, Parcours sein, 6, avenue de Bourgogne, 54519 Vandœuvre-lès-Nancy cedex, France.
| | - Julia Salleron
- Cellule data biostatistique, Institut de cancérologie de Lorraine Alexis-Vautrin, 6, avenue de Bourgogne, 54519 Vandœuvre-lès-Nancy, France
| | - Emmanuel Desandes
- Service en charge des données de santé, Institut de cancérologie de Lorraine Alexis-Vautrin, Parcours sein, 6, avenue de Bourgogne, 54519 Vandoeuvre-lès-Nancy cedex, France
| | - Anne Lesur
- Institut de cancérologie de Lorraine Alexis-Vautrin, Parcours sein, 6, avenue de Bourgogne, 54519 Vandœuvre-lès-Nancy cedex, France
| |
Collapse
|
35
|
Nusbaum JS, Mirza I, Shum J, Freilich RW, Cohen RE, Pillinger MH, Izmirly PM, Buyon JP. Sex Differences in Systemic Lupus Erythematosus: Epidemiology, Clinical Considerations, and Disease Pathogenesis. Mayo Clin Proc 2020; 95:384-394. [PMID: 32029091 DOI: 10.1016/j.mayocp.2019.09.012] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 08/17/2019] [Accepted: 09/20/2019] [Indexed: 12/12/2022]
Abstract
Systemic lupus erythematosus (SLE) is a chronic, multiorgan, systemic autoimmune disease that is more common in women than men and is typically diagnosed during reproductive age, necessitating sex-specific considerations in care. In women there is no substantive evidence to suggest that SLE reduces fertility, but subfertility may occur as a result of active disease, immunosuppressive drugs, and age-related declines in fertility related to delays in childbearing. Although pregnancy outcomes have improved, SLE still poses risks in pregnancy that contribute to poorer maternal and fetal outcomes. Cyclophosphamide, an important agent for the treatment of severe or life-threatening lupus, may adversely affect fertility, particularly with increases in dose and patient age. Fertility preservation techniques are therefore an important consideration for women and men before cytotoxic treatment. There is mixed evidence as to whether exogenous estrogen in the form of oral contraceptive pills or hormone replacement therapy may increase the risk for the development of SLE, but among women with SLE already diagnosed, combined oral contraceptive pills and hormone replacement therapy do not confer risk for severe flare and remain important in reproductive care. The higher incidence of SLE in women may nonetheless be attributable to effects of endogenous estrogen, as well as failures in X chromosome inactivation, increased Toll-like receptor gene products, and changes in microRNA function. A greater appreciation of the biological underpinnings and consequences of sex differences in SLE may lead to more targeted treatments and improved outcomes for patients with SLE.
Collapse
Affiliation(s)
- Julie S Nusbaum
- Division of Rheumatology, Department of Medicine, New York University School of Medicine, New York.
| | - Ibraheem Mirza
- Division of Rheumatology, Department of Medicine, New York University School of Medicine, New York
| | - Justine Shum
- Division of Rheumatology, Department of Medicine, New York University School of Medicine, New York
| | - Robert W Freilich
- Division of Rheumatology, Department of Medicine, New York University School of Medicine, New York
| | - Rebecca E Cohen
- Division of Rheumatology, Department of Medicine, New York University School of Medicine, New York
| | - Michael H Pillinger
- Division of Rheumatology, Department of Medicine, New York University School of Medicine, New York
| | - Peter M Izmirly
- Division of Rheumatology, Department of Medicine, New York University School of Medicine, New York
| | - Jill P Buyon
- Division of Rheumatology, Department of Medicine, New York University School of Medicine, New York
| |
Collapse
|
36
|
Fertility preservation in patients undergoing gonadotoxic therapy or gonadectomy: a committee opinion. Fertil Steril 2019; 112:1022-1033. [DOI: 10.1016/j.fertnstert.2019.09.013] [Citation(s) in RCA: 161] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 09/11/2019] [Indexed: 02/08/2023]
|
37
|
Robson D, Phua C, Howard R, Marren A. Fertility preservation in oncology patients: A literature review examining current fertility preservation techniques and access to oncofertility services in Australia. Aust N Z J Obstet Gynaecol 2019; 60:18-26. [PMID: 31617210 DOI: 10.1111/ajo.13081] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 08/19/2019] [Accepted: 09/11/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND In Australia, between the years 2010 and 2014, over 4500 adolescents and young adults (15-25 years old) were diagnosed with cancer. Treatment regimens are often gonadotoxic and are well known to induce ovarian and testicular failure. Oncofertility is an emerging discipline in obstetrics and gynaecology which seeks to preserve and restore the reproductive future of cancer patients. AIM To perform a systematic literature review to assess the current fertility preservation techniques available to patients and examine access and uptake of fertility preservation in Australia. MATERIALS AND METHODS Electronic databases, including Medline, Cochrane Review, SCOPUS and CINHAL, were searched for peer-reviewed publications and national guidelines examining oncofertility practices from 2008 to July 2018. Three hundred and seventy-five articles were initially screened, with 158 articles for full text review and an additional five clinical guidelines were identified. RESULTS There is a paucity of Australian data on oncofertility with <50% of data included for analysis reflecting the Australian experience. The majority of primary research included retrospective papers with small cohort numbers. Key areas addressed included live birth outcomes, uptake of services and patient and physician perspectives on fertility preservation. Few articles sought to examine the positive and negative side effects of fertility preservation in oncology, social challenges of oncofertility and access to services worldwide. CONCLUSION Oncofertility is an emerging discipline which seeks to provide safe, efficient and effective fertility preservation options for young adults and adolescents diagnosed with cancer. A multi-disciplinary approach with collaborative communication with oncologists is key to providing this service within Australia.
Collapse
Affiliation(s)
- Danielle Robson
- Royal Prince Alfred Hospital for Women and Babies, Sydney, New South Wales, Australia
| | - Cheryl Phua
- Royal Prince Alfred Hospital for Women and Babies, Sydney, New South Wales, Australia.,Genea LTD, Sydney, New South Wales, Australia
| | - Robyn Howard
- Royal Prince Alfred Hospital for Women and Babies, Sydney, New South Wales, Australia
| | - Anthony Marren
- Royal Prince Alfred Hospital for Women and Babies, Sydney, New South Wales, Australia.,Genea LTD, Sydney, New South Wales, Australia.,The Institute of Academic Surgery, Royal Prince Alfred Hospital & the University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
38
|
Taylan E, Oktay K. Fertility preservation in gynecologic cancers. Gynecol Oncol 2019; 155:522-529. [PMID: 31604663 DOI: 10.1016/j.ygyno.2019.09.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/03/2019] [Accepted: 09/09/2019] [Indexed: 12/11/2022]
Abstract
An increasing number of women in modern societies are delaying childbearing beyond the age of 35, and gynecologic cancers affect a significant proportion of reproductive age women who wish to preserve fertility for a future chance of childbearing. As a result, providing treatment options for fertility preservation in women with gynecologic cancer has become a crucial component of cancer survivorship care. In this review article, we discussed the current knowledge on fertility-sparing surgical approaches, as well as assisted reproductive technologies that can be utilized to preserve reproductive potential in women with cervical, endometrial, and ovarian cancer. A brief section on fertility preservation in pediatric gynecologic malignancies is also provided.
Collapse
Affiliation(s)
- Enes Taylan
- Women's Cancer Program at Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Laboratory of Molecular Reproduction and Fertility Preservation, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA.
| | - Kutluk Oktay
- Laboratory of Molecular Reproduction and Fertility Preservation, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA; Innovation Institute for Fertility Preservation and IVF, New York, NY, USA; Ovarian Transplantation Program, NYU Winthrop Hospital, Mineola, NY, USA
| |
Collapse
|
39
|
Ferraro E, Trapani D, Marrucci E, Curigliano G. Evaluating triptorelin as a treatment option for breast cancer. Expert Opin Pharmacother 2019; 20:1809-1818. [PMID: 31500470 DOI: 10.1080/14656566.2019.1650020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Introduction: Triptorelin is a luteinizing hormone-releasing hormone analog (LH-RHa) inducing ovarian function suppression (OFS). It is approved by FDA and EMA in association with tamoxifen or aromatase inhibitor (AI) and with fulvestrant and palbociclib in premenopausal women with hormone receptor (HR)-positive breast cancer. Its potential role to preserve ovarian function during chemotherapy has also been recently clarified. Areas covered: Several studies have investigated the role of adding OFS to tamoxifen and aromatase inhibitors as adjuvant treatment in early breast cancer. The addition of triptorelin is not free from adverse events as the combination with tamoxifen and exemestane resulted in an increase of endocrine-deprivation symptoms. Clinical trials have explored the combination of LH-RHa with chemotherapy in fertility preservation, demonstrating no detrimental effect on patients' oncological outcome. This is all discussed in this evaluation. Expert opinion: Triptorelin represents a standard-of-care in premenopausal women with HR-positive breast cancer and in some cases of male breast cancer. In the adjuvant setting, a personalized approach is required to combine LH-RHa with the right partner considering the risk of recurrence and the toxicity profile. LH-RHa may be offered to breast cancer patients in the hope of reducing the likelihood of chemotherapy-induced ovarian insufficiency.
Collapse
Affiliation(s)
- Emanuela Ferraro
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS , Milan , Italy.,Department of Oncology and Hemato-Oncology, University of Milan , Milan , Italy
| | - Dario Trapani
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS , Milan , Italy.,Department of Oncology and Hemato-Oncology, University of Milan , Milan , Italy
| | - Eleonora Marrucci
- Campus Bio-Medico University of Rome, Rome, Italy; Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS , Milan , Italy
| | - Giuseppe Curigliano
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS , Milan , Italy.,Department of Oncology and Hemato-Oncology, University of Milan , Milan , Italy
| |
Collapse
|
40
|
Blumenfeld Z. Fertility Preservation Using GnRH Agonists: Rationale, Possible Mechanisms, and Explanation of Controversy. CLINICAL MEDICINE INSIGHTS. REPRODUCTIVE HEALTH 2019; 13:1179558119870163. [PMID: 31488958 PMCID: PMC6710670 DOI: 10.1177/1179558119870163] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 07/18/2019] [Indexed: 12/15/2022]
Abstract
The only clinically accepted method of fertility preservation in young women facing gonadotoxic chemo- and/or radiotherapy for malignant or autoimmune diseases is cryopreservation of embryos or unfertilized ova, whereas cryopreservation of ovarian tissue for future reimplantation, or in vitro maturation of follicles, and the use of gonadotropin-releasing hormone agonists (GnRHa) are still considered investigational, by several authorities. Whereas previous publications have raised the fear of GnRHa's possible detrimental effects in patients with hormone receptor-positive breast cancers, recent randomized controlled trials (RCTs) have shown that it either improves or does not affect disease-free survival (DFS) in such patients. This review summarizes the pros and cons of GnRHa co-treatment for fertility preservation, suggesting 5 theoretical mechanisms for GnRHa action: (1) simulating the prepubertal hypogonadotropic milieu, (2) direct effect on GnRH receptors, (3) decreased ovarian perfusion, (4) upregulation of an ovarian-protecting molecule such as sphingosine-1-phosphate, and (5) protecting a possible germinative stem cell. We try to explain the reasons for the discrepancy between most publications that support the use of GnRHa for fertility preservation and the minority of publications that did not support its efficiency.
Collapse
Affiliation(s)
- Zeev Blumenfeld
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
41
|
Poggio F, Lambertini M, Bighin C, Conte B, Blondeaux E, D'Alonzo A, Dellepiane C, Buzzatti G, Molinelli C, Boccardo F, Del Mastro L. Potential Mechanisms of Ovarian Protection with Gonadotropin-Releasing Hormone Agonist in Breast Cancer Patients: A Review. CLINICAL MEDICINE INSIGHTS. REPRODUCTIVE HEALTH 2019; 13:1179558119864584. [PMID: 31391786 PMCID: PMC6669835 DOI: 10.1177/1179558119864584] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 06/25/2019] [Indexed: 01/22/2023]
Abstract
The use of chemotherapy in premenopausal cancer patients may lead to chemotherapy-induced premature ovarian failure. Pharmacological temporary ovarian suppression obtained with the gonadotropin-releasing hormone agonist (GnRHa) administered concomitantly with chemotherapy has been investigated as a technique capable to reduce the gonadotoxicity, reducing the risk of developing premature menopause. In recent years, important evidence has become available on the efficacy and safety of this strategy that should now be considered a standard option for ovarian function preservation in premenopausal breast cancer patients. However, in women interested in fertility preservation, this is not an alternative to cryopreservation strategies, which remains the first option to be proposed. The purpose of this review is to summarize the mechanisms of GnRHa in the preservation of fertility in premenopausal cancer patient candidates to receive chemotherapy, highlighting the areas of doubt that require further investigation.
Collapse
Affiliation(s)
| | - Matteo Lambertini
- Department of Medical Oncology, Clinica di Oncologia Medica, IRCCS San Martino Hospital, Genova, Italy.,Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genova, Genova, Italy
| | - Claudia Bighin
- UO Breast Unit, IRCCS San Martino Hospital, IST, Genova, Italy
| | - Benedetta Conte
- UO Breast Unit, IRCCS San Martino Hospital, IST, Genova, Italy
| | - Eva Blondeaux
- UO Breast Unit, IRCCS San Martino Hospital, IST, Genova, Italy
| | | | | | - Giulia Buzzatti
- UO Breast Unit, IRCCS San Martino Hospital, IST, Genova, Italy
| | | | - Francesco Boccardo
- Department of Medical Oncology, Clinica di Oncologia Medica, IRCCS San Martino Hospital, Genova, Italy.,Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genova, Genova, Italy
| | - Lucia Del Mastro
- UO Breast Unit, IRCCS San Martino Hospital, IST, Genova, Italy.,Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genova, Genova, Italy
| |
Collapse
|
42
|
Zhong Y, Lin Y, Cheng X, Huang X, Zhou Y, Mao F, Wang Y, Guan J, Shen S, Xu Y, Peng L, Li Y, Cao X, Sun Q. GnRHa for Ovarian Protection and the Association between AMH and Ovarian Function during Adjuvant Chemotherapy for Breast Cancer. J Cancer 2019; 10:4278-4285. [PMID: 31413747 PMCID: PMC6691711 DOI: 10.7150/jca.31859] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 05/19/2019] [Indexed: 11/16/2022] Open
Abstract
Background: Chemotherapy impairs ovarian function in premenopausal breast cancer patients. Many breast cancer patients experience menopause earlier and therefore lose their reproductive abilities. The protective effect of gonadotropin-releasing hormone agonist (GnRha) upon the ovary is clearly apparent for hormone receptor (HR) negative patients, although the available data is not consistent for the patient body as a whole when considered regardless of HR status. It is also unknown whether levels of Anti-Mullerian Hormone (AMH) can reflect the influence of chemotherapy upon the ovary. Methods: We randomly assigned 98 premenopausal breast cancer patients regardless HR-positive or -negative to receive either standard chemotherapy with GnRHa (GnRHa group) or standard chemotherapy without GnRHa (control group). Our primary end point was ovarian failure rate (OVF) at 1 year. In addition, we observed the change of AMH level during chemotherapy and the association between AMH and OVF. Results: OVF was significantly lower (44.7%) in the GnRHa group than in the control group (80.6%; P=0.002). Median AMH levels were significantly higher before chemotherapy when compared to after 1/2cycles of chemotherapy, both in the GnRHa group (1.86ng/ml vs 0.12ng/ml; P=0.000) and in the control group (1.57ng/ml vs 0.10ng/ml; P=0.000). OVF was 91.3% in the AMH baseline level <1.1ng/ml group and 63.5% in the AMH baseline level >1.1ng/ml group (P=0.013). Conclusion: Data showed that GnRHa may have a protective effect on young breast cancer patients regardless of HR during chemotherapy. AMH could reflect changes of OVF during chemotherapy and predict OVF after chemotherapy.
Collapse
Affiliation(s)
- Ying Zhong
- Department of Breast Disease, Peking Union Medical College Hospital, Shuaifuyuan, Wangfujing, Beijing 100730, China
| | - Yan Lin
- Department of Breast Disease, Peking Union Medical College Hospital, Shuaifuyuan, Wangfujing, Beijing 100730, China
| | - Xinqi Cheng
- Department of Clinical laboratory, Peking Union Medical College Hospital, Shuaifuyuan, Wangfujing, Beijing 100730, China
| | - Xin Huang
- Department of Breast Disease, Peking Union Medical College Hospital, Shuaifuyuan, Wangfujing, Beijing 100730, China
| | - Yidong Zhou
- Department of Breast Disease, Peking Union Medical College Hospital, Shuaifuyuan, Wangfujing, Beijing 100730, China
| | - Feng Mao
- Department of Breast Disease, Peking Union Medical College Hospital, Shuaifuyuan, Wangfujing, Beijing 100730, China
| | - Yajing Wang
- Department of Clinical laboratory, Peking Union Medical College Hospital, Shuaifuyuan, Wangfujing, Beijing 100730, China
| | - Jinghong Guan
- Department of Breast Disease, Peking Union Medical College Hospital, Shuaifuyuan, Wangfujing, Beijing 100730, China
| | - Songjie Shen
- Department of Breast Disease, Peking Union Medical College Hospital, Shuaifuyuan, Wangfujing, Beijing 100730, China
| | - Yali Xu
- Department of Breast Disease, Peking Union Medical College Hospital, Shuaifuyuan, Wangfujing, Beijing 100730, China
| | - Li Peng
- Department of Breast Disease, Peking Union Medical College Hospital, Shuaifuyuan, Wangfujing, Beijing 100730, China
| | - Yan Li
- Department of Breast Disease, Peking Union Medical College Hospital, Shuaifuyuan, Wangfujing, Beijing 100730, China
| | - Xi Cao
- Department of Breast Disease, Peking Union Medical College Hospital, Shuaifuyuan, Wangfujing, Beijing 100730, China
| | - Qiang Sun
- Department of Breast Disease, Peking Union Medical College Hospital, Shuaifuyuan, Wangfujing, Beijing 100730, China
| |
Collapse
|
43
|
Wang SY, Wang S. [Anti-Müllerian hormone as a new marker of the ovarian reserve function preservation by goserelin during (neo)adjuvant chemotherapy for young breast cancer patients]. JOURNAL OF PEKING UNIVERSITY. HEALTH SCIENCES 2019; 51:536-541. [PMID: 31209428 DOI: 10.19723/j.issn.1671-167x.2019.03.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To observe the dynamic change of anti-Müllerian hormone (AMH) in 1 year after chemotherapy which is the best biochemical marker of ovarian reserve in reproductive medicine setting and to evaluate the effect of gonadotropin-releasing hormone agonist (GnRHa)goserelin to prevent ovarian reserve function during (neo)adjuvant chemotherapy for young breast cancer patients. METHODS Between December 2015 and June 2017, 101 breast cancer patients of age ≤ 45 years with stages I to III had been enrolled. The patients were assigned without interference to receive either (neo) adjuvant chemotherapy with goserelin (goserelin group) or without goserelin (chemotherapy group) as their own selection. AMH and menstrual status were evaluated before, during and 0.5 year, 1 year after chemotherapy. Primary end point was the incidence of low AMH value (<0.4 μg/L) at the end of 1 year. Secondary end point was the incidence of amenorrhea (the absence of menses in the preceding 12 months after assignment). RESULTS In the study, 51 patients chose to join the chemotherapy group, while the other 50 patients selected goserelin to preserve their ovarian reserve function. More unmarried or childless, hormone receptors negative,receiving breast conservation therapy patients with earlier stage selected goserelin before chemotherapy. The incidence of low AMH value was significantly higher in chemotherapy group than in goserelin group (74.5% vs. 38.0%, P<0.001) in 1 year after chemotherapy. The incidence of amenorrhea was consistent with AMH (56.9% vs. 24.0%, P=0.001). And more patients' menstruation (78.9% vs. 54.5%) and AMH value (71.0% vs. 53.8%) recovered in goserelin group within 6 months after chemotherapy. In subgroup analysis, AMH and menstruation seemingly recovered more in goserelin group independent of age, chemotherapy regimen and use of tamoxifen. Especially, AMH value of 36.4% (8/22) patients in chemotherapy group and 18.4% (7/38) patients in goserelin group still maintained low level (<0.4 μg /L) although their menstruation had recovered 1 year after chemotherapy. In addition, 41 patients (20 patients in chemotherapy group, 21 patients in goserelin group) could be evaluated for the dynamic change of AMH and menstrual status during chemotherapy. The mean level of AMH in chemotherapy group declined rapidly to very low level before the 3rd cycle, while 70% of the patients kept presence of menstruation. At the same time, the mean level of AMH in goserelin group was still above 0.4 μg /L, but all of the patients had menopause. CONCLUSION Our study has offered evidence that Goserelin with chemotherapy could protect against ovarian reserve failure for young breast cancer patients, now that more patients' AMH value recovered earlier who had selected co-treatment. AMH may be a more precise marker than menstrual status to clinically evaluate ovarian reserve function pre-, during and post- chemotherapy.
Collapse
Affiliation(s)
- S Y Wang
- Department of Breast Surgery, Peking University People's Hospital, Beijing 100044, China
| | - S Wang
- Department of Breast Surgery, Peking University People's Hospital, Beijing 100044, China
| |
Collapse
|
44
|
Zheng F, Zhu B, Feng Q, Wu L, Cui Y, Liu Y, Wang Y. Protective effect of gonadotropin-releasing hormone agonist against chemotherapy-induced ovarian dysfunction: A meta-analysis. Oncol Lett 2019; 17:5319-5326. [PMID: 31186748 PMCID: PMC6507318 DOI: 10.3892/ol.2019.10252] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 12/19/2018] [Indexed: 11/06/2022] Open
Abstract
The protective effects of gonadotropin-releasing hormone agonist (GnRHa) against ovarian chemotherapy induced-toxicity have not completely been demonstrated and the impact of chemotherapy on ovarian dysfunction remains unclear. The present meta-analysis aimed to evaluate the efficiency of GnRHa and to determine whether GnRHa could influence the long-term survival rate of patients with cancer. A total of 12 clinical randomized controlled trials were included, consisting of 1,413 patients who were divided into the GnRHa group (n=705) and the control group (n=708). The meta-analysis revealed that GnRHa may significantly improve the menstrual function recovery rate in patients who received chemotherapy [RR=1.29, 95% confidence interval (CI)=1.09-1.54, P=0.004] and reduce the rate of premature ovarian failure (RR=0.47, 95% CI=0.31-0.71, P=0.0004). However, it had no effect on the pregnancy rate (RR=1.40, 95% CI=0.98-1.98, P=0.06), on the rate of disease-free survival and overall survival of patients (disease-free survival rate: RR=1.04, 95% CI=0.95-1.13, P=0.40; overall survival rate: RR=1.02, 95% CI=0.90-1.16, P=0.72). In conclusion, GnRHa may reduce chemotherapy-induced ovarian dysfunction without compromising or influencing the therapeutic effects of chemotherapy.
Collapse
Affiliation(s)
- Fei Zheng
- Department of Obstetrics and Gynecology, Family Planning Service Center of Yiwu Maternity and Child Health Care Hospital, Yiwu, Zhejiang 322000, P.R. China
| | - Bin Zhu
- Department of Obstetrics and Gynecology, Family Planning Service Center of Yiwu Maternity and Child Health Care Hospital, Yiwu, Zhejiang 322000, P.R. China
| | - Qingjing Feng
- Department of Galactophore, Family Planning Service Center of Yiwu Maternity and Child Health Care Hospital, Yiwu, Zhejiang 322000, P.R. China
| | - Lili Wu
- Department of Obstetrics and Gynecology, Family Planning Service Center of Yiwu Maternity and Child Health Care Hospital, Yiwu, Zhejiang 322000, P.R. China
| | - Yuechong Cui
- Department of Human Health and Human Service, Family Planning Service Center of Yiwu Maternity and Child Health Care Hospital, Yiwu, Zhejiang 322000, P.R. China
| | - Yumo Liu
- Department of Public Health, Southwest Medical University, Luzhou, Sichuan 646600, P.R. China
| | - Yingqian Wang
- Department of Obstetrics and Gynecology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050035, P.R. China
| |
Collapse
|
45
|
Abstract
Breast cancer is the most commonly diagnosed cancer in women. As long-term survival rates have improved, there has been a concurrent increase in quality of life considerations, of which fertility preservation is of utmost importance. A number of fertility preservation options are available to women diagnosed with breast cancer, including administration of a GnRH agonist during chemotherapy in an attempt to minimize ovarian damage, oocyte or embryo cryopreservation prior to the administration of chemotherapy, in vitro maturation of oocytes or ovarian tissue cryopreservation. The safety of pregnancy after a diagnosis of breast cancer has been confirmed in numerous studies.
Collapse
Affiliation(s)
- Rachael J Rodgers
- School of Women's and Children's Health University of New South Wales, Sydney, Australia -
| |
Collapse
|
46
|
Abdel-Razeq H. Gonadotropin-releasing hormone agonists during chemotherapy for ovarian function and fertility preservation for patients with early-stage breast cancer. Cancer Manag Res 2019; 11:4273-4282. [PMID: 31190993 PMCID: PMC6514123 DOI: 10.2147/cmar.s204069] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 03/27/2019] [Indexed: 01/09/2023] Open
Abstract
Breast cancer remains the most common cancer among women worldwide. Many patients, especially in our region, are affected while young and during their child-bearing years. Chemotherapy, more commonly used in this age group, may result in premature ovarian failure and thus negatively impact their fertility. Several fertility-preservation methods are currently in use in this age group. Unfertilized ova cryopreservation and in vitro fertilization plus embryo cryopreservation are widely used in clinical practice. More recently, ovarian tissue cryopreservation is gaining in popularity. Several clinical trials and meta-analyses have shown that premenopausal women who received ovarian function suppression with gonadotropin-releasing hormone agonists while on chemotherapy were less likely to experience ovarian failure and had higher rates of menses resumption compared to those who did not. Some studies have also shown higher rates of successful pregnancies among treated patients. Given the conflicting results of the reported clinical trials and even the many published meta-analyses, this approach continues to be controversial and should only be used when other established fertility preservation methods cannot be utilized. The current review seeks to provide an updated summary on this controversial topic by reviewing all recently published clinical trials and meta-analyses.
Collapse
Affiliation(s)
- Hikmat Abdel-Razeq
- Department of Internal Medicine, Section of Hematology and Medical Oncology, King Hussein Cancer Center and School of Medicine, University of Jordan, Amman, Jordan
| |
Collapse
|
47
|
Shah NM, Scott DM, Kandagatla P, Moravek MB, Cobain EF, Burness ML, Jeruss JS. Young Women with Breast Cancer: Fertility Preservation Options and Management of Pregnancy-Associated Breast Cancer. Ann Surg Oncol 2019; 26:1214-1224. [PMID: 30680478 PMCID: PMC6458084 DOI: 10.1245/s10434-019-07156-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Breast cancer is the most common malignancy diagnosed in women of childbearing age. A breast cancer diagnosis in this young patient population can be uniquely complex to navigate when considering the potential impact of fertility loss associated with specific gonadotoxic therapies. Another unique challenge for young breast cancer patients is pregnancy-associated breast cancer (PABC), which occurs in approximately 1 of every 3000 pregnancies. Pregnancy adds a layer of complexity to breast cancer treatment planning as many therapies can affect the developing fetus. These two clinical challenges require nuanced multidisciplinary approaches to facilitate optimal treatment outcomes. We sought to review and summarize the management strategy options for both fertility preservation and PABC. METHODS A guideline and literature review was performed for fertility preservation, young patients with breast cancer, and pregnancy-associated breast cancer. RESULTS Fertility preservation options, both established and experimental, are detailed. Suggested clinical practice guidelines for PABC are also presented, which delineate breast cancer treatment recommendations based on pregnancy trimester. CONCLUSION A multidisciplinary approach to patient care, including oncologists and early referral to reproductive specialists, can provide young breast cancer patients with options for fertility preservation. Under the guidance of a multidisciplinary treatment team, PABC can also be diagnosed and treated to permit the best possible outcomes for the mother and the developing fetus.
Collapse
Affiliation(s)
- Nikita M Shah
- Division of Surgical Oncology, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Dana M Scott
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
| | - Pridvi Kandagatla
- Division of Surgical Oncology, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Department of Surgery, Henry Ford Health System/Wayne State University, Detroit, MI, USA
| | - Molly B Moravek
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
| | - Erin F Cobain
- Division of Medical Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Monika L Burness
- Division of Medical Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Jacqueline S Jeruss
- Division of Surgical Oncology, Department of Surgery, University of Michigan, Ann Arbor, MI, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
48
|
Lambertini M, Richard F, Nguyen B, Viglietti G, Villarreal-Garza C. Ovarian Function and Fertility Preservation in Breast Cancer: Should Gonadotropin-Releasing Hormone Agonist be administered to All Premenopausal Patients Receiving Chemotherapy? CLINICAL MEDICINE INSIGHTS. REPRODUCTIVE HEALTH 2019; 13:1179558119828393. [PMID: 30886529 PMCID: PMC6410390 DOI: 10.1177/1179558119828393] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 01/10/2019] [Indexed: 11/15/2022]
Abstract
Chemotherapy-induced premature ovarian insufficiency (POI) is one of the potential drawbacks of chemotherapy use of particular concern for newly diagnosed premenopausal breast cancer patients. Temporary ovarian suppression obtained pharmacologically with the administration of a gonadotropin-releasing hormone agonist (GnRHa) during chemotherapy has been specifically developed as a method to counteract chemotherapy-induced gonadotoxicity with the main goal of diminishing the risk of POI. In recent years, important clinical evidence has become available on the efficacy and safety of this strategy that should now be considered a standard option for ovarian function preservation in premenopausal breast cancer patients, including women who are not interested in conceiving after treatment or that would not be candidates for fertility preservation strategies because of their age. Nevertheless, in women interested in fertility preservation, this is not an alternative to gamete cryopreservation, which remains as the first option to be offered. In this setting, temporary ovarian suppression with GnRHa during chemotherapy should be also proposed following gamete cryopreservation or to women who have no access, refuse, or have contraindications to surgical fertility preservation techniques. In this article, we present an overview about the role of temporary ovarian suppression with GnRHa during chemotherapy in breast cancer patients by addressing the available clinical evidence with the aim of identifying both the best candidates for the use of this strategy and the still existing gray zones requiring further investigation.
Collapse
Affiliation(s)
- Matteo Lambertini
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, Ospedale Policlinico San Martino, Genova, Italy.,Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - François Richard
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles (U.L.B), Brussels, Belgium
| | - Bastien Nguyen
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles (U.L.B), Brussels, Belgium
| | - Giulia Viglietti
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles (U.L.B), Brussels, Belgium
| | - Cynthia Villarreal-Garza
- Centro de Cancer de Mama del Hospital Zambrano Hellion, Tecnologico de Monterrey, Nuevo Leon, Mexico.,Departamento de Investigacion y de Tumores Mamarios del Instituto Nacional de Cancerologia, Mexico City, Mexico
| |
Collapse
|
49
|
Chen H, Xiao L, Li J, Cui L, Huang W. Adjuvant gonadotropin-releasing hormone analogues for the prevention of chemotherapy-induced premature ovarian failure in premenopausal women. Cochrane Database Syst Rev 2019; 3:CD008018. [PMID: 30827035 PMCID: PMC6397718 DOI: 10.1002/14651858.cd008018.pub3] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND This is an update of the original review published in the Cochrane Database of Systematic Reviews 2011, Issue 11, and updated in 2015, Issue 4.Chemotherapy has significantly improved prognosis for women with malignant and some non-malignant conditions. This treatment, however, is associated with ovarian toxicity. The use of gonadotropin-releasing hormone (GnRH) analogues, both agonists and antagonists, may have a protective effect on the ovaries. The primary mechanism of action of GnRH analogues is to suppress the gonadotropin levels to simulate pre-pubertal hormonal milieu and subsequently prevent primordial follicles from maturation and therefore decrease the number of follicles that are more vulnerable to chemotherapy. OBJECTIVES To assess the efficacy and safety of GnRH analogues given before or in parallel to chemotherapy to prevent chemotherapy-related ovarian damage in premenopausal women with malignant or non-malignant conditions. SEARCH METHODS The search was run for the original review in July 2011, and for the first update in July 2014. For this update we searched the following databases in November 2018: the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, Embase, and the Chinese Biomedicine Database (CBM). SELECTION CRITERIA Randomised controlled trials (RCTs), in all languages, which examined the effect of GnRH analogues for chemotherapy-induced ovarian failure in premenopausal women, were eligible for inclusion in the review. DATA COLLECTION AND ANALYSIS Two review authors independently extracted data and assessed trial quality using the Cochrane 'Risk of bias' tool. We analysed binary data using risk ratios (RRs) with 95% confidence intervals (CI) and for continuous data, we used the standardized mean difference (SMD) to combine trials. We applied the random-effects model in our analyses. We used the GRADE approach to produce a 'Summary of findings' table for our main outcomes of interest. MAIN RESULTS We included 12 RCTs involving 1369 women between the ages of 12 and 51.1 years. Participants were diagnosed with breast malignancy, ovarian malignancy, or Hodgkin's lymphoma, and most of them received alkylating, or platinum complexes, based chemotherapy. The included studies were funded by a university (n = 1), research centres (n = 4), and pharmaceutical companies (n = 1). Trials were at high or unclear risk of bias.Comparison 1: GnRH agonist plus chemotherapy versus chemotherapy aloneThe incidence of menstruation recovery or maintenance was 178 of 239 (74.5%) in the GnRH agonist group and 110 of 221 (50.0%) in the control group during a follow-up period no longer than 12 months (RR 1.60, 95% CI 1.14 to 2.24; 5 studies, 460 participants; I2 = 79%; low-certainty evidence), with an overall effect favouring treatment with GnRH agonist (P = 0.006). However, we observed no difference during a follow-up period longer than 12 months between these two groups (P = 0.24). In the GnRH agonist group, 326 of 447 participants had menstruation recovery or maintenance (72.9%) in comparison to the control group, in which 276 of 422 participants had menstruation recovery or maintenance (65.4%) during a follow-up period longer than 12 months (RR 1.08, 95% CI 0.95 to 1.22; 8 studies, 869 participants; I2 = 56%; low-certainty evidence).The incidence of premature ovarian failure was 43 of 401 (10.7%) in the GnRH agonist group and 96 of 379 (25.3%) in the control group (RR 0.44, 95% CI 0.31 to 0.61; 4 studies, 780 participants; I2 = 0%; moderate-certainty evidence), with an overall effect favouring treatment with GnRH agonist (P < 0.00001).The incidence of pregnancy was 32 of 356 (9.0%) in the GnRH agonist group and 22 of 347 (6.3%) in the control group (RR 1.59, 95% CI 0.93 to 2.70; 7 studies, 703 participants; I2 = 0%; low-certainty evidence), with no difference between groups (P = 0.09). However, we are cautious about this conclusion because there were insufficient data about whether the participants intended to become pregnant.The incidence of ovulation was 29 of 47 (61.7%) in the GnRH agonist group and 12 of 48 (25.0%) in the control group (RR 2.47, 95% CI 1.43 to 4.26; 2 studies, 95 participants; I2 = 0%; low-certainty evidence) with an overall effect favouring treatment with GnRH (P = 0.001).The most common adverse effects of GnRH analogues included hot flushes, vaginal dryness, urogenital symptoms, and mood swings. The pooled analysis of safety data showed no difference in adverse effects between GnRH agonist group and control group.Comparison 2: GnRH agonist-antagonist cotreatment plus chemotherapy versus chemotherapy aloneOnly one RCT discussed GnRH agonist-antagonist cotreatment. The limited evidence showed the incidence of menstruation recovery or maintenance was 20 of 25 (80%) in both cotreatment group and control group during a 12-month follow-up period (RR 1.00, 95% CI 0.76 to 1.32; 50 participants; very low-certainty evidence), with no difference between groups (P = 1.00). In the cotreatment group, 13 of 25 participants had menstruation recovery or maintenance (52.0%) in comparison to the control group, in which 14 of 25 participants had menstruation recovery or maintenance (56.0%) during a follow-up period longer than 12 months (RR 0.93, 95% CI 0.56 to 1.55; 50 participants; very low-certainty evidence), with no difference between groups (P = 0.78). The incidence of pregnancy was 1 of 25 (4.0%) in the cotreatment group and 0 of 25 (0%) in the control group (RR 3.00, 95% CI 0.13 to 70.30; 50 participants; very low-certainty evidence), with no difference between groups (P = 0.49). AUTHORS' CONCLUSIONS GnRH agonist appears to be effective in protecting the ovaries during chemotherapy, in terms of maintenance and resumption of menstruation, treatment-related premature ovarian failure and ovulation. Evidence for protection of fertility was insufficient and needs further investigation. Evidence was also insufficient to assess the effect of GnRH agonist and GnRH antagonist cotreatment on ovarian protection against chemotherapy. The included studies differed in some important aspects of design, and most of these studies had no age-determined subgroup analysis. Large and well-designed RCTs with longer follow-up duration should be conducted to clarify the effects of GnRH analogues in preventing chemotherapy-induced ovarian failure, especially on different age groups or different chemotherapy regimens. Furthermore, studies should address the effects on pregnancy rates and anti-tumour therapy.
Collapse
Affiliation(s)
- Hengxi Chen
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, No. 17, Section Three, Ren Min Nan Lu Avenue, Chengdu, Sichuan, China, 610041
| | | | | | | | | |
Collapse
|
50
|
Venturini M, Bergamini A, Perani L, Sanchez AM, Rossi EG, Colarieti A, Petrone M, De Cobelli F, Del Maschio A, Viganò P, Mangili G, Candiani M, Tacchetti C, Esposito A. Contrast-enhanced ultrasound for ovary assessment in a murine model: preliminary findings on the protective role of a gonadotropin-releasing hormone analogue from chemotherapy-induced ovarian damage. Eur Radiol Exp 2018; 2:44. [PMID: 30564987 PMCID: PMC6298912 DOI: 10.1186/s41747-018-0076-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Accepted: 10/26/2018] [Indexed: 11/27/2022] Open
Abstract
The prolonged, gonadotoxic effect of chemotherapy can finally lead to infertility in female cancer survivors. There is controversial evidence regarding the protective role of gonadotropin-releasing hormone analogue (GnRH-a) on chemotherapy-induced ovarian damage. In the present study on a murine model, ultrasound (US) and contrast-enhanced US (CEUS) were firstly used to characterise ovarian glands in normal conditions to validate a preclinical model. In addition, preliminary findings were obtained on anatomical and vascular ovarian changes induced by GnRH-a based on decapeptyl administration. Ovaries were accurately assessed with US and CEUS in a murine model placed in prone position, providing quantitative and reproducible information. Ovaries were identified in 40/40 cases and CEUS analysis was successfully performed in 20/20 cases with 100% technical success. A statistically significant increase of the diameter of the dominant follicle at US and a statistically significant reduced vascularisation at CEUS in decapeptyl-treated mice compared to untreated control mice were recorded. Further studies using US and CEUS in the murine model combining GnRH-a and chemotherapeutic agents will be needed to obtain more translational information useful for clinical practice.
Collapse
Affiliation(s)
- Massimo Venturini
- Department of Radiology, San Raffaele Scientific Institute, Milan, Italy. .,Experimental Imaging Center, San Raffaele Scientific Institute, Milan, Italy.
| | - Alice Bergamini
- Obstetrics and Gynecology Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Laura Perani
- Experimental Imaging Center, San Raffaele Scientific Institute, Milan, Italy
| | - Ana Maria Sanchez
- Obstetrics and Gynecology Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Elena Giulia Rossi
- Obstetrics and Gynecology Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Anna Colarieti
- Department of Radiology, San Raffaele Scientific Institute, Milan, Italy
| | - Micaela Petrone
- Obstetrics and Gynecology Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Francesco De Cobelli
- Department of Radiology, San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Alessandro Del Maschio
- Department of Radiology, San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Paola Viganò
- Obstetrics and Gynecology Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Giorgia Mangili
- Obstetrics and Gynecology Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Massimo Candiani
- Obstetrics and Gynecology Unit, San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Carlo Tacchetti
- Experimental Imaging Center, San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Antonio Esposito
- Department of Radiology, San Raffaele Scientific Institute, Milan, Italy.,Experimental Imaging Center, San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|