1
|
Xie Q, Xie Y, Shi Y, Quan X, Yang X. Impact of haemostasis methods during ovarian cystectomy on ovarian reserve: a pairwise and network meta-analysis. J OBSTET GYNAECOL 2024; 44:2320294. [PMID: 38406841 DOI: 10.1080/01443615.2024.2320294] [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: 07/06/2023] [Accepted: 02/02/2024] [Indexed: 02/27/2024]
Abstract
BACKGROUND Haemostasis during ovarian cystectomy is reported to damage the ovarian reserve, but the comparative impacts of three haemostasis methods (bipolar energy, suture and haemostatic sealant) on ovarian reserve in patients with ovarian cysts are not well known. METHODS The Cochrane Library, PubMed and Web of Science databases were searched from the date of inception of the database to June 2022 for literature exploring the impact of haemostasis methods during ovarian cystectomy on ovarian reserve. A traditional meta-analysis was performed using Review Manager software. A network meta-analysis (NMA) was performed using Stata and GemTC software. RESULTS The direct meta-analysis comparison indicated that the mean postoperative reduction of anti-Müllerian hormone (AMH) level was significantly higher in the electrocoagulation (bipolar) group than suture and haemostatic sealant group, both in the overall group and subgroup of women with ovarian endometrioma. In NMA, the reduction of postoperative AMH levels in the electrocoagulation (bipolar) group was higher than the suture group at 6 months with a statistical significance, and at 1, 3 and 12 months without a significant difference. The difference in the postoperative decrease of AMH level did not reach statistical significance between suture and sealant, coagulation and haemostatic sealant. The comprehensive ranking results revealed that suture treatment was, with the highest probability, beneficial to the protection of the ovarian reserve. CONCLUSIONS There was insufficient research to detect the optimal haemostasis method for ovarian reserve preservation in ovarian cystectomy. Nevertheless, haemostasis by electrocoagulation (bipolar) should be avoided when possible, and the suture might be considered as the best choice.
Collapse
Affiliation(s)
- Qin Xie
- Department of Obstetrics and Gynecology, Xiangyang Central Hospital, The Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, PR China
| | - Yue Xie
- Department of Obstetrics and Gynecology, Xiangyang Central Hospital, The Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, PR China
| | - Ying Shi
- Department of Obstetrics and Gynecology, Xiangyang Central Hospital, The Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, PR China
| | - Xiaozhen Quan
- Department of Obstetrics and Gynecology, Xiangyang Central Hospital, The Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, PR China
| | - Xuezhou Yang
- Department of Obstetrics and Gynecology, Xiangyang Central Hospital, The Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, PR China
| |
Collapse
|
2
|
Génard-Walton M, Angot E, Monfort C, Rouget F, Warembourg C, Giton F, Lainé F, Gaudreau E, Cordier S, Kvaskoff M, Chevrier C, Garlantézec R. Prenatal exposure to persistent organic pollutants and its impact on the ovarian reserve at 12 years old in the PELAGIE mother-child cohort. ENVIRONMENTAL RESEARCH 2024; 262:119959. [PMID: 39276833 DOI: 10.1016/j.envres.2024.119959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/17/2024]
Abstract
Although the ovarian reserve is constituted in utero, the literature on the effects of persistent organic pollutants (POPs) during this vulnerable period on the ovarian reserve later in life is limited. We investigated whether cord blood concentrations of POPs were associated with decreased anti-Müllerian hormone (AMH, a marker of the ovarian reserve) levels in girls at the age of 12. We included 239 girls from the French mother-child PELAGIE cohort. POP concentrations of 14 organochlorine pesticides, 17 polychlorinated biphenyls (PCBs), 5 polybrominated diphenyl ethers, and 9 per-polyfluoroalkyl substances were measured on cord blood sampled at birth. During a follow-up study at 12 years old, blood samples were collected to measure AMH levels. Single-exposure associations were examined with multivariable linear regression models adjusted a priori for potential confounders. Stratification on menarche status was also performed. Mixture effects were investigated using quantile g-computation and Bayesian kernel machine regression. Overall, 16 POPs were measured in at least 30% of samples. No significant associations were found in multivariable linear regressions, except for the third tercile of exposure to PCB 180 which was statistically significantly associated with an increase in AMH levels at 12 years old (Tercile 2 v. Tercile 1: 0.13 ng/mL, 95% CI = -0.29, 0.56; Tercile 3 v. Tercile 1: 0.51 ng/mL, 95% CI = 0.02, 0.99). Additionally, in post-menarcheal girls (N = 104) only, the second tercile of p,p'-DDE was statistically significantly associated with decreased AMH levels at 12 years old (Tercile 2 v. Tercile 1: -0.61 ng/mL, 95% CI = -1.16, -0.05, Tercile 3 v. Tercile 1: 0.02 ng/mL, 95% CI = -0.51, 0.54). Both mixture models returned null associations. Despite the limited associations observed in this study, we recommend exploring these associations in larger mother-child cohorts and at older ages.
Collapse
Affiliation(s)
- Maximilien Génard-Walton
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000, Rennes, France.
| | - Elisabeth Angot
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Christine Monfort
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Florence Rouget
- Univ Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Charline Warembourg
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Frank Giton
- AP-HP, Pôle Biologie-Pathologie Henri Mondor, Créteil, France; Inserm IMRB, Faculté de Santé, Créteil, France
| | | | - Eric Gaudreau
- Centre de Toxicologie Du Québec (CTQ), Institut National de Santé Publique Du Québec (INSPQ), Québec, Québec, Canada
| | - Sylvaine Cordier
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Marina Kvaskoff
- Université Paris-Saclay, UVSQ, Inserm, Gustave Roussy, Équipe "Exposome Et Hérédité", CESP UMR1018, 94805, Villejuif, France
| | - Cécile Chevrier
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Ronan Garlantézec
- Univ Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| |
Collapse
|
3
|
Cipres DT, Gordon CM. Primary Ovarian Insufficiency, Bone Health, and Other Outcomes in Adolescents. Obstet Gynecol Clin North Am 2024; 51:663-678. [PMID: 39510737 PMCID: PMC11566969 DOI: 10.1016/j.ogc.2024.08.004] [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] [Indexed: 11/15/2024]
Abstract
Adolescents with primary ovarian insufficiency (POI) frequently present with arrested pubertal development or amenorrhea. Early evaluation of menstrual irregularities can avoid a delayed diagnosis. There are various genetic, autoimmune, and iatrogenic causes of POI, although most of the cases will not have an identified cause. Prompt initiation of hormone replacement therapy will restore developmentally appropriate pubertal progression, establishment of menses, and optimization of bone and cardiovascular health. The diagnosis of POI is often unexpected and life-altering for an adolescent and has broad health and psychosocial implications that are best approached with empathy, educational resources, and engagement of multidisciplinary specialists.
Collapse
Affiliation(s)
- Danielle T Cipres
- Division of Gynecology, Department of Surgery, Boston Children's Hospital, 333 Longwood Avenue, 5th Floor, Boston, MA 02115, USA; Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA.
| | - Catherine M Gordon
- Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health, NICHD Office of the Clinical Director, 10 CRC, Room 5-2583, 10 Center Drive, MSC 1109, Bethesda, MD 20892, USA
| |
Collapse
|
4
|
Li Y, Wang L, Liu J, Nie G, Yang H. Huyang Yangkun formula regulates the mitochondria pathway of ovarian granulosa cell apoptosis through FTO/m6A-P53 pathway. Front Pharmacol 2024; 15:1491546. [PMID: 39584135 PMCID: PMC11581872 DOI: 10.3389/fphar.2024.1491546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 10/25/2024] [Indexed: 11/26/2024] Open
Abstract
Background Premature ovarian insufficiency (POI) presents a significant challenge to female reproductive health. The Huyang Yangkun Formula (HYF), a traditional Chinese medicinal formulation, has been utilized in clinical settings for the treatment of POI for over a decade. Nevertheless, the therapeutic application of HYF is considerably constrained by the lack of clarity regarding its underlying mechanism of action. Methods The experimental procedures entailed administering VCD to female Sprague-Dawley rats at a dosage of 160 mg/kg/day over a period of 15 days, succeeded by a 100-day treatment with HYF. Blood serum samples were collected and analyzed using ELISA to quantify the concentrations of Anti-Müllerian Hormone (AMH), Follicle-Stimulating Hormone (FSH), and Estradiol (E2). The levels of N6-methyladenosine (m6A) were assessed through Dot blot analysis and liquid chromatography-tandem mass spectrometry (LC-MS/MS). Western blotting was employed to validate the differential expression of m6A-related catalytic enzymes and apoptosis-related regulators, including BCL-2, BCL-XL, and MCL-1, which may be implicated in the effects of HYF. Certain shRNA-COV434 cell line was constructed for the exploration of molecular mechanism, and then the potential targets were finally verified by MeRIP-qPCR. Results HYF has been identified as having a significant influence on the development of residual ovarian follicles in rats with POI, especially during the initial stages. It was observed that HYF facilitates the progression of escaping antral follicles to full maturation. Additionally, HYF exhibited the capacity to enhance the proliferation of COV434, a human ovarian granulosa cell line, while concurrently inhibiting apoptosis within these cells. Notably, HYF treatment resulted in the downregulation of apoptotic proteins, including BCL-XL, cleaved-caspase 9, cleaved-caspase 3, and Bcl-2. Concurrently, m6A modification is implicated in the regulation of HYF. Both in vitro and in vivo studies indicate that FTO may play a role in the anti-apoptotic mechanisms mediated by m6A in ovarian granulosa cells influenced by HYF. Moreover, employing qPCR and MeRIP-qPCR techniques, P53 has been identified as the target gene for m6A modification mediated by FTO. Conclusion These findings suggest that HYF holds promise as a potential treatment for POI and provide a more comprehensive understanding of the mechanism by which HYF operates, specifically its ability to prevent the BCL-2 mitochondrial apoptosis pathway mediated by P53 in ovarian granulosa cells of POI rats by regulating FTO/m6A-Tp53.
Collapse
Affiliation(s)
- Yang Li
- Department of Gynaecology, The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lingdi Wang
- Department of Gynecology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jian Liu
- Department of Gynaecology, The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guangning Nie
- Department of Gynaecology, The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hongyan Yang
- Department of Gynaecology, The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China
| |
Collapse
|
5
|
Dunlop CE, Anderson RA. Clinical dilemmas in ovarian tissue cryopreservation. Fertil Steril 2024; 122:559-564. [PMID: 38825305 DOI: 10.1016/j.fertnstert.2024.05.167] [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: 05/05/2024] [Revised: 05/26/2024] [Accepted: 05/29/2024] [Indexed: 06/04/2024]
Abstract
Ovarian tissue cryopreservation (OTC) is increasingly offered globally as a fertility preservation strategy for both postpubertal women and prepubertal girls, with subsequent reimplantation of cryopreserved ovarian cortex resulting in a rapidly growing number of live births. There remains very limited evidence of efficacy from tissue stored when the patient was prepubertal or from conditions affecting the ovary directly, e.g., Turner syndrome. Although OTC is becoming a more established practice, several clinical dilemmas remain from a practical and ethical standpoint. This review discusses the challenges regarding optimal patient selection for the procedure, the use of OTC in patients with a poor prognosis, the potential of reimplantation of tissue contaminated with malignant cells, and the role of OTC in those with an intrinsic ovarian disorder.
Collapse
Affiliation(s)
- Cheryl E Dunlop
- Obstetrics & Gynaecology Department, Edinburgh Royal Infirmary, Edinburgh, United Kingdom.
| | - Richard A Anderson
- Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
6
|
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
|
7
|
Bhuiyan J, Kavarthapu R, Soliman M, Brown GT, Yano JC, Dowlut-McElroy T, Gomez-Lobo V. Histologic analysis of gonadal tissue in patients with Turner syndrome and Y chromosome material (Response to commentary). J Pediatr Urol 2024:S1477-5131(24)00446-7. [PMID: 39299877 DOI: 10.1016/j.jpurol.2024.08.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 08/16/2024] [Indexed: 09/22/2024]
Affiliation(s)
- Julia Bhuiyan
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA.
| | - Raghuveer Kavarthapu
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Mary Soliman
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | | | - Jacqueline C Yano
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Tazim Dowlut-McElroy
- Pediatric and Adolescent Gynecology, Department of Surgery, Children's Mercy Hospital, Kansas City, MO, USA
| | - Veronica Gomez-Lobo
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| |
Collapse
|
8
|
Drechsel KCE, Broer SL, Stoutjesdijk FS, van Dulmen-den Broeder E, Beishuizen A, Wallace WH, Körholz D, Mauz-Körholz C, Hasenclever D, Cepelova M, Uyttebroeck A, Ronceray L, Twisk JWR, Kaspers GJL, Veening MA. The impact of treatment for childhood classical Hodgkin lymphoma according to the EuroNet-PHL-C2 protocol on serum anti-Müllerian Hormone. Hum Reprod 2024; 39:1701-1711. [PMID: 38794915 PMCID: PMC11291946 DOI: 10.1093/humrep/deae112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 04/23/2024] [Indexed: 05/26/2024] Open
Abstract
STUDY QUESTION What is the impact of the EuroNet-PHL-C2 treatment protocol for children with classical Hodgkin lymphoma (cHL) on gonadal function in girls, based on assessment of serum anti-Müllerian hormone (AMH)? SUMMARY ANSWER Serum AMH levels decreased after induction chemotherapy and increased during subsequent treatment and 2 years of follow-up, with lowest levels in patients treated for advanced stage cHL. WHAT IS KNOWN ALREADY Treatment for cHL, particularly alkylating agents and pelvic irradiation, can be gonadotoxic and result in premature reduction of primordial follicles in females. The current EuroNet-PHL-C2 trial aims to reduce the use of radiotherapy in standard childhood cHL treatment, by intensifying chemotherapy. This study aims to assess the gonadotoxic effect of the EuroNet-PHL-C2 protocol. STUDY DESIGN, SIZE, DURATION This international, prospective, multicenter cohort study is embedded in the EuroNet-PHL-C2 trial, an European phase-3 treatment study evaluating the efficacy of standard cHL treatment with OEPA-COPDAC-28 (OEPA: vincristine, etoposide, prednisone, and doxorubicin; COPDAC-28: cyclophosphamide, vincristine, prednisone, and dacarbazine) versus intensified OEPA-DECOPDAC-21 (DECOPDAC-21: COPDAC with additional doxorubicin and etoposide and 25% more cyclophosphamide) in a randomized setting. Participants were recruited between January 2017 and September 2021. PARTICIPANTS/MATERIALS, SETTING, METHODS Female patients aged ≤18 years, treated according to the EuroNet-PHL-C2 protocol for cHL were recruited across 18 sites in the Netherlands, Belgium, Germany, Austria, and Czech Republic. All parents and patients (aged ≥12 years old) provided written informed consent. Serum AMH levels and menstrual cycle characteristics were evaluated over time (at diagnosis, one to three times during treatment and 2 up to 5 years post-diagnosis) and compared between treatment-levels (TL1, TL2, and TL3) and treatment-arms (OEPA-COPDAC-28 and OEPA-DECOPDAC-21). Serum samples obtained from patients after receiving pelvic radiotherapy were excluded from the main analyses. MAIN RESULTS AND THE ROLE OF CHANCE A total of 104 females, with median age at diagnosis of 15.6 years (IQR 13.7; 17.0), were included in the analysis. Ninety-nine were (post)pubertal. Eighteen girls were diagnosed with an early stage of cHL (TL1) and 86 with intermediate or advanced stage disease (50 TL2 and 36 TL3, 66% received COPDAC-28 and 34% DECOPDAC-21). Five patients received pelvic radiotherapy. Median AMH level at diagnosis was 1.7 µg/l (IQR 0.9; 2.7). After two courses of OEPA chemotherapy, AMH levels decreased substantially in all patients (98% <0.5 µg/l), followed by a significant increase during the consolidation treatment and follow-up. After 2 years, 68% of patients reached their baseline AMH value, with overall median recovery of 129% (IQR 75.0; 208.9) compared to baseline measurement. Five patients (7%) had AMH <0.5 µg/l. In patients treated for advanced stage disease, AMH levels remained significantly lower compared to early- or intermediate stage disease, with median serum AMH of 1.3 µg/l (IQR 0.8; 2.1) after 2 years. Patients who received DECOPDAC-21 consolidation had lower AMH levels during treatment than patients receiving COPDAC-28, but the difference was no longer statistically significant at 2 years post-diagnosis. Of the 35 postmenarchal girls who did not receive hormonal co-treatment, 19 (54%) experienced treatment-induced amenorrhea, two girls had persisting amenorrhea after 2 years. LIMITATIONS, REASONS FOR CAUTION The studied population comprises young girls with diagnosis of cHL often concurring with pubertal transition, during which AMH levels naturally rise. There was no control population, while the interpretation of AMH as a biomarker during childhood is complex. The state of cHL disease may affect AMH levels at diagnosis, potentially complicating assessment of AMH recovery as a comparison with baseline AMH. The current analysis included data up to 2-5 years post-diagnosis. WIDER IMPLICATIONS OF THE FINDINGS The current PANCARE guideline advises to use the cyclophosphamide-equivalent dose score (CED-score, as an estimation of cumulative alkylating agent exposure) with a cut-off of 6000 mg/m2 to identify females aged <25 years at high risk of infertility. All treatment-arms of the EuroNet-PHL-C2 protocol remain below this cut-off, and based on this guideline, girls treated for cHL should therefore be considered low-risk of infertility. However, although we observed an increase in AMH after chemotherapy, it should be noted that not all girls recovered to pre-treatment AMH levels, particularly those treated for advanced stages of cHL. It remains unclear how our measurements relate to age-specific expected AMH levels and patterns. Additional (long-term) data are needed to explore clinical reproductive outcomes of survivors treated according to the EuroNet-PHL-C2 protocol. STUDY FUNDING/COMPETING INTEREST(S) The fertility add-on study was funded by the Dutch charity foundation KiKa (project 257) that funds research on all forms of childhood cancer. C.M-K., D.K., W.H.W., D.H., M.C., A.U., and A.B. were involved in the development of the EuroNet-PHL-C2 regimen. The other authors indicated no potential conflicts of interest. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- K C E Drechsel
- Pediatric Oncology, Cancer Center Amsterdam, Emma Children’s Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Paediatric Haemato-Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Cancer Center Amsterdam, Treament and quality of life, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands
| | - S L Broer
- Department of Reproductive Medicine & Gynecology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - F S Stoutjesdijk
- Pediatric Oncology, Cancer Center Amsterdam, Emma Children’s Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - E van Dulmen-den Broeder
- Pediatric Oncology, Cancer Center Amsterdam, Emma Children’s Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - A Beishuizen
- Department of Paediatric Haemato-Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Haematology/Oncology, Erasmus MC—Sophia Children’s Hospital, Rotterdam, The Netherlands
| | - W H Wallace
- Department of Haematology/Oncology, Royal Hospital for Sick Children, Edinburgh, UK
| | - D Körholz
- Department of Pediatric Hematology and Oncology, Universitätsklinikum Giessen und Marburg GmbH, Standort Giessen—Zentrum für Kinderheilkunde und Jugendmedizin, Giessen, Germany
| | - C Mauz-Körholz
- Department of Pediatric Hematology and Oncology, Universitätsklinikum Giessen und Marburg GmbH, Standort Giessen—Zentrum für Kinderheilkunde und Jugendmedizin, Giessen, Germany
- Clinic for Paediatric and Adolescent Medicine, Medical Faculty of the Martin, Luther University of Halle, Halle, Germany
| | - D Hasenclever
- Institut für Medizinische Informatik, Statistik und Epidemiologie, Universität Leipzig, Leipzig, Germany
| | - M Cepelova
- Department of Pediatric Hematology and Oncology, Faculty Hospital Motol and 2nd Medical Faculty, Charles University, Prague, Czech Republic
| | - A Uyttebroeck
- Department of Paediatric Haematology and Oncology, KU Leuven, UZ Leuven, Leuven, Belgium
| | - L Ronceray
- Pediatric Hematology and Oncology, St Anna Children's Hospital, Medical University of Vienna, Wien, Austria
| | - J W R Twisk
- Department of Epidemiology and Data Science, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - G J L Kaspers
- Pediatric Oncology, Cancer Center Amsterdam, Emma Children’s Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Paediatric Haemato-Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - M A Veening
- Pediatric Oncology, Cancer Center Amsterdam, Emma Children’s Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Paediatric Haemato-Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| |
Collapse
|
9
|
Decanter C, Elefant E, Poirot C, Courbiere B. What reproductive follow-up for adolescent and young women after cancer? A review. Reprod Biomed Online 2024; 49:103891. [PMID: 38761433 DOI: 10.1016/j.rbmo.2024.103891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 01/02/2024] [Accepted: 02/09/2024] [Indexed: 05/20/2024]
Abstract
Fertility capacity has been shown to be one of the main concerns of young cancer survivors. Gonadotoxic treatments may lead to both premature ovarian failure and/or infertility. This review aimed to define which, and when, reproductive indicators should be followed-up to help doctors to counsel patients regarding their fertility and ovarian function, and to determine if a second stage of fertility preservation after the end of cancer treatment is clinically relevant. Longitudinal assessment of anti-Müllerian hormone (AMH) concentrations during cancer treatment indicates the degree of follicular depletion, and allows discrimination between low and high gonadotoxic treatments. Sustained low AMH concentrations after treatment, especially in the case of alkylating protocols, may reduce the duration of the conception window significantly, and expose the patient to the risk of premature ovarian failure. It remains unknown whether this may impact further fertility capacity because of the lack of systematic follow-up of adolescent and young adult (AYA) women after chemo-radiotherapy. It appears that dedicated reproductive follow-up of AYA women under cancer treatment is needed to refine fertility preservation strategies, and to determine if low AMH concentrations after treatment impact the chance of pregnancy in this specific survivor population.
Collapse
Affiliation(s)
- C Decanter
- CHU Lille, ART and Fertility Preservation Department, Lille, France; ONCOLille Cancer Institute, Lille, France.
| | - E Elefant
- Reference Centre for Teratogenic Agents, Hospital Armand Trousseau, Paris, France; Faculty of Medicine, Sorbonne University, Paris, France
| | - C Poirot
- Department of Haematology, Adolescents and Young Adults, Fertility Preservation, Assistance Publique des Hôpitaux de Paris, Saint Louis Hospital, Paris, France; Medecine Sorbonne University, Paris, France
| | - B Courbiere
- Department of Gynaecology-Obstetric and Reproductive Medicine-Fertility Preservation, AP-HM, Hôpital La Conception, Marseille, France; Aix-Marseille Université, Avignon Université, Avignon, France
| |
Collapse
|
10
|
Magaton IM, Arecco L, Mariamidze E, Jankovic K, Stana M, Buzzatti G, Trevisan L, Scavone G, Ottonello S, Fregatti P, Massarotti C, von Wolff M, Lambertini M. Fertility and Pregnancy-Related Issues in Young BRCA Carriers With Breast Cancer. Breast Cancer (Auckl) 2024; 18:11782234241261429. [PMID: 38882447 PMCID: PMC11179469 DOI: 10.1177/11782234241261429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 05/06/2024] [Indexed: 06/18/2024] Open
Abstract
Approximately 10% to 15% of breast cancer cases in young women are diagnosed in patients harbouring germline (g) pathogenic or likely pathogenic variants (PVs) in the BReast CAncer 1 (BRCA1) or BReast CAncer 2 (BRCA2) genes. Preclinical and clinical studies showed a potential negative effect of germline BRCA1/2 (gBRCA1/2) PVs on ovarian reserve and reproductive potential, even before starting anticancer therapies. The aim of this article is to summarize the current literature on the fertility potential of young gBRCA1/2 PVs carriers with breast cancer and the risk of gonadotoxicity associated with anticancer treatments. Moreover, we describe the available evidence on the efficacy of fertility preservation techniques in young gBRCA1/2 PVs carriers and the safety data on having a pregnancy after breast cancer treatment.
Collapse
Affiliation(s)
- Isotta Martha Magaton
- Division of Gynaecological Endocrinology and Reproductive Medicine, University Women's Hospital, Inselspital, Bern, Switzerland
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Luca Arecco
- Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genoa, Genoa, Italy
| | - Elene Mariamidze
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Medical Oncology & Hematology, Todua Clinic, Tbilisi, Georgien
| | - Kristina Jankovic
- Department of Medical Oncology, University Clinic Center Nis, Nis, Serbia
| | - Mihaela Stana
- Department of Medical Oncology, Elysee Hospital, Alba Iulia, Romania
| | - Giulia Buzzatti
- Department of Medical Oncology, Unit of Hereditary Cancer, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Lucia Trevisan
- Department of Medical Oncology, Unit of Hereditary Cancer, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Graziana Scavone
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Silvia Ottonello
- Departent of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Piero Fregatti
- Department of Surgery, U.O.C. Clinica di Chirurgia Senologica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Surgical Sciences and Integrated Diagnostic (DISC), School of Medicine, University of Genoa, Genoa, Italy
| | - Claudia Massarotti
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics and Maternal and Child Health (DINOGMI Department), University of Genoa, Genoa, Italy
- Academic Unit of Obstetrics and Gynecology, Maternal-Child Department, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Michael von Wolff
- Division of Gynaecological Endocrinology and Reproductive Medicine, University Women's Hospital, Inselspital, Bern, Switzerland
| | - Matteo Lambertini
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genoa, Genoa, Italy
| |
Collapse
|
11
|
Foster KL, Lee DJ, Witchel SF, Gordon CM. Ovarian Insufficiency and Fertility Preservation During and After Childhood Cancer Treatment. J Adolesc Young Adult Oncol 2024; 13:377-388. [PMID: 38265460 DOI: 10.1089/jayao.2023.0111] [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] [Indexed: 01/25/2024] Open
Abstract
Premature ovarian insufficiency (POI) is one of many potential long-term consequences of childhood cancer treatment in females. Causes of POI in this patient population can include chemotherapy, especially alkylating agents, and radiation therapy. Rarely, ovarian tumors lead to ovarian dysfunction. POI can manifest as delayed pubertal development, irregular menses or amenorrhea, and infertility. This diagnosis often negatively impacts emotional health due to the implications of impaired ovarian function after already enduring treatment for a primary malignancy. The emerging adult may be challenged by the impact on energy level, quality of life, and fertility potential. POI can also lead to low bone density and compromised skeletal strength. This review discusses the health consequences of POI in childhood cancer survivors (CCS). We also explore the role of fertility preservation for CCS, including ovarian tissue cryopreservation and other available options. Lastly, knowledge gaps are identified that will drive a future research agenda.
Collapse
Affiliation(s)
- Kayla L Foster
- Texas Children's Cancer and Hematology Center, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Danielle J Lee
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Selma F Witchel
- Division of Pediatric Endocrinology, Department of Pediatrics, UPMC Children's Hospital, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Catherine M Gordon
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
12
|
Lai THT, Lau LSK, Ngu SF, Chu MYM, Chan KKL, Ng EHY, Ngan HYS, Li RHW, Tse KY. Comparison of the multiples of the median of serum anti-müllerian hormone and pregnancy outcomes in patients with gestational trophoblastic disease: A case-control study. Cancer Med 2024; 13:e7134. [PMID: 38545760 PMCID: PMC10973878 DOI: 10.1002/cam4.7134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 03/04/2024] [Accepted: 03/09/2024] [Indexed: 04/04/2024] Open
Abstract
INTRODUCTION Chemotherapy is crucial in treating gestational trophoblastic neoplasia (GTN), but its impact on gonadotoxicity is unclear. MATERIALS AND METHODS This case-control study included 57 GTN patients and 19 age-matched patients with molar pregnancies (MP) in 2012-2018. Multiples of the median (MoM) of the serum AMH levels were compared between the two groups, and between patients using single-agent and combination chemotherapy, at baseline, 6, 12, and 24 months after treatment. Their pregnancy outcomes were also compared. RESULTS There was no significant difference in the MoM of serum AMH between GTN and MP groups at all time points. Single-agent chemotherapy did not adversely affect the MoM. However, those receiving combination chemotherapy had lower MoM than those receiving single-agent chemotherapy at all time points. The trend of decline from the baseline was marginally significant in patients with combination chemotherapy, but the drop was only significant at 12 months (Z = -2.69, p = 0.007) but not at 24 months (Z = -1.90; p = 0.058). Multivariable analysis revealed that combination chemotherapy did not affect the MoM. There was no significant difference in the 4-year pregnancy rate and the livebirth rate between the single-agent and combination groups who attempting pregnancy, but it took 1 year longer to achieve the first pregnancy in the combination group compared to the single-agent group (2.88 vs. 1.88 years). CONCLUSION This study showed combination chemotherapy led to a decreasing trend of MoM of serum AMH especially at 12 months after treatment, but the drop became static at 24 months. Although pregnancy is achievable, thorough counseling is still needed in this group especially those wish to achieve pregnancy 1-2 years after treatment or with other risk factors.
Collapse
Affiliation(s)
| | - Lesley Suk Kwan Lau
- Division of Gynaecological Oncology, Department of Obstetrics and Gynaecology, School of Clinical MedicineHong KongChina
| | - Siew Fei Ngu
- Department of Obstetrics and GynaecologyQueen Mary HospitalHong KongChina
- Division of Gynaecological Oncology, Department of Obstetrics and Gynaecology, School of Clinical MedicineHong KongChina
| | - Man Yee Mandy Chu
- Department of Obstetrics and GynaecologyQueen Mary HospitalHong KongChina
- Division of Gynaecological Oncology, Department of Obstetrics and Gynaecology, School of Clinical MedicineHong KongChina
| | - Karen Kar Loen Chan
- Department of Obstetrics and GynaecologyQueen Mary HospitalHong KongChina
- Division of Gynaecological Oncology, Department of Obstetrics and Gynaecology, School of Clinical MedicineHong KongChina
| | - Ernest Hung Yu Ng
- Department of Obstetrics and GynaecologyQueen Mary HospitalHong KongChina
- Division of Reproductive Medicine, Department of Obstetrics and Gynaecology, School of Clinical MedicineThe University of Hong KongHong KongChina
| | - Hextan Yuen Sheung Ngan
- Department of Obstetrics and GynaecologyQueen Mary HospitalHong KongChina
- Division of Gynaecological Oncology, Department of Obstetrics and Gynaecology, School of Clinical MedicineHong KongChina
| | - Raymond Hang Wun Li
- Department of Obstetrics and GynaecologyQueen Mary HospitalHong KongChina
- Division of Reproductive Medicine, Department of Obstetrics and Gynaecology, School of Clinical MedicineThe University of Hong KongHong KongChina
| | - Ka Yu Tse
- Department of Obstetrics and GynaecologyQueen Mary HospitalHong KongChina
- Division of Gynaecological Oncology, Department of Obstetrics and Gynaecology, School of Clinical MedicineHong KongChina
| |
Collapse
|
13
|
Barcellini A, Cassani C, Orlandi E, Nappi RE, Broglia F, Delmonte MP, Molinelli S, Vai A, Vitolo V, Gronchi A, D'Ambrosio G, Cobianchi L, Fiore MR. Is motherhood still possible after pelvic carbon ion radiotherapy? A promising combined fertility-preservation approach. TUMORI JOURNAL 2024; 110:132-138. [PMID: 38183176 DOI: 10.1177/03008916231218794] [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] [Indexed: 01/07/2024]
Abstract
INTRODUCTION Preserving the endocrine and reproductive function in young female cancer patients undergoing pelvic radiation is a significant challenge. While the photon beam radiation's adverse effects on the uterus and ovaries are well established, the impact of pelvic carbon ion radiotherapy on women's reproductive function is largely unexplored. Strategies such as oocyte cryopreservation and ovarian transposition are commonly recommended for safeguarding future fertility. METHODS This study presents a pioneering case of successful pregnancy after carbon ion radiotherapy for locally advanced sacral chondrosarcoma. RESULTS A multidisciplinary approach facilitated the displacement of ovaries and uterus before carbon ion radiotherapy, resulting in the preservation of endocrine and reproductive function. CONCLUSION The patient achieved optimal oncological response and delivered a healthy infant following the completion of cancer treatment.
Collapse
Affiliation(s)
- Amelia Barcellini
- Radiation Oncology Unit, Clinical Department, CNAO National Center for Oncological Hadrontherapy, Pavia, Italy
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| | - Chiara Cassani
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
- Unit of Obstetrics and Gynecology, Foundation IRCCS Polyclinic San Matteo, Pavia, Italy
| | - Ester Orlandi
- Radiation Oncology Unit, Clinical Department, CNAO National Center for Oncological Hadrontherapy, Pavia, Italy
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Rossella E Nappi
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
- Research Center for Reproductive Medicine, Gynecological Endocrinology and Menopause, Foundation IRCCS Polyclinic San Matteo, Pavia, Italy
| | - Federica Broglia
- Department of Anesthesia and Intensive Care, Unit of Obstetric Anesthesia, Foundation IRCCS Polyclinic San Matteo, Pavia, Italy
| | - Maria Paola Delmonte
- Department of Anesthesia and Intensive Care, Unit of Obstetric Anesthesia, Foundation IRCCS Polyclinic San Matteo, Pavia, Italy
| | - Silvia Molinelli
- Medical Physics Unit, CNAO National Center for Oncological Hadrontherapy, Pavia, Italy
| | - Alessandro Vai
- Medical Physics Unit, CNAO National Center for Oncological Hadrontherapy, Pavia, Italy
| | - Viviana Vitolo
- Radiation Oncology Unit, Clinical Department, CNAO National Center for Oncological Hadrontherapy, Pavia, Italy
| | - Alessandro Gronchi
- Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Gioacchino D'Ambrosio
- Department of Molecular Medicine, Anatomic Pathology Unit, University of Pavia and Foundation IRCCS Polyclinic San Matteo, Pavia, Italy
| | - Lorenzo Cobianchi
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
- Department of General Surgery, Foundation IRCCS Polyclinic San Matteo, Pavia, Italy
- ITIR-Institute for Transformative Innovation Research, University of Pavia, Pavia, Italy
| | - Maria Rosaria Fiore
- Radiation Oncology Unit, Clinical Department, CNAO National Center for Oncological Hadrontherapy, Pavia, Italy
| |
Collapse
|
14
|
Hickey M, Basu P, Sassarini J, Stegmann ME, Weiderpass E, Nakawala Chilowa K, Yip CH, Partridge AH, Brennan DJ. Managing menopause after cancer. Lancet 2024; 403:984-996. [PMID: 38458217 DOI: 10.1016/s0140-6736(23)02802-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/18/2023] [Accepted: 12/11/2023] [Indexed: 03/10/2024]
Abstract
Globally, 9 million women are diagnosed with cancer each year. Breast cancer is the most commonly diagnosed cancer worldwide, followed by colorectal cancer in high-income countries and cervical cancer in low-income countries. Survival from cancer is improving and more women are experiencing long-term effects of cancer treatment, such as premature ovarian insufficiency or early menopause. Managing menopausal symptoms after cancer can be challenging, and more severe than at natural menopause. Menopausal symptoms can extend beyond hot flushes and night sweats (vasomotor symptoms). Treatment-induced symptoms might include sexual dysfunction and impairment of sleep, mood, and quality of life. In the long term, premature ovarian insufficiency might increase the risk of chronic conditions such as osteoporosis and cardiovascular disease. Diagnosing menopause after cancer can be challenging as menopausal symptoms can overlap with other common symptoms in patients with cancer, such as fatigue and sexual dysfunction. Menopausal hormone therapy is an effective treatment for vasomotor symptoms and seems to be safe for many patients with cancer. When hormone therapy is contraindicated or avoided, emerging evidence supports the efficacy of non-pharmacological and non-hormonal treatments, although most evidence is based on women older than 50 years with breast cancer. Vaginal oestrogen seems safe for most patients with genitourinary symptoms, but there are few non-hormonal options. Many patients have inadequate centralised care for managing menopausal symptoms after cancer treatment, and more information is needed about cost-effective and patient-focused models of care for this growing population.
Collapse
Affiliation(s)
- Martha Hickey
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne and the Royal Women's Hospital, Melbourne, VIC, Australia.
| | - Partha Basu
- Early Detection, Prevention and Infections Branch, International Agency for Research on Cancer, WHO, Lyon, France
| | - Jenifer Sassarini
- Department of Obstetrics and Gynaecology, School of Gynaecology, University of Glasgow, Glasgow, UK
| | - Mariken E Stegmann
- Department of Primary and Long-term Care, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | | | | | - Cheng-Har Yip
- Department of Surgery, University of Malaya, Kuala Lumpur, Malaysia
| | - Ann H Partridge
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Donal J Brennan
- Gynaecological Oncology Group, UCD School of Medicine, Mater Misericordiae University Hospital, Dublin, Ireland; Systems Biology Ireland, UCD School of Medicine, Dublin, Ireland
| |
Collapse
|
15
|
Yang EH, Strohl HB, Su HI. Fertility preservation before and after cancer treatment in children, adolescents, and young adults. Cancer 2024; 130:344-355. [PMID: 37962199 PMCID: PMC10841998 DOI: 10.1002/cncr.35108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/06/2023] [Accepted: 10/19/2023] [Indexed: 11/15/2023]
Abstract
Fertility is a top concern for many survivors of cancer diagnosed as children, adolescents and young adults (CAYA). Fertility preservation (FP) treatments are effective, evidence-based interventions to support their family building goals. Fertility discussions are a part of quality oncology care throughout the cancer care continuum. For nearly 2 decades, clinical guidelines recommend counseling patients about the possibility of infertility promptly at diagnosis and offering FP options and referrals as indicated. Multiple guidelines now recommend post-treatment counseling. Infertility risks differ by cancer treatments and age, rendering risk stratification a central part of FP care. To support FP decision-making, online tools for female risk estimation are available. At diagnosis, females can engage in mature oocyte/embryo cryopreservation, ovarian tissue cryopreservation, ovarian suppression with GnRH agonists, in vitro oocyte maturation, and/or conservative management for gynecologic cancers. Post-treatment, several populations may consider undergoing oocyte/embryo cryopreservation. Male survivors' standard of care FP treatments center on sperm cryopreservation before cancer treatment and do not have the same post-treatment indication for additional gamete cryopreservation. In practice, FP care requires systemized processes to routinely screen for FP needs, bridge oncology referrals to fertility, offer timely fertility consultations and access to FP treatments, and support financial navigation. Sixteen US states passed laws requiring health insurers to provide insurance benefits for FP treatments, but variation among the laws and downstream implementation are barriers to accessing FP treatments. To preserve the reproductive futures of CAYA survivors, research is needed to improve risk stratification, FP options, and delivery of FP care.
Collapse
Affiliation(s)
- Emily H. Yang
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego
| | | | - H. Irene Su
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Sciences, Moores Cancer Center, University of California, San Diego
| |
Collapse
|
16
|
Omranipour R, Ahmadi-Harchegani F, Saberi A, Moini A, Shiri M, Jalaeefar A, Arian A, Seifollahi A, Madani M, Eslami B, Alipour S. A New Model Including AMH Cut-off Levels to Predict Post-treatment Ovarian Function in Early Breast Cancer: A Prospective Cohort Study. ARCHIVES OF IRANIAN MEDICINE 2024; 27:96-104. [PMID: 38619033 PMCID: PMC11017260 DOI: 10.34172/aim.2024.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 12/24/2023] [Indexed: 04/16/2024]
Abstract
BACKGROUND Breast cancer (BC) treatment decreases fertility capacity, but unnecessary fertility preservation procedures in women who would not be infertile after treatment would be a waste of time and resources and could cause the unwarranted exposure of cancer cells to exogenous sex hormones. It has been largely shown that post-treatment ovarian reserve is directly associated with pre-treatment anti-mullerian hormone levels (AMH0). A threshold for AMH0, or a model including AMH0 and patient characteristics that could distinguish the patients who will be infertile after treatments, still needs to be defined. Accordingly, this study was performed to specifically target this high-priority concern. METHODS Women≤45 years old with newly diagnosed non-metastatic BC were entered in this multicenter prospective cohort study. AMH0 and two-year post-treatment AMH (AMH2) were measured, and hormonal patient features were recorded as well. Receiver operating characteristic (ROC) curve analysis, decision tree (DT), and random forest analyses were performed to find a cut-off point for AMH0 and define a model involving related features for the prediction of AMH2. RESULTS The data from 84 patients were analyzed. ROC curve analysis revealed that AMH0>3 ng/mL (Area under the curve=0.69, 95% CI: 0.54‒0.84) was the best indicator for predicting AMH2≥0.7 (sensitivity=79%, specificity=60%). The best model detected by DT and random forest for predicting an AMH2>0.7 with a probability of 93% consisted of a combination of AMH0>3.3, menarche age<14, and age<31. CONCLUSION This combination model can be used to withhold fertility preservation procedures in BC patients. Performing larger studies is suggested to further test this model.
Collapse
Affiliation(s)
- Ramesh Omranipour
- Breast Diseases Research Center (BDRC), Cancer Institute, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Oncologic Surgery, Cancer Institute, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Cancer Control Research Center, Cancer Control Foundation, Iran University of Medical Sciences, Tehran, Iran
| | | | - Azin Saberi
- Department of Surgery, Arash Women’s Hospital, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ashraf Moini
- Breast Diseases Research Center (BDRC), Cancer Institute, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Infertility, Arash Women’s Hospital, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Endocrinology and Female Infertility at Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Mostafa Shiri
- Faculty of Mathematical Sciences, Shahid Beheshti University, Tehran, Iran
| | - Amirmohsen Jalaeefar
- Department of Oncologic Surgery, Cancer Institute, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Arvin Arian
- Department of Radiology, Cancer Institute, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Tehran, Iran
| | - Akram Seifollahi
- Department of Pathology, Arash Women’s Hospital, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahshad Madani
- Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Bita Eslami
- Breast Diseases Research Center (BDRC), Cancer Institute, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sadaf Alipour
- Breast Diseases Research Center (BDRC), Cancer Institute, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Surgery, Arash Women’s Hospital, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Iwase A, Asada Y, Sugishita Y, Osuka S, Kitajima M, Kawamura K. Anti-Müllerian hormone for screening, diagnosis, evaluation, and prediction: A systematic review and expert opinions. J Obstet Gynaecol Res 2024; 50:15-39. [PMID: 37964401 DOI: 10.1111/jog.15818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 10/12/2023] [Indexed: 11/16/2023]
Abstract
AIM To present evidence-based recommendations for anti-Müllerian hormone (AMH) measurement as an ovarian reserve test. METHODS A systematic literature search for the clinical utility of AMH was conducted in PubMed from its inception to August 2022 to identify studies, including meta-analyses, reviews, randomized controlled trials, and clinical trials, followed by an additional systematic search using keywords. Based on this evidence, an expert panel developed clinical questions (CQs). RESULTS A total of 1895 studies were identified and 95 articles were included to establish expert opinions subdivided into general population, infertility treatment, primary ovarian insufficiency, polycystic ovary syndrome, surgery, and oncofertility. We developed 13 CQs and 1 future research question with levels of evidence and recommendations. CONCLUSION The findings of the current systematic review covered the clinical utility of AMH including its screening, diagnosis, evaluation, and prediction. Although some clinical implications of AMH remain debatable, these expert opinions may help promote a better understanding of AMH and establish its clinical significance.
Collapse
Affiliation(s)
- Akira Iwase
- Department of Obstetrics and Gynecology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | | | - Yodo Sugishita
- Department of Frontier Medicine, Institute of Medical Science, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Satoko Osuka
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Michio Kitajima
- Department of Obstetrics and Gynecology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kazuhiro Kawamura
- Department of Obstetrics and Gynaecology, Juntendo University Faculty of Medicine, Tokyo, Japan
| |
Collapse
|
18
|
He CY, Lee DJ, Foster KL, Gordon CM. Impact of ovarian insufficiency on bone health in childhood cancer survivors: Two cases. Bone 2024; 178:116930. [PMID: 37844715 DOI: 10.1016/j.bone.2023.116930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 09/14/2023] [Accepted: 10/08/2023] [Indexed: 10/18/2023]
Abstract
PURPOSE To investigate the skeletal phenotype of adolescent girls with premature ovarian insufficiency (POI). METHODS Data are presented from two adolescent girls who participated in a clinical research protocol to evaluate axial bone mineral density (BMD) (via dual-energy x-ray absorptiometry, DXA) and appendicular bone density, microarchitecture, and strength (via high-resolution peripheral quantitative computed tomography, HRpQCT). Anthropometric data were also obtained, and pubertal staging was performed by a clinician. RESULTS Both cases presented with an undetectable estradiol concentration and an elevated follicle stimulating hormone (FSH), meeting the criteria for POI. Each also received alkylating agents as part of their chemotherapy and radiotherapy, but in different locations as one presented with stage IV neuroblastoma and the other, metastatic medulloblastoma. Both had a low BMD of the axial and appendicular skeleton, as well as microarchitectural changes of the latter. The low BMD Z-score (<-2.0) seen when interpreting their DXA measurements for chronological age improved when adjusted for short stature, but it was not normalized. Lastly, most variables obtained by HRpQCT were abnormal for each participant, indicating that appendicular bone structure and strength were compromised. CONCLUSIONS Chemotherapy and radiation affect growth, puberty, and bone accrual deleteriously. However, as these cases show, POI in an adolescent is not always classic primary ovarian insufficiency. Adolescents with brain cancer can present with signs of estrogen deficiency but may not be able to secrete FSH to the extent of elevation typically seen in long-term cancer survivors. Estrogen deficiency is almost universally present in either clinical setting and prompt recognition facilitates early provision of hormone replacement therapy that may then allow for a resumption of bone accrual as an adolescent approaches her peak bone mass.
Collapse
Affiliation(s)
- Cara Y He
- Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States of America.
| | - Danielle J Lee
- USDA/ARS Children's Nutrition Research Center, 1100 Bates Avenue, Houston, TX 77030, United States of America.
| | - Kayla L Foster
- Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States of America; Section of Hematology/Oncology, Texas Children's Hospital, 6621 Fannin Street, Houston, TX 77030, United States of America.
| | - Catherine M Gordon
- Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States of America; USDA/ARS Children's Nutrition Research Center, 1100 Bates Avenue, Houston, TX 77030, United States of America.
| |
Collapse
|
19
|
Im C, Lu Z, Mostoufi-Moab S, Delaney A, Yu L, Baedke JL, Han Y, Sapkota Y, Yasui Y, Chow EJ, Howell RM, Bhatia S, Hudson MM, Ness KK, Armstrong GT, Nathan PC, Yuan Y. Development and validation of age-specific risk prediction models for primary ovarian insufficiency in long-term survivors of childhood cancer: a report from the Childhood Cancer Survivor Study and St Jude Lifetime Cohort. Lancet Oncol 2023; 24:1434-1442. [PMID: 37972608 PMCID: PMC10842148 DOI: 10.1016/s1470-2045(23)00510-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND Female survivors of childhood cancer are at risk for primary ovarian insufficiency (POI), defined as the cessation of gonadal function before the age of 40 years. We aimed to develop and validate models to predict age-specific POI risk among long-term survivors of childhood cancer. METHODS To develop models to predict age-specific POI risk for the ages of 21-40 years, we used data from the Childhood Cancer Survivor Study (CCSS). Female survivors aged 18 years or older at their latest follow-up, with self-reported menstrual history information and free of subsequent malignant neoplasms within 5 years of diagnosis, were included. We evaluated models that used algorithms based on statistical or machine learning to consider all predictors, including cancer treatments. Cross-validated prediction performance metrics (eg, area under the receiver operating characteristic curve [AUROC]) were compared to select the best-performing models. For external validation of the models, we used data from 5-year survivors in the St Jude Lifetime Cohort (SJLIFE) with ovarian status clinically ascertained using hormone measurements (menopause defined by follicle stimulating hormone >30 mIU/mL and oestradiol <17 pg/mL) and medical chart or questionnaire review. We also evaluated an SJLIFE-based polygenic risk score for POI among 1985 CCSS survivors with genotype data available. FINDINGS 7891 female CCSS survivors (922 with POI) were included in the development of the POI risk prediction model, and 1349 female SJLIFE survivors (101 with POI) were included in the validation study. Median follow-up from cancer diagnosis was 23·7 years (IQR 18·3-30·0) in CCSS and 15·1 years (10·4-22·9) in SJLIFE. Between the ages of 21 and 40 years, POI prevalence increased from 7·9% (95% CI 7·3-8·5) to 18·6% (17·3-20·0) in CCSS and 7·3% (5·8-8·9) to 14·9% (11·6-19·1) in SJLIFE. Age-specific logistic regression models considering ovarian radiation dosimetry or prescribed pelvic and abdominal radiation dose, along with individual chemotherapy predictors, performed well in CCSS. In the SJLIFE validation, the prescribed radiation dose model performed well (AUROC 0·88-0·95), as did a simpler model that considered any exposures to pelvic or abdominal radiotherapy or alkylators (0·82-0·90). Addition of the polygenic risk predictor significantly improved the average positive predictive value (from 0·76 [95% CI 0·63-0·89] to 0·87 [0·80-0·94]; p=0·029) among CCSS survivors treated with ovarian radiation and chemotherapy. INTERPRETATION POI risk prediction models using treatment information showed robust prediction performance in adult survivors of childhood cancer. FUNDING Canadian Institutes of Health Research, US National Cancer Institute.
Collapse
Affiliation(s)
- Cindy Im
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Zhe Lu
- School of Public Health, University of Alberta, Edmonton, AB, Canada
| | - Sogol Mostoufi-Moab
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Angela Delaney
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, TN, USA; Division of Endocrinology, Department of Pediatric Medicine, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Lin Yu
- School of Public Health, University of Alberta, Edmonton, AB, Canada
| | - Jessica L Baedke
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Yutong Han
- School of Public Health, University of Alberta, Edmonton, AB, Canada
| | - Yadav Sapkota
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Yutaka Yasui
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Eric J Chow
- Public Health Sciences and Clinical Research Divisions, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Rebecca M Howell
- Department of Radiation Physics, University of Texas at MD Anderson Cancer Center, Houston, TX, USA
| | - Smita Bhatia
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Melissa M Hudson
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, TN, USA; Department of Oncology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Kirsten K Ness
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Gregory T Armstrong
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, TN, USA; Department of Oncology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Paul C Nathan
- Division of Hematology/Oncology, The Hospital for Sick Children, Toronto, Canada
| | - Yan Yuan
- School of Public Health, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
20
|
Ding X, Lv S, Guo Z, Gong X, Wang C, Zhang X, Meng K. Potential Therapeutic Options for Premature Ovarian Insufficiency: Experimental and Clinical Evidence. Reprod Sci 2023; 30:3428-3442. [PMID: 37460850 DOI: 10.1007/s43032-023-01300-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 07/10/2023] [Indexed: 12/03/2023]
Abstract
Premature ovarian insufficiency (POI) is a condition in which a woman experiences premature decline in ovarian function before the age of 40 years, manifested by menstrual disorders, decreased fertility, and possibly postmenopausal symptoms such as insomnia, hot flashes, and osteoporosis, and is one of the predominant clinical syndromes leading to female infertility. Genetic, immunologic, iatrogenic and other factors, alone or in combination, have been reported to trigger POI, yet the etiology remains unknown in most cases. The main methods currently used clinically to ameliorate menopausal symptoms due to hypoestrogenemia in POI patients are hormone replacement therapy, while the primary methods available to address infertility in POI patients are oocyte donation and cryopreservation techniques, both of which have limitations to some degree. In recent years, researchers have continued to explore more efficient and safe therapies, and have achieved impressive results in preclinical trials. In this article, we will mainly review the three most popular therapies and their related signaling pathways published in the past ten years, with the aim of providing ideas for clinical applications.
Collapse
Affiliation(s)
- Xuechun Ding
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, China
- College of Second Clinical Medical, Jining Medical University, Jining, China
| | - Shenmin Lv
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, China
- College of Second Clinical Medical, Jining Medical University, Jining, China
| | - Zhipeng Guo
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, China
- College of Second Clinical Medical, Jining Medical University, Jining, China
| | - Xiaowei Gong
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, China
- College of Second Clinical Medical, Jining Medical University, Jining, China
| | - Caiqin Wang
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, China
- College of Second Clinical Medical, Jining Medical University, Jining, China
| | - Xiaoyan Zhang
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, China
- College of Basic Medicine, Jining Medical University, Jining, China
| | - Kai Meng
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, China.
- Lin He's Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining, China.
| |
Collapse
|
21
|
Garrido Colino C, González Urdiales P, Molinés Honrubia A, Ortega Acosta MJ, García Abos M. Primary ovarian insufficiency in cancer survivors: Keys to optimal management. An Pediatr (Barc) 2023; 99:385-392. [PMID: 37993293 DOI: 10.1016/j.anpede.2023.11.007] [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: 05/22/2023] [Accepted: 09/24/2023] [Indexed: 11/24/2023] Open
Abstract
INTRODUCTION Primary ovarian insufficiency (POI) carries significant morbidity, causing infertility, sexual disfunction, decreased bone density, cardiovascular risk, emotional distress and early mortality. OBJECTIVE To know the incidence and current management of POI in childhood/adolescent solid tumour survivors. MATERIAL AND METHODS We conducted a multicentre observational study. It included female patients aged 12-18 years with a diagnosis of solid tumour and meeting clinical or biochemical criteria for POI. The risk was estimated based on the criteria of the Pediatric Initiative Network of the Oncofertility Consortium. RESULTS We found an incidence of 1.5 (30 cases of POI): The median age at the time of the event was 14 years (standard deviation, 2.09). The solid tumours associated most frequently with POI were Ewing sarcoma and brain and germ cell tumours. Eighty-three percent of patients did not undergo fertility preservation. Sixty-three percent reported not having undergone menarche at the time of ovarian failure. Ninety-seven percent were at high risk of gonadal toxicity, yet 47% were not monitored before the diagnosis. The median time elapsed to the occurrence of the event was 43.5 months after diagnosis and 29.5 months after completing treatment. The Kaplan-Meier curves showed that approximately 30% of POI cases developed within 2 years of diagnosis and that women at Tanner stage 1 developed insufficiency later than women at Tanner stage 5. CONCLUSIONS There is room for improvement in the follow-up of women at risk of POI in Spain. The tools currently available facilitate risk assessment at the time of treatment planning and allow the implementation of monitoring, education, early diagnosis, fertility preservation, and replacement therapy as needed. All of this would achieve significant improvement in health outcomes.
Collapse
Affiliation(s)
- Carmen Garrido Colino
- Servicio de Pediatría, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.
| | | | - Antonio Molinés Honrubia
- Servicio de Hematología, Hospital Insular Las Palmas de Gran Canarias, Las Palmas de Gran Canaria, Las Palmas, Spain
| | | | - Mirian García Abos
- Servicio de Pediatría, Hospital Universitario Donostia, Donostia, Gipuzkoa, Spain
| |
Collapse
|
22
|
Iwase A, Hasegawa Y, Tsukui Y, Kobayashi M, Hiraishi H, Nakazato T, Kitahara Y. Anti-Müllerian hormone beyond an ovarian reserve marker: the relationship with the physiology and pathology in the life-long follicle development. Front Endocrinol (Lausanne) 2023; 14:1273966. [PMID: 38027144 PMCID: PMC10657644 DOI: 10.3389/fendo.2023.1273966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Anti-Müllerian hormone (AMH), an indirect indicator of the number of remaining follicles, is clinically used as a test for ovarian reserve. Typically, a decline suggests a decrease in the number of remaining follicles in relation to ovarian toxicity caused by interventions, which may implicate fertility. In contrast, serum AMH levels are elevated in patients with polycystic ovary syndrome. AMH is produced primarily in the granulosa cells of the preantral and small antral follicles. Thus it varies in association with folliculogenesis and the establishment and shrinking of the follicle cohort. Ovarian activity during the female half-life, from the embryonic period to menopause, is based on folliculogenesis and maintenance of the follicle cohort, which is influenced by developmental processes, life events, and interventions. AMH trends over a woman's lifetime are associated with in vivo follicular cohort transitions that cannot be observed directly.
Collapse
Affiliation(s)
- Akira Iwase
- Department of Obstetrics and Gynecology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | | | | | | | | | | | | |
Collapse
|
23
|
Drechsel KCE, Broer SL, Stoutjesdijk FS, Twisk JWR, van den Berg MH, Lambalk CB, van Leeuwen FE, Overbeek A, van den Heuvel-Eibrink MM, van Dorp W, de Vries ACH, Loonen JJ, van der Pal HJ, Kremer LC, Tissing WJ, Versluys B, Kaspers GJL, van Dulmen-den Broeder E, Veening MA. Clinical and self-reported markers of reproductive function in female survivors of childhood Hodgkin lymphoma. J Cancer Res Clin Oncol 2023; 149:13677-13695. [PMID: 37522923 PMCID: PMC10590326 DOI: 10.1007/s00432-023-05035-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: 05/22/2023] [Accepted: 06/28/2023] [Indexed: 08/01/2023]
Abstract
PURPOSE To evaluate the impact of treatment for Hodgkin lymphoma (HL) on clinical reproductive markers and pregnancy outcomes. METHODS This study was embedded within the DCOG LATER-VEVO study; a Dutch, multicenter, retrospective cohort study between 2004 and 2014. Serum anti-Müllerian hormone (AMH), follicle stimulating hormone (FSH), inhibin B, antral follicle count (AFC), and self-reported (first) pregnancy outcomes were evaluated in female childhood HL survivors and controls. RESULTS 84 HL survivors and 798 controls were included, aged 29.6 and 32.7 years old at time of assessment. Median age at HL diagnosis was 13.4 years. Cyclophosphamide equivalent dose (CED-score) exceeded 6000 mg/m2 in 56 women and 14 survivors received pelvic irradiation. All clinical markers were significantly deteriorated in survivors (odds-ratio for low AMH (< p10) 10.1 [95% CI 4.9; 20.6]; low AFC (< p10) 4.6 [95% CI 2.1; 9.9]; elevated FSH (> 10 IU/l) 15.3 [95% CI 5.7; 41.1], low Inhibin B (< 20 ng/l) 3.6 [ 95% CI 1.7; 7.7], p < 0.001). Pregnancy outcomes were comparable between survivors and controls (± 80% live birth, ± 20% miscarriage). However, survivors were significantly younger at first pregnancy (27.0 years vs 29.0 years, P = 0.04). Adjusted odds-ratio for time to pregnancy > 12 months was 2.5 [95% CI 1.1; 5.6] in survivors, p = 0.031. Adverse outcomes were specifically present after treatment with procarbazine and higher CED-score. CONCLUSION HL survivors appear to have an impaired ovarian reserve. However, chance to achieve pregnancy seems reassuring at a young age. Additional follow-up studies are needed to assess fertile life span and reproductive potential of HL survivors, in particular for current HL treatments that are hypothesized to be less gonadotoxic.
Collapse
Affiliation(s)
- K C E Drechsel
- Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric Oncology, Amsterdam, The Netherlands.
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.
- Cancer Center Amsterdam, Amsterdam UMC, Location VUmc, Amsterdam, Netherlands.
| | - S L Broer
- Department of Reproductive Medicine & Gynecology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - F S Stoutjesdijk
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - J W R Twisk
- Department of Epidemiology and Data Science, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - M H van den Berg
- Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric Oncology, Amsterdam, The Netherlands
| | - C B Lambalk
- Department of Obstetrics and Gynaecology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - F E van Leeuwen
- Department of Epidemiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - A Overbeek
- Department of Obstetrics and Gynaecology, Northwest Clinics, Alkmaar, The Netherlands
| | - M M van den Heuvel-Eibrink
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Paediatric Hemato-Oncology, Erasmus MC - Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - W van Dorp
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - A C H de Vries
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Paediatric Hemato-Oncology, Erasmus MC - Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - J J Loonen
- Department of Hematology, Radboudumc Center of Expertise for Cancer Survivorship, Radboud University Medical Center, Nijmegen, The Netherlands
| | - H J van der Pal
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - L C Kremer
- Cancer Center Amsterdam, Amsterdam UMC, Location VUmc, Amsterdam, Netherlands
| | - W J Tissing
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - B Versluys
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Heamatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | - G J L Kaspers
- Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric Oncology, Amsterdam, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - E van Dulmen-den Broeder
- Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric Oncology, Amsterdam, The Netherlands
| | - M A Veening
- Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric Oncology, Amsterdam, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| |
Collapse
|
24
|
Cui W, Rocconi RP, Thota R, Anderson RA, Bruinooge SS, Comstock IA, Denduluri N, Gassman A, Gralow J, Hutt KJ, Amiri-Kordestani L, Lambertini M, Leighton J, Lu KH, Mostoufi-Moab S, Pollastro T, Pradhan S, Saber H, Schenkel C, Spratt D, Wedam S, Phillips KA. Measuring ovarian toxicity in clinical trials: an American Society of Clinical Oncology research statement. Lancet Oncol 2023; 24:e415-e423. [PMID: 37797647 DOI: 10.1016/s1470-2045(23)00390-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/24/2023] [Accepted: 08/03/2023] [Indexed: 10/07/2023]
Abstract
Anticancer agents can impair ovarian function, resulting in premature menopause and associated long-term health effects. Ovarian toxicity is not usually adequately assessed in trials of anticancer agents, leaving an important information gap for patients facing therapy choices. This American Society of Clinical Oncology (ASCO) statement provides information about the incorporation of ovarian toxicity measures in trial design. ASCO recommends: (1) measurement of ovarian toxicity in relevant clinical trials of anticancer agents that enrol post-pubertal, pre-menopausal patients; (2) collection of ovarian function measures at baseline and at 12-24 months after anticancer agent cessation, as a minimum, and later in line with the trial schedule; and (3) assessment of both clinical measures and biomarkers of ovarian function. ASCO recognises that routine measurement of ovarian toxicity and function in cancer clinical trials will add additional complexity and burden to trial resources but asserts that this issue is of such importance to patients that it cannot continue to be overlooked.
Collapse
Affiliation(s)
- Wanda Cui
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Rodney P Rocconi
- The University of Mississippi Medical Center, Cancer Center & Research Institute, Jackson, MS, USA
| | | | - Richard A Anderson
- Medical Research Council Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | | | - Ioanna A Comstock
- Office of Rare Diseases, Pediatrics, Urologic and Reproductive Medicine, Division of Urology, Obstetrics, and Gynecology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | | | - Audrey Gassman
- Office of Rare Diseases, Pediatrics, Urologic and Reproductive Medicine, Division of Urology, Obstetrics, and Gynecology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Julie Gralow
- American Society of Clinical Oncology, Alexandria, VA, USA
| | - Karla J Hutt
- Development and Stem Cell Program and Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Laleh Amiri-Kordestani
- Division of Oncology 1, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Matteo Lambertini
- Department of Internal Medicine and Medical Specialties, School of Medicine, University of Genova, Genoa, Italy; Department of Medical Oncology, UO Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - John Leighton
- Division of Hematology Oncology Toxicity, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Karen H Lu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sogol Mostoufi-Moab
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Shan Pradhan
- Office of Oncologic Diseases, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Haleh Saber
- Division of Hematology Oncology Toxicity, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | | | - Daniel Spratt
- Moffitt Cancer Center and Research Institute, Cleveland, OH, USA
| | - Suparna Wedam
- Division of Oncology 1, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Kelly-Anne Phillips
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia; Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
25
|
Minoia C, Viviani S, Silvestris E, Palini S, Parissone F, De Palma G, Fedina A, Cormio G, Guarini A, Gini G, Montano L, Merli F, Peccatori FA. Fertility preservation and monitoring in adult patients diagnosed with lymphoma: consensus-based practical recommendations by the Fondazione Italiana Linfomi & Società Italiana della Riproduzione Umana. Front Oncol 2023; 13:1252433. [PMID: 37766870 PMCID: PMC10520955 DOI: 10.3389/fonc.2023.1252433] [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] [Received: 07/03/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
Introduction Fertility preservation (FP) and monitoring has considerable relevance in the multidisciplinary approach to cancer patients. In these consensus-based practical recommendations, the scientific societies Fondazione Italiana Linfomi (FIL) and Società Italiana della Riproduzione Umana (SIRU) reviewed the main aspects and identified the optimal paths which aim to preserve and monitor fertility in patients diagnosed with lymphoma at the different phases of the disease and during long-term survivorship. Methods For the Panel, eleven experts were selected for their expertise in research and clinical practice on onco-fertility and lymphoma. The Panel's activity was supervised by a chairman. A series of rank-ordering key questions were proposed according to their clinical relevance and discussed among the Panel, focusing on patients diagnosed with non-Hodgkin's lymphomas and Hodgkin lymphoma. Agreement among all the Panelists on the content and terminology of the statements was evaluated by a web-based questionnaire according to the Delphi methodology. Results From the literature review a total of 78 questions or sentences, divided into the 6 areas of interest, were identified. By applying the Gwet's AC, k was: Section 1: 0,934 (Very good); Section 2: 0,958 (Very good); Section 3: 0,863 (Very good); Section 4: 0,649 (Good); Section 5: 0,936 (Very good); Section 6 raw agreement 100%. Two rounds of Delphi allowed to provide the maximum agreement. All statements were newly discussed in a round robin way and confirmed for the drafting of the final recommendations. Discussion These recommendations would be useful for onco-hematologists, gynecologists, urologists, and general practice physicians who take care of young lymphoma patients to guarantee an evidence-based oncofertility assessment and treatment during the oncologic pathway.
Collapse
Affiliation(s)
- Carla Minoia
- Hematology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Simonetta Viviani
- Division of Onco-Hematology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Erica Silvestris
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Simone Palini
- Physiopathology of Reproduction Unit, Cervesi Hospital, Cattolica, Italy
| | - Francesca Parissone
- Department of Obstetrics and Gynecology, Azienda Ospedaliera Universitaria Integrata di Verona, Università di Verona, Verona, Italy
| | - Giuseppe De Palma
- Institutional BioBank, Experimental Oncology and Biobank Management Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Anna Fedina
- Data Office Fondazione Italiana Linfomi, Alessandria, Italy
| | - Gennaro Cormio
- IRCCS Istituto Tumori Departiment of Interdisciplinary Medicine (DIM), University of Bari, Bari, Italy
| | - Attilio Guarini
- Hematology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Guido Gini
- Clinic of Hematology Azienda Ospedaliera Universitaria (AOU) delle Marche, Ancona, Italy
| | - Luigi Montano
- Andrology Unit and Service of Lifestyle Medicine in UroAndrology, Local Health Authority (ASL), Salerno, Italy
| | - Francesco Merli
- Hematology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | | |
Collapse
|
26
|
Stern E, Ben-Ami M, Gruber N, Toren A, Caspi S, Abebe-Campino G, Lurye M, Yalon M, Modan-Moses D. Hypothalamic-pituitary-gonadal function, pubertal development, and fertility outcomes in male and female medulloblastoma survivors: a single-center experience. Neuro Oncol 2023; 25:1345-1354. [PMID: 36633935 PMCID: PMC10326472 DOI: 10.1093/neuonc/noad009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Endocrine deficiencies, including hypothalamic-pituitary-gonadal axis (HPGA) impairment, are common in survivors of childhood and adolescent medulloblastoma. Still, data regarding pubertal development and fecundity are limited, and few studies assessed HPGA function in males. We aimed to describe HPGA function in a large cohort of patients with medulloblastoma. METHODS A retrospective study comprising all 62 medulloblastoma patients treated in our center between 1987 and 2021, who were at least 2 years from completion of therapy. HPGA function was assessed based on clinical data, biochemical markers, and questionnaires. RESULTS Overall, 76% of female patients had clinical or biochemical evidence of HPGA dysfunction. Biochemical evidence of diminished ovarian reserve was seen in all prepubertal girls (n = 4). Among the males, 34% had clinical or biochemical evidence of gonadal dysfunction, 34% had normal function, and 29% were age-appropriately clinically and biochemically prepubertal. The difference between males and females was significant (P = .003). Cyclophosphamide-equivalent dose was significantly associated with HPGA function in females, but not in males. There was no association between HPGA dysfunction and other endocrine deficiencies, length of follow-up, weight status, and radiation treatment protocol. Two female and 2 male patients achieved successful pregnancies, resulting in 6 live births. CONCLUSIONS HPGA dysfunction is common after treatment for childhood medulloblastoma. This is seen more in females, likely due to damage to the ovaries from spinal radiotherapy. Our findings may assist in counseling patients and their families regarding risk to future fertility and need for fertility preservation.
Collapse
Affiliation(s)
- Eve Stern
- Pediatric Endocrinology and Diabetes Unit, The Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Ramat-Gan, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michal Ben-Ami
- Pediatric Endocrinology and Diabetes Unit, The Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Ramat-Gan, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Noah Gruber
- Pediatric Endocrinology and Diabetes Unit, The Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Ramat-Gan, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Amos Toren
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Division of Pediatric Hematology-Oncology, The Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Ramat-Gan, Israel
| | - Shani Caspi
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Division of Pediatric Hematology-Oncology, The Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Ramat-Gan, Israel
| | - Gadi Abebe-Campino
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Division of Pediatric Hematology-Oncology, The Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Ramat-Gan, Israel
| | - Michal Lurye
- Division of Pediatric Hematology-Oncology, The Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Ramat-Gan, Israel
| | - Michal Yalon
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Division of Pediatric Hematology-Oncology, The Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Ramat-Gan, Israel
| | - Dalit Modan-Moses
- Pediatric Endocrinology and Diabetes Unit, The Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Ramat-Gan, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
27
|
Zhou XY, Yang YZ, Zhang J, Zhang XF, Liu YD, Wang Z, Chen SL. Elevated cell-free mitochondria DNA level of patients with premature ovarian insufficiency. BMC Pregnancy Childbirth 2023; 23:462. [PMID: 37349693 DOI: 10.1186/s12884-023-05769-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 06/09/2023] [Indexed: 06/24/2023] Open
Abstract
BACKGROUND Premature ovarian insufficiency (POI) patients present with a chronic inflammatory state. Cell-free mitochondria DNA (cf-mtDNA) has been explored as a reliable biomarker for estimating the inflammation-related disorders, however, the cf-mtDNA levels in POI patients have never been measured. Therefore, in the presenting study, we aimed to evaluate the levels of cf-mtDNA in plasma and follicular fluid (FF) of POI patients and to determine a potential role of cf-mtDNA in predicting the disease progress and pregnancy outcomes. METHODS We collected plasma and FF samples from POI patients, biochemical POI (bPOI) patients and control women. Quantitative real-time PCR was used to measure the ratio of mitochondrial genome to nuclear genome of cf-DNAs extracted from the plasma and FF samples. RESULTS The plasma cf-mtDNA levels, including COX3, CYB, ND1 and mtDNA79, were significantly higher in overt POI patients than those in bPOI patients or control women. The plasma cf-mtDNA levels were weakly correlated with ovarian reserve, and could not be improved by regular hormone replacement therapy. The levels of cf-mtDNA in FF, rather than those in plasma, exhibited the potential to predict the pregnancy outcomes, although they were comparable among overt POI, bPOI and control groups. CONCLUSIONS The increased plasma cf-mtDNA levels in overt POI patients indicated its role in the progress of POI and the FF cf-mtDNA content may hold the value in predicting pregnancy outcomes of POI patients.
Collapse
Affiliation(s)
- Xing-Yu Zhou
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Northern Road, Guangzhou, 510515, Guangdong, China
| | - Yi-Zhen Yang
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Northern Road, Guangzhou, 510515, Guangdong, China
| | - Jun Zhang
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Northern Road, Guangzhou, 510515, Guangdong, China
| | - Xiao-Fei Zhang
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Northern Road, Guangzhou, 510515, Guangdong, China
| | - Yu-Dong Liu
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Northern Road, Guangzhou, 510515, Guangdong, China
| | - Zhe Wang
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Northern Road, Guangzhou, 510515, Guangdong, China
| | - Shi-Ling Chen
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Northern Road, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
28
|
Slonim M, Peate M, Merigan K, Lantsberg D, Anderson RA, Stern K, Gook D, Jayasinghe Y. Ovarian stimulation and oocyte cryopreservation in females and transgender males aged 18 years or less: a systematic review. Front Endocrinol (Lausanne) 2023; 14:1146476. [PMID: 37404308 PMCID: PMC10315913 DOI: 10.3389/fendo.2023.1146476] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 05/15/2023] [Indexed: 07/06/2023] Open
Abstract
Background Fertility preservation is an important healthcare focus in the paediatric and adolescent population when gonadotoxic treatments are required. Ovarian stimulation (OS) resulting in oocyte cryopreservation is a well-established fertility preservation option in the adult population. It's utility, however, is little known in young patients. The purpose of this review was to synthesise the available literature on OS in patients ≤18 years old, to identify gaps in current research and provide suggestions for future research directions. Methods Using PRISMA guidelines, a systematic review of the literature was performed for all relevant full-text articles published in English in Medline, Embase, the Cochrane Library and Google Scholar databases. The search strategy used a combination of subject headings and generic terms related to the study topic and population. Two reviewers independently screened studies for eligibility, extracted data and assessed the risk of bias. Characteristics of the studies, objectives and key findings were extracted and summarised in a narrative synthesis. Results Database search and manual review identified 922 studies, 899 were eliminated based on defined exclusion criteria. Twenty-three studies were included and comprised 468 participants aged ≤18 years who underwent OS (median 15.2, range 7-18 years old). Only three patients were premenarchal, and four patients were on treatment to suppress puberty. Patients had OS for a broad range of indications including oncology treatment, transgender care and Turner syndrome. A total of 488 cycles of OS were completed, with all but 18 of these cycles (96.3%) successfully resulting in cryopreserved mature oocytes (median 10 oocytes, range 0-35). Fifty-three cycles (9.8%) were cancelled. Complications were rare (<1%). One pregnancy was reported from a female who had OS aged 17 years old. Conclusion This systematic review demonstrates that OS and oocyte cryopreservation is achievable in young females however there are only a few cases in the literature describing OS in premenarcheal children or those who have suppressed puberty. There is little proof that OS can lead to pregnancy in adolescents, and no proof that this can be achieved in premenarchal girls. Therefore it should be regarded as an innovative procedure for adolescents and experimental for premenarcheal girls. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=265705, identifier CRD42021265705.
Collapse
Affiliation(s)
- Marnie Slonim
- Oncofertility Program and Department of Gynaecology, Royal Children’s Hospital, Melbourne, VIC, Australia
| | - Michelle Peate
- Department of Obstetrics and Gynaecology, Royal Women's Hospital, The University of Melbourne, Melbourne, VIC, Australia
| | - Kira Merigan
- Department of Obstetrics and Gynaecology, Royal Women’s Hospital, Melbourne, VIC, Australia
| | | | - Richard A. Anderson
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| | - Kate Stern
- Department of Obstetrics and Gynaecology, Royal Women’s Hospital, Melbourne, VIC, Australia
- Melbourne IVF, Melbourne, VIC, Australia
| | - Debra Gook
- Department of Obstetrics and Gynaecology, Royal Women's Hospital, The University of Melbourne, Melbourne, VIC, Australia
- Melbourne IVF, Melbourne, VIC, Australia
| | - Yasmin Jayasinghe
- Oncofertility Program and Department of Gynaecology, Royal Children’s Hospital, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Royal Women's Hospital, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
29
|
Germeyer A, Nawroth F. [Indication and implementation of fertility preservation measures in female cancer patients]. DERMATOLOGIE (HEIDELBERG, GERMANY) 2023:10.1007/s00105-023-05171-0. [PMID: 37314451 DOI: 10.1007/s00105-023-05171-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Accepted: 05/04/2023] [Indexed: 06/15/2023]
Abstract
The aspects of fertility preservation in women prior to surgical, gonadotoxic or radiation therapy represent a challenging topic in many disciplines and often in an interdisciplinary setting. Within an often short period of time, individual counselling and consideration must be given as to whether fertility-protective measures are useful. The implementation is ultimately decided by the patient. A prerequisite for helpful counselling is knowledge about the potential effects of cancer treatment on ovarian function as well as the implementation and potential individual benefits of fertility-protective measures. Networks such as FertiPROTEKT Netzwerk e. V. are helpful for orientation in terms of content and timely implementation of counselling and corresponding measures.
Collapse
Affiliation(s)
- Ariane Germeyer
- Gynäkologische Endokrinologie und Fertilitätsstörungen, Universitäts-Frauenklinik Heidelberg, Im Neuenheimer Feld 440, 69120, Heidelberg, Deutschland.
| | - Frank Nawroth
- Facharzt-Zentrum für Kinderwunsch, Pränatale Medizin, Endokrinologie und Osteologie, amedes MVZ Hamburg, Hamburg, Deutschland
| |
Collapse
|
30
|
Su C, Zhang R, Zhang X, Lv M, Liu X, Ao K, Hao J, Mu YL. Dingkun Pill modulate ovarian function in chemotherapy-induced premature ovarian insufficiency mice by regulating PTEN/PI3K/AKT/FOXO3a signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2023:116703. [PMID: 37257704 DOI: 10.1016/j.jep.2023.116703] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/22/2023] [Accepted: 05/29/2023] [Indexed: 06/02/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Dingkun Pill (DKP) is a traditional Chinese medicine that has been shown to have beneficial effects on reproductive function. However, the specific mechanism underlying its effect on POI is not well understood. AIM OF THE STUDY To investigate the effect of different doses of Dingkun Pill on ovarian function in cyclophosphamide (CTX)-induced premature ovarian insufficiency (POI) mice and to explore its molecular mechanism through PTEN/PI3K/AKT/FOXO3a signaling pathway. This study will provide valuable insights into the potential clinical application of Dingkun Pill for the treatment of POI. MATERIALS AND METHODS Fifty female ICR mice were randomly divided into normal control (NC) group, model control (MC) group, and Dingkun Pill low, medium, high dose (DKP-L, M, H) groups. Mice were injected with CTX to construct the POI model. Mice in the DKP-L, M, and H groups were given 0.9 g/kg, 1.8 g/kg, and 3.6 g/kg of Dingkun Pill suspension for 21 days, respectively. Mice in the NC and MC groups were given the same amount of normal saline by gavage. Changes in body weight, estrous cycle and gonadal index were observed in each group of mice. Serum levels of FSH, LH, E2 and AMH were detected by ELISA. Hematoxylin-eosin (HE) staining observed the changes of ovarian pathological morphology and follicle counts at all levels. qRT-PCR was used to measure the levels of the PTEN and FOXO3a genes in ovarian tissue. The expression of PTEN/PI3K/AKT/FOXO3a signaling pathway related proteins were detected by Western-blot and immunohistochemistry(IHC). RESULTS In POI mice, Dingkun Pill increased body weight, promoted the recovery of estrous cycle, increased ovarian index, and improved pathological morphology of the ovaries. The FSH level decreased in the medium dose group (P < 0.05), the LH level reduced significantly in the medium and high dose groups (P < 0.01), and the E2 level in the high dose group increased (P < 0.05). There was no significant difference in AMH levels across all dose groups. The number of growing follicles improved at all levels in the low and medium dose groups, but declined significantly in the high dose group. However, the number of corpus luteum increased significantly in the high dose group (P < 0.001), and the atretic follicles in the three dose groups decreased. Results from qRT-PCR, Western-blot and IHC showed that the moderate dose of Dingkun Pill suppressed the levels of the p-PI3K and p-AKT proteins by upregulating the expression of PTEN in the ovarian tissues of POI mice, thereby inhibiting the expression of the key protein p-FOXO3a. However, the inhibitory effect of the higher dose may be less than that of the lower and intermediate dose groups. CONCLUSIONS The Dingkun Pill modulated hormonal levels, promoted follicle growth and induced ovulation in mice with CTX-induced POI, with better results in the low and moderate dose groups. Its mechanism may be related to the regulation of the PTEN/PI3K/AKT/FOXO3a signaling pathway.
Collapse
Affiliation(s)
- Chan Su
- Department of Gynecology, Provincial Hospital, Affiliated to Shandong First Medical University, Jinan, 250098, China; The Second Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, 250013, China
| | - Ruihong Zhang
- Key Laboratory of Experimental Teratology, Ministry of Education, Department of Histology and Embryology, School of Medicine, Shandong University, Jinan, 250012, China
| | - Xiujuan Zhang
- Department of Gynecology, Provincial Hospital, Affiliated to Shandong First Medical University, Jinan, 250098, China
| | - Mengxiao Lv
- Department of Gynecology, Provincial Hospital, Affiliated to Shandong First Medical University, Jinan, 250098, China
| | - Xiang Liu
- The Second Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, 250013, China
| | - Kai Ao
- The Second Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, 250013, China
| | - Jing Hao
- Key Laboratory of Experimental Teratology, Ministry of Education, Department of Histology and Embryology, School of Medicine, Shandong University, Jinan, 250012, China.
| | - Yu-Lan Mu
- Department of Gynecology, Provincial Hospital, Affiliated to Shandong First Medical University, Jinan, 250098, China.
| |
Collapse
|
31
|
Himpe J, Lammerant S, Van den Bergh L, Lapeire L, De Roo C. The Impact of Systemic Oncological Treatments on the Fertility of Adolescents and Young Adults-A Systematic Review. Life (Basel) 2023; 13:life13051209. [PMID: 37240854 DOI: 10.3390/life13051209] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/11/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND Over the past decades, advancements in oncological treatments have led to major improvements in survival. Particularly for adolescents and young adults (AYAs), fertility is an important concern in cancer survivorship. The purpose of the review is to provide physicians with a practical overview of the current knowledge about the impact of systemic oncological treatments on the fertility of female and male AYAs. METHODS A systematic review was performed based on relevant articles obtained from 4 databases up until 31 December 2022. RESULTS The mechanisms of gonadotoxicity and the concurrent risk is described for the following categories: chemotherapy, targeted therapy and immunotherapy. For the category "chemotherapy", the specific effects and risks are listed for the different classes and individual chemotherapeutics. In the category "targeted therapy", a distinction was made between tyrosine kinase inhibitors (TKIs) and monoclonal antibodies. Information concerning immunotherapy is scarce. CONCLUSIONS The effects of chemotherapy on fertility are well investigated, but even in this category, results can be conflicting. Insufficient data are available on the fertility effects of targeted therapy and immunotherapy to draw definitive conclusions. More research is needed for these therapies and their evolving role in treating cancers in AYAs. It would be useful to include fertility endpoints in clinical trials that evaluate new and existing oncological treatments.
Collapse
Affiliation(s)
- Justine Himpe
- Department of Medical Oncology, Ghent University Hospital, 9000 Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Sander Lammerant
- Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Lore Van den Bergh
- Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Lore Lapeire
- Department of Medical Oncology, Ghent University Hospital, 9000 Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
- AYA Research Centre and Hub (ARCH), Ghent University, 9000 Ghent, Belgium
| | - Chloë De Roo
- AYA Research Centre and Hub (ARCH), Ghent University, 9000 Ghent, Belgium
- Department of Reproductive Medicine, Ghent University Hospital, 9000 Ghent, Belgium
| |
Collapse
|
32
|
Han E, Seifer DB. Oocyte Cryopreservation for Medical and Planned Indications: A Practical Guide and Overview. J Clin Med 2023; 12:jcm12103542. [PMID: 37240648 DOI: 10.3390/jcm12103542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Oocyte cryopreservation (OC) is the process in which ovarian follicles are stimulated, the follicular fluid is retrieved, and mature oocytes are isolated and vitrified. Since the first successful pregnancy utilizing previously cryopreserved oocytes in 1986, OC has become increasingly utilized as an option for future biologic children in patients facing gonadotoxic therapies, such as for the treatment of cancer. Planned OC, also termed elective OC, is growing in popularity as a means to circumvent age-related fertility decline. In this narrative review, we describe both medically indicated and planned OC, focusing on the physiology of ovarian follicular loss, OC technique and risks, timing of when OC should be performed, associated financial considerations, and outcomes.
Collapse
Affiliation(s)
- Eric Han
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510, USA
| | - David B Seifer
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
33
|
Chen L, Dong Z, Chen X. Fertility preservation in pediatric healthcare: a review. Front Endocrinol (Lausanne) 2023; 14:1147898. [PMID: 37206440 PMCID: PMC10189781 DOI: 10.3389/fendo.2023.1147898] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 04/12/2023] [Indexed: 05/21/2023] Open
Abstract
Survival rates for children and adolescents diagnosed with malignancy have been steadily increasing due to advances in oncology treatments. These treatments can have a toxic effect on the gonads. Currently, oocyte and sperm cryopreservation are recognized as well-established and successful strategies for fertility preservation for pubertal patients, while the use of gonadotropin-releasing hormone agonists for ovarian protection is controversial. For prepubertal girls, ovarian tissue cryopreservation is the sole option. However, the endocrinological and reproductive outcomes after ovarian tissue transplantation are highly heterogeneous. On the other hand, immature testicular tissue cryopreservation remains the only alternative for prepubertal boys, yet it is still experimental. Although there are several published guidelines for navigating fertility preservation for pediatric and adolescent patients as well as transgender populations, it is still restricted in clinical practice. This review aims to discuss the indications and clinical outcomes of fertility preservation. We also discuss the probably effective and efficient workflow to facilitate fertility preservation.
Collapse
Affiliation(s)
- Lin Chen
- Reproductive Medical Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zirui Dong
- Department of Obstetrics and Gynecology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Xiaoyan Chen
- Maternal-Fetal Medicine Institute, Shenzhen Baoan Women’s and Children’s Hospital, Shenzhen University, Shenzhen, China
- The Fertility Preservation Research Center, Department of Obstetrics and Gynecology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
34
|
Fraire-Zamora JJ, Sharma K, Ammar OF, Massarotti C, Ali ZE, Telfer EE, Williams S, Ata B, Liperis G. Mind the gap: deciphering the role of anti-Müllerian hormone in follicular development-from animal studies toward clinical application. Hum Reprod 2023:7131353. [PMID: 37075310 DOI: 10.1093/humrep/dead075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Indexed: 04/21/2023] Open
Affiliation(s)
| | - Kashish Sharma
- HealthPlus Fertility and Women's Health Centre, Abu Dhabi, UAE
| | - Omar F Ammar
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, UK
| | - Claudia Massarotti
- Academic Unit of Obstetrics and Gynecology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- DINOGMI Department, University of Genova, Genova, Italy
| | - Zoya E Ali
- Research & Development Department, Hertility Health Limited, London, UK
| | - Evelyn E Telfer
- University of Edinburgh Institute of Cell Biology, Edinburgh, UK
| | - Suzannah Williams
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, UK
| | - Baris Ata
- Obstetrics and Gynecology Department, Koc University, Istanbul, Turkey
- ART Fertility Clinics, Dubai, United Arab Emirates
| | | |
Collapse
|
35
|
Ruan X, Cheng J, Du J, Jin F, Gu M, Ju R, Wu Y, Li L, Wang Y, Jiang L, Yang Y, Li Y, Wang Z, Ma J, Zhang M, Mueck AO. Ovarian tissue cryopreservation in the pediatric with rare diseases- experience from China's first and the largest ovarian tissue cryobank. Front Endocrinol (Lausanne) 2023; 14:1137940. [PMID: 37077363 PMCID: PMC10106563 DOI: 10.3389/fendo.2023.1137940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 03/22/2023] [Indexed: 04/05/2023] Open
Abstract
Background There is limited information about the efficacy of ovarian tissue cryopreservation (OTC) in children. In the present study, we report eight patients with rare diseases who underwent OTC in China's first and largest ovarian tissue cryobank. Procedure Data from girls with rare diseases who underwent OTC between September 2020 and November 2022 were retrospectively analyzed. We also compared the number of cryopreserved cortex pieces, follicle number, and AMH in those with rare diseases and age-matched children with non-rare diseases who also underwent OTC in our cryobank. Results The median age of the children was 5.88 ± 3.52 (range 2-13) years old. Unilateral oophorectomy was undertaken via laparoscopy in all of the children. The diseases in the 8 patients were: 4 mucopolysaccharidoses (MPS I two cases, IVA two cases), 1 Diamond-Blackfan anemia (DBA), 1 Fanconi anemia (FA), 1 hyperimmunoglobulin E syndrome (HIES), 1 Niemann-Pick disease. The number of cryopreserved cortex pieces was 17.13 ± 6.36, and the follicle count per 2 mm biopsy was 447.38 ± 524.35. No significant difference in age, the count of cryopreserved cortex pieces, follicle number per 2 mm biopsy, and AMH level was seen between the 20 children with non-rare diseases and those with rare diseases. Conclusions The reports help practitioners counsel girls with rare diseases about fertility preservation. The demand for OTC in pediatrics will likely grow as a standard of care.
Collapse
Affiliation(s)
- Xiangyan Ruan
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
- Department for Women's Health, University Women’s Hospital and Research Center for Women’s Health, University of Tuebingen, Tuebingen, Germany
| | - Jiaojiao Cheng
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Juan Du
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Fengyu Jin
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Muqing Gu
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Rui Ju
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yurui Wu
- Department of Thoracic Surgery and Surgical Oncology, Children’s Hospital, Capital Institute of Pediatrics, Beijing, China
| | - Long Li
- Department of Pediatric Surgery, Children’s Hospital, Capital Institute of Pediatrics, Beijing, China
| | - Yuejiao Wang
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Lingling Jiang
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yu Yang
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yanqiu Li
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Zecheng Wang
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Jun Ma
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Mingzhen Zhang
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Alfred O. Mueck
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
- Department for Women's Health, University Women’s Hospital and Research Center for Women’s Health, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
36
|
Razeti MG, Soldato D, Arecco L, Levaggi A, Puglisi S, Solinas C, Agostinetto E, Spinaci S, Lapuchesky L, Genova C, Massarotti C, Lambertini M. Approaches to Fertility Preservation for Young Women With Breast Cancer. Clin Breast Cancer 2023; 23:241-248. [PMID: 36710145 DOI: 10.1016/j.clbc.2023.01.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 12/23/2022] [Accepted: 01/10/2023] [Indexed: 01/15/2023]
Abstract
In patients with early breast cancer, the combination of different systemic treatment strategies, including chemotherapy, endocrine therapy, targeted therapy, and more recently also immunotherapy has demonstrated to significantly improve their survival outcomes. However, this gain is often obtained at the cost of higher toxicity calling for the need of increased attention toward survivorship-related issues, including fertility preservation in young women. According to available guidelines, health care providers should offer oncofertility counseling to all patients with cancer diagnosed at reproductive age. Counselling should focus on the risk of gonadotoxicity of anticancer treatments and on the access to fertility preservation techniques. However, several surveys have demonstrated suboptimal implementation of these recommendations. This review aims at summarizing the available evidence on oncofertility to guide health care providers involved in the management of young women with breast cancer. Available and effective options for fertility preservation include oocyte/embryo cryopreservation or ovarian tissue cryopreservation. Patient, disease, and treatment characteristics should be carefully considered when offering these strategies. Ovarian function preservation with gonadotrophin-releasing hormone agonists during chemotherapy should be discussed and offered to every premenopausal woman concerned about developing premature ovarian insufficiency and independently of her wish to preserve fertility. Current available data confirm that pregnancy occurring after proper treatment for breast cancer is safe, both in terms of long-term clinical outcomes and for the babies. Fertility preservation and pregnancy desire should be pivotal components of the multimodal management of breast cancer in young women, and require a multidisciplinary approach based on close collaborations between oncologists and fertility specialists.
Collapse
Affiliation(s)
- Maria G Razeti
- Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - Davide Soldato
- Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - Luca Arecco
- Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - Alessia Levaggi
- Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Silvia Puglisi
- Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - Cinzia Solinas
- Medical Oncology, AOU Cagliari, Policlinico Duilio Casula, Monserrato, Cagliari, Italy
| | - Elisa Agostinetto
- Academic Trials Promoting Team, Institut Jules Bordet, Université Libre de Bruxelles (U.L.B.), Brussels, Belgium
| | | | - Laura Lapuchesky
- Medical Oncology, Instituto Alexander Fleming, Ciudad Autónoma De Buenos Aires, Argentina
| | - Carlo Genova
- Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - Claudia Massarotti
- Physiopatology of Human Reproduction Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics and Maternal-Child Health (DiNOGMI Department), School of Medicine, University of Genova, Genova, Italy
| | - Matteo Lambertini
- Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy.
| |
Collapse
|
37
|
Drechsel KCE, Pilon MCF, Stoutjesdijk F, Meivis S, Schoonmade LJ, Wallace WHB, van Dulmen-den Broeder E, Beishuizen A, Kaspers GJL, Broer SL, Veening MA. Reproductive ability in survivors of childhood, adolescent, and young adult Hodgkin lymphoma: a review. Hum Reprod Update 2023:7034966. [PMID: 36779325 DOI: 10.1093/humupd/dmad002] [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: 07/14/2022] [Revised: 12/21/2022] [Indexed: 02/14/2023] Open
Abstract
BACKGROUND Owing to a growing number of young and adolescent Hodgkin lymphoma (HL) survivors, awareness of (long-term) adverse effects of anticancer treatment increases. The risk of impaired reproductive ability is of great concern given its impact on quality of life. There is currently no review available on fertility after childhood HL treatment. OBJECTIVE AND RATIONALE The aim of this narrative review was to summarize existing literature on different aspects of reproductive function in male and female childhood, adolescent, and young adult HL survivors. SEARCH METHODS PubMed and EMBASE were searched for articles evaluating fertility in both male and female HL survivors aged <25 years at diagnosis. In females, anti-Müllerian hormone (AMH), antral follicle count, premature ovarian insufficiency (POI), acute ovarian failure, menstrual cycle, FSH, and pregnancy/live births were evaluated. In males, semen-analysis, serum FSH, inhibin B, LH, testosterone, and reports on pregnancy/live births were included. There was profound heterogeneity among studies and a lack of control groups; therefore, no meta-analyses could be performed. Results were presented descriptively and the quality of studies was not assessed individually. OUTCOMES After screening, 75 articles reporting on reproductive markers in childhood or adolescent HL survivors were included. Forty-one papers reported on 5057 female HL survivors. The incidence of POI was 6-34% (median 9%; seven studies). Signs of diminished ovarian reserve or impaired ovarian function were frequently seen (low AMH 55-59%; median 57%; two studies. elevated FSH 17-100%; median 53%; seven studies). Most survivors had regular menstrual cycles. Fifty-one studies assessed fertility in 1903 male HL survivors. Post-treatment azoospermia was highly prevalent (33-100%; median 75%; 29 studies). Long-term follow-up data were limited, but reports on recovery of semen up to 12 years post-treatment exist. FSH levels were often elevated with low inhibin B (elevated FSH 0-100%; median 51.5%; 26 studies. low inhibin B 19-50%; median 45%; three studies). LH and testosterone levels were less evidently affected (elevated LH 0-57%, median 17%; 21 studies and low testosterone 0-43%; median 6%; 15 studies). In both sexes, impaired reproductive ability was associated with a higher dose of cumulative chemotherapeutic agents and pelvic radiotherapy. The presence of abnormal markers before treatment indicated that the disease itself may also negatively affect reproductive function (Females: AMH<p10 9%; one study and Males: azoospermia 0-50%; median 10%; six studies). Reports on chance to achieve pregnancy during survivorship are reassuring, although studies had their limitations and the results are difficult to evaluate. In the end, a diminished ovarian reserve does not exclude the chance of a live birth, and males with aberrant markers may still be able to conceive. WIDER IMPLICATIONS This review substantiates the negative effect of HL treatment on gonadal function and therefore young HL survivors should be counseled regarding their future reproductive life, and fertility preservation should be considered. The current level of evidence is insufficient and additional trials on the effects of HL and (current) treatment regimens on reproductive function are needed. In this review, we make a recommendation on reproductive markers that could be assessed and the timing of (repeated) measurements.
Collapse
Affiliation(s)
- Katja C E Drechsel
- Pediatric Oncology, Cancer Center Amsterdam, Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.,Cancer Center Amsterdam, Amsterdam UMC, Location VUmc, VU Amsterdam, Amsterdam, The Netherlands
| | - Maxime C F Pilon
- Pediatric Oncology, Cancer Center Amsterdam, Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Francis Stoutjesdijk
- Pediatric Oncology, Cancer Center Amsterdam, Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Salena Meivis
- Pediatric Oncology, Cancer Center Amsterdam, Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Linda J Schoonmade
- Medical Library, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | | - Eline van Dulmen-den Broeder
- Pediatric Oncology, Cancer Center Amsterdam, Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Auke Beishuizen
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.,Department of Haematology/Oncology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Gertjan J L Kaspers
- Pediatric Oncology, Cancer Center Amsterdam, Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Simone L Broer
- Department of Reproductive Medicine & Gynecology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Margreet A Veening
- Pediatric Oncology, Cancer Center Amsterdam, Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| |
Collapse
|
38
|
Lambertini M, Ceppi M, Anderson RA, Cameron DA, Bruzzone M, Franzoi MA, Massarotti C, El-Abed S, Wang Y, Lecocq C, Nuciforo P, Rolyance R, Pusztai L, Sohn J, Latocca MM, Arecco L, Pistilli B, Ruddy KJ, Ballestrero A, Del Mastro L, Peccatori FA, Partridge AH, Saura C, Untch M, Piccart M, Di Cosimo S, de Azambuja E, Demeestere I. Impact of Anti-HER2 Therapy Alone and With Weekly Paclitaxel on the Ovarian Reserve of Young Women With HER2-Positive Breast Cancer. J Natl Compr Canc Netw 2023; 21:33-41.e16. [PMID: 36634607 DOI: 10.6004/jnccn.2022.7065] [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: 05/29/2022] [Accepted: 08/09/2022] [Indexed: 01/13/2023]
Abstract
BACKGROUND The potential gonadotoxicity of anti-HER2 agents remains largely unknown, and limited, conflicting evidence exists for taxanes. Antimüllerian hormone (AMH) is an established biomarker of ovarian reserve that may aid in quantifying anticancer treatment-induced gonadotoxicity. PATIENTS AND METHODS The present biomarker analysis of the randomized phase III neoadjuvant NeoALTTO trial included premenopausal women aged ≤45 years at diagnosis of HER2-positive early breast cancer with available frozen serum samples at baseline (ie, before anticancer treatments), at week 2 (ie, the "biological window" of anti-HER2 therapy alone), and/or at the time of surgery (ie, after completing paclitaxel + anti-HER2 therapy, before starting adjuvant chemotherapy). RESULTS The present analysis included 130 patients with a median age of 38 years (interquartile ratio [IQR], age 33-42 years). AMH values at the 3 time points differed significantly (P<.001). At baseline, median AMH levels were 1.29 ng/mL (IQR, 0.56-2.62 ng/mL). At week 2, a small but significant reduction in AMH levels was observed (median, 1.10 ng/mL; IQR, 0.45-2.09 ng/mL; P<.001). At surgery, a larger significant decline in AMH levels was observed (median, 0.01 ng/mL; IQR, 0.01-0.03 ng/mL; P<.001). Although the type of anti-HER2 treatment (trastuzumab and/or lapatinib) did not seem to impact the results, age and pretreatment ovarian reserve had a major influence on treatment-induced gonadotoxicity risk. CONCLUSIONS This NeoALTTO biomarker analysis showed that anti-HER2 therapies alone had limited gonadotoxicity but that the addition of weekly paclitaxel resulted in marked AMH decline with possible negative implications for subsequent ovarian function and fertility.
Collapse
Affiliation(s)
- Matteo Lambertini
- School of Medicine, University of Genova, Genova, Italy.,UOC Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | | | - Richard A Anderson
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - David A Cameron
- Institute of Genomics and Cancer, The University of Edinburgh, Edinburgh, United Kingdom
| | | | | | - Claudia Massarotti
- IRCCS Ospedale Policlinico San Martino, Genova, Italy.,University of Genova, Genova, Italy
| | | | | | | | - Paolo Nuciforo
- Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, SOLTI BC Cooperative Group, Barcelona, Spain
| | | | | | - Joohyuk Sohn
- Yonsei Cancer Center, Yonsei University Health System, Seoul, Korea
| | - Maria Maddalena Latocca
- School of Medicine, University of Genova, Genova, Italy.,UOC Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Luca Arecco
- School of Medicine, University of Genova, Genova, Italy.,UOC Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | | | | | - Alberto Ballestrero
- School of Medicine, University of Genova, Genova, Italy.,IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Lucia Del Mastro
- School of Medicine, University of Genova, Genova, Italy.,UOC Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | | | - Ann H Partridge
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Cristina Saura
- Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, SOLTI BC Cooperative Group, Barcelona, Spain
| | | | - Martine Piccart
- Institut Jules Bordet, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | | - Evandro de Azambuja
- Institut Jules Bordet, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Isabelle Demeestere
- Fertility Clinic, CUB-Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
39
|
Osuka S, Kasahara Y, Iyoshi S, Sonehara R, Myake N, Muraoka A, Nakamura T, Iwase A, Kajiyama H. Follicle development and its prediction in patients with primary ovarian insufficiency: Possible treatments and markers to maximize the ability to conceive with residual follicles. Reprod Med Biol 2023; 22:e12556. [PMID: 38144239 PMCID: PMC10746865 DOI: 10.1002/rmb2.12556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 11/16/2023] [Accepted: 12/11/2023] [Indexed: 12/26/2023] Open
Abstract
Background Primary ovarian insufficiency (POI) is characterized by the development of hypergonadotropic hypogonadism before 40 years of age and leads to intractable infertility. Although in vitro fertilization and embryo transfer with donated eggs enables pregnancy, not a few patients desire pregnancy using their oocytes. However, follicular development is rare and unpredictable in patients with POI. Thus, there is a need for treatments that promote the development of residual follicles and methods to accurately predict infrequent ovulation. Methods This review discusses the effects of various treatments for obtaining eggs from POI patients. Furthermore, this study focused a potential marker for predicting follicular growth in patients with POI. Main Findings Different treatments such as hormone-replacement therapy, dehydroepiandrosterone supplementation, platelet-rich plasma injection, and in vitro activation have shown varying degrees of effectiveness in retrieving oocytes from patients with POI. To predict follicle development in the cycle, elevated serum estradiol and reduced follicle-stimulating hormone (FSH) levels are important. However, these markers are not always reliable under continuous estradiol-replacement therapy. As a novel marker for predicting follicle growth, serum anti-Müllerian hormone (AMH) levels, measured using the picoAMH enzyme-linked immunosorbent assay, were found to predict follicle growth in patients and the cycle. Conclusion This review highlights the challenges and available interventions for achieving pregnancy using a patient's oocytes in cases of POI. We believe that a combination of currently available treatments and prediction methods is the best strategy to enable patients with POI to conceive using their own eggs. Although AMH levels may predict follicle growth, further research is necessary to improve the chances of successful follicular development and conception in patients with POI.
Collapse
Affiliation(s)
- Satoko Osuka
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
| | | | - Shohei Iyoshi
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
- Institute for Advanced ResearchNagoya UniversityNagoyaJapan
| | - Reina Sonehara
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Natsuki Myake
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Ayako Muraoka
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
- Division of Perinatology, Center for Maternal‐Neonatal CareNagoya University HospitalNagoyaJapan
| | - Tomoko Nakamura
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
- Division of Perinatology, Center for Maternal‐Neonatal CareNagoya University HospitalNagoyaJapan
| | - Akira Iwase
- Department of Obstetrics and GynecologyGunma University Graduate School of MedicineMaebashiJapan
| | - Hiroaki Kajiyama
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
| |
Collapse
|
40
|
Wang X, Wang S, Yao S, Shi W, Ma K. The clinical characteristics and treatment of ovarian malignant mesoderm mixed tumor: a systematic review. J Ovarian Res 2022; 15:104. [PMID: 36114551 PMCID: PMC9482291 DOI: 10.1186/s13048-022-01037-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/30/2022] [Indexed: 12/02/2022] Open
Abstract
Background Ovarian malignant mesoderm mixed tumor (OMMMT) is a rare clinical entity. To provide reference for the treatment and prognosis of OMMMT, we analyzed the clinical features, pathology and molecular biology characteristic of published cases. Methods The English and Chinese reported cases of OMMMT were selected from PubMed, Clinical Trials.gov and CNKI database from 2000 to December 15th, 2021 following the PRISMA guidelines. Results A total of 63 literatures including 199 OMMMT cases were included. The average age of patients at diagnosis was 56.46 years, the highest incidence age was 60-65 years, and 82% of them were menopausal women. Most patients were diagnosed in FIGO III stage (59.64%). The most common symptom of OMMMT was abdominal pain (60.5%). 61.6% of patients were accompanied by ascites, while ascites was not associated with metastatic tumor and local recurrence. The CA125 of 88.68% patients increased. The most common reported carcinomatous component and sarcomatous component were serous adenocarcinoma (44.96%) and chondrosarcoma (24.81%), respectively. Initial treatment included surgery (94.97%) and taxanes-based (55.10%) or platinum-based (85.71%) chemotherapy regimens. The median survival time of patients was 20 months. Heterologous sarcoma component did not shorten life expectancy. The optimal ovarian tumor cell debulking surgery (OOTCDS), radiotherapy and chemotherapy could significantly prolong the median survival time of patients. Furthermore, platinum drugs could significantly prolong the survival time after comparing various chemotherapy schemes. Besides, the combination of platinum and taxanes was therapeutically superior to the combination of platinum and biological alkylating agents. Conclusion The OOTCDS and platinum-based chemotherapy regimen can improve the prognosis of OMMMT. Targeted therapy might become a new research direction in the future. Since the elderly patients are the majority, the toxicity of new drugs on the elderly patients is more noteworthy. Supplementary Information The online version contains supplementary material available at 10.1186/s13048-022-01037-6.
Collapse
|
41
|
Lambertini M, Marrocco C, Spinaci S, Demeestere I, Anderson RA. Risk of gonadotoxicity with immunotherapy and targeted agents remains an unsolved but crucial issue. Eur J Clin Invest 2022; 52:e13779. [PMID: 35342940 DOI: 10.1111/eci.13779] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/15/2022] [Accepted: 03/26/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Matteo Lambertini
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Camilla Marrocco
- Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Stefano Spinaci
- Division of Breast Surgery, Ospedale Villa Scassi, Genova, Italy
| | - Isabelle Demeestere
- Fertility clinic, CUB-Erasme Hospital, Université Libre de Bruxelles (U.L.B.), Brussels, Belgium
| | - Richard A Anderson
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
42
|
Anderson RA, Cui W. Improving analysis of ovarian function and female fertility in cancer survivors. Fertil Steril 2022; 117:1057-1058. [PMID: 35512967 DOI: 10.1016/j.fertnstert.2022.02.031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 02/25/2022] [Indexed: 01/10/2023]
Affiliation(s)
- Richard A Anderson
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Wanyuan Cui
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
43
|
Kuang X, Tang Y, Xu H, Ji M, Lai D. The Evaluation of Ovarian Function Recovery Following Treatment of Primary Ovarian Insufficiency: A Systematic Review. Front Endocrinol (Lausanne) 2022; 13:855992. [PMID: 35573993 PMCID: PMC9095968 DOI: 10.3389/fendo.2022.855992] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 03/28/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Primary ovarian insufficiency (POI) is gaining awareness as its prevalence increases and its effect on patients is extremely negative. To date, several therapies have been designed to treat POI, but the conclusions are conflicting, in part, due to inconsistent evaluation methods. Thus, we explore a multi-index of ovarian function assessment methods to evaluate the recovery of ovarian function after various therapies in order to evaluate effectiveness in a more comprehensive manner. AIM The purpose of this review is to assess the effectiveness of various therapies to recover ovarian function in patients with POI. The primary outcome measures were anti-Müllerian hormone (AMH) levels, follicle stimulating hormone (FSH) levels, and antral follicle count (AFC). The secondary outcomes included the change of mean ovarian volume, menstruation recovery, and pregnancy rate. METHODS Our systematic searching including PubMed, Web of Science, Cochrane, and Embase databases was conducted to find all human clinical trial articles published from January 2000 to April 2021 and related to POI treatment, including the keywords: POI, AFC, and hormones. All prospective and retrospective studies exploring ovarian function recovery that include AFC, AMH levels, and FSH levels evolution throughout treatment were included. All patients included in the studies met the POI criteria described by the European Society for Human Reproductive Embryology (ESHRE) guideline. RESULTS Six studies were selected based on the criteria: one randomized controlled trial and five observational studies. Among them, two studies focused on the intraovarian platelet-rich plasma (PRP) infusion treatment, two studies focused on dehydroepiandrosterone (DHEA) supplements, one study focused on hormone replacement therapy (HRT), and one study focused on autologous adipose-derived stromal cells (ADSCs) treatment. There was insufficient scientific evidence that any approach could help ovarian function recovery in patients with POI because the ovarian function markers in each study had inconsistent changes with 26 patients (6.2%) reporting spontaneous pregnancy. CONCLUSION Serum AMH levels, FSH levels, and AFC are sensitive indicators and reflect the evolution of ovarian function. Large randomized controlled trials are necessary, and the data on ovarian function should be collected comprehensively to evaluate the effectiveness of a variety of treatments.
Collapse
Affiliation(s)
- Xiaojun Kuang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- School of Biomedical Engineering, Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Yongzhe Tang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Hong Xu
- School of Biomedical Engineering, Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Min Ji
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- *Correspondence: Min Ji, ; Dongmei Lai,
| | - Dongmei Lai
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- *Correspondence: Min Ji, ; Dongmei Lai,
| |
Collapse
|