1
|
Raveendran C, Meloot SS, Yadev IP. Clinical and biochemical amenorrhea in premenopausal patients with breast cancer treated with chemotherapy - a prospective cohort study. BMC Cancer 2025; 25:753. [PMID: 40264070 PMCID: PMC12012977 DOI: 10.1186/s12885-025-14159-z] [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: 11/04/2024] [Accepted: 04/15/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Patients with breast cancer receiving chemotherapy frequently experience amenorrhea due to the cytotoxic effects of drugs, resulting in ovarian suppression and impacting reproductive health. This study aimed to prospectively investigate the incidence of clinical amenorrhea defined as the cessation of menstrual bleeding for at least three consecutive months and and biochemical amenorrhea, characterized by serum estradiol levels < 20 pg/mL and FSH levels ≥ 40 mIU/mL, using clinical and biochemical methods to determine their relation and identify the predictive factors for early amenorrhea. METHODS This longitudinal study was conducted at a tertiary care hospital with 76 premenopausal patients with breast cancer who received chemotherapy. The patients were followed up for two years with monthly clinical assessments of amenorrhea status for a year and biannual hormonal assessments of serum estradiol and follicle-stimulating hormone (FSH) levels. The incidence of clinical and biochemical amenorrhea was estimated and explored to determine any association between the two factors and the impact of risk factors. RESULTS The rates of clinical and biochemical amenorrhea were 84.2% and 78.9%, respectively. The median time for clinical amenorrhea was 8 ( 95% CI, 7.83-8.17) months and 18 (95% CI, 17.90-18.10) months for biochemical amenorrhea. A significant association was observed between clinical and biochemical amenorrhea (P =.0022). The estradiol and FSH levels were initially in the premenopausal range and reached postmenopausal values by the end of the study period. Age, BMI, chemotherapy regimen, hormonal treatment, and biochemical amenorrhea were not predictive of the time to clinical amenorrhea. CONCLUSIONS The occurrence of chemotherapy-related amenorrhea in premenopausal patients with breast cancer was high. Clinical amenorrhea is a reliable early indicator of biochemical amenorrhea. Regular menstrual and hormonal assessments are essential in patients with breast cancer undergoing chemotherapy. Future studies with larger cohorts are required to explore predictive factors associated with chemotherapy-related amenorrhea.
Collapse
Affiliation(s)
- Ciniraj Raveendran
- Department of Radiation Oncology, Government Medical College Thiruvananthapuram, Thiruvananthapuram, PIN-695011, Kerala State, India.
| | - Suma Susan Meloot
- Department of Radiation Oncology, Government Medical College Thiruvananthapuram, Thiruvananthapuram, PIN-695011, Kerala State, India
| | - I P Yadev
- Department of General Surgery, Government Medical College Kollam, Kollam City, PIN-691574, Kerala State, India
| |
Collapse
|
2
|
Lee MJ, Jung JJ, Cheun JH, Kang E, Kim HK, Lee HB, Moon HG, Han W. Comparison of oncological outcomes of premenopausal with ovarian function suppression versus postmenopausal women in ER+/HER2- breast cancer. Breast 2025; 81:104449. [PMID: 40120519 PMCID: PMC11976225 DOI: 10.1016/j.breast.2025.104449] [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: 02/05/2025] [Revised: 03/15/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND The Rx for positive node endocrine-responsive breast cancer trial highlighted that premenopausal (PRE) women who underwent chemotherapy exhibited superior survival rates compared to postmenopausal (POST) counterparts, but showed worse survival without chemotherapy. This raises the question whether application of ovarian function suppression (OFS) in PRE women aligns with their cancer biology, treatment response, and outcomes observed in POST women. METHODS Data from the Seoul National University Hospital breast cancer cohort focusing on patients with stage pT1-3, pN0-1, estrogen receptor-positive (ER+), and HER2-negative breast cancer were analyzed. Survival outcomes, including invasive disease-free survival (iDFS) and distant relapse-free survival (DRFS), were compared between PRE women receiving OFS and POST women, with chemotherapy usage as a stratification factor. Propensity score matching was performed. RESULT We analyzed 3483 patients, comprising 2901 POST and 582 PRE women with OFS. In the cohort without chemotherapy, the 10-year iDFS rates were 90.3 % and 88.3 % (hazard ratio [HR], 1.32; p = 0.16), and 10-year DRFS rates were 94.3 % and 96.1 % (HR, 0.78; p = 0.41) for POST and PRE women with OFS, respectively. Among women treated with chemotherapy, 10-year iDFS rates were 83.0 % and 79.5 % (HR, 1.21; p = 0.37), and DRFS rates were 86.7 % and 85.7 % (HR, 1.14; p = 0.58) for POST and PRE women with OFS, respectively. These results remained consistent after PSM. CONCLUSION Oncological outcomes of PRE women receiving OFS were comparable to those of POST women with ER+ and HER2-early breast cancer, irrespective of chemotherapy administration.
Collapse
Affiliation(s)
- Min Jung Lee
- Department of surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ji-Jung Jung
- Department of surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jong-Ho Cheun
- Department of Surgery, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Eunhye Kang
- Department of surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hong-Kyu Kim
- Department of surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea; Cancer Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Han-Byoel Lee
- Department of surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea; Cancer Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hyeong-Gon Moon
- Department of surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea; Cancer Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Wonshik Han
- Department of surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea; Cancer Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.
| |
Collapse
|
3
|
Bornes L, van Winden LJ, Geurts VCM, de Bruijn B, Azarang L, Lanfermeijer M, Caruso M, Proost N, Boeije M, Lohuis JO, Belthier G, Noguera Delgado E, de Gruil N, Kroep JR, van de Ven M, Menezes R, Wesseling J, Kok M, Linn S, Broeks A, van Rossum HH, Scheele CLGJ, van Rheenen J. The oestrous cycle stage affects mammary tumour sensitivity to chemotherapy. Nature 2025; 637:195-204. [PMID: 39633046 PMCID: PMC11666466 DOI: 10.1038/s41586-024-08276-1] [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: 08/24/2021] [Accepted: 10/23/2024] [Indexed: 12/07/2024]
Abstract
The response of breast cancer to neoadjuvant chemotherapy (NAC) varies substantially, even when tumours belong to the same molecular or histological subtype1. Here we identify the oestrous cycle as an important contributor to this heterogeneity. In three mouse models of breast cancer, we show reduced responses to NAC when treatment is initiated during the dioestrus stage, when compared with initiation during the oestrus stage. Similar findings were observed in retrospective premenopausal cohorts of human patients. Mechanistically, the dioestrus stage exhibits systemic and localized changes, including (1) an increased number of cells undergoing epithelial-to-mesenchymal transition linked to chemoresistance2-4 and (2) decreased tumour vessel diameter, suggesting potential constraints to drug sensitivity and delivery. In addition, an elevated presence of macrophages, previously associated with chemoresistance induction5, characterizes the dioestrus phase. Whereas NAC disrupts the oestrous cycle, this elevated macrophage prevalence persists and depletion of macrophages mitigates the reduced therapy response observed when initiating treatment during dioestrus. Our data collectively demonstrate the oestrous cycle as a crucial infradian rhythm determining chemosensitivity, warranting future clinical studies to exploit optimal treatment initiation timing for enhanced chemotherapy outcomes.
Collapse
Affiliation(s)
- Laura Bornes
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Lennart J van Winden
- Laboratory of Clinical Chemistry and Hematology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Veerle C M Geurts
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Beaunelle de Bruijn
- VIB Center for Cancer Biology, KU Leuven Department of Oncology, Leuven, Belgium
| | - Leyla Azarang
- Biostatistics Centre & Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Mirthe Lanfermeijer
- Laboratory of Clinical Chemistry and Hematology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Marika Caruso
- VIB Center for Cancer Biology, KU Leuven Department of Oncology, Leuven, Belgium
| | - Natalie Proost
- Mouse Clinic for Cancer and Aging (MCCA) Preclinical Intervention Unit, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Manon Boeije
- Mouse Clinic for Cancer and Aging (MCCA) Preclinical Intervention Unit, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jeroen O Lohuis
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Guillaume Belthier
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Eulàlia Noguera Delgado
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Nadia de Gruil
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Judith R Kroep
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Marieke van de Ven
- Mouse Clinic for Cancer and Aging (MCCA) Preclinical Intervention Unit, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Renee Menezes
- Biostatistics Centre & Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jelle Wesseling
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Pathology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, the Netherlands
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Marleen Kok
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Sabine Linn
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Pathology, Utrecht University Medical Center, Utrecht, the Netherlands
| | - Annegien Broeks
- Core Facility Molecular Pathology & Biobanking, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Huub H van Rossum
- Laboratory of Clinical Chemistry and Hematology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | - Jacco van Rheenen
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
4
|
Tan TW, Tan HL, Chung YC. Effectiveness of resistance training in preventing sarcopenia among breast cancer patients undergoing chemotherapy: A systematic review and meta-analysis. Worldviews Evid Based Nurs 2024; 21:687-694. [PMID: 39572015 DOI: 10.1111/wvn.12756] [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: 08/26/2023] [Revised: 10/19/2024] [Accepted: 10/21/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND Breast cancer patients undergoing chemotherapy experience body composition changes impacting treatment outcomes. The role of resistance training in mitigating chemotherapy-induced sarcopenia in breast cancer patients is not well defined. AIMS This study aims to assess the efficacy of resistance training in preventing sarcopenia among breast cancer patients undergoing chemotherapy. METHODS A systematic search was conducted across PubMed, EMBASE, Medline, the Cochrane Library, and CINAHL until May 5, 2023. Selected literature focused on the effects of resistance training on body fat, muscle mass, muscle strength, and physical performance in breast cancer patients undergoing chemotherapy. Cochrane Risk of Bias tool version 2.0 was employed for quality assessment, and data were analyzed using Comprehensive Meta-Analysis version 2.0. RESULTS Eleven randomized controlled trials (RCTs) showed that resistance training had a significant positive impact on reducing body fat (SMD = -0.250, 95% CI [-0.450, -0.050]), increasing lean body mass (SMD = 0.374, 95% CI [0.178, 0.571]), and enhancing handgrip strength at both the affected site (SMD = 0.326, 95% CI [0.108, 0.543]) and the nonaffected site (SMD = 0.276, 95% CI [0.059, 0.492]). Additionally, significant improvements were observed in leg press strength (SMD = 0.598, 95% CI [0.401, 0.796]) and overall physical performance (SMD = 0.671, 95% CI [0.419, 0.923]). LINKING EVIDENCE TO ACTION Resistance training is a recommended intervention for reducing body fat, increasing muscle mass, muscle strength, and enhancing physical performance in breast cancer patients undergoing chemotherapy. Ideal low-intensity resistance training programs span 8-24 weeks, with 20-to-90-min sessions 2-4 times weekly. Regimens generally entail 8-12 repetitions at 40%-90% of one-repetition maximum test, with free-weight resistance training targeting major muscle groups yielding substantial benefits. Further research should explore outcomes across different chemotherapy phases and investigate long-term resistance training effects for a comprehensive view.
Collapse
Affiliation(s)
- Ting-Wan Tan
- Department of Nursing, Hsinchu Mackay Memorial Hospital, Hsinchu, Taiwan
- Department of Nursing, Yuanpei University of Medical Technology, Hsinchu, Taiwan
| | - Han-Ling Tan
- Department of Orthopaedic Surgery, National Orthopaedic Centre of Excellence in Research and Learning (NOCERAL), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Yu-Chu Chung
- Department of Nursing, Yuanpei University of Medical Technology, Hsinchu, Taiwan
| |
Collapse
|
5
|
Feng Y, Zheng H, Yin C, Liang D, Zhang S, Chen J, Mai F, Lan Z, Zhu M, Mai Z, Shen S, Jayawardana T, Wu R, Tang W, Zhang R, He X, Zheng S, Hu Q, Han Y, Yang Y, Gong S, Wang Z, El-Omar EM, Luo W, Chen X, Chen G, Li P, Chen X. β-resorcylic acid released by Limosilactobacillus reuteri protects against cisplatin-induced ovarian toxicity and infertility. Cell Rep Med 2024; 5:101678. [PMID: 39096912 PMCID: PMC11384965 DOI: 10.1016/j.xcrm.2024.101678] [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: 05/08/2024] [Revised: 06/07/2024] [Accepted: 07/11/2024] [Indexed: 08/05/2024]
Abstract
Chemotherapy-induced premature ovarian insufficiency (CIPOI) triggers gonadotoxicity in women undergoing cancer treatment, leading to loss of ovarian reserves and subfertility, with no effective therapies available. In our study, fecal microbiota transplantation in a cisplatin-induced POI mouse model reveals that a dysbiotic gut microbiome negatively impacts ovarian health in CIPOI. Multi-omics analyses show a significant decrease in Limosilactobacillus reuteri and its catabolite, β-resorcylic acid , in the CIPOI group in comparison to healthy controls. Supplementation with L. reuteri or β-RA mitigates cisplatin-induced hormonal disruptions, morphological damages, and reductions in follicular reserve. Most importantly, β-RA pre-treatment effectively preserves oocyte function, embryonic development, and fetus health, thereby protecting against chemotherapy-induced subfertility. Our results provide evidence that β-RA suppresses the nuclear accumulation of sex-determining region Y-box 7, which in turn reduces Bcl-2-associated X activation and inhibits granulosa cell apoptosis. These findings highlight the therapeutic potential of targeting the gut-ovary axis for fertility preservation in CIPOI.
Collapse
Affiliation(s)
- Yinglin Feng
- Central Laboratory of the Medical Research Center, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province, China; Department of Obstetrics and Gynecology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province, China; Department of Obstetrics, Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan, Guangdong Province, China
| | - Huimin Zheng
- Central Laboratory of the Medical Research Center, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province, China
| | - Chunhua Yin
- Department of Gynecology and Obstetrics, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Dong Liang
- Department of Obstetrics and Gynecology, The First People's Hospital of Foshan, Foshan, Guangdong Province, China
| | - Siyou Zhang
- Department of Obstetrics and Gynecology, The First People's Hospital of Foshan, Foshan, Guangdong Province, China
| | - Jingrui Chen
- Department of Obstetrics and Gynecology, The First People's Hospital of Foshan, Foshan, Guangdong Province, China
| | - Feihong Mai
- Institute of Ecological Science, School of Life Science, South China Normal University, Guangzhou, Guangdong Province, China
| | - Zixin Lan
- The Second Clinical Medical College, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Menglin Zhu
- The Second Clinical Medical College, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zhensheng Mai
- Department of Obstetrics and Gynecology, The First People's Hospital of Foshan, Foshan, Guangdong Province, China
| | - Sj Shen
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campuses, UNSW Sydney, Sydney, NSW, Australia
| | - Thisun Jayawardana
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campuses, UNSW Sydney, Sydney, NSW, Australia
| | - Rong Wu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Wenli Tang
- Central Laboratory of the Medical Research Center, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province, China
| | - Renfang Zhang
- Department of Gynecology and Obstetrics, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Xiaoyun He
- Department of Gynecology and Obstetrics, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Shanshan Zheng
- Health Medical Center, The First People's Hospital of Foshan, Foshan, Guangdong Province, China
| | - Qian Hu
- Institute of Translational Medicine, The First People's Hospital of Foshan, Foshan, Guangdong Province, China
| | - Yubin Han
- Department of Obstetrics and Gynecology, The First People's Hospital of Foshan, Foshan, Guangdong Province, China
| | - Yuanhao Yang
- Mater Research Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia; Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Shenhai Gong
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Zhang Wang
- Institute of Ecological Science, School of Life Science, South China Normal University, Guangzhou, Guangdong Province, China
| | - Emad M El-Omar
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campuses, UNSW Sydney, Sydney, NSW, Australia
| | - Wei Luo
- Institute of Translational Medicine, The First People's Hospital of Foshan, Foshan, Guangdong Province, China
| | - Xueqin Chen
- Central Laboratory of the Medical Research Center, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province, China; Department of Obstetrics and Gynecology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province, China.
| | - Guoqiang Chen
- Institute of Translational Medicine, The First People's Hospital of Foshan, Foshan, Guangdong Province, China.
| | - Pan Li
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campuses, UNSW Sydney, Sydney, NSW, Australia.
| | - Xia Chen
- Central Laboratory of the Medical Research Center, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province, China; Department of Obstetrics and Gynecology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province, China.
| |
Collapse
|
6
|
Raimondo D, Raffone A, Neola D, Genovese F, Travaglino A, Aguzzi A, De Gobbi V, Virgilio A, Di Santo S, Vicenti R, Magnani V, Guida M, Pippucci T, Seracchioli R. Molecular Factors Predicting Ovarian Chemotoxicity in Fertile Women: A Systematic Review. Cancers (Basel) 2024; 16:2793. [PMID: 39199566 PMCID: PMC11352339 DOI: 10.3390/cancers16162793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/28/2024] [Accepted: 08/07/2024] [Indexed: 09/01/2024] Open
Abstract
Background: Recent advances in cancer diagnosis and treatment have significantly improved survival rates among women of reproductive age facing cancer. However, the potential iatrogenic loss of fertility caused by chemotherapeutic agents underscores the need to understand and predict chemotherapy-induced ovarian damage. This study addresses this gap by systematically reviewing the literature to investigate genetic markers associated with chemotherapy-induced ovarian failure (CIOF). Objective: The primary objective is to identify genetic markers linked to CIOF, contributing to a comprehensive understanding of the factors influencing fertility preservation in female cancer survivors. Methods: A systematic review was conducted using PubMed, EMBASE, Web of Science, Scopus, and OVID electronic databases from inception through December 2023. Studies were included if they featured genomic assessments of genes or polymorphisms related to CIOF in women with histologically confirmed tumors. Exclusion criteria comprised in vitro and animal studies, reviews, and pilot studies. The resulting four human-based studies were scrutinized for insights into genetic influences on CIOF. Results: Of the 5179 articles initially identified, four studies met the inclusion criteria, focusing on alkylating agents, particularly cyclophosphamide, and anthracyclines. Su et al. explored CYP3A41B variants, revealing modified associations with CIOF based on age. Charo et al. investigated GSTA1 and CYP2C19 polymorphisms, emphasizing the need to consider age and tamoxifen therapy in assessing associations. Oktay et al. delved into the impact of BRCA mutations on anti-Müllerian hormone (AMH) levels post-chemotherapy, supported by in vitro assays. Van der Perk et al. focused on childhood cancer survivors and revealed significant associations of CYP3A43 and CYP2B6*2 SNPs with AMH levels. Conclusions: This systematic review analyzes evidence regarding genetic markers influencing CIOF, emphasizing the complex interplay of age, specific genetic variants, and chemotherapy regimens. The findings underscore the need for a personalized approach in assessing CIOF risk, integrating genetic markers with traditional ovarian reserve testing. The implications of this study extend to potential advancements in fertility preservation strategies, offering clinicians a comprehensive baseline assessment for tailored interventions based on each patient's unique genetic profile. Further research is essential to validate these findings and establish a robust framework for integrating genetic markers into clinical practice.
Collapse
Affiliation(s)
- Diego Raimondo
- Division of Gynecology and Human Reproduction Physiopathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy (A.A.); (V.D.G.); (S.D.S.); (R.V.); (V.M.); (R.S.)
| | - Antonio Raffone
- Department of Woman, Child, and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy;
| | - Daniele Neola
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (F.G.); (M.G.)
| | - Federica Genovese
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (F.G.); (M.G.)
| | - Antonio Travaglino
- Unit of Pathology, Department of Medicine and Technological Innovation, University of Insubria, 21100 Varese, Italy;
| | - Alberto Aguzzi
- Division of Gynecology and Human Reproduction Physiopathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy (A.A.); (V.D.G.); (S.D.S.); (R.V.); (V.M.); (R.S.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy;
| | - Valeria De Gobbi
- Division of Gynecology and Human Reproduction Physiopathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy (A.A.); (V.D.G.); (S.D.S.); (R.V.); (V.M.); (R.S.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy;
| | - Agnese Virgilio
- Division of Gynecology and Human Reproduction Physiopathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy (A.A.); (V.D.G.); (S.D.S.); (R.V.); (V.M.); (R.S.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy;
| | - Sara Di Santo
- Division of Gynecology and Human Reproduction Physiopathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy (A.A.); (V.D.G.); (S.D.S.); (R.V.); (V.M.); (R.S.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy;
| | - Rossella Vicenti
- Division of Gynecology and Human Reproduction Physiopathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy (A.A.); (V.D.G.); (S.D.S.); (R.V.); (V.M.); (R.S.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy;
| | - Valentina Magnani
- Division of Gynecology and Human Reproduction Physiopathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy (A.A.); (V.D.G.); (S.D.S.); (R.V.); (V.M.); (R.S.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy;
| | - Maurizio Guida
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (F.G.); (M.G.)
| | - Tommaso Pippucci
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy;
| | - Renato Seracchioli
- Division of Gynecology and Human Reproduction Physiopathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy (A.A.); (V.D.G.); (S.D.S.); (R.V.); (V.M.); (R.S.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy;
| |
Collapse
|
7
|
Marmé F, Martin M, Untch M, Thode C, Bonnefoi H, Kim SB, Bear H, Mc Carthy N, Gelmon K, García-Sáenz JA, Kelly CM, Reimer T, Valota O, Toi M, Rugo HS, Gnant M, Makris A, Bassy M, Zhang Z, Furlanetto J, Nekljudova V, Loibl S. Palbociclib combined with endocrine treatment in hormone receptor-positive, HER2-negative breast cancer patients with high relapse risk after neoadjuvant chemotherapy: subgroup analyses of premenopausal patients in PENELOPE-B. ESMO Open 2024; 9:103466. [PMID: 38838498 PMCID: PMC11190462 DOI: 10.1016/j.esmoop.2024.103466] [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: 12/19/2023] [Revised: 04/15/2024] [Accepted: 04/19/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND The PENELOPE-B study demonstrated that the addition of 1-year post-neoadjuvant palbociclib to endocrine therapy (ET) in patients with high-risk early breast cancer (BC) did not improve invasive disease-free survival (iDFS) compared to placebo. Here, we report results for premenopausal women. PATIENTS AND METHODS Patients with hormone receptor-positive, human epidermal growth factor receptor 2-negative BC at high risk of relapse [defined as no pathological complete response after neoadjuvant chemotherapy and a clinical, pathological stage, estrogen receptor, grading (CPS-EG) score ≥3 or 2/ypN+] were randomized to receive 13 cycles of palbociclib or placebo + standard ET. Ovarian function (OF) was evaluated by centrally assessed estradiol, follicle-stimulating hormone and anti-Müllerian hormone serum levels. RESULTS Overall, 616 of 1250 randomized patients were premenopausal; of these, 30.0% were <40 years of age, 47.4% had four or more metastatic lymph nodes, and 58.2% had a CPS-EG score ≥3. 66.1% of patients were treated with tamoxifen alone, and 32.9% received ovarian function suppression (OFS) in addition to either tamoxifen or aromatase inhibitor (AI). After a median follow-up of 42.8 months (97.2% completeness) no difference in iDFS between palbociclib and placebo was observed [hazard ratio = 0.95, 95% confidence interval (CI) 0.69-1.30, P = 0.737]. The estimated 3-year iDFS rate was marginally higher in the palbociclib arm (80.6% versus 78.3%). Three year iDFS was higher in patients receiving AI than tamoxifen plus OFS or tamoxifen alone (86.0% versus 78.6% versus 78.0%). Patients receiving tamoxifen plus OFS showed a favorable iDFS with palbociclib (83.0% versus 74.1%, hazard ratio = 0.52, 95% CI 0.27-1.02, P = 0.057). Hematologic adverse events were more frequent with palbociclib (76.1% versus 1.9% grade 3-4, P < 0.001). Palbociclib seems not to negatively impact the OF throughout the treatment period. CONCLUSIONS In premenopausal women, who received tamoxifen plus OFS as ET, the addition of palbociclib to ET results in a favorable iDFS. The safety profile seems favorable and in contrast to chemotherapy palbociclib does not impact OF throughout the treatment period.
Collapse
Affiliation(s)
- F Marmé
- Medical Faculty Mannheim, Heidelberg University, University Hospital Mannheim, Mannheim, Germany.
| | - M Martin
- Instituto de Investigacion Sanitaria Gregorio Marañon, CIBERONC, Universidad Complutense, Madrid; Spanish Breast Cancer Group, GEICAM, Madrid, Spain
| | - M Untch
- Helios Kliniken Berlin-Buch, Berlin
| | - C Thode
- Amedes MVZ Wagnerstibbe für Laboratoriumsmedizin, Medizinische Mikrobiologie und Immunologie, Göttingen, Germany
| | - H Bonnefoi
- Institut Bergonié and Université de Bordeaux INSERM U916, Bordeaux, France
| | - S-B Kim
- Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - H Bear
- Division of Surgical Oncology, Massey Cancer Center, Virginia Commonwealth University, VCU Health, Richmond, USA
| | - N Mc Carthy
- Breast Cancer Trials Australia and New Zealand and University of Queensland, Icon Cancer Centre Wesley, Auchenflower, Australia
| | - K Gelmon
- BC Cancer Agency, Vancouver, Canada
| | - J A García-Sáenz
- Service de Oncología Médica, Hospital Clínico San Carlos, Madrid, Spain
| | - C M Kelly
- Mater Private Hospital, Dublin and Cancer Trials, Dublin, Ireland
| | - T Reimer
- Department of Obstetrics and Gynecology, University of Rostock, Rostock, Germany
| | | | - M Toi
- Breast Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - H S Rugo
- University of California San Francisco Comprehensive Cancer Center, San Francisco, USA
| | - M Gnant
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - A Makris
- Institute of Cancer Research, Mount Vernon Cancer Centre, Northwood, UK
| | - M Bassy
- Amedes MVZ Wagnerstibbe für Laboratoriumsmedizin, Medizinische Mikrobiologie und Immunologie, Göttingen, Germany
| | | | | | | | - S Loibl
- German Breast Group, Neu-Isenburg, Germany. https://twitter.com/GBG_Forschung
| |
Collapse
|
8
|
Roof KA, Andre KE, Modesitt SC, Schirmer DA. Maximizing ovarian function and fertility following chemotherapy in premenopausal patients: Is there a role for ovarian suppression? Gynecol Oncol Rep 2024; 53:101383. [PMID: 38633671 PMCID: PMC11021951 DOI: 10.1016/j.gore.2024.101383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/19/2024] Open
Abstract
As more premenopausal patients undergo fertility preserving cancer treatments, there is an increased need for fertility counseling and ovarian sparing strategies. Many patients receive gonadotoxic chemotherapeutic agents which can put them at risk of primary ovarian insufficiency or profoundly diminished ovarian reserve. Traditionally, estradiol and follicle stimulating hormone (FSH) values have been used to evaluate ovarian function but more recently, reproductive endocrinologists have been proponents of anti-mullerian hormone (AMH) as a validated measure of ovarian potential. While the gold standard for fertility preservation remains oocyte cryopreservation, data suggest there may be additional interventions that can mitigate the gonadotoxic effects of chemotherapeutic agents. The main objectives of this focused review were to quantify the risk of primary ovarian failure associated with the most common chemotherapies used in treatment of gynecologic cancers and to evaluate and recommend potential interventions to mitigate toxic effects on ovarian function. Chemotherapeutic agents can cause direct loss of oocytes and primordial follicles as well as stromal and vascular atrophy and the extent is dependent upon mechanism of action and age of the patient. The risk of ovarian failure is the highest with alkylating agents (42.2 %), anthracyclines (<10-34 % in patients under 40 years versus 98 % in patients aged 40-49), taxanes (57.1 %) and platinum agents (50 %). Multiple trials demonstrate that gonadotropin releasing hormone (GnRH) agonists, when administered concurrently with chemotherapy, may have protective effects, with more patients experiencing resumption of a regular menstruation pattern and recovering ovarian function more quickly post-treatment. Premenopausal patients receiving chemotherapy for the treatment of gynecologic cancers should receive adequate counseling on the potential adverse effects on their fertility. Although oocyte cryopreservation remains the gold standard for fertility preservation, there is some evidence to suggest that GNRH agonists could help maintain and preserve ovarian function and should be considered.
Collapse
Affiliation(s)
- Kelsey A. Roof
- Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Kerri E. Andre
- Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Susan C. Modesitt
- Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA, United States
| | - D. Austin Schirmer
- Division of Reproductive Endocrinology, Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
9
|
Łubik-Lejawka D, Gabriel I, Marzec A, Olejek A. Oncofertility as an Essential Part of Comprehensive Cancer Treatment in Patients of Reproductive Age, Adolescents and Children. Cancers (Basel) 2024; 16:1858. [PMID: 38791937 PMCID: PMC11119835 DOI: 10.3390/cancers16101858] [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: 03/31/2024] [Revised: 05/08/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
The number of children, adolescents and young adults diagnosed with cancer has been rising recently. Various oncological treatments have a detrimental effect on female fertility, and childbearing becomes a major issue during surveillance after recovery. This review discusses the impact of oncological treatments on the ovarian reserve with a thorough explanation of oncologic treatments' effects and modes of oncofertility procedures. The aim of this review is to help clinicians in making an informed decision about post-treatment fertility in their patients. Ultimately, it may lead to improved overall long-term outcomes among young populations suffering from cancer.
Collapse
Affiliation(s)
| | | | | | - Anita Olejek
- Department of Gynaecology, Obstetrics and Oncological Gynaecology in Bytom, Medical University of Silesia, 40-055 Katowice, Poland; (D.Ł.-L.); (I.G.); (A.M.)
| |
Collapse
|
10
|
Link-Rachner CS, Göbel A, Jaschke NP, Rachner TD. Endocrine health in survivors of adult-onset cancer. Lancet Diabetes Endocrinol 2024; 12:350-364. [PMID: 38604215 DOI: 10.1016/s2213-8587(24)00088-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 03/11/2024] [Accepted: 03/11/2024] [Indexed: 04/13/2024]
Abstract
Long-term survivors of cancer (ie, the patient who is considered cured or for whom the disease is under long-term control and unlikely to recur) are at an increased risk of developing endocrine complications such as hypothalamic-pituitary dysfunctions, hypogonadisms, osteoporosis, or metabolic disorders, particularly when intensive tumour-directed therapies are applied. Symptom severity associated with these conditions ranges from mild and subclinical to highly detrimental, affecting individual health and quality of life. Although they are usually manageable, many of these endocrine pathologies remain underdiagnosed and untreated for years. To address this challenge, a higher degree of awareness, standardised screening tools, comprehensible treatment algorithms, and a close collaborative effort between endocrinologists and oncologists are essential to early identify patients who are at risk, and to implement appropriate treatment protocols. This Review highlights common symptoms and conditions related to endocrine disorders among survivors of adult-onset cancer, provides a summary of the currently available practice guidelines, and proposes a practical approach to diagnose affected patients among this group.
Collapse
Affiliation(s)
- Cornelia S Link-Rachner
- Division of Haematology and Oncology, Department of Medicine I, Technische Universität Dresden, Dresden, Germany
| | - Andy Göbel
- Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany; German Cancer Consortium (DKTK), partner site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nikolai P Jaschke
- Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany; German Cancer Consortium (DKTK), partner site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Tilman D Rachner
- Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany; German Cancer Consortium (DKTK), partner site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
11
|
Alalawy AI, Sakran M, Alzuaibr FM, Alotaibi MA, El-Hefnawy ME, Hazazi AY, El-Gendy SM, Aidy EA, Effat H, Ismail DF, Hessien M. Inhibition of Drp1 orchestrates the responsiveness of breast cancer cells to paclitaxel but insignificantly relieves paclitaxel-related ovarian damage in mice. Sci Rep 2023; 13:22782. [PMID: 38129495 PMCID: PMC10739747 DOI: 10.1038/s41598-023-49578-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 12/09/2023] [Indexed: 12/23/2023] Open
Abstract
Chemoresistance and chemotherapy-related ovarian damage are well-reported in breast cancer (BC) young patients. Herein, the inhibition of the mitochondrial fission was invested to explore its chemosensitizing role in Paclitaxel (PTX)-resistant cells, and its ability to restore the ovarian integrity in mice receiving PTX or cisplatin chemotherapy. To establish these aims, PTX-resistance was generated in BC cells, which were treated with PTX in combination with Drp1 deficiency, via mdivi-1, or Drp1-specific siRNA transfection. Furthermore, the alterations in the ovarian structure and the endocrine-related hormones were explored in mice receiving repetitive doses of PTX or cisplatin. We found that combining PTX with mdivi-1 improved cell responsiveness to PTX, induced apoptosis- and autophagy-mediated cell death, and relieved cellular oxidative stress. Additionally, the expression of PCNA1 and cyclin B1 genes were downregulated, meanwhile, p53, p21, and mitochondrial fusion proteins (Mfu1&Mfu2) were increased. The in vivo investigations in mice demonstrated that PTX induced gonadotoxic damage similar to cisplatin, whereas dual treatment of mice with PTX+ mdivi-1 failed to restore their normal follicular count and the circulating levels of E2 and AMH hormones. These results suggested that combining Drp1 inhibition with PTX resensitized breast cancer cells to PTX but failed to offer enough protection against chemotherapy-related gonadotoxicity.
Collapse
Affiliation(s)
- Adel I Alalawy
- Department of Biochemistry, Faculty of Science, University of Tabuk, 71491, Tabuk, Saudi Arabia
| | - Mohamed Sakran
- Department of Biochemistry, Faculty of Science, University of Tabuk, 71491, Tabuk, Saudi Arabia
- Division of Biochemistry, Faculty of Science, Tanta University, Tanta City, 31512, Egypt
| | - Fahad M Alzuaibr
- Biology Department, Faculty of Science, Tabuk University, Tabuk, Saudi Arabia
| | - Maeidh A Alotaibi
- King Faisal Medical Complex Laboratory, Ministry of Health, Taif, Saudi Arabia
| | - Mohamed E El-Hefnawy
- Division of Biochemistry, Faculty of Science, Tanta University, Tanta City, 31512, Egypt
- Department of Chemistry, Rabigh College of Sciences and Arts, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdulelah Y Hazazi
- Department of Biochemistry, Faculty of Science, University of Tabuk, 71491, Tabuk, Saudi Arabia
| | - Saad M El-Gendy
- Department of Cancer Biology, National Cancer Institute, Cairo University, Giza, Egypt
| | - Esraa A Aidy
- Department of Cancer Biology, National Cancer Institute, Cairo University, Giza, Egypt
| | - Heba Effat
- Department of Cancer Biology, National Cancer Institute, Cairo University, Giza, Egypt
| | - Doha F Ismail
- Department of Cancer Biology, National Cancer Institute, Cairo University, Giza, Egypt
| | - Mohamed Hessien
- Division of Biochemistry, Faculty of Science, Tanta University, Tanta City, 31512, Egypt.
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31512, Egypt.
| |
Collapse
|
12
|
Ünal Ç, Ordu Ç, Özmen T, İlgun AS, Çelebi F, Baysal B, Özkurt E, Duymaz T, Erdoğan İyigün Z, Kurt S, Öztürk MA, Pilancı KN, Alço G, Yararbaş K, Kayan Tapan T, Güven DC, Soybir G, Özmen V. Evaluation of Anti-Mullerian Hormone Levels, Antral Follicle Counts, and Mean Ovarian Volumes in Chemotherapy-Induced Amenorrhea among Breast Cancer Patients: A Prospective Clinical Study. Curr Oncol 2023; 30:9217-9229. [PMID: 37887566 PMCID: PMC10605884 DOI: 10.3390/curroncol30100666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/14/2023] [Accepted: 10/17/2023] [Indexed: 10/28/2023] Open
Abstract
Estradiol (E2), a follicle-stimulating hormone (FSH), AMH, and inhibin B levels, along with AFC and MOV, are used to determine ovarian reserve in pre-menopausal women. Studies have shown that AMH levels are more sensitive than those of E2, FSH, and inhibin B and that AFC and MOV can be used to evaluate ovarian reserve. AMH, AFC, and MOV measurements were performed before and after adjuvant SC in 3-month periods for one year. Patients were classified as experiencing chemotherapy-induced amenorrhea (CIA) if they did not have menstrual cycles for a period of six months or longer following the conclusion of their chemotherapy treatment. We aimed to evaluate the factors affecting chemotherapy-induced amenorrhea in breast cancer patients treated with adjuvant chemotherapy and the performance of baseline measurements of AMH, AFC, and MOV to predict chemotherapy-induced amenorrhea. The effects of different chemotherapy regimens on the AMH level, AFC, and MOV in CIA patients were investigated. Seventy-one patients were eligible for this study, and the median age was 38 years (range: 23-45). The median follow-up was 37 months (range: 20-51), and CIA developed in 62% of the patients. The AMH level and AFC were significantly decreased one year after SC (p < 0.0001), whereas MOV was not (p = 0.507). AMH levels before chemotherapy (median: 1.520 vs. 0.755, p = 0.001) and at the end of the first year (median: 0.073 vs. 0.010, p = 0.030) and pre-treatment AFC (median: 12 vs. 4.50, p = 0.026) were lower in patients with CIA compared to those without CIA. The AMH levels before SC were the most valuable and earliest factor for predicting CIA development. In addition, there was no difference between the chemotherapy regimens (including or not including taxane) in terms of CIA development.
Collapse
Affiliation(s)
- Çağlar Ünal
- Division of Medical Oncology, Department of Internal Medicine, Kartal Dr. Lütfi Kırdar City Hospital, Istanbul 34865, Turkey
| | - Çetin Ordu
- Division of Medical Oncology, Department of Internal Medicine, Gayrettepe Florence Nightingale Hospital, Istanbul 34349, Turkey;
| | - Tolga Özmen
- Division of Gastrointestinal and Oncologic Surgery, Harvard Medical School, Boston, MA 02115, USA;
- Division of Gastrointestinal and Oncologic Surgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | - Filiz Çelebi
- Department of Radiology, Yeditepe University Hospital, Istanbul 34755, Turkey;
| | - Bülent Baysal
- Department of Obstetrics and Gynecology, İstanbul Florence Nightingale Hospital, Istanbul 34381, Turkey;
| | - Enver Özkurt
- Department of General Surgery, İstanbul Florence Nightingale Hospital, Istanbul 34381, Turkey;
| | - Tomris Duymaz
- Department of Physiotherapy and Rehabilitation, Faculty of Health Sciences, Istanbul Bilgi University, Istanbul 34060, Turkey;
| | - Zeynep Erdoğan İyigün
- Department of Physical Theraphy and Rehabilitation, Göztepe Medical Park Hospital, Istanbul 34732, Turkey;
| | - Sevgi Kurt
- Department of Plastic Surgery, İstanbul Florence Nightingale Hospital, Istanbul 34381, Turkey;
| | | | - Kezban Nur Pilancı
- Division of Medical Oncology, Department of Internal Medicine, Memorial Bahçelievler Hospital, Istanbul 34180, Turkey;
| | - Gül Alço
- Department of Radiation Oncology, Gayrettepe Florence Nightingale Hospital, Istanbul 34349, Turkey;
| | - Kanay Yararbaş
- Department of Medical Genetics, Demiroglu Bilim University, Istanbul 34394, Turkey;
| | - Tuba Kayan Tapan
- Department of Nutrition and Dietetic, Faculty of Health Science, Demiroglu Bilim University, Istanbul 34394, Turkey;
| | - Deniz Can Güven
- Division of Medical Oncology, Department of Internal Medicine, Elazıg Fethi Sekin City Hospital, Elazıg 23280, Turkey;
| | - Gürsel Soybir
- Department of General Surgery, Memorial Şişli Hospital, Istanbul 34384, Turkey;
| | - Vahit Özmen
- Department of General Surgery, İstanbul School of Medicine, İstanbul University, Istanbul 34093, Turkey;
| |
Collapse
|
13
|
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: 9] [Impact Index Per Article: 4.5] [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
|
14
|
Dixit N, Braaten KP, Taylor C, Nekhlyudov L. Reproductive choice is supportive care in cancer. Support Care Cancer 2023; 31:249. [PMID: 37017781 DOI: 10.1007/s00520-023-07715-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 03/28/2023] [Indexed: 04/06/2023]
Affiliation(s)
- Niharika Dixit
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA.
| | - Kari P Braaten
- Department of Obstetrics and Gynecology, Brigham and Women's Hospital, Boston, MA, USA
| | | | | |
Collapse
|
15
|
Kuang X, Wei L, Huang Y, Ji M, Tang Y, Wei B, Yang S, Lai D, Xu H. Development of a digital anti-Müllerian hormone immunoassay: ultrasensitive, accurate and practical strategy for reduced ovarian reserve monitoring and assessment. Talanta 2023; 253:123970. [PMID: 36206626 DOI: 10.1016/j.talanta.2022.123970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 09/18/2022] [Accepted: 09/24/2022] [Indexed: 12/13/2022]
Abstract
Anti-Müllerian hormone (AMH) is an ideal biomarker for the assessment of ovarian reserve. However, its application in determining ovarian reserve reduction is restricted due to the low sensitivity of existing AMH assays. Herein, a homebrew ultrasensitive digital AMH assay (UD-AMH) was established based on a single-molecule array (SiMoA, HD-X platform), and the analytical performance of UD-AMH was evaluated systematically. The limit of detection (LoD) and limit of quantitation (LoQ) of UD-AMH were 0.13 and 0.14 pg/mL, respectively, which is approximately 100-fold higher than that of the current reported general clinical AMH assay. A comparison study showed a high correlation, with r = 0.988 for the Beckman Access AMH assay and r = 0.945 for the Kangrun AMH assay. In addition, we found that the AMH concentrations of premature ovarian insufficiency (POI) patients were very low (2.59 (0.86, 31.79) pg/mL) and similar to those of perimenopausal women (2.37 (0.65, 35.88) pg/mL) but significantly higher than those of menopausal women (0.43 (0.28, 1.17) pg/mL). Furthermore, we observed that the AMH concentration of most hormone therapy (HT) treated POI patients decreased sharply, suggesting that the ovarian reserve of POI patients declines over time even under HT-treatment.
Collapse
Affiliation(s)
- Xiaojun Kuang
- School of Biomedical Engineering, Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China; 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
| | - Liutong Wei
- 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
| | - Yuanxin Huang
- 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
| | - 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
| | - 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
| | - Bing Wei
- 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
| | - Shuang Yang
- Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - 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.
| | - Hong Xu
- School of Biomedical Engineering, Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
16
|
Qing T, Karn T, Rozenblit M, Foldi J, Marczyk M, Shan NL, Blenman K, Holtrich U, Kalinsky K, Meric-Bernstam F, Pusztai L. Molecular differences between younger versus older ER-positive and HER2-negative breast cancers. NPJ Breast Cancer 2022; 8:119. [PMID: 36344517 PMCID: PMC9640562 DOI: 10.1038/s41523-022-00492-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 10/26/2022] [Indexed: 11/09/2022] Open
Abstract
The RxPONDER and TAILORx trials demonstrated benefit from adjuvant chemotherapy in patients age ≤ 50 with node-positive breast cancer and Recurrence Score (RS) 0-26, and in node-negative disease with RS 16-25, respectively, but no benefit in older women with the same clinical features. We analyzed transcriptomic and genomic data of ER+/HER2- breast cancers with in silico RS < 26 from TCGA (n = 530), two microarray cohorts (A: n = 865; B: n = 609), the METABRIC (n = 867), and the SCAN-B (n = 1636) datasets. There was no difference in proliferation-related gene expression between age groups. Older patients had higher mutation burden and more frequent ESR1 copy number gain, but lower frequency of GATA3 mutations. Younger patients had higher rate of ESR1 copy number loss. In all datasets, younger patients had significantly lower mRNA expression of ESR1 and ER-associated genes, and higher expression of immune-related genes. The ER- and immune-related gene signatures showed negative correlation and defined three subpopulations in younger women: immune-high/ER-low, immune-intermediate/ER-intermediate, and immune-low/ER-intermediate. We hypothesize that in immune-high cancers, the cytotoxic effect of chemotherapy may drive the benefit, whereas in immune-low/ER-intermediate cancers chemotherapy induced ovarian suppression may play important role.
Collapse
Affiliation(s)
- Tao Qing
- Breast Medical Oncology, School of Medicine, Yale University, New Haven, CT, USA
| | - Thomas Karn
- Department of Gynecology and Obstetrics, Goethe-University Frankfurt, Frankfurt, Germany
| | - Mariya Rozenblit
- Breast Medical Oncology, School of Medicine, Yale University, New Haven, CT, USA
| | - Julia Foldi
- Breast Medical Oncology, School of Medicine, Yale University, New Haven, CT, USA
| | - Michal Marczyk
- Breast Medical Oncology, School of Medicine, Yale University, New Haven, CT, USA
- Department of Data Science and Engineering, Silesian University of Technology, Gliwice, Poland
| | - Naing Lin Shan
- Breast Medical Oncology, School of Medicine, Yale University, New Haven, CT, USA
| | - Kim Blenman
- Breast Medical Oncology, School of Medicine, Yale University, New Haven, CT, USA
| | - Uwe Holtrich
- Department of Gynecology and Obstetrics, Goethe-University Frankfurt, Frankfurt, Germany
| | - Kevin Kalinsky
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lajos Pusztai
- Breast Medical Oncology, School of Medicine, Yale University, New Haven, CT, USA.
| |
Collapse
|
17
|
Lan A, Jin Y, Wang Y, Ding N, Wang Y, Dai Y, Jiang L, Tang Z, Peng Y, Liu S. Association of serum reproductive hormones changes after neoadjuvant chemotherapy with hormone receptors expression alterations and survival outcomes in breast cancer. Front Surg 2022; 9:947218. [PMID: 36117838 PMCID: PMC9470751 DOI: 10.3389/fsurg.2022.947218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/15/2022] [Indexed: 11/30/2022] Open
Abstract
Purpose This study aimed to determine the effect of neoadjuvant chemotherapy (NAC) on circulating levels of reproductive hormones and evaluate the correlation of hormone changes after NAC with hormone receptors expression alterations and relapse-free survival (RFS) outcomes in breast cancer. Methods Information from 181 breast cancer patients who received NAC was retrospectively analyzed. For hormones parameters, the median and interquartile range (IQR) were provided at baseline and the end of NAC then was compared by Wilcoxon signed-rank test. Categorical variables were represented as numbers and percentages and were compared via two-sided chi-square and Fisher's tests. The RFS outcomes were compared between patients according to hormone changes using the log-rank test. Univariate and multivariate survival analyses with hazard ratios (HR) and 95% confidence intervals (95% CI) were carried out using Cox regression. Results Sex steroids including estradiol, progesterone, testosterone, and dehydroepiandrosterone sulfate (DHEAS) levels decreased significantly after NAC among both premenopausal and postmenopausal patients (all P < 0.05). Decreased estradiol levels were associated with reduced progesterone receptor (PR) expression (P = 0.030). In multivariate survival analysis, the non-decreased progesterone level was strongly associated with worse RFS (non-decreased vs. decreased, HR = 7.178, 95% CI 2.340–22.019, P = 0.001). Patients with decreased progesterone levels exhibited better 3-year RFS compared with those with non-decreased (87.6% vs. 58.3%, log-rank, P = 0.001). Conclusion Multiple reproductive hormone levels were influenced by NAC. The change in estradiol level had a positive connection with PR expression alteration. Furthermore, an inverse association between the change in progesterone level and RFS outcomes was found. These findings may provide a theoretical basis for pre-operative endocrine therapy combined with NAC in breast cancer patients.
Collapse
|
18
|
Construction of esterase-responsive hyperbranched polyprodrug micelles and their antitumor activity in vitro. E-POLYMERS 2022. [DOI: 10.1515/epoly-2022-0047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
This research constructs an esterase-responsive hyperbranched polyprodrug nano pharmaceutical and investigates their antitumor activity. Polyprodrug micelle was prepared by one-pot method based on glutathione (GSH), doxorubicin (DOX), and polyethylene glycol (PEG) under the catalyst of N,N-dicyclohexylcarbodiimide (DCC), 4-dimethylaminopyridine (DMAP), and 1-hydroxybenzotriazole (HOBt). The polyprodrug was characterized by nuclear magnetic resonance (NMR), Fourier transform infrared spectrometer (FT-IR), ultraviolet-visible spectrophotometer (UV-Vis), dynamic light scattering (DLS), and transmission electron microscope (TEM), respectively. The antitumor activity of polyprodrug micelle was evaluated by Hela cell and the distributions of micelles in cells were observed by fluorescent microscope. The NMR and FT-IR confirmed that the DOX-GSH-PEG polyprodrug was successfully synthesized. The drug loading rate is 10.21% and particle size is 106.4 ± 1 nm with a narrowed polydispersity (PDI = 0.145). The DLS showed that the micelles were stable during 7 days at 25°C. The drug release results showed that the micelles could be esterase-responsive disrupted, and the drug release rate could reach 43% during 72 h. Cell uptake and cell viability demonstrated that the micelles could distribute to cell nuclei during 8 h and induce cell apoptosis during 48 h. Overall, these hyperbranched polyprodrug micelles prepared by one-pot method could be esterase-responsive disrupted and release the antitumor drugs in a high esterase environment for cancer therapy in vitro. These results confirm that DOX-GSH-PEG is an effective nanomedicine in vitro and the endogenous-based strategy with one-pot synthesis to construct esterase-responsive polyprodrug would probably be a preferred choice in the future.
Collapse
|
19
|
Pre-treatment circulating reproductive hormones levels predict pathological and survival outcomes in breast cancer submitted to neoadjuvant chemotherapy. Int J Clin Oncol 2022; 27:899-910. [PMID: 35239089 DOI: 10.1007/s10147-022-02141-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 02/03/2022] [Indexed: 12/14/2022]
Abstract
PURPOSE This study aimed to evaluate the correlation of pre-treatment circulating reproductive hormones levels with pathological and survival outcomes in breast cancer patients received neoadjuvant chemotherapy (NAC). METHODS Information from 196 premenopausal and 137 postmenopausal breast cancer patients who received NAC were retrospectively analyzed. Treatment response to NAC, with odds ratios (OR) and 95% confidence intervals (95% CI) was estimated using logistic regression adjusted for key confounders. Survival outcomes with hazard ratios (HR) and 95% CI were estimated using Cox regression adjusted for key confounders. The Kaplan-Meier method was applied in the survival analysis. RESULTS Premenopausal patients with lower testosterone levels (OR = 0.996, 95% CI 0.992-0.999, P = 0.026), and postmenopausal patients with higher follicle-stimulating hormone (FSH) levels (OR = 1.045, 95% CI 1.014-1.077, P = 0.005) were likely to achieve pathological complete response (pCR). In multivariate survival analysis, the lowest tertile (T) progesterone was associated with worse overall survival (OS) in premenopausal patients (T2 vs T1, HR = 0.113, 95% CI 0.013-0.953, P = 0.045; T3 vs T1, HR = 0.109, 95% CI 0.013-0.916, P = 0.041). Premenopausal patients with the lowest tertile progesterone exhibited worse 3-year OS compared with those with higher tertiles (72.9% vs 97.4%, log-rank, P = 0.007). CONCLUSION Pre-treatment testosterone and FSH are significant independent predictors for pCR to NAC in premenopausal and postmenopausal patients, respectively. Low progesterone levels are correlated with worse OS in premenopausal patients. These findings may provide a theoretical basis for pre-operative endocrine therapy combined with NAC in breast cancer.
Collapse
|
20
|
Wu C, Wu T, Chen D, Wei S, Tang W, Xue L, Xiong J, Huang Y, Guo Y, Chen Y, Wu M, Wang S. The effects and mechanism of taxanes on chemotherapy-associated ovarian damage: A review of current evidence. Front Endocrinol (Lausanne) 2022; 13:1025018. [PMID: 36531475 PMCID: PMC9756165 DOI: 10.3389/fendo.2022.1025018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/09/2022] [Indexed: 11/25/2022] Open
Abstract
Chemotherapy is often a cause of premature ovarian insufficiency and infertility since the ovarian follicles are extremely sensitive to the effects of chemotherapeutic agents. Different chemotherapeutic agents with varying mechanisms of action may damage ovarian function differently. Taxanes are widely used in clinical cancer treatment, but the specific reproductive toxicological information is still controversial. This review described the impact and duration of taxanes on ovarian function in women and analyzed the possible reasons for different conclusions. Furthermore, the toxicity of taxanes on ovarian function and its possible mechanisms were discussed. The potential protective strategies and agents against ovarian damage induced by taxanes are also reviewed.
Collapse
Affiliation(s)
- Chuqing Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Tong Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Dan Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Simin Wei
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Weicheng Tang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Liru Xue
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Jiaqiang Xiong
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yibao Huang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Yican Guo
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Ying Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Meng Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
- *Correspondence: Shixuan Wang, ; Meng Wu,
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
- *Correspondence: Shixuan Wang, ; Meng Wu,
| |
Collapse
|