1
|
Signorino C, Bencini E, Tondo A, Scalini P, Tamburini A, Casini T, Sardi I, Stagi S. Ovarian function in adolescents and young adults undergoing cancer treatment: biochemical and ultrasound marker analysis. Endocr Connect 2025; 14:e240511. [PMID: 39887687 PMCID: PMC11896650 DOI: 10.1530/ec-24-0511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 01/07/2025] [Accepted: 01/31/2025] [Indexed: 02/01/2025]
Abstract
Objective Gonadal dysfunction is a major late complication after cancer diagnosis and treatment. We aimed to study the prevalence of premature ovarian insufficiency (POI) and the potential reduction of ovarian reserve in a cohort of adolescent and young adult (AYA) patients undergoing cancer treatments, evaluating ovarian function and reserve markers. We also aimed to analyze how these markers are related to each other and to treatment-related risk factors. Methods We performed a retrospective study, including all female AYA patients undergoing cancer treatment during childhood or adolescence, who visited our pediatric gynecology outpatient clinic between January 1, 2018, and August 30, 2023. Serum levels of anti-Mullerian hormone (AMH) and follicle-stimulating hormone (FSH), antral follicle count (AFC) and mean ovarian volume were evaluated. The correlations between these markers and how their alterations are related to treatment-related risk factors were analyzed. Results The prevalence of POI in 95 patients was 18.9%. A significant positive correlation was observed between AMH levels and both AFC and ovarian volume. AMH was the most reliable serum marker for ovarian function in terms of POI. Independent risk factors for ovarian dysfunction in relation to all the markers analyzed were hematopoietic stem cell transplantation (HSCT) and high doses of alkylating agents (≥6000 mg/m2). Conclusion Gonadal dysfunction and infertility are quite common in AYA patients undergoing cancer treatment. High-dose alkylating agents and HSCT are the independent risk factors. AMH and FSH values, AFC and mean ovarian volume provide different but consistent information for closely monitoring patients after cancer treatment.
Collapse
Affiliation(s)
- Claudia Signorino
- Auxo-Endocrinology Unit, Meyer Children's Hospital IRCCS, Florence, Italy
- Pediatrics and Neonatology Unit, Santo Stefano Hospital, AUSL Toscana Centro, Prato, Italy
| | - Erica Bencini
- Pediatric Gynecology, Prenatal Diagnosis and Congenital Defects Unit, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Annalisa Tondo
- Department of Pediatric Oncology and Haematology, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Perla Scalini
- Department of Pediatric Oncology and Haematology, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Angela Tamburini
- Department of Pediatric Oncology and Haematology, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Tommaso Casini
- Department of Pediatric Oncology and Haematology, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Iacopo Sardi
- Neuro-Oncology Unit, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Stefano Stagi
- Auxo-Endocrinology Unit, Meyer Children's Hospital IRCCS, Florence, Italy
- Department of Health Sciences, University of Florence, Florence, Italy
| |
Collapse
|
2
|
Bourdais A, Viard P, Bormann J, Sesboüé C, Guerrier D, Therville N, Guillermet-Guibert J, Carroll J, Halet G. Distinct requirements for PI3K isoforms p110α and p110δ for PIP3 synthesis in mouse oocytes and early embryos. Development 2025; 152:dev204398. [PMID: 39982048 DOI: 10.1242/dev.204398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 02/09/2025] [Indexed: 02/22/2025]
Abstract
The phosphoinositide 3-kinase (PI3K)/Akt pathway is thought to regulate key steps of mammalian oogenesis, such as dormant oocyte awakening during follicular activation, meiotic resumption and oocyte maturation. Supporting evidence is, however, indirect, as oocyte PI3K activation has never been formally demonstrated, and the PI3K isoforms involved have not been revealed. Here, we employed fluorescent PIP3 biosensors to characterize PI3K dynamics in mouse oocytes and we investigated the contribution of the PI3K isoform p110α by conditional genetic ablation. Prophase oocytes showed baseline PI3K/Akt activation that could be further stimulated by adding Kit ligand. Contrary to previous reports, maternal PI3K proved dispensable for oocyte maturation in vitro, yet it was required for PIP3 synthesis in early embryos. We further show that oocyte p110α is not essential for oogenesis and female fertility. Accordingly, our data suggest that Kit ligand activates isoform p110δ for PIP3 synthesis in oocytes. In contrast, constitutive PIP3 synthesis in early embryos is achieved by maternal p110α acting redundantly with p110δ. This study highlights the relevance of PIP3 biosensors in establishing the dynamics, mechanisms and roles of maternal PI3K signaling during mammalian oogenesis.
Collapse
Affiliation(s)
- Anne Bourdais
- Université de Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Patricia Viard
- Université de Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Jenny Bormann
- Department of Chemical Biology, ZMB, Faculty of Biology, University of Duisburg-Essen, Universitätsstrasse 2, 45117 Essen, Germany
| | - Côme Sesboüé
- Université de Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Daniel Guerrier
- Université de Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Nicole Therville
- CRCT, Université de Toulouse, INSERM, CNRS, Université Toulouse III-Paul Sabatier, Centres de Recherches en Cancérologie de Toulouse, Toulouse, France
| | - Julie Guillermet-Guibert
- CRCT, Université de Toulouse, INSERM, CNRS, Université Toulouse III-Paul Sabatier, Centres de Recherches en Cancérologie de Toulouse, Toulouse, France
| | - John Carroll
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Guillaume Halet
- Université de Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000 Rennes, France
| |
Collapse
|
3
|
Bahmanpour S, Ameri N, Zareifard N, Karimi F. The Protective Effect of GnRH Agonist Triptorelin on the Histomorphometric Parameters of the Utero-ovarian Tissue in the Doxorubicin- and Cyclophosphamide-treated Mice. Cell Biochem Biophys 2025; 83:573-586. [PMID: 39244688 DOI: 10.1007/s12013-024-01487-3] [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] [Accepted: 08/09/2024] [Indexed: 09/10/2024]
Abstract
One of the common side effects of chemotherapy drugs is ovarian failure and uterine dysfunction, which can occur after the administration of doxorubicin and/or cyclophosphamide. In clinics, gonadotropin-releasing hormone agonists (GnRHa) are used to modulate the toxic effect of chemotherapy and intercept infertility with some controversy and limited histological knowledge. This study aimed to evaluate the serological and histological features of protective effects of triptorelin, (GnRHa), on utero-ovarian tissue in the mice treated with cyclophosphamide and/or doxorubicin. Forty-eight female BALB/c mice were randomly divided into 8 groups as follows: Group I: normal saline; Group II: triptorelin; Group III: cyclophosphamide; Group IV: doxorubicin; Group V: cyclophosphamide + doxorubicin; and Groups VI, VII, and VIII: after injection of cyclophosphamide, doxorubicin, or cyclophosphamide + doxorubicin, administration of triptorelin (1 mg/kg; intraperitoneally) for 15 consecutive days, respectively. On the 21st day, the ovaries and uterine horns were dissected and weighed. Then, tissue processing and staining were performed for further histological and stereological studies. Triptorelin treatment in the damaged groups significantly increased the number of primordial and pre-antral follicles and granulosa cells. It decreased the number of atretic follicles compared to cyclophosphamide and/or doxorubicin-treated groups (P < 0.05). Triptorelin also significantly improved the volume of the ovary, cortex, medulla, oocytes in the primordial and antral follicles, uterus, endometrium, myometrium, uterine glands, and endometrial blood vessels in the damaged groups (P < 0.05). Triptorelin treatment prevents the destructive effects of cyclophosphamide and/or doxorubicin on utero-ovarian tissue.
Collapse
Affiliation(s)
- Soghra Bahmanpour
- Anatomy Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negin Ameri
- Anatomy Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nehleh Zareifard
- Anatomy Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Karimi
- Anatomy Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
4
|
Cao H, Qiu C, Fang A, Shang J, Xu W, He L, Duan X, Zhang Q, Yu C. Extensive homologous recombination safeguards oocyte genome integrity in mammals. Nucleic Acids Res 2025; 53:gkae1304. [PMID: 39797737 PMCID: PMC11724361 DOI: 10.1093/nar/gkae1304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 12/12/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
Meiosis in mammalian oocytes is interrupted by a prolonged arrest at the germinal vesicle stage, during which oocytes have to repair DNA lesions to ensure genome integrity or otherwise undergo apoptosis. The FIRRM/FLIP-FIGNL1 complex dissociates RAD51 from the joint DNA molecules in both homologous recombination (HR) and DNA replication. However, as a type of non-meiotic, non-replicative cells, whether this RAD51-dismantling mechanism regulates genome integrity in oocytes remains elusive. Here, we show that FIRRM/FLIP is required for disassembly of RAD51-filaments and maintenance of genome integrity in oocytes. Deletion of FIRRM in oocytes leads to formation of massive nuclear RAD51 foci in oocytes of primordial follicles and activated follicles in mice. These RAD51 foci colocalize with the sites of DNA damage repair, as indicated by RPA2 and EdU, suggesting substantial DNA damage and extensive HR in oocytes. Especially in fully-grown FIRRM-deleted oocytes, RAD51 forms a net-like structure. As a consequence, FIRRM-deleted females are infertile due to aberrant homologous chromosome segregation at metaphase I and primordial follicle insufficiency at young adulthood. Hence, our study demonstrates the physiological importance of HR in maintaining genome integrity in oocytes.
Collapse
Affiliation(s)
- Huiwen Cao
- MOE Key Laboratory of Biosystems Homeostasis and Protection, College of Life Sciences, Zhejiang University, No.866 Yuhangtang Road, 310058, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, No.718 East Haizhou Road, 314499, Haining, China
| | - Cheng Qiu
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, No.718 East Haizhou Road, 314499, Haining, China
| | - Anxuan Fang
- MOE Key Laboratory of Biosystems Homeostasis and Protection, College of Life Sciences, Zhejiang University, No.866 Yuhangtang Road, 310058, Hangzhou, China
| | - Jianzhou Shang
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, No.666 Wusu Street, 311300, Hangzhou, China
| | - Wei Xu
- MOE Key Laboratory of Biosystems Homeostasis and Protection, College of Life Sciences, Zhejiang University, No.866 Yuhangtang Road, 310058, Hangzhou, China
| | - Lugeng He
- Department of Urology, the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, No.79 Qinchun Road, 310003,Hangzhou, China
| | - Xing Duan
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, No.666 Wusu Street, 311300, Hangzhou, China
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, No.87 Dingjiaqiao Street, 210009, Nanjing, China
| | - Qianting Zhang
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, No.718 East Haizhou Road, 314499, Haining, China
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, No.88 Jiefang Road, 310009, Hangzhou, China
| | - Chao Yu
- MOE Key Laboratory of Biosystems Homeostasis and Protection, College of Life Sciences, Zhejiang University, No.866 Yuhangtang Road, 310058, Hangzhou, China
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University, School of Medicine, No.3 Qinchun Road, 310009, Hangzhou, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, No.3 Qinchun Road, 310009, Hangzhou, China
| |
Collapse
|
5
|
Mazza T, Scalise M, Console L, Galluccio M, Giangregorio N, Tonazzi A, Pochini L, Indiveri C. Carnitine traffic and human fertility. Biochem Pharmacol 2024; 230:116565. [PMID: 39368751 DOI: 10.1016/j.bcp.2024.116565] [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: 07/31/2024] [Revised: 09/18/2024] [Accepted: 10/01/2024] [Indexed: 10/07/2024]
Abstract
Carnitine is a vital molecule in human metabolism, prominently involved in fatty acid β-oxidation within mitochondria. Predominantly sourced from dietary intake, carnitine also derives from endogenous synthesis. This review delves into the complex network of carnitine transport and distribution, emphasizing its pivotal role in human fertility. Together with its role in fatty acid oxidation, carnitine modulates the acety-CoA/CoA ratio, influencing carbohydrate metabolism, lipid biosynthesis, and gene expression. The intricate regulation of carnitine homeostasis involves a network of membrane transporters, notably OCTN2, which is central in its absorption, reabsorption, and distribution. OCTN2 dysfunction, results in Primary Carnitine Deficiency (PCD), characterized by systemic carnitine depletion and severe clinical manifestations, including fertility issues. In the male reproductive system, carnitine is crucial for sperm maturation and motility. In the female reproductive system, carnitine supports mitochondrial function necessary for oocyte quality, folliculogenesis, and embryonic development. Indeed, deficiencies in carnitine or its transporters have been linked to asthenozoospermia, reduced sperm quality, and suboptimal fertility outcomes in couples. Moreover, the antioxidant properties of carnitine protect spermatozoa from oxidative stress and help in managing conditions like polycystic ovary syndrome (PCOS) and endometriosis, enhancing sperm viability and fertilization potential of oocytes. This review summarizes the key role of membrane transporters in guaranteeing carnitine homeostasis with a special focus on the implications in fertility and possible treatments of infertility and other related disorders.
Collapse
Affiliation(s)
- Tiziano Mazza
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, Arcavacata di Rende 87036, Italy
| | - Mariafrancesca Scalise
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, Arcavacata di Rende 87036, Italy
| | - Lara Console
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, Arcavacata di Rende 87036, Italy
| | - Michele Galluccio
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, Arcavacata di Rende 87036, Italy
| | - Nicola Giangregorio
- CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnology (IBIOM), via Amendola 122/O, Bari 70126, Italy
| | - Annamaria Tonazzi
- CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnology (IBIOM), via Amendola 122/O, Bari 70126, Italy
| | - Lorena Pochini
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, Arcavacata di Rende 87036, Italy; CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnology (IBIOM), via Amendola 122/O, Bari 70126, Italy.
| | - Cesare Indiveri
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, Arcavacata di Rende 87036, Italy; CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnology (IBIOM), via Amendola 122/O, Bari 70126, Italy.
| |
Collapse
|
6
|
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
|
7
|
Teixeira De Oliveira J, Brito Tecchio K, Silva Lopes M, Nunes Andrade S, Iara Maciel De Azambuja Ribeiro R, Varotti FDP, Barbosa De Oliveira R, Henrique Ribeiro Viana G, J Da Silva Vieira Dos Santos V, Vieira Dos Santos F. In vitro evaluation of the selective cytotoxicity and genotoxicity of three synthetic ortho-nitrobenzyl derivatives in human cancer cell lines, with and without metabolic activation. Drug Chem Toxicol 2024; 47:404-415. [PMID: 38949608 DOI: 10.1080/01480545.2023.2184478] [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: 07/20/2022] [Accepted: 02/18/2023] [Indexed: 03/11/2023]
Abstract
Although the presence of nitro groups in chemicals can be recognized as structural alerts for mutagenicity and carcinogenicity, nitroaromatic compounds have attracted considerable interest as a class of agents that can serve as source of potential new anticancer agents. In the present study, the in vitro cytotoxicity, genotoxicity, and mutagenicity of three synthetic ortho-nitrobenzyl derivatives (named ON-1, ON-2 and ON-3) were evaluated by employing human breast and ovarian cancer cell lines. A series of biological assays was carried out with and without metabolic activation. Complementarily, computational predictions of the pharmacokinetic properties and druglikeness of the compounds were performed in the Swiss ADME platform. The MTT assay showed that the compounds selectively affected selectively the cell viability of cancer cells in comparison with a nontumoral cell line. Additionally, the metabolic activation enhanced cytotoxicity, and the compounds affected cell survival, as demonstrated by the clonogenic assay. The comet assay, the cytokinesis-block micronucleus assay, and the immunofluorescence of the γ-H2AX foci formation assay have that the compounds caused chromosomal damage to the cancer cells, with and without metabolic activation. The results obtained in the present study showed that the compounds assessed were genotoxic and mutagenic, inducing double-strand breaks in the DNA structure. The high selectivity indices observed for the compounds ON-2 and ON-3, especially after metabolic activation with the S9 fraction, must be highlighted. These experimental biological results, as well as the theoretical properties predicted for the compounds have shown that they are promising anticancer candidates to be exploited in additional studies.
Collapse
Affiliation(s)
- Júlia Teixeira De Oliveira
- Laboratório de Biologia Celular e Mutagênese (LaBCeM), Universidade Federal de São João del Rei (UFSJ), Divinópolis, Brazil
- Núcleo de Pesquisa em Química Biológica (NQBio), Universidade Federal de São João del Rei (UFSJ), Divinópolis, Brazil
| | - Kimberly Brito Tecchio
- Laboratório de Biologia Celular e Mutagênese (LaBCeM), Universidade Federal de São João del Rei (UFSJ), Divinópolis, Brazil
- Núcleo de Pesquisa em Química Biológica (NQBio), Universidade Federal de São João del Rei (UFSJ), Divinópolis, Brazil
| | - Marcela Silva Lopes
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Silmara Nunes Andrade
- Núcleo de Pesquisa em Química Biológica (NQBio), Universidade Federal de São João del Rei (UFSJ), Divinópolis, Brazil
| | | | - Fernando De Pilla Varotti
- Núcleo de Pesquisa em Química Biológica (NQBio), Universidade Federal de São João del Rei (UFSJ), Divinópolis, Brazil
| | - Renata Barbosa De Oliveira
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | | | - Fabio Vieira Dos Santos
- Laboratório de Biologia Celular e Mutagênese (LaBCeM), Universidade Federal de São João del Rei (UFSJ), Divinópolis, Brazil
- Núcleo de Pesquisa em Química Biológica (NQBio), Universidade Federal de São João del Rei (UFSJ), Divinópolis, Brazil
| |
Collapse
|
8
|
Carmi YK, Agbarya A, Khamaisi H, Farah R, Shechtman Y, Korobochka R, Gopas J, Mahajna J. Ovarian cancer ascites confers platinum chemoresistance to ovarian cancer cells. Transl Oncol 2024; 44:101939. [PMID: 38489872 PMCID: PMC10955424 DOI: 10.1016/j.tranon.2024.101939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 03/01/2024] [Accepted: 03/11/2024] [Indexed: 03/17/2024] Open
Abstract
Ovarian cancer (OC), the second most common form of gynecologic malignancy, has a poor prognosis and is often discovered in the late stages. Platinum-based chemotherapy is the first line of therapy. Nevertheless, treatment OC has proven challenging due to toxicity and the development of acquired resistance to therapy. Tumor microenvironment (TME) has been associated with platinum chemoresistance. Malignant ascites has been used as OC tumor microenvironment and its ability to induce platinum chemoresistance has been investigated. Our results suggest that exposure to OC ascites induces platinum chemoresistance in 11 of 13 cases (85 %) on OC cells. In contrast, 75 % of cirrhotic ascites (3 of 4) failed to confer platinum chemoresistance to OC cells. Cytokine array analysis revealed that IL -6 and to a lesser extent HGF were enriched in OC ascites, whereas IL -22 was enriched in cirrhotic ascites. Pharmaceutical inhibitors targeting the IL -6/ JAK pathway were mildly effective in overcoming platinum chemoresistance induced by malignant ascites. In contrast, crizotinib, an HGF/c- MET inhibitor, and 2-hydroxyestradiol (2HE2) were effective in restoring platinum chemosensitivity to OC. Our results demonstrate the importance of OC ascites in supporting platinum chemoresistance and the potential of combination therapy to restore chemosensitivity of OC cells.
Collapse
Affiliation(s)
- Yifat Koren Carmi
- Department of Nutrition and Natural Products, Migal - Galilee Research Institute, Kiryat Shmona, Israel; Shraga Segal Department of Microbiology, Immunology and Genetics, and Department of Oncology, Soroka University Medical Center, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Abed Agbarya
- Oncology Department, Bnai Zion MC, Haifa, Israel
| | - Hazem Khamaisi
- Department of Nutrition and Natural Products, Migal - Galilee Research Institute, Kiryat Shmona, Israel
| | - Raymond Farah
- Department of Internal Medicine, Ziv Medical Center, Safed, Israel
| | | | | | - Jacob Gopas
- Shraga Segal Department of Microbiology, Immunology and Genetics, and Department of Oncology, Soroka University Medical Center, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Jamal Mahajna
- Department of Nutrition and Natural Products, Migal - Galilee Research Institute, Kiryat Shmona, Israel; Department of Biotechnology, Tel-Hai College, Kiryat Shmona, Israel.
| |
Collapse
|
9
|
Helgadottir H, Matikas A, Fernebro J, Frödin JE, Ekman S, Rodriguez-Wallberg KA. Fertility and reproductive concerns related to the new generation of cancer drugs and the clinical implication for young individuals undergoing treatments for solid tumors. Eur J Cancer 2024; 202:114010. [PMID: 38520926 DOI: 10.1016/j.ejca.2024.114010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/03/2024] [Accepted: 03/05/2024] [Indexed: 03/25/2024]
Abstract
The treatment landscape of solid tumors has changed markedly in the last years. Molecularly targeted treatments and immunotherapies have been implemented and have, in many cancers, lowered the risk of relapse and prolonged survival. Patients with tumors harboring specific targetable molecular alterations or mutations are often of a younger age, and hence future fertility and family building can be important concerns in this group. However, there are great uncertainties regarding the effect of the new drugs on reproductive functions, including fertility, pregnancy and lactation and how young patients with cancers, both women and men should be advised. The goal with this review is to gather the current knowledge regarding oncofertility and the different novel therapies, including immune checkpoint inhibitors, antibody-drug conjugates, small molecules and monoclonal antibody targeted therapies. The specific circumstances and reproductive concerns in different patient groups where novel treatments have been broadly introduced are also discussed, including those with melanoma, lung, breast, colorectal and gynecological cancers. It is clear, that more awareness is needed regarding potential drug toxicity on reproductive tissues, and it is of essence that individuals are informed based on current expertise and on available fertility preservation methods.
Collapse
Affiliation(s)
- Hildur Helgadottir
- Department of Oncology and Pathology, Karolinska Institutet, 171 77 Stockholm, Sweden; Skin Cancer Centrum, Theme Cancer, Karolinska University Hospital, 171 76 Stockholm, Sweden.
| | - Alexios Matikas
- Department of Oncology and Pathology, Karolinska Institutet, 171 77 Stockholm, Sweden; Breast Center, Karolinska Comprehensive Cancer Center and Karolinska University Hospital, Stockholm, Sweden
| | - Josefin Fernebro
- Department of Oncology and Pathology, Karolinska Institutet, 171 77 Stockholm, Sweden; Division of Gynecological Cancer, Department of Pelvic Cancer, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Jan-Erik Frödin
- Department of Oncology and Pathology, Karolinska Institutet, 171 77 Stockholm, Sweden; Division of Gastrointestinal Oncology, Department of Upper abdomen, Karolinska University Hospital, Sweden
| | - Simon Ekman
- Department of Oncology and Pathology, Karolinska Institutet, 171 77 Stockholm, Sweden; Thoracic Oncology Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Kenny A Rodriguez-Wallberg
- Department of Oncology and Pathology, Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Reproductive Medicine, Division of Gynecology and Reproduction Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
10
|
Nascimento BC, Ferreira CS, Oliveira SP, Pereira LAAC, Lopes GA, Nogueira JM, Paula RS, Jorge EC, Campos-Junior PHA. Naproxen administration affects murine late folliculogenesis, reduces granulosa cell proliferation and the number of ovulated oocytes. Reprod Toxicol 2024; 124:108527. [PMID: 38160782 DOI: 10.1016/j.reprotox.2023.108527] [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: 05/26/2023] [Revised: 12/14/2023] [Accepted: 12/19/2023] [Indexed: 01/03/2024]
Abstract
Naproxen reduces the production of prostaglandins via inhibition of the cyclooxygenase. Studies have shown that its administration in women can be related to failed ovulation. Therefore, preclinical investigations must be performed in order to investigate its effects in experimental models. Thus, the aim of this study was to evaluate the effects of naproxen on murine folliculogenesis, ovulation, and female fertility. Female C57BL/6 mice (n = 128 - 6 weeks old) were divided into Control, low (10 mg/kg), and high naproxen (50 mg/kg) groups, who were treated for 8 days and directed to morphofunctional analyses. Follicular quantification showed a reduced percentage of antral follicles in naproxen-treated animals. These treated animals also showed smaller oocytes included in secondary and antral follicles, and the diameter of secondary and antral follicles was also reduced. A reduction in the percentage of Ki67-positive granulosa cells was observed in treated animals that also showed down-regulation of Igf1r compared to control. After an ovarian stimulation protocol, naproxen-treated animals showed a reduction in the percentage of secondary and antral follicles, a reduced number of ovulated oocytes and, corpora lutea, and an increased number of failed ovulations. Finally, naproxen-treated animals also showed a reduction in mating index and pregnancy rate. Our findings suggested that, in mice, naproxen administration (eight days treatment) negatively affects molecular and morphological aspects related to late folliculogenesis, ovulation, and fertility.
Collapse
Affiliation(s)
- Bernardo Camara Nascimento
- Laboratory for Reproductive Biology Research, Department of Natural Sciences, Federal University of São João del Rei, São João Del-Rei, MG, Brazil
| | - Camila Stefane Ferreira
- Laboratory for Reproductive Biology Research, Department of Natural Sciences, Federal University of São João del Rei, São João Del-Rei, MG, Brazil
| | - Stella Pollyanne Oliveira
- Laboratory for Reproductive Biology Research, Department of Natural Sciences, Federal University of São João del Rei, São João Del-Rei, MG, Brazil
| | | | - Guilherme Antonio Lopes
- Laboratory for Reproductive Biology Research, Department of Natural Sciences, Federal University of São João del Rei, São João Del-Rei, MG, Brazil
| | - Júlia Meireles Nogueira
- Departament of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Rayan Silva Paula
- Departament of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Erika Cristina Jorge
- Departament of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Paulo Henrique Almeida Campos-Junior
- Laboratory for Reproductive Biology Research, Department of Natural Sciences, Federal University of São João del Rei, São João Del-Rei, MG, Brazil.
| |
Collapse
|
11
|
Emori C, Boucher Z, Bolcun-Filas E. CHEK2 signaling is the key regulator of oocyte survival after chemotherapy. SCIENCE ADVANCES 2023; 9:eadg0898. [PMID: 37862420 PMCID: PMC10588956 DOI: 10.1126/sciadv.adg0898] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 09/06/2023] [Indexed: 10/22/2023]
Abstract
Cancer treatments can damage the ovarian follicle reserve, leading to primary ovarian insufficiency and infertility among survivors. Checkpoint kinase 2 (CHEK2) deficiency prevents elimination of oocytes in primordial follicles in female mice exposed to radiation and preserves their ovarian function and fertility. Here, we demonstrate that CHEK2 also coordinates the elimination of oocytes after exposure to standard-of-care chemotherapy drugs. CHEK2 activates two downstream targets-TAp63 and p53-which direct oocyte elimination. CHEK2 knockout or pharmacological inhibition preserved ovarian follicle reserve after radiation and chemotherapy. However, the lack of specificity for CHEK2 among available inhibitors limits their potential for clinical development. These findings demonstrate that CHEK2 is a master regulator of the ovarian cellular response to damage caused by radiation and chemotherapy and warrant the development of selective inhibitors specific to CHEK2 as a potential avenue for ovario-protective treatments.
Collapse
Affiliation(s)
- Chihiro Emori
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 5650871, Japan
| | - Zachary Boucher
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | | |
Collapse
|
12
|
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
|
13
|
Telfer EE, Grosbois J, Odey YL, Rosario R, Anderson RA. Making a good egg: human oocyte health, aging, and in vitro development. Physiol Rev 2023; 103:2623-2677. [PMID: 37171807 PMCID: PMC10625843 DOI: 10.1152/physrev.00032.2022] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 05/03/2023] [Accepted: 05/06/2023] [Indexed: 05/13/2023] Open
Abstract
Mammalian eggs (oocytes) are formed during fetal life and establish associations with somatic cells to form primordial follicles that create a store of germ cells (the primordial pool). The size of this pool is influenced by key events during the formation of germ cells and by factors that influence the subsequent activation of follicle growth. These regulatory pathways must ensure that the reserve of oocytes within primordial follicles in humans lasts for up to 50 years, yet only approximately 0.1% will ever be ovulated with the rest undergoing degeneration. This review outlines the mechanisms and regulatory pathways that govern the processes of oocyte and follicle formation and later growth, within the ovarian stroma, through to ovulation with particular reference to human oocytes/follicles. In addition, the effects of aging on female reproductive capacity through changes in oocyte number and quality are emphasized, with both the cellular mechanisms and clinical implications discussed. Finally, the details of current developments in culture systems that support all stages of follicle growth to generate mature oocytes in vitro and emerging prospects for making new oocytes from stem cells are outlined.
Collapse
Affiliation(s)
- Evelyn E Telfer
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Johanne Grosbois
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Yvonne L Odey
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Roseanne Rosario
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Richard A Anderson
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
14
|
Tsafrir A, BenHaroush A, Hyman J, Karavani G, Imbar T. What are the most common controversial clinical issues in fertility preservation? A content analysis of a collaborative professional online consultation group. Reprod Biol Endocrinol 2023; 21:77. [PMID: 37620900 PMCID: PMC10463612 DOI: 10.1186/s12958-023-01122-5] [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: 06/20/2023] [Accepted: 07/26/2023] [Indexed: 08/26/2023] Open
Abstract
RESEARCH QUESTION Clinicians involved in fertility preservation (FP) are often required to make prompt and consequential decisions despite the absence of evidence-based data. We established a collaborative professional online consultation group for fertility preservation issues. We sought to determine the main controversial clinical issues in FP as raised by participants of this group. DESIGN Content analysis of a dedicated community of practice interacting via a messaging application (WhatsApp) and a survey of group participants. RESULTS Between January 2019 and July 2022, group members posed 39 clinical questions which were discussed and debated by the group. Common themes included management of oncofertility cases (33%), potential gonadotoxicity of various therapies (23%), fertility preservation in women and girls with premature ovarian insufficiency (POI) (18%), and technical aspects of ovarian tissue cryopreservation (10%). All but one query received prompt response (mean time for first response for 95% of queries 7.1 ± 9.0 min) from a mean of 5.4 ± 3.2 members. An anonymous online survey of group members was conducted during August 2022 (n = 31, response rate 94%). The majority of respondents stated they gained knowledge and assistance in clinical decision making from participation in the discussion group (90% and 58% of respondents, respectively). CONCLUSIONS Management of clinical oncofertility cases, potential gonadotoxic effect of therapeutics and fertility preservation in women and girls with POI were the most common controversial issues in our fertility preservation community of practice. Intra-professional collaborative communication via a messaging application can aid in clinical management of fertility preservation and augment clinician's knowledge.
Collapse
Affiliation(s)
- Avi Tsafrir
- IVF unit, Department of Obstetrics and Gynecology, Shaare Zedek Medical Center, Jerusalem, Israel.
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Avi BenHaroush
- IVF unit, Rabin Medical Center, Petah Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jordana Hyman
- IVF unit, Department of Obstetrics and Gynecology, Shaare Zedek Medical Center, Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Gilad Karavani
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Fertility preservation unit, Department of Obstetrics and Gynecology Hadassah Medical Center, Jerusalem, Israel
| | - Tal Imbar
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Fertility preservation unit, Department of Obstetrics and Gynecology Hadassah Medical Center, Jerusalem, Israel
| |
Collapse
|
15
|
Yang H, Li X, Yang W. Advances in targeted therapy and immunotherapy for esophageal cancer. Chin Med J (Engl) 2023; 136:1910-1922. [PMID: 37403208 PMCID: PMC10431250 DOI: 10.1097/cm9.0000000000002768] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Indexed: 07/06/2023] Open
Abstract
ABSTRACT Esophageal cancer (EC) is one of the most common aggressive malignant tumors in the digestive system with a severe epidemiological situation and poor prognosis. The early diagnostic rate of EC is low, and most EC patients are diagnosed at an advanced stage. Multiple multimodality treatments have gradually evolved into the main treatment for advanced EC, including surgery, chemotherapy, radiotherapy, targeted therapy, and immunotherapy. And the emergence of targeted therapy and immunotherapy has greatly improved the survival of EC patients. This review highlights the latest advances in targeted therapy and immunotherapy for EC, discusses the efficacy and safety of relevant drugs, summarizes related important clinical trials, and tries to provide references for therapeutic strategy of EC.
Collapse
Affiliation(s)
- Haiou Yang
- Cancer center, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi 030032, China
| | - Xuewei Li
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Wenhui Yang
- Department of Gastroenterology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi 030001, China
| |
Collapse
|
16
|
Alesi LR, Nguyen QN, Stringer JM, Winship AL, Hutt KJ. The future of fertility preservation for women treated with chemotherapy. REPRODUCTION AND FERTILITY 2023; 4:RAF-22-0123. [PMID: 37068157 PMCID: PMC10235927 DOI: 10.1530/raf-22-0123] [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: 11/14/2022] [Accepted: 04/17/2023] [Indexed: 04/19/2023] Open
Abstract
Cytotoxic chemotherapies have been a mainstay of cancer treatment, but are associated with numerous systemic adverse effects, including impacts to fertility and endocrine health. Irreversible ovarian damage and follicle depletion are side-effects of chemotherapy that can lead to infertility and premature menopause, both being major concerns of young cancer patients. Notably, many women will proceed with fertility preservation, but unfortunately existing strategies don't entirely solve the problem. Most significantly, oocyte and embryo freezing do not prevent cancer treatment-induced ovarian damage from occurring, which may result in the impairment of long-term hormone production. Unfortunately, loss of endogenous endocrine function is not fully restored by hormone replacement therapy. Additionally, while GnRH agonists are standard care for patients receiving alkylating chemotherapy to lessen the risk of premature menopause, their efficacy is incomplete. The lack of more broadly effective options stems, in part, from our poor understanding of how different treatments damage the ovary. Here, we summarise the impacts of two commonly utilised chemotherapies - cyclophosphamide and cisplatin - on ovarian function and fertility, and discuss the mechanisms underpinning this damage. Additionally, we critically analyse current research avenues in the development of novel fertility preservation strategies, with a focus on fertoprotective agents.
Collapse
Affiliation(s)
- Lauren R Alesi
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Quynh-Nhu Nguyen
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Paediatric Integrated Cancer Service, VIC, Australia
| | - Jessica M Stringer
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Amy L Winship
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Karla J Hutt
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| |
Collapse
|
17
|
Sellami I, Beau I, Sonigo C. Chemotherapy and female fertility. ANNALES D'ENDOCRINOLOGIE 2023; 84:382-387. [PMID: 36967045 DOI: 10.1016/j.ando.2023.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Chemotherapy to treat cancer is usually responsible for early ovarian follicle depletion. Ovarian damage induced by cancer treatments frequently results in infertility in surviving patients of childbearing age. Several fertility preservation techniques have been developed. Nowadays, oocyte or embryo cryopreservation with or without ovarian stimulation and cryopreservation of the ovarian cortex are the most commonly used. However, these methods may be difficult to implement in some situations, and subsequent use of the cryopreserved germ cells remains uncertain, with no guarantee of pregnancy. Improved knowledge of the molecular mechanisms and signaling pathways involved in chemotherapy-induced ovarian damage is therefore necessary, to develop new strategies for fertility preservation. The effects of various chemotherapies have been studied in animal models or in vitro on ovarian cultures, suggesting various mechanisms of gonadotoxicity. Today the challenge is to develop molecules and techniques to limit the negative impact of chemotherapy on the ovaries, using experimental models, especially in animals. In this review, the various theories concerning ovarian damage induced by chemotherapy will be reviewed and emerging approaches for ovarian protection will be explained.
Collapse
Affiliation(s)
- Ines Sellami
- Department of Reproductive Medicine and Fertility Preservation, Université Paris-Saclay, Assistance publique Hôpitaux de Paris, Antoine Beclere Hospital, 92140, Clamart, France; Université Paris Saclay, Inserm, physiologie et physiopathologie endocrinienne, 94276, Le Kremlin-Bicêtre, France
| | - Isabelle Beau
- Université Paris Saclay, Inserm, physiologie et physiopathologie endocrinienne, 94276, Le Kremlin-Bicêtre, France
| | - Charlotte Sonigo
- Department of Reproductive Medicine and Fertility Preservation, Université Paris-Saclay, Assistance publique Hôpitaux de Paris, Antoine Beclere Hospital, 92140, Clamart, France; Université Paris Saclay, Inserm, physiologie et physiopathologie endocrinienne, 94276, Le Kremlin-Bicêtre, France.
| |
Collapse
|
18
|
Zhou Z, Jin H, Xu J. A gene signature driven by abnormally methylated DEGs was developed for TP53 wild-type ovarian cancer samples by integrative omics analysis of DNA methylation and gene expression data. ANNALS OF TRANSLATIONAL MEDICINE 2023; 11:20. [PMID: 36760264 PMCID: PMC9906212 DOI: 10.21037/atm-22-5764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 12/27/2022] [Indexed: 01/15/2023]
Abstract
Background Integrated omics analysis based on transcriptome and DNA methylation data combined with machine learning methods is very promising for the diagnosis, prognosis, and classification of cancer. In this study, the DNA methylation and gene expression data of ovarian cancer (OC) were analyzed to identify abnormally methylated differentially expressed genes (DEGs), screen potential therapeutic agents for OC, and construct a risk model based on the abnormally methylated DEGs to predict patient prognosis. Methods The gene expression and DNA methylation data of primary OC samples with tumor protein 53 (TP53) wild-type and normal samples were obtained from The Cancer Genome Atlas (TCGA) database. DEGs with aberrant methylation were analyzed by screening the intersection between DEGs and differentially methylated genes (DMGs). We attempted to search for potential drugs targeting DEGs with aberrant methylation by employing a network medicine framework. A gene signature based on the DEGs with aberrant methylation was constructed by regularized least absolute shrinkage and selection operator (LASSO) regression analysis. Results A total of 440 aberrant methylated DEGs were screened. Based on their gene expression profiles and methylation data from different regions, the results of both discriminative pattern recognition analysis and principal component analysis (PCA) showed a significant separation between tumor tissue and healthy ovarian tissue. In total, 126 potential therapeutic drugs were identified for OC by network-based proximity analysis. Five genes were identified in 440 aberrant methylated DEGs, which formed an aberrant methylated DEGs-driven gene signature. This signature could significantly distinguish the different overall survivals (OS) of OC patients and showed better predictive performance in both the training and validation sets. Conclusions In this study, the DNA methylation and gene expression data of OC were analyzed to identify abnormally methylated DEGs and potential therapeutic drugs, and a gene signature based on five aberrant methylation DEGs was constructed, which could better predict the risk of death in patients.
Collapse
Affiliation(s)
- Zhu Zhou
- Gynaecology Department, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Hang Jin
- Gynaecology Department, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Jian Xu
- Reproductive Center, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| |
Collapse
|