1
|
Timme K, González-Alvarez ME, Keating AF. Pre-pubertal obesity compromises ovarian oxidative stress, DNA repair and chemical biotransformation. Toxicol Appl Pharmacol 2024; 489:116981. [PMID: 38838792 DOI: 10.1016/j.taap.2024.116981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/22/2024] [Accepted: 05/25/2024] [Indexed: 06/07/2024]
Abstract
Obesity in adult females impairs fertility by altering oxidative stress, DNA repair and chemical biotransformation. Whether prepubertal obesity results in similar ovarian impacts is under-explored. The objective of this study was to induce obesity in prepubertal female mice and assess puberty onset, follicle number, and abundance of oxidative stress, DNA repair and chemical biotransformation proteins basally and in response to 7,12-dimethylbenz(a)anthracene (DMBA) exposure. DMBA is a polycyclic aromatic hydrocarbon that has been shown to be ovotoxic. Lactating dams (C57BL6J) were fed either a normal rodent containing 3.5% kCal from fat (lean), or a high fat diet comprised of 60% kCal from fat, and 9% kCal from sucrose. The offspring were weaned onto the diet of their dam and exposed at postnatal day 35 to either corn oil or DMBA (1 mg/kg) for 7 d via intraperitoneal injection. Mice on the HFD had reduced (P < 0.05) age at puberty onset as measured by vaginal opening but DMBA did not impact puberty onset. Heart, spleen, kidney, uterus and ovary weight were increased (P < 0.05) by obesity and liver weight was increased (P < 0.05) by DMBA exposure in obese mice. Follicle number was largely unaffected by obesity or DMBA exposure, with the exception of primary follicle number, which were higher (P < 0.05) in lean DMBA exposed and obese control relative to lean control mice. There were also greater numbers (P < 0.05) of corpora lutea in obese relative to lean mice. In lean mice, DMBA exposure reduced (P < 0.05) the level of CYP2E1, EPHX1, GSTP1, BRCA1, and CAT but this DMBA-induced reduction was absent in obese mice. Basally, obesity reduced (P < 0.05) the abundance of CYP2E1, EPHX1, GSTP1, BRCA1, SOD1 and CAT. There was greater (P < 0.05) fibrotic staining in obese DMBA-exposed ovaries and PPP2CA was decreased (P < 0.05) in growing follicles by both obesity and DMBA exposure. Thus, prepubertal obesity alters the capacity of the ovary to respond to DNA damage, ovotoxicant exposure and oxidative stress.
Collapse
Affiliation(s)
- Kelsey Timme
- Department of Animal Science, Iowa State University, Ames, IA, USA
| | | | - Aileen F Keating
- Department of Animal Science, Iowa State University, Ames, IA, USA.
| |
Collapse
|
2
|
Bedrick BS, Kohn TP, Pecker LH, Christianson MS. Fertility preservation for pediatric patients with hemoglobinopathies: Multidisciplinary counseling needed to optimize outcomes. Front Endocrinol (Lausanne) 2022; 13:985525. [PMID: 36353243 PMCID: PMC9638952 DOI: 10.3389/fendo.2022.985525] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 09/30/2022] [Indexed: 01/19/2023] Open
Abstract
Hemoglobinopathies are autosomal recessive disorders that occur when genetic mutations negatively impact the function of hemoglobin. Common hemoglobinopathies that are clinically significant include sickle cell disease, alpha thalassemia, and beta thalassemia. Advancements in disease-modifying and curative treatments for the common hemoglobinopathies over the past thirty years have led to improvements in patient quality of life and longevity for those who are affected. However, the diseases, their treatments and cures pose infertility risks, making fertility preservation counseling and treatment an important part of the contemporary comprehensive patient care. Sickle cell disease negatively impacts both male and female infertility, primarily by testicular failure and decreased ovarian reserve, respectively. Fertility in both males and females with beta thalassemia major are negatively impacted by iron deposition due to chronic blood transfusions. Hematopoietic stem cell transplant (HSCT) is currently the only curative treatment for SCD and transfusion dependent beta thalassemia. Many of the conditioning regimens for HSCT contain chemotherapeutic agents with known gonadotoxicity and whole-body radiation. Although most clinical studies on toxicity and impact of HSCT on long-term health do not evaluate fertility, gonadal failure is common. Male fertility preservation modalities that exist prior to gonadotoxic treatment include sperm banking for pubertal males and testicular cryopreservation for pre-pubertal boys. For female patients, fertility preservation options include oocyte cryopreservation and ovarian tissue cryopreservation. Oocyte cryopreservation requires controlled ovarian hyperstimulation (COH) with ten to fourteen days of intensive monitoring and medication administration. This is feasible once the patient has undergone menarche. Follicular growth is monitored via transvaginal or transabdominal ultrasound, and hormone levels are monitored through frequent blood work. Oocytes are then harvested via a minimally invasive approach under anesthesia. Complications of COH are more common in patients with hemoglobinopathies. Ovarian hyperstimulation syndrome creates a greater risk to patients with underlying vascular, pulmonary, and renal injury, as they may be less able to tolerate fluids shifts. Thus, it is critical to monitor patients undergoing COH closely with close collaboration between the hematology team and the reproductive endocrinology team. Counseling patients and families about future fertility must take into consideration the patient's disease, treatment history, and planned treatment, acknowledging current knowledge gaps.
Collapse
Affiliation(s)
- Bronwyn S. Bedrick
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Taylor P. Kohn
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Lydia H. Pecker
- Department of Medicine, Division of Adult Hematology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mindy S. Christianson
- Department of Gynecology and Obstetrics, Division of Reproductive Endocrinology and Infertility, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
3
|
Kim S, Lee S, Park HT, Song JY, Kim T. Genomic Consideration in Chemotherapy-Induced Ovarian Damage and Fertility Preservation. Genes (Basel) 2021; 12:1525. [PMID: 34680919 PMCID: PMC8535252 DOI: 10.3390/genes12101525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 09/25/2021] [Accepted: 09/25/2021] [Indexed: 11/25/2022] Open
Abstract
Chemotherapy-induced ovarian damage and fertility preservation in young patients with cancer are emerging disciplines. The mechanism of treatment-related gonadal damage provides important information for targeting prevention methods. The genomic aspects of ovarian damage after chemotherapy are not fully understood. Several studies have demonstrated that gene alterations related to follicular apoptosis or accelerated follicle activation are related to ovarian insufficiency and susceptibility to ovarian damage following chemotherapy. This may accelerate follicular apoptosis and follicle reservoir utilization and damage the ovarian stroma via multiple molecular reactions after chemotherapy. This review highlights the importance of genomic considerations in chemotherapy-induced ovarian damage and multidisciplinary oncofertility strategies for providing high-quality care to young female cancer patients.
Collapse
Affiliation(s)
- Seongmin Kim
- Gynecologic Cancer Center, CHA Ilsan Medical Center, CHA University College of Medicine, 1205 Jungang-ro, Ilsandong-gu, Goyang-si 10414, Korea;
| | - Sanghoon Lee
- Department of Obstetrics and Gynecology, Korea University College of Medicine, 73 Inchon-ro, Seongbuk-gu, Seoul 02841, Korea; (H.-T.P.); (J.-Y.S.); (T.K.)
| | - Hyun-Tae Park
- Department of Obstetrics and Gynecology, Korea University College of Medicine, 73 Inchon-ro, Seongbuk-gu, Seoul 02841, Korea; (H.-T.P.); (J.-Y.S.); (T.K.)
| | - Jae-Yun Song
- Department of Obstetrics and Gynecology, Korea University College of Medicine, 73 Inchon-ro, Seongbuk-gu, Seoul 02841, Korea; (H.-T.P.); (J.-Y.S.); (T.K.)
| | - Tak Kim
- Department of Obstetrics and Gynecology, Korea University College of Medicine, 73 Inchon-ro, Seongbuk-gu, Seoul 02841, Korea; (H.-T.P.); (J.-Y.S.); (T.K.)
| |
Collapse
|
4
|
Kim S, Kim SW, Han SJ, Lee S, Park HT, Song JY, Kim T. Molecular Mechanism and Prevention Strategy of Chemotherapy- and Radiotherapy-Induced Ovarian Damage. Int J Mol Sci 2021; 22:ijms22147484. [PMID: 34299104 PMCID: PMC8305189 DOI: 10.3390/ijms22147484] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 07/12/2021] [Indexed: 12/14/2022] Open
Abstract
Fertility preservation is an emerging discipline, which is of substantial clinical value in the care of young patients with cancer. Chemotherapy and radiation may induce ovarian damage in prepubertal girls and young women. Although many studies have explored the mechanisms implicated in ovarian toxicity during cancer treatment, its molecular pathophysiology is not fully understood. Chemotherapy may accelerate follicular apoptosis and follicle reservoir utilization and damage the ovarian stroma via multiple molecular reactions. Oxidative stress and the radiosensitivity of oocytes are the main causes of gonadal damage after radiation treatment. Fertility preservation options can be differentiated by patient age, desire for conception, treatment regimen, socioeconomic status, and treatment duration. This review will help highlight the importance of multidisciplinary oncofertility strategies for providing high-quality care to young female cancer patients.
Collapse
Affiliation(s)
- Seongmin Kim
- Gynecologic Cancer Center, CHA Ilsan Medical Center, CHA University College of Medicine, 1205 Jungang-ro, Ilsandong-gu, Goyang-si 10414, Korea;
| | - Sung-Woo Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea; (S.-W.K.); (S.-J.H.)
| | - Soo-Jin Han
- Department of Obstetrics and Gynecology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea; (S.-W.K.); (S.-J.H.)
| | - Sanghoon Lee
- Department of Obstetrics and Gynecology, Korea University College of Medicine, 73 Inchon-ro, Seongbuk-gu, Seoul 02841, Korea; (H.-T.P.); (J.-Y.S.); (T.K.)
- Correspondence: ; Tel.: +82-2-920-6773
| | - Hyun-Tae Park
- Department of Obstetrics and Gynecology, Korea University College of Medicine, 73 Inchon-ro, Seongbuk-gu, Seoul 02841, Korea; (H.-T.P.); (J.-Y.S.); (T.K.)
| | - Jae-Yun Song
- Department of Obstetrics and Gynecology, Korea University College of Medicine, 73 Inchon-ro, Seongbuk-gu, Seoul 02841, Korea; (H.-T.P.); (J.-Y.S.); (T.K.)
| | - Tak Kim
- Department of Obstetrics and Gynecology, Korea University College of Medicine, 73 Inchon-ro, Seongbuk-gu, Seoul 02841, Korea; (H.-T.P.); (J.-Y.S.); (T.K.)
| |
Collapse
|
5
|
Brancati S, Gozzo L, Longo L, Vitale DC, Russo G, Drago F. Fertility Preservation in Female Pediatric Patients With Cancer: A Clinical and Regulatory Issue. Front Oncol 2021; 11:641450. [PMID: 33796467 PMCID: PMC8008167 DOI: 10.3389/fonc.2021.641450] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 01/29/2021] [Indexed: 12/22/2022] Open
Abstract
Fertility preservation represents one important goal of cancer patients’ management due to the high impact on health and quality of life of survivors. The available preventive measures cannot be performed in all patients and are not feasible in all health-care facilities. Therefore, the pharmacological treatment with GnRHa has become a valuable non-invasive and well-tolerated alternative, especially in those who cannot access to cryopreservation options due to clinical and/or logistic issues. Supporting data demonstrate a significant advantage for the survivors who received GnRHa in the long-term maintenance of ovarian function and preservation of fertility. The prevention of the risk of ovarian failure with GnRHa is a typical off-label use, defined as the administration of a medicinal product not in accordance with the authorized product information. Italy has officially recognized the off-label use of GnRHa in adult women at risk of premature and permanent menopause following chemotherapy. However, fertility preservation still represents an unmet medical need in adolescents who cannot access to other treatment options.
Collapse
Affiliation(s)
- Serena Brancati
- Clinical Pharmacology Unit/Regional Pharmacovigilance Centre, University Hospital of Catania, Catania, Italy
| | - Lucia Gozzo
- Clinical Pharmacology Unit/Regional Pharmacovigilance Centre, University Hospital of Catania, Catania, Italy.,Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Laura Longo
- Clinical Pharmacology Unit/Regional Pharmacovigilance Centre, University Hospital of Catania, Catania, Italy
| | - Daniela Cristina Vitale
- Clinical Pharmacology Unit/Regional Pharmacovigilance Centre, University Hospital of Catania, Catania, Italy
| | - Giovanna Russo
- Pediatric Onco-Hematology, University Hospital of Catania, Catania, Italy
| | - Filippo Drago
- Clinical Pharmacology Unit/Regional Pharmacovigilance Centre, University Hospital of Catania, Catania, Italy.,Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.,Centre for Research and Consultancy in HTA and drug Regulatory Affairs (CERD), University of Catania, Catania, Italy
| |
Collapse
|
6
|
Yoo M, Tanaka T, Konishi H, Tanabe A, Taniguchi K, Komura K, Hayashi M, Ohmichi M. The Protective Effect of Testosterone on the Ovarian Reserve During Cyclophosphamide Treatment. Onco Targets Ther 2020; 13:2987-2995. [PMID: 32308430 PMCID: PMC7152736 DOI: 10.2147/ott.s242703] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/27/2020] [Indexed: 11/23/2022] Open
Abstract
Introduction Cyclophosphamide, which is widely used to treat malignant disease, causes ovarian follicular atresia, which leads to premature ovarian insufficiency. The present study evaluated the protective effect of testosterone in preventing the decline in the ovarian reserve during cyclophosphamide treatment. Methods Using the COV434 human granulosa cell line, the protective effect of testosterone against cyclophosphamide was evaluated by immunocytochemistry, Western blotting and an MTS assay. The follicles in mouse ovaries and serum anti-Mullerian hormone were also assessed to evaluate the effects of testosterone. Results Testosterone suppressed the decrease in cell viability and apoptosis caused by cyclophosphamide treatment in vitro. In vivo, the number of atretic follicles in the mouse ovary was significantly lower in the testosterone plus cyclophosphamide group than in the cyclophosphamide alone group (p=0.03). The serum anti-Mullerian hormone was significantly higher in the testosterone plus cyclophosphamide group than in the cyclophosphamide alone group (16.2 [9.7–22.6]) vs 11.2 [8.9–12.1], p<0.01). The rate of cleaved Caspase-3 expression in the testosterone plus cyclophosphamide group was lower than that in the cyclophosphamide alone group (28.4% vs 48.6%, p=0.03). Conclusion These findings indicated that testosterone has the potential to prevent ovarian damage induced by cyclophosphamide by protecting granulosa cells from cyclophosphamide-induced apoptosis.
Collapse
Affiliation(s)
- Masae Yoo
- Department of Obstetrics and Gynecology
| | - Tomohito Tanaka
- Department of Obstetrics and Gynecology.,Translational Research Program, Osaka Medical College, Takatsuki, Japan
| | | | | | - Kohei Taniguchi
- Translational Research Program, Osaka Medical College, Takatsuki, Japan
| | - Kazumasa Komura
- Translational Research Program, Osaka Medical College, Takatsuki, Japan
| | | | | |
Collapse
|
7
|
Akahori T, Woods DC, Tilly JL. Female Fertility Preservation through Stem Cell-based Ovarian Tissue Reconstitution In Vitro and Ovarian Regeneration In Vivo. CLINICAL MEDICINE INSIGHTS. REPRODUCTIVE HEALTH 2019; 13:1179558119848007. [PMID: 31191070 PMCID: PMC6540489 DOI: 10.1177/1179558119848007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 03/27/2019] [Indexed: 12/13/2022]
Abstract
Historically, approaches designed to offer women diagnosed with cancer the prospects of having a genetically matched child after completion of their cytotoxic treatments focused on the existing oocyte population as the sole resource available for clinical management of infertility. In this regard, elective oocyte and embryo cryopreservation, as well as autologous ovarian cortical tissue grafting posttreatment, have gained widespread support as options for young girls and reproductive-age women who are faced with cancer to consider. In addition, the use of ovarian protective therapies, including gonadotropin-releasing hormone agonists and sphingosine-1-phosphate analogs, has been put forth as an alternative way to preserve fertility by shielding existing oocytes in the ovaries in vivo from the side-effect damage caused by radiotherapy and many chemotherapeutic regimens. This viewpoint changed with the publication of now numerous reports that adult ovaries of many mammalian species, including humans, contain a rare population of oocyte-producing germ cells-referred to as female germline or oogonial stem cells (OSCs). This new line of study has fueled research into the prospects of generating new oocytes, rather than working with existing oocytes, as a novel approach to sustain or restore fertility in female cancer survivors. Here, we overview the history of work from laboratories around the world focused on improving our understanding of the biology of OSCs and how these cells may be used to reconstitute "artificial" ovarian tissue in vitro or to regenerate damaged ovarian tissue in vivo as future fertility-preservation options.
Collapse
Affiliation(s)
- Taichi Akahori
- Laboratory for Aging and Infertility Research, Department of Biology, Northeastern University, Boston, MA, USA.,On leave from the Department of Obstetrics and Gynecology, Saitama Medical Center, Saitama Medical University, Saitama, Japan
| | - Dori C Woods
- Laboratory for Aging and Infertility Research, Department of Biology, Northeastern University, Boston, MA, USA
| | - Jonathan L Tilly
- Laboratory for Aging and Infertility Research, Department of Biology, Northeastern University, Boston, MA, USA
| |
Collapse
|
8
|
Wang Y, Chen H, Ju K, Kopp MF, Johnson SB, Farrell KK, Yuan G, Ataman LM, Zheng W, Woodruff TK, Xiao S. Female oncofertility attitude and knowledge: a survey of reproductive health professionals in Shanghai, China. Future Oncol 2019; 15:371-379. [PMID: 30620219 PMCID: PMC6462868 DOI: 10.2217/fon-2018-0428] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 09/21/2018] [Indexed: 12/13/2022] Open
Abstract
AIM To evaluate the female oncofertility attitude and knowledge of reproductive health professionals in China. METHODS An online survey was distributed to reproductive health professionals in Shanghai, China. RESULTS Female professionals were more likely to consider that cancer patients would want to preserve their fertility. Participants with higher educational background tended to have a more positive attitude toward oncofertility. The majority of the participants (71.0%) obtained a fair or low level of oncofertility knowledge, and only 25.3% of them received scores at the 'good knowledge' level. CONCLUSION There are significant gaps in the current oncofertility knowledge among reproductive health professionals in China, suggesting an urgent, unmet need for establishing an interdisciplinary fertility preservation training and service system.
Collapse
Affiliation(s)
- Yuqing Wang
- Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
- School of Public Health, Nanjing Medical University, Nanjing, 21009, PR China
| | - Hanyi Chen
- Science Research & Information Management Section, Shanghai Pudong New Area Center for Disease Control & Prevention, Shanghai, 200136, PR China
- Pudong Institute of Preventive Medicine, Fudan University, Shanghai, 20032, PR China
| | - Ke Ju
- Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
- School of Public Health, Nanjing Medical University, Nanjing, 21009, PR China
| | - Megan F Kopp
- Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - Sarah B Johnson
- Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - Katherine K Farrell
- Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - Gehui Yuan
- Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
- School of Public Health, Nanjing Medical University, Nanjing, 21009, PR China
| | - Lauren M Ataman
- Department of Obstetrics & Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Weiwei Zheng
- Key Laboratory of Public Health Safety, Ministry of Education, Shanghai, 20032, PR China
- Institution for Water Pollution & Health Research, School of Public Health, Fudan University, Shanghai, 20032, PR China
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 20032, PR China
| | - Teresa K Woodruff
- Department of Obstetrics & Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Shuo Xiao
- Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| |
Collapse
|
9
|
Smith KL, Gracia C, Sokalska A, Moore H. Advances in Fertility Preservation for Young Women With Cancer. Am Soc Clin Oncol Educ Book 2018; 38:27-37. [PMID: 30231357 DOI: 10.1200/edbk_208301] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Female patients of reproductive age with cancer often require treatment that can compromise their future fertility. Treatment-related infertility is an important cancer survivorship issue and is associated with depression and diminished quality of life. Recent advances in reproductive health care provide the opportunity to preserve fertility prior to the initiation of cancer therapy. Clinical guidelines recommend that oncology providers counsel patients about the risk of treatment-related infertility and fertility preservation options, and that they refer those who are interested in fertility preservation to fertility specialists. Guidelines endorse the use of assisted reproductive techniques (ART) provided by reproductive endocrinologists to preserve fertility in young female patients with cancer. In addition, ovarian suppression with gonadotropin-releasing hormone (GnRH) agonists may be considered for ovarian protection during chemotherapy. This article reviews currently available and emerging ART for fertility preservation in female patients of reproductive age with cancer and current data supporting the use of ovarian suppression for ovarian protection during chemotherapy in this population. We also review the uptake of fertility services and discuss barriers to fertility preservation in female patients of reproductive age with cancer.
Collapse
Affiliation(s)
- Karen Lisa Smith
- From The Johns Hopkins University School of Medicine, Baltimore, MD; Hospital of the University of Pennsylvania, Philadelphia, PA; Cleveland Clinic, Cleveland, OH
| | - Clarisa Gracia
- From The Johns Hopkins University School of Medicine, Baltimore, MD; Hospital of the University of Pennsylvania, Philadelphia, PA; Cleveland Clinic, Cleveland, OH
| | - Anna Sokalska
- From The Johns Hopkins University School of Medicine, Baltimore, MD; Hospital of the University of Pennsylvania, Philadelphia, PA; Cleveland Clinic, Cleveland, OH
| | - Halle Moore
- From The Johns Hopkins University School of Medicine, Baltimore, MD; Hospital of the University of Pennsylvania, Philadelphia, PA; Cleveland Clinic, Cleveland, OH
| |
Collapse
|
10
|
Li Z, Liu H, Lang J, Zhang G, He Z. Effects of cisplatin on surgically induced endometriosis in a rat model. Oncol Lett 2018; 16:5282-5290. [PMID: 30250597 PMCID: PMC6144798 DOI: 10.3892/ol.2018.9275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 05/22/2018] [Indexed: 12/19/2022] Open
Abstract
Research has strongly suggested that the features of endometriosis serve as a precursor lesion of ovarian cancer. Cisplatin (CDDP) is the preferred drug against these cancer types. The present study investigated the effects of CDDP on surgically induced endometriosis in a rat model. Endometriosis was surgically induced by the autologous transplantation of endometrial tissue. A total of 36 model rats were randomly divided into three groups. The rats in Group 1 (control group, n=12) received no medication. The rats in Group 2 (n=12) and Group 3 (n=12) were administered 35 mg/m2 CDDP and 70 mg/m2 CDDP, respectively, every four days. All rats were treated for a total of 24 days. The growth and histologic scores of the implants were calculated. The expression of protein markers, including vascular endothelial growth factor (VEGF), aromatase P450 (P450arom), transforming growth factor-β (TGF-β) and matrix metalloproteinase (MMP)-2, were assessed using immunohistochemistry, an enzyme-linked immunosorbent assay and western blot analysis. Following CDDP treatment, the mean implant sizes were significantly reduced in Groups 2 and 3 compared with the control group (P=0.01). The mean histologic scores were also significantly lower in Groups 2 and 3. Furthermore, the protein expression of VEGF, P450arom, TGF-β and MMP-2 was significantly lower in Groups 2 and 3 when compared with the control group. A loss of hair was observed in 4 rats, which only occurred in Group 3. A dose-dependent effect was observed in the two CDDP-treated groups. In conclusion, the expression of proliferation- and angiogenesis-associated proteins was significantly lower following treatment with CDDP. CDDP caused a significant regression in the size of the endometriotic implants and induced atrophy of these lesions in rats.
Collapse
Affiliation(s)
- Zhanfei Li
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China.,Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Beijing 100005, P.R. China
| | - Huibing Liu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jinghe Lang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Beijing 100005, P.R. China
| | - Guorui Zhang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Beijing 100005, P.R. China
| | - Zhengxing He
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Beijing 100005, P.R. China
| |
Collapse
|
11
|
Zhou X, He W, Zhu R, Zheng Y. Impact of GnRHa pretreatment on the autotransplatation efficacy of cytopreserved rat ovarian tissue. Biomed Pharmacother 2016; 86:136-142. [PMID: 27960135 DOI: 10.1016/j.biopha.2016.11.070] [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: 07/17/2016] [Revised: 11/12/2016] [Accepted: 11/16/2016] [Indexed: 11/28/2022] Open
Abstract
Pre-therapeutic cryopreservation of ovarian tissue and subsequent transplantation after disease remission has recently emerged as an option to restore endocrinal function and preserve fertility for patients subjected to gonadotoxic treatments such as chemotherapy. However, the relatively low survival rate of follicles after grafting the frozen-thawed ovarian tissue largely limited the application of this technique. Gonadotropin-releasing hormone analogues (GnRHa), owing to their endocrinal regulatory effect, have been successfully used for fertility preservation against gonadotoxic conditions such as cytotoxic agents based anti-cancer therapy. In this study, we evaluated the potential protective effect of precedent GnRHa treatment before cryopreservation on freezing-thawing related follicular damages using a rat autoxenograft model. We have observed that GnRHa significantly increases the fraction of follicles with normal morphology, while presenting negnigible effect on the physiological recovery of the grafted follicular tissue, as demonstrated by the estrous cycle, folliculogenesis and post-autograft vascularization. Our data implicates that GnRHa pretreatment may effectively increase the efficacy of cryopreservation and the subsequent successful rate of transplantation.
Collapse
Affiliation(s)
- Xinhui Zhou
- Department of Gynecology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Weihua He
- Department of Gynecology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Rong Zhu
- Department of Gynecology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Yulong Zheng
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China.
| |
Collapse
|
12
|
Rossi V, Lispi M, Longobardi S, Mattei M, Di Rella F, Salustri A, De Felici M, Klinger FG. LH prevents cisplatin-induced apoptosis in oocytes and preserves female fertility in mouse. Cell Death Differ 2016; 24:72-82. [PMID: 27689876 PMCID: PMC5260508 DOI: 10.1038/cdd.2016.97] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 07/18/2016] [Accepted: 07/20/2016] [Indexed: 01/09/2023] Open
Abstract
Premature ovarian failure and female infertility are frequent side effects of anticancer therapies, owing to the extreme sensitivity of the ovarian reserve oocytes to the damaging effects of irradiation and chemotherapy on DNA. We report here a robust protective effect of luteinizing hormone (LH) on the primordial follicle pool of prepubertal ovaries against the cisplatin (Cs)-induced apoptosis. In vitro LH treatment of prepubertal ovarian fragments generated anti-apoptotic signals by a subset of ovarian somatic cells expressing LH receptor (LHR) through cAMP/PKA and Akt pathways. Such signals, reducing the oocyte level of pro-apoptotic TAp63 protein and favoring the repair of the Cs-damaged DNA in the oocytes, prevented their apoptosis. Noteworthy, in vivo administration to prepubertal female mice of a single dose of LH together with Cs inhibited the depletion of the primordial follicle reserve caused by the drug and preserved their fertility in reproductive age, preventing significant alteration in the number of pregnancy and of delivered pups. In conclusion, these findings establish a novel ovoprotective role for LH and further support the very attracting prospective to use physiological 'fertoprotective' approaches for preventing premature infertility and risks linked to precocious menopause in young patients who survived cancer after chemotherapy.
Collapse
Affiliation(s)
- Valerio Rossi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Monica Lispi
- Medical Affair Department Fertility TA, Merck-Serono SAS, Rome, Italy
| | | | - Maurizio Mattei
- STA, Department of Biology, University of Rome 'Tor Vergata', Rome, Italy
| | - Francesca Di Rella
- UOC Oncologia Medica Senologica, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione G. Pascale, Naples, Italy
| | - Antonietta Salustri
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Massimo De Felici
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Francesca G Klinger
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
13
|
Biswas B, Bakhshi S. Management of Ewing sarcoma family of tumors: Current scenario and unmet need. World J Orthop 2016; 7:527-538. [PMID: 27672565 PMCID: PMC5027007 DOI: 10.5312/wjo.v7.i9.527] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 06/21/2016] [Accepted: 07/13/2016] [Indexed: 02/06/2023] Open
Abstract
Ewing sarcoma family tumors (ESFT) are heterogeneous, aggressive group of disease with peak incidence in adolescent and young adults. The outcome has been improved dramatically from 10% with surgery and radiotherapy alone to 65%-70% now, in localized disease, with the introduction of chemotherapy. Chemotherapy regimen evolved from single agent to multiagent with effort of many cooperative clinical trials over decades. The usual treatment protocol include introduction of multi-agent chemotherapy in neoadjuvant setting to eradicate systemic disease with timely incorporation of surgery and/or radiotherapy as local treatment modality and further adjuvant chemotherapy to prevent recurrence. Risk adapted chemotherapy in neoadjuvant and adjuvant setting along with radiotherapy has been used in many international collaborative trials and has resulted in improved outcome, more so in patients with localized disease. The role of high dose chemotherapy with stem cell rescue is still debatable. The outcome of patients with metastatic disease is dismal with long term outcome ranges from 20%-40% depending on the sites of metastasis and intensity of treatment. There is a huge unmet need to improve outcome further, more so in metastatic setting. Novel therapy targeting the molecular pathways and pathogenesis of ESFT is very much required. Here we have discussed the current standard of management in patients with ESFT, investigational targeted or novel therapies along with future promises.
Collapse
|
14
|
Metallinou C, Asimakopoulos B, Schröer A, Nikolettos N. Gonadotropin-Releasing Hormone in the Ovary. Reprod Sci 2016; 14:737-49. [DOI: 10.1177/1933719107310707] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Chryssa Metallinou
- Department of Physiology, School of Medicine, Democritus University of Thrace, Dragana, Greece
| | - Byron Asimakopoulos
- Department of Physiology, School of Medicine, Democritus University of Thrace, Dragana, Greece
| | - Andreas Schröer
- Department of Obstetrics/Gynecology, University Klinik of Schleswig-Holstein, Lübeck, Germany
| | - Nikos Nikolettos
- Department of Physiology, School of Medicine, Democritus University of Thrace, Dragana, Greece
| |
Collapse
|
15
|
Blumenfeld Z, Avivi I, Ritter M, Rowe JM. Preservation of Fertility and Ovarian Function and Minimizing Chemotherapy-Induced Gonadotoxicity in Young Women. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/107155769900600502] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Z. Blumenfeld
- Reproductive Endocrinology and Infertility Section, Departments of Obstetrics and Gynecology and Hematology, Rambam Medical Center, Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | | | | | - J. M. Rowe
- Reproductive Endocrinology and Infertility Section, Departments of Obstetrics and Gynecology and Hematology, Rambam Medical Center, Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
16
|
Silva CAA, Brunner HI. Review: Gonadal functioning and preservation of reproductive fitness with juvenile systemic lupus erythematosus. Lupus 2016; 16:593-9. [PMID: 17711894 DOI: 10.1177/0961203307077538] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Increased survival of children with juvenile systemic lupus erythematosus (jSLE) and improved prognosis have led to a change in the long-term health issues arising for jSLE patients. Preservation of gonadal functioning and fertility are of increasing importance for young adults with jSLE. Events during childhood, such as exposure to alkylating agents, may compromise the reproductive potential. Even in the absence of gonadotoxic therapies, fertility may be decreased through organs specific involvement with jSLE. Strategies to preserve the reproductive potential of girl and boys with jSLE are discussed. Lupus (2007) 16, 593—599.
Collapse
Affiliation(s)
- C A A Silva
- Pediatric Rheumatology Unit, Children's Hospital, and Division of Rheumatology of University of São Paulo, São Paulo, Brazil
| | | |
Collapse
|
17
|
Phelan R, Mann E, Napurski C, DeFor TE, Petryk A, Miller WP, Wagner JE, Verneris MR, Smith AR. Ovarian function after hematopoietic cell transplantation: a descriptive study following the use of GnRH agonists for myeloablative conditioning and observation only for reduced-intensity conditioning. Bone Marrow Transplant 2016; 51:1369-1375. [PMID: 27272448 DOI: 10.1038/bmt.2016.150] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 04/21/2016] [Accepted: 04/22/2016] [Indexed: 12/11/2022]
Abstract
Gonadal failure is a health and quality-of-life concern in hematopoietic cell transplant (HCT) survivors. While ovarian dysfunction is nearly universal following myeloablative (MA) conditioning, the risk is unclear after reduced-intensity conditioning (RIC). Gonadotropin-releasing hormone agonists decrease ovarian failure rates following conventional chemotherapy, but little is known about its effectiveness with HCT. We investigated the impact of leuprolide on ovarian function after MA conditioning and monitored ovarian function after RIC in this descriptive pilot study. Post-menarchal females <50 years undergoing HCT with adequate baseline ovarian function (follicle-stimulating hormone (FSH) level <40 mIU/mL and normal menstruation) were eligible. Prior to MA conditioning, leuprolide was administered. Those undergoing RIC were observed. FSH was measured at various time points. Seventeen women aged 12-45 years were evaluated (7 in the intervention group and 10 in the observation group). Compared to the historical high rate of ovarian failure after MA conditioning, 3 of 7 evaluable Lupron recipients had ovarian failure at a median of 703 days post transplant. Ovarian failure occurred in 1 of 10 recipients of RIC at a median follow-up of 901 days. In conclusion, leuprolide may protect ovarian function after MA conditioning. Additionally, RIC with cyclophosphamide, fludarabine and low-dose TBI has a low risk of ovarian failure.
Collapse
Affiliation(s)
- R Phelan
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - E Mann
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - C Napurski
- Cancer Survivorship Program, University of Minnesota, Minneapolis, MN, USA
| | - T E DeFor
- BMT Research Program, University of Minnesota, Minneapolis, MN, USA
| | - A Petryk
- Division of Pediatric Endocrinology, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - W P Miller
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - J E Wagner
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - M R Verneris
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - A R Smith
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
18
|
Lee PA, Houk CP. Gonadotropin-releasing hormone analog therapy for central precocious puberty and other childhood disorders affecting growth and puberty. ACTA ACUST UNITED AC 2016; 5:287-96. [PMID: 17002488 DOI: 10.2165/00024677-200605050-00003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Gonadotropin-releasing hormone (GnRH) analog therapy relies primarily on the ability of these compounds to bind to and modulate GnRH-receptor activity. GnRH analogs have been used in pediatric patients where endogenous gonadotropin release is undesirable or potentially harmful, such as in: (i) patients with central precocious puberty (CPP); (ii) healthy short children where pubertal delay would provide an opportunity to supplement pre-pubertal linear growth; and (iii) children with malignancies and other disorders where treatment requires the use of gonadotoxic compounds. In the first two groups of patients, GnRH agonists may be used alone or in conjunction with somatropin (growth hormone [GH]) to prevent early skeletal maturation and increase the subsequent adult height, while in the latter case, GnRH agonists are used alone or in conjunction with GnRH antagonists in an attempt to preserve gonadal function.In children and adolescents with CPP, timely use of GnRH agonists alone can result in an adult height within the genetic potential of the individual (target height); however, minimal height is gained when GnRH agonist therapy is commenced after a marked advancement of skeletal age. This provides the rationale for combined therapy with GnRH agonists and somatropin in such patients, and studies have shown improved growth with this approach compared with GnRH agonists alone. Combination therapy with GnRH agonists and somatropin has also been shown to increase adult heights to a greater extent than GnRH agonists alone in pediatric patients with concomitant CPP and GH deficiency, those with idiopathic short stature, and those born small for gestational age; however, such combination therapy has shown no increased benefit over somatropin alone in pediatric patients with GH deficiency. Limited results in children and adolescents with congenital adrenal hyperplasia and chronic primary hypothyroidism have also shown increased growth rates, while no growth benefit was seen in pediatric renal transplant recipients.GnRH analogs also have potential as gonadoprotective agents; studies of GnRH agonists used alone and in combination with GnRH antagonists in women undergoing cytotoxic therapy have shown increased preservation of reproductive potential in patients who were receiving GnRH analog therapy versus those who were not.The adverse effects of GnRH analogs mainly consist of menopausal-like complaints. Increases in bodyweight and body mass index in children receiving GnRH agonist therapy have been shown; however, these increases do not persist after discontinuation of therapy. Adult bone mineral density and fertility are also not adversely affected by childhood GnRH agonist therapy.GnRH analog therapy appears to be both well tolerated and effective in pediatric patients, as it allows the preservation or improvement of adult height, and shows no longstanding negative effects on body composition, bone density, reproductive function, or endocrine physiology. These agents may also be useful for preservation of gonadal function in children and adolescents undergoing cytotoxic therapy.
Collapse
Affiliation(s)
- Peter A Lee
- Department of Pediatrics, Penn State College of Medicine, The Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | | |
Collapse
|
19
|
Aihara T, Toyama T, Takahashi M, Yamamoto Y, Hara F, Akabane H, Fujisawa T, Ishikawa T, Nagai S, Nakamura R, Tsurutani J, Ito Y, Mukai H. The Japanese Breast Cancer Society Clinical Practice Guideline for systemic treatment of breast cancer, 2015 edition. Breast Cancer 2016; 23:329-42. [DOI: 10.1007/s12282-016-0670-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 12/18/2015] [Indexed: 02/07/2023]
|
20
|
Gris-Martínez JM, Trillo-Urrutia L, Gómez-Cabeza JJ, Encabo-Duró G. Efecto protector de los análogos de la GnRH sobre la capacidad reproductiva de las mujeres en edad fértil con neoplasia o enfermedad autoinmunitaria tratadas con fármacos citotóxicos. Resultado final de un ensayo clínico fase II. Med Clin (Barc) 2016; 146:97-103. [DOI: 10.1016/j.medcli.2015.04.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 04/26/2015] [Accepted: 04/30/2015] [Indexed: 12/24/2022]
|
21
|
Kanter M, Sapmaz-Metin M, Serez B. Effects of GnRHa on early embryonic development in mice receiving cyclophosphamide. Arch Gynecol Obstet 2015; 293:203-209. [DOI: 10.1007/s00404-015-3831-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 07/27/2015] [Indexed: 10/23/2022]
|
22
|
GNRH agonists and antagonists in rescue for cyclophosphamide-induced ovarian damage: friend or foe? Arch Gynecol Obstet 2014; 291:1403-10. [DOI: 10.1007/s00404-014-3564-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 11/25/2014] [Indexed: 10/24/2022]
|
23
|
The feasibility and efficacy of gonadotropin-releasing hormone agonists for prevention of chemotherapy induced ovarian failure in patient with gynecological malignancies. Obstet Gynecol Sci 2014; 57:478-83. [PMID: 25469336 PMCID: PMC4245341 DOI: 10.5468/ogs.2014.57.6.478] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 05/21/2014] [Accepted: 06/13/2014] [Indexed: 11/23/2022] Open
Abstract
Objective To assess the effects of a gonadotropin-releasing hormone agonist (GnRH-a) depot (Leuprolide acetate) in women with gynecologic cancer receiving chemotherapy while taking a continuous add-back on the prevention of premature ovarian failure. Methods Fourteen premenopausal patients with gynecological malignancies who had undergone conservation of ovaries surgery received a GnRH-a depot plus add-back until chemotherapy was completed. Four weeks thereafter, a hormonal profile (follicle stimulating hormone) was measured. Results The mean follicle stimulating hormone level was 15.8 IU/L. All patients exhibited a restoration of ovarian failure during follow-up. One patient became pregnant during the follow-up period. Conclusion In the short term, GnRH-a appears to protect ovarian function and ability to achieve pregnancy following chemotherapy. The result of our study needs further elucidation in a large randomized controlled trial.
Collapse
|
24
|
Muñoz E, Portela S, Remohi J, Pellicer A. Pregnancy in patients with premature ovarian failure. ACTA ACUST UNITED AC 2014. [DOI: 10.1586/17474108.2.6.711] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
25
|
Rody A, Loibl S, von Minckwitz G, Kaufmann M. Use of goserelin in the treatment of breast cancer. Expert Rev Anticancer Ther 2014; 5:591-604. [PMID: 16111461 DOI: 10.1586/14737140.5.4.591] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Gonadotropin-releasing hormone analogs are, alongside tamoxifen, one of the most commonly used drugs in the treatment of pre-/perimenopausal endocrine-responsive breast cancer. Goserelin, as a principal agent of this class of drugs, is mainly investigated in clinical trials. The indirect comparison of goserelin with tamoxifen as a single drug in the adjuvant setting showed similar efficacy. Furthermore, goserelin is as effective as cyclophosphamide, methotrexate and 5-fluorouracil chemotherapy, and total endocrine blockade as a combination of gonadotropin-releasing hormone analog and tamoxifen showed a comparable benefit with anthracycline-containing adjuvant chemotherapy. Goserelin administered after cessation of chemotherapy leads to a further improvement and may be equieffective as tamoxifen or a combination of both. Data concerning taxane-based and dose-dense chemotherapy as well as combination of gonadotropin-releasing hormone analogs with third-generation aromatase inhibitors are still lacking (ongoing suppression of ovarian function, tamoxifen and exemestane, and premenopausal endocrine-responsive chemotherapy trials). Moreover, duration of therapy with gonadotropin-releasing hormone analogs (2-3 years or longer) is still a matter of debate. Palliative endocrine treatment is standard in the first-line therapy of patients without life-threatening disease and endocrine-responsive breast cancer. Treatment decisions depend upon adjuvant endocrine pretreatment. Clinical data regarding ovarian protection by synchronous use of gonadotropin-releasing hormone in young breast cancer patients receiving chemotherapy are incoherent.
Collapse
Affiliation(s)
- Achim Rody
- Department of Obstetrics and Gynecology, JW Goethe-University, Theodor-Stern-Kai 7, D-60590 Frankfurt, Germany.
| | | | | | | |
Collapse
|
26
|
Torrealday S, Lalioti MD, Guzeloglu-Kayisli O, Seli E. Characterization of the gonadotropin releasing hormone receptor (GnRHR) expression and activity in the female mouse ovary. Endocrinology 2013; 154:3877-87. [PMID: 23913446 PMCID: PMC3776864 DOI: 10.1210/en.2013-1341] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
GnRH agonists (GnRHa) are increasingly used for fertility preservation in women undergoing gonadotoxic chemotherapy. However, the protective mechanisms of action for these compounds have not yet been elucidated. In this study, we aimed to determine whether GnRHa have a direct effect on ovarian granulosa cells. GnRH receptor (GnRHR) expression was determined in mouse somatic and gonadal tissues including granulosa/cumulus cells and oocytes using quantitative RT-PCR and immunohistochemistry. Granulosa cells were isolated from mouse ovaries primed with pregnant mare serum gonadotropin. Response to GnRHa in cultured granulosa cells was assessed by determining the increase of intracellular cAMP and by assessing phosphorylation of downstream mediators of GnRH signaling: ERK and p38. To measure intracellular cAMP in our system, the cells were transfected with a cAMP-responsive luciferase reporter plasmid and stimulated with GnRHa. For all experiments, pituitary tissue and/or the αT3-1 mouse pituitary cell line were used as controls. GnRHR mRNA and protein were detected in mouse ovaries, granulosa/cumulus cells, and oocytes. After GnRHa stimulation at various time intervals, we were unable to detect a cAMP increase or activation of the ERK or p38 signaling pathway in cultured primary mouse granulosa cells, whereas activation was detected in the control αT3-1 mouse pituitary cells. In this study, we have not detected activation of the canonical GnRH signaling pathways in mouse ovarian somatic cells. Our findings suggest that the mechanism of action of GnRHa in the ovary is either below the detection level of our experimental design or is different from that in the pituitary.
Collapse
Affiliation(s)
- Saioa Torrealday
- MD, Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, 310 Cedar Street, LSOG 304B, New Haven, Connecticut 06520-8063.
| | | | | | | |
Collapse
|
27
|
Song G, Gao H, Yuan Z. Effect of leuprolide acetate on ovarian function after cyclophosphamide-doxorubicin-based chemotherapy in premenopausal patients with breast cancer: results from a phase II randomized trial. Med Oncol 2013; 30:667. [PMID: 23904400 DOI: 10.1007/s12032-013-0667-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 07/12/2013] [Indexed: 01/02/2023]
Abstract
Previous studies provided inconclusive evidence for the effectiveness of gonadotropin-releasing hormone analogue on ovarian function protection against chemotherapy-induced genotoxicity in premenopausal patients. This study was designed to examine the efficacy of leuprolide acetate on ovarian function preservation in patients with breast cancer. A total of 220 patients were recruited in this prospective clinical trial and were assigned randomly to receive cyclophosphamide-doxorubicin-based chemotherapy only or chemotherapy plus leuprolide acetate. Resumption of menses or premenopausal levels of both follicle-stimulating hormone (FSH) and estradiol (E2) within 12 months after the end of chemotherapy were considered as effective ovarian preservation. A total of 183 patients were considered evaluable (94 in chemotherapy-only group and 89 in chemotherapy plus leuprolide acetate group). At the end of follow-up, 27 patients in chemotherapy group and 15 in chemotherapy plus leuprolide acetate group resumed menses; seven patients in chemotherapy group and 14 in chemotherapy plus leuprolide acetate group restored premenopausal levels of FSH and E2. The median time to resume menses was 9.2 months for patients in chemotherapy plus leuprolide acetate group and was not reached in chemotherapy-only group. In addition, our results demonstrated that age and chemotherapy doses made no significant difference in the occurrence of premature menopause. The leuprolide acetate treatment simultaneously with cyclophosphamide-doxorubicin-based chemotherapy reduced the risk of developing premature menopause in premenopausal patients with breast cancer.
Collapse
Affiliation(s)
- Guiping Song
- Department of Pharmacy, Jiangyin Hospital Affiliated to Nanjing University of Traditional Chinese Medicine, Jiangyin, Jiangsu, China.
| | | | | |
Collapse
|
28
|
Gadducci A, Tana R, Sergiampietri C, Guiggi I. Fertility outcome of breast cancer and Hodgkin's lymphoma female survivors: a growing clinical challenge for gynecologists and oncologists. Gynecol Endocrinol 2013; 29:729-34. [PMID: 23751054 DOI: 10.3109/09513590.2013.797402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The issue of taking into consideration future fertility in young women with breast cancer and Hodgkin's lymphoma [HL] will become more and more common and represent a growing clinical challenge for gynecologists and oncologists. The present paper will review literature data on the attempts of preventing chemotherapy-induced ovarian damage in these women and on their fertility outcome. Gonadotropin-releasing hormone [Gn-RH] agonists have been widely investigated as agents able to prevent ovarian failure in animal models and in humans. The majority of the studies on women with breast cancer and HL have shown a protective effect of Gn-RH agonists. A recent meta-analysis of five randomized trials, including 528 premenopausal breast cancer patients, revealed that relative risk [RR] of developing premature ovarian failure within one year was 0.40 (95% confidence interval [CI] = 0.21-0.75) for the women who received Gn-RH agonists with chemotherapy compared to those who received chemotherapy alone. However, the concurrent administration of Gn-RH agonists during chemotherapy appeared to have no effect on spontaneous pregnancy rates. Limited information are available about pregnancies in breast cancer and HL survivors, but the current literature appears to show no apparent increase in pregnancy complications, spontaneous abortions, or congenital abnormalities compared to general obstetric population.
Collapse
Affiliation(s)
- Angiolo Gadducci
- Department of Clinical and Experimental Medicine, Division of Gynecology and Obstetrics, University of Pisa, Italy.
| | | | | | | |
Collapse
|
29
|
Katz D, Mazeh H, Meirovitz A, Uziely B, Rottenberg Y, Allweis TM, Revel A, Peretz T. Women's post-chemotherapy parity is affected by offspring number and marital status. Breast 2013; 22:362-6. [DOI: 10.1016/j.breast.2012.09.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Revised: 09/04/2012] [Accepted: 09/23/2012] [Indexed: 10/27/2022] Open
|
30
|
Li X, Kang X, Deng Q, Cai J, Wang Z. Combination of a GnRH agonist with an antagonist prevents flare-up effects and protects primordial ovarian follicles in the rat ovary from cisplatin-induced toxicity: a controlled experimental animal study. Reprod Biol Endocrinol 2013; 11:16. [PMID: 23452939 PMCID: PMC3598983 DOI: 10.1186/1477-7827-11-16] [Citation(s) in RCA: 32] [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: 07/16/2012] [Accepted: 01/31/2013] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND With the continuous improvement of surgery and chemotherapeutic treatments, many tumour patients increasingly achieve long-term survival and can even be completely cured. However, platinum-containing drugs, which are widely used to treat a variety of types of cancer, cause menstrual disorders and ovarian failure, which in turn lead to infertility. Thus far, gonadotropin releasing hormone (GnRH) agonist (GnRHa) and antagonist (GnRHant) are reported to act as protective agents of the ovary in chemotherapy through the inhibition of the female gonadal axis. Nevertheless, they both have disadvantages that limit their use. GnRHa causes a flare-up effect during the first week after administration, and no long-acting GnRHant agent is available. GnRHa combined with GnRHant may prevent the flare-up effect of GnRHa and rapidly inhibit the female gonadal axis. Several clinical studies with small sample sizes have reported controversial conclusions. In this strictly controlled animal study, we investigated the advantages of combination treatment with GnRHa and GnRHant. METHODS Rats aged 12 weeks were divided into six groups: Control, cisplatin (CDDP), GnRHa, GnRHant, Combination (sht, short-term) and Combination (lng, long-term) of GnRHa and GnRHant. The last four groups received Triptorelin (1 mg/kg·d, for 14 days), Cetrorelix (0.5 mg/kg·d, for 10 days), a combination of Triptorelin (1 mg/kg·d, for 10 days) and Cetrorelix (0.5 mg/kg·d, for 10 days) in the long-term group and for 3 days in the short-term group. The Control and CDDP groups received saline (1 ml/kg·d, for 10 day). Then, all groups apart from the Control group received cisplatin (1 mg/kg·d, for 10 days), and the Control group received another 10 days of saline as described above. Blood samples were collected to detect the serum levels of E2, LH and FSH. Observation of oestrous cyclicity was also performed after drug administration. Finally, bilateral ovaries were collected for histological study and follicle counting. RESULTS We observed a flare-up effect in rats treated with GnRHa, but not in any of the combination groups. The percentage of normal cyclicity increased from 0% in the CDDP group to 25.0%, 33.3%, 66.7% and 41.7%, in the GnRHa, GnRHant, combination (lng) and combination (sht) groups, respectively. Pretreatment with GnRHa, GnRHant and combination (lng) significantly protected the primordial follicles from destruction by preserving 57.6%, 63.4%, 87.1% and 60.4% of the follicles, respectively. CONCLUSIONS The combination of a GnRH agonist with antagonist completely prevented the flare-up effect and enhanced the protective effect of the ovary from cisplatin-induced gonadotoxicity in rats.
Collapse
Affiliation(s)
- Xiaoyan Li
- Department of Obstetrics and Gynecology, Union hospita, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Kang
- Department of Obstetrics and Gynecology, Union hospita, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingchun Deng
- Department of Obstetrics and Gynecology, Union hospita, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Cai
- Department of Obstetrics and Gynecology, Union hospita, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zehua Wang
- Department of Obstetrics and Gynecology, Union hospita, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
31
|
Wong M, O'Neill S, Walsh G, Smith I. Goserelin with chemotherapy to preserve ovarian function in pre-menopausal women with early breast cancer: menstruation and pregnancy outcomes. Ann Oncol 2013; 24:133-8. [DOI: 10.1093/annonc/mds250] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
32
|
Kishk EAF, Mohammed Ali MH. Effect of a gonadotropin-releasing hormone analogue on cyclophosphamide-induced ovarian toxicity in adult mice. Arch Gynecol Obstet 2012; 287:1023-9. [PMID: 23224696 DOI: 10.1007/s00404-012-2658-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Accepted: 11/22/2012] [Indexed: 10/27/2022]
Abstract
PURPOSE To evaluate the possible protective effect of low and high dose of triptorelin, a GnRH analogue, on cyclophosphamide-induced ovarian toxicity in adult female mice. METHODS Thirty-six sexually mature, virgin, female mice were divided randomly into six groups of six each: control group, low-dose triptorelin (TL) group, high-dose triptorelin (TH) group, cyclophosphamide (CPA) group, low-dose triptorelin plus cyclophosphamide (TL + CPA) group and high-dose triptorelin plus cyclophosphamide (T + CPA) group. Mice in both the TL + CPA and the TH + CPA groups were injected with 3.8 and 38 mg/kg of triptorelin subcutaneously, respectively. Four weeks later, mice in the CPA, TL + CPA and TH + CPA groups were injected with cyclophosphamide, intraperitoneally, at a dose of 50 mg/kg. Ovaries were removed 4 weeks later and processed for light microscopic examinations. RESULTS Obvious destruction of ovarian structure and significant depletion of primordial, primary, secondary and antral follicles were demonstrated in the CPA group and compared with the control group, the difference was statistically highly significant (p < 0.001), affirming the ovarian toxicity of cyclophosphamide. In the TL + CPA group, there was a significant increase in primordial, primary, secondary and antral follicles compared with the CPA group (p < 0.05), showing the effect of triptorelin on ovarian protection. Regarding, the high-dose GnRH agonist the difference was statistically highly significant for primordial and primary follicles (p < 0.001). CONCLUSIONS This study has showed a dose-dependent protective effect of GnRH analogue on ovarian reserve against ovarian toxic chemotherapy, thus demonstrating an important role of GnRH analogues in fertility preservation.
Collapse
Affiliation(s)
- Eman A Fouad Kishk
- Department of Obstetrics and Gynecology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.
| | | |
Collapse
|
33
|
Demeestere I, Brice P, Peccatori FA, Kentos A, Gaillard I, Zachee P, Casasnovas RO, Van Den Neste E, Dechene J, De Maertelaer V, Bron D, Englert Y. Gonadotropin-releasing hormone agonist for the prevention of chemotherapy-induced ovarian failure in patients with lymphoma: 1-year follow-up of a prospective randomized trial. J Clin Oncol 2012; 31:903-9. [PMID: 23129737 DOI: 10.1200/jco.2012.42.8185] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
PURPOSE To assess the efficacy of gonadotropin-releasing hormone agonist (GnRHa) in preventing chemotherapy-induced ovarian failure in patients treated for Hodgkin or non-Hodgkin lymphoma within the setting of a multicenter, randomized, prospective trial. PATIENTS AND METHODS Patients age 18 to 45 years were randomly assigned to receive either the GnRHa triptorelin plus norethisterone (GnRHa group) or norethisterone alone (control group) concomitantly with alkylating agents containing chemotherapy. The primary end point was the premature ovarian failure (POF) rate (follicle-stimulating hormone [FSH] ≥ 40 IU/L) after 1 year of follow-up. RESULTS Eighty-four of 129 randomly assigned patients completed the 1-year follow-up. The mean FSH values were higher in the control group than in the GnRHa group during chemotherapy; however, this difference was no longer observed after 6 months of follow-up. After 1 year, 20% and 19% of patients in the GnRHa and control groups, respectively, exhibited POF (P = 1.00). More than half of patients in each group completely restored their ovarian function (FSH < 10 IU/L), but the anti-Müllerian hormone values were higher in the GnRHa group than in the control group (1.4 ± 0.35 v 0.5 ± 0.15 ng/mL, respectively; P = .040). The occurrence of adverse events was similar in both groups with the exception of metrorrhagia, which was more frequently observed in the control group than the GnRHa group (38.4% v 15.6%, respectively; P = .024). CONCLUSION Approximately 20% of patients in both groups exhibited POF after 1 year of follow-up. Triptorelin was not associated with a significant decreased risk of POF in young patients treated for lymphoma but may provide protection of the ovarian reserve.
Collapse
|
34
|
Biasoli I, Falorio S, Luminari S, Spector N, Federico M. Fertility in female survivors of Hodgkin's lymphoma. Rev Bras Hematol Hemoter 2012; 34:48-53. [PMID: 23049384 PMCID: PMC3459604 DOI: 10.5581/1516-8484.20120014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Accepted: 12/04/2011] [Indexed: 11/27/2022] Open
Abstract
Currently, Hodgkin's lymphoma is one of the most curable types of cancer. Patients are often young and so the long-term morbidities of treatment have become of increasing concern. Among these, infertility is one of the most challenging consequences for patients in reproductive age. Premature ovarian failure in premenopausal women is a serious long-term sequel of the toxicity of chemotherapy. The main consequence of this syndrome is infertility, but women also present other symptoms related to estrogen deprivation. Different rates of impaired gonadal function are reported, depending on the patient's age, stage of disease, dose and intensity of chemotherapy and the use of radiation therapy. The most established strategy in female infertility is cryopreservation of embryos after in vitro fertilization. Additionally, the use of oral contraceptives or gonadotropinreleasing hormone analogs (GnRH-a) during treatment is under study. This review will provide a general overview of the main studies conducted to evaluate the infertility rate among female Hodgkin's lymphoma survivors and risk factors associated to treatment, different end-point definitions for evaluating fertility and also a brief description of the available strategies for fertility preservation.
Collapse
Affiliation(s)
- Irene Biasoli
- Hematology and Pathology Services, Hospital Universitário, Universidade Federal do Rio de Janeiro - UFRJ, Rio de Janeiro, RJ, Brazil
| | | | | | | | | |
Collapse
|
35
|
Falorio S, Biasoli I, Luminari S, Quintana G, Musso M, Dell'Olio M, Specchia MR, di Renzo N, Cesaretti M, Buda G, Vallisa D, Mannina D, Andriani A, Chiattone CS, Delamain MT, de Souza CA, Spector N, Angrilli F, Federico M. Risk factors for impaired gonadal function in female Hodgkin lymphoma survivors: final analysis of a retrospective multicenter joint study from Italian and Brazilian Institutions. Hematol Oncol 2012; 31:72-8. [DOI: 10.1002/hon.2029] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Revised: 08/19/2012] [Accepted: 09/05/2012] [Indexed: 11/11/2022]
Affiliation(s)
- Simona Falorio
- Dipartimento di Ematologia; Ospedale Civile Spirito Santo; Pescara; Italy
| | - Irene Biasoli
- Department of Medicine, University Hospital and School of Medicine; Universidade Federal do Rio de Janeiro; Rio de Janeiro; Brazil
| | - Stefano Luminari
- Dipartimento di Oncologia ed Ematologia; Università degli studi di Modena; Modena; Italy
| | | | - Maurizio Musso
- Dipartimento di Ematologia; Ospedale La Maddalena; Palermo; Italy
| | - Matteo Dell'Olio
- Dipartimento di Ematologia; Ospedale Casa Sollievo della Sofferenza; San Giovanni Rotondo; Italy
| | | | | | - Marina Cesaretti
- Dipartimento di Oncologia ed Ematologia; Università degli studi di Modena; Modena; Italy
| | - Gabriele Buda
- Dipartimento di Ematologia; Università di Pisa; Pisa; Italy
| | | | - Donato Mannina
- Divisione di Ematologia; Ospedale Papardo; Messina; Italy
| | | | | | - Márcia Torresan Delamain
- Center of Hematology and Hemotherapy, Department of Internal Medicine, Faculty of Medicine; State University of Campinas; Campinas-SP; Brazil
| | - Cármino A de Souza
- Center of Hematology and Hemotherapy, Department of Internal Medicine, Faculty of Medicine; State University of Campinas; Campinas-SP; Brazil
| | - Nelson Spector
- Department of Medicine, University Hospital and School of Medicine; Universidade Federal do Rio de Janeiro; Rio de Janeiro; Brazil
| | - Francesco Angrilli
- Dipartimento di Ematologia; Ospedale Civile Spirito Santo; Pescara; Italy
| | - Massimo Federico
- Dipartimento di Oncologia ed Ematologia; Università degli studi di Modena; Modena; Italy
| |
Collapse
|
36
|
Marder W, McCune WJ, Wang L, Wing JJ, Fisseha S, McConnell DS, Christman GM, Somers EC. Adjunctive GnRH-a treatment attenuates depletion of ovarian reserve associated with cyclophosphamide therapy in premenopausal SLE patients. Gynecol Endocrinol 2012; 28:624-7. [PMID: 22296584 PMCID: PMC3396751 DOI: 10.3109/09513590.2011.650752] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND We measured antimullerian hormone (AMH), a marker of ovarian reserve, in women with lupus treated with cyclophosphamide (CYC) (group I), CYC plus gonadotropin-releasing hormone agonist (GnRH-a) (group II) or neither (group III). We hypothesized that AMH would be diminished in women exposed to CYC versus women receiving adjunctive GnRH-a treatment or no CYC exposure. METHODS Forty-eight premenopausal lupus patients were retrospectively divided into three treatment groups: CYC alone (group I, n = 11), CYC + GnRH-a (group II, n = 10) and neither (group III, n = 27). Serum AMH levels between groups were compared using a nonparametric test (Wilcoxon rank-sum). Multiple linear regression adjusting for age was performed. RESULTS AMH (ng/mL) levels at the last collection were significantly lower in group I versus group III (mean ± SD: 0.18 ± 0.20 group I vs 1.33 ± 1.59 group III; p = 0.015), and versus group II (mean ± SD: 0.86 ± 1.06; p = 0.018). When centered on age 30 years, average AMH levels for group I, group II and group III were 0.20, 0.44 and 1.00, respectively. When adjusted for age, AMH between all groups was significantly different (p<0.0001). CONCLUSION Posttreatment AMH levels were significantly higher among patients receiving CYC + GnRH-a compared to CYC alone, suggesting that GnRH-a coadministration mitigates CYC-induced ovarian injury.
Collapse
Affiliation(s)
- W Marder
- Department of Internal Medicine, Division of Rheumatology, University of Michigan, Ann Arbor, MI 48109-5358, USA.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Chuai Y, Xu X, Wang A. Preservation of fertility in females treated for cancer. Int J Biol Sci 2012; 8:1005-12. [PMID: 22904668 PMCID: PMC3421231 DOI: 10.7150/ijbs.4800] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2012] [Accepted: 07/29/2012] [Indexed: 12/17/2022] Open
Abstract
Advancements of diagnosis and treatment have substantially improved cancer survival rates in the last few decades. The increasing number of survivors focuses attention on long-term effects caused by cancer treatment and its impact on quality of life. Ovarian failure is one of the major sequelae of cytotoxic chemotherapy and/or radiotherapy in female children and reproductive-age women. Oncologists should address the patients about fertility preservation options before therapy. Embryo cryopreservation is the only well-established method for females in preserving fertility; however other strategies including ovarian suppression, ovarian transposition and cryopreservation of oocytes and ovarian tissue are still experimental. Patients need advice and to know which are the most practical options for them. This article reviews the available fertility preservation methods in women, and the related issues including normal physiology of the ovary, effect of anticancer therapy on fertility, role of the oncologist and ethics. We performed a MEDLINE search from 1971 to 2011 in a similar way as Jensen et al. 2011, using the following MeSH terms: antineoplastic agents; ovarian failure; premature; infertility, female; fertility preservation; child and cancer; reproductive technologies, assisted.
Collapse
Affiliation(s)
- Yunhai Chuai
- Department of Obstetrics and Gynecology, Navy General Hospital, Beijing, China
| | | | | |
Collapse
|
38
|
Gnaneswaran S, Deans R, Cohn RJ. Reproductive late effects in female survivors of childhood cancer. Obstet Gynecol Int 2012; 2012:564794. [PMID: 22619680 PMCID: PMC3352687 DOI: 10.1155/2012/564794] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Revised: 12/27/2011] [Accepted: 01/25/2012] [Indexed: 01/18/2023] Open
Abstract
Childhood cancer treatments can cause female reproductive late effects. Radiation to the hypothalamic-pituitary-ovarian axis is associated with altered menarche, miscarriage, and implantation failure. Patients who receive chemotherapy and/or ovarian radiation are at risk of premature ovarian failure; the risk increases with increasing radiation dose, alkylating agent score, combination therapy, and older age at treatment. Ovarian reserve may be assessed using antimullerian hormone assay and ultrasound measurements of ovarian volume and antral follicle count; however, their efficacy is poorly established in this cohort. Fertility preservation options including cryopreservation, oophoropexy, and gonadotropin-releasing hormone analogues may be initiated prior to treatment, although most are still considered experimental. Uterine radiation has been linked to pregnancy complications including miscarriage, premature delivery, stillbirth, low-birth-weight and small-for-gestational-age infants. This paper summarises the literature on female reproductive late effects. The information should facilitate counseling and management of female survivors throughout their reproductive lives.
Collapse
Affiliation(s)
| | - Rebecca Deans
- University of New South Wales, Sydney, NSW 2052, Australia
- Royal Hospital for Women, Barker Street, Randwick, NSW 2031, Australia
- Centre for Children's Cancer and Blood Disorders, Sydney Children's Hospital, High Street, Randwick, NSW 2031, Australia
| | - Richard J. Cohn
- University of New South Wales, Sydney, NSW 2052, Australia
- Centre for Children's Cancer and Blood Disorders, Sydney Children's Hospital, High Street, Randwick, NSW 2031, Australia
| |
Collapse
|
39
|
Carp HJ, Selmi C, Shoenfeld Y. The autoimmune bases of infertility and pregnancy loss. J Autoimmun 2012; 38:J266-74. [PMID: 22284905 DOI: 10.1016/j.jaut.2011.11.016] [Citation(s) in RCA: 153] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2011] [Revised: 11/28/2011] [Accepted: 11/28/2011] [Indexed: 11/26/2022]
|
40
|
Bhattacharya P, Keating AF. Impact of environmental exposures on ovarian function and role of xenobiotic metabolism during ovotoxicity. Toxicol Appl Pharmacol 2012; 261:227-35. [PMID: 22531813 DOI: 10.1016/j.taap.2012.04.009] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 04/04/2012] [Accepted: 04/05/2012] [Indexed: 11/19/2022]
Abstract
The mammalian ovary is a heterogeneous organ and contains oocyte-containing follicles at varying stages of development. The most immature follicular stage, the primordial follicle, comprises the ovarian reserve and is a finite number, defined at the time of birth. Depletion of all follicles within the ovary leads to reproductive senescence, known as menopause. A number of chemical classes can destroy follicles, thus hastening entry into the menopausal state. The ovarian response to chemical exposure can determine the extent of ovotoxicity that occurs. Enzymes capable of bioactivating as well as detoxifying xenobiotics are expressed in the ovary and their impact on ovotoxicity has been partially characterized for trichloroethylene, 7,12-dimethylbenz[a]anthracene, and 4-vinylcyclohexene. This review will discuss those studies, as well as illustrate where knowledge gaps remain for chemicals that have also been established as ovotoxicants.
Collapse
|
41
|
Kim CH, Jeon GH. Fertility preservation in female cancer patients. ISRN OBSTETRICS AND GYNECOLOGY 2012; 2012:807302. [PMID: 22462006 PMCID: PMC3302115 DOI: 10.5402/2012/807302] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Accepted: 11/29/2011] [Indexed: 11/25/2022]
Abstract
With improved survival rates among cancer patients, fertility preservation is now being recognized as an issue of great importance. There are currently several methods of fertility preservation available in female cancer patients and the options and techniques via assisted reproduction and cryopreservation are increasing, but some are still experimental and continues to be evaluated. The established means of preserving fertility include embryo cryopreservation, gonadal shielding during radiation therapy, ovarian transposition, conservative gynecologic surgery such as radical trachelectomy, donor embryos/oocytes, gestational surrogacy, and adoption. The experimental methods include oocyte cryopreservation, ovarian cryopreservation and transplantation, in vitro maturation, and ovarian suppression. With advances in methods for the preservation of fertility, providing information about risk of infertility and possible options of fertility preservation to all young patients with cancer, and discussing future fertility with them should be also considered as one of the important parts of consultation at the time of cancer diagnosis.
Collapse
Affiliation(s)
- Chung-Hoon Kim
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Asan Medical Center, College of Medicine, University of Ulsan, 388-1 Poognap-dong, Songpa-gu, Seoul 138-736, Republic of Korea
| | | |
Collapse
|
42
|
Cheng YC, Takagi M, Milbourne A, Champlin RE, Ueno NT. Phase II study of gonadotropin-releasing hormone analog for ovarian function preservation in hematopoietic stem cell transplantation patients. Oncologist 2012; 17:233-8. [PMID: 22282904 DOI: 10.1634/theoncologist.2011-0205] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
PURPOSE Premature ovarian failure occurs in 40%-70% of patients who receive conventional chemotherapy alone. However, the incidence is higher, 70%-100%, in patients who undergo myeloablative chemotherapy with hematopoietic stem cell transplantation (HSCT). Gonadotropin-releasing hormone (GnRH) analogs, such as leuprolide, in a continuous-release formulation, may protect the ovaries from the gonadotoxic effects of chemotherapy. In non-HSCT settings, GnRH analogs have reduced the risk for premature ovarian failure to <10%. We conducted a phase II clinical trial based on the hypothesis that giving leuprolide before conditioning chemotherapy in HSCT patients reduces premature ovarian failure incidence. PATIENTS AND METHODS Eligible patients were women aged ≤40 years who were HSCT candidates, were premenopausal, and had both follicle-stimulating hormone (FSH) and luteinizing hormone (LH) levels ≤20 IU/L. Two 22.5-mg leuprolide doses were delivered in 3-month depot i.m. injections, the first within 2 months before HSCT. Patients were monitored for menstruation return, and ovarian function tests (FSH, LH, and estradiol) were done every 2 months starting 90 days after the last leuprolide dose. RESULTS Sixty eligible patients were enrolled, 59 underwent HSCT, and 44 were evaluable (median age, 25 years; median follow-up, 355 days). Only seven of 44 patients (16%) regained ovarian function. Of the 33 who received myeloablative regimens, six (18%) regained ovarian function. However, among the 11 who received nonmyeloablative regimens, only one (9%) regained ovarian function (p = .66). CONCLUSION Leuprolide did not preserve ovarian function in patients who underwent HSCT using either myeloablative or nonmyeloablative regimens. Other measures that protect ovarian function need to be investigated.
Collapse
Affiliation(s)
- Yee Chung Cheng
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | | | | | | | | |
Collapse
|
43
|
Binart N, Sauvat F. [Cryopreserved ovaries: models of experimental grafting]. Biol Aujourdhui 2012; 205:235-43. [PMID: 22251858 DOI: 10.1051/jbio/2011019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Indexed: 01/16/2023]
Abstract
Childhood cancers account for 1% of malignant tumors. As a result of advances in treatment, almost 80% of children and adolescents who currently receive a diagnosis of cancer become long-term survivors. Assessment of potential for fertility preservation should thus be a systematic element of care for children treated for a malignant tumor (high-dose chemotherapy with alkylizing agents, radiation therapy including the gonads) or those receiving hematopoietic stem cell grafts for malignant or benign disease (sickle-cell anemia, immune deficit). Potential adverse consequences of treatment include impaired puberty and fertility due to gonadal removal, genital tract injury or damage to germ cells from adjuvant therapy. Advances in assisted reproductive technologies have led to new possibilities for the prevention and treatment of infertility. Among them, cryopreservation of ovarian tissue appears to be the most promising, or perhaps the only one available before puberty with encouraging results. Nevertheless the uncertainties, or even risks, related to these treatments, should not be neglected. We review experimental data in mouse and sheep animal models. The results demonstrate that immature ovarian grafting can restore spontaneous puberty and fertility in both models. This study addresses the very important issue of epigenetics, and provides valuable information for the study of ovarian transplantation, suggesting that these procedures do not perturb normal epigenetic marks. These results are highly relevant to the question of immature cortex reimplantation.
Collapse
|
44
|
Munster PN, Moore AP, Ismail-Khan R, Cox CE, Lacevic M, Gross-King M, Xu P, Carter WB, Minton SE. Randomized trial using gonadotropin-releasing hormone agonist triptorelin for the preservation of ovarian function during (neo)adjuvant chemotherapy for breast cancer. J Clin Oncol 2012; 30:533-8. [PMID: 22231041 DOI: 10.1200/jco.2011.34.6890] [Citation(s) in RCA: 172] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Chemotherapy-induced amenorrhea is a serious concern for women undergoing cancer therapy. This prospective randomized trial evaluated the use of gonadotropin-releasing hormone (GnRH) analog triptorelin to preserve ovarian function in women treated with chemotherapy for early-stage breast cancer. PATIENTS AND METHODS Premenopausal women age 44 years or younger were randomly assigned to receive either triptorelin or no triptorelin during (neo)adjuvant chemotherapy and were further stratified by age (< 35, 35 to 39, > 39 years), estrogen receptor status, and chemotherapy regimen. Objectives included the resumption of menses and serial monitoring of follicle-stimulating hormone (FSH) and inhibin A and B levels. RESULTS Targeted for 124 patients with a planned 5-year follow-up, the trial was stopped for futility after 49 patients were enrolled (median age, 39 years; range, 21 to 43 years); 47 patients were treated according to assigned groups with four cycles of adriamycin plus cyclophosphamide alone or followed by four cycles of paclitaxel or six cycles of fluorouracil, epirubicin, and cyclophosphamide. Menstruation resumed in 19 (90%) of 21 patients in the control group and in 23 (88%) of 26 in the triptorelin group (P= .36). Menses returned after a median of 5.8 months (range, 1 to 19 months) after completion of chemotherapy in the triptorelin versus 5.0 months (range, 0 to 28 months) in the control arm (P= .58). Two patients (age 26 and 35 years at random assignment) in the control group had spontaneous pregnancies with term deliveries. FSH and inhibin B levels correlated with menstrual status. CONCLUSION When stratified for age, estrogen receptor status, and treatment regimen, amenorrhea rates on triptorelin were comparable to those seen in the control group.
Collapse
|
45
|
Fertility Preservation Options for Females. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 732:9-28. [DOI: 10.1007/978-94-007-2492-1_2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
46
|
Chen H, Li J, Cui T, Hu L. Adjuvant gonadotropin-releasing hormone analogues for the prevention of chemotherapy induced premature ovarian failure in premenopausal women. Cochrane Database Syst Rev 2011:CD008018. [PMID: 22071842 DOI: 10.1002/14651858.cd008018.pub2] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Chemotherapy has significantly improved prognosis for patients with malignant and some non-malignant conditions. This treatment, however, is associated with ovarian toxicity and gonadotropin-releasing hormone (GnRH) analogues may have a protective effect on the ovaries. The mechanism of action of GnRH is based on suppression of the gonadotropin levels to simulate pre-pubertal hormonal milieu and decrease utero-ovarian perfusion. OBJECTIVES To assess the efficacy and safety of GnRH analogues given before or in parallel to chemotherapy to prevent chemotherapy-related ovarian damage in premenopausal women with malignant or non-malignant conditions. SEARCH METHODS We searched the Cochrane Gynaecological Cancer Group Specialized Register (up to July 2011), the Cochrane Central Register of Controlled Trials (CENTRAL) (The Cochrane Library, Issue 2, 2011); MEDLINE (1950 to July 2011); EMBASE (1980 to July 2011); and the Chinese Biomedicine Database (CBM) (1976 to July 2011). SELECTION CRITERIA Randomized controlled trials (RCTs), in all languages, which examined the effect of GnRH analogues for chemotherapy-induced ovarian failure in premenopausal women, were eligible for inclusion in the review. DATA COLLECTION AND ANALYSIS The review authors independently extracted data and assessed trial quality using the Cochrane risk of bias tool. We analyzed binary data using risk ratios (RRs) with 95% confidence intervals (CI) and for continuous data, we used the standardized mean difference (SMD) to combine trials. As there was substantial difference in the types of chemotherapy used, we applied the random-effects model in our analyses. We contacted study authors for additional information. MAIN RESULTS Included studies in this review showed that intramuscular/subcutaneous administration of GnRH agonists was effective in protecting menstruation and ovulation after chemotherapy (resumed menses: RR 1.90, 95% CI 1.30 to 2.79; amenorrhoea: RR 0.08, 95% CI 0.01 to 0.58; ovulation: RR 2.70, 95% CI 1.52 to 4.79), whereas intranasal administration of GnRH agonists had no protective effect on ovaries (resumed menses: RR 0.75, 95% CI 0.33 to 1.72; ovulation: RR 1.13, 95% CI 0.20 to 6.24). Pregnancy rates were not significantly different between groups (intramuscular/subcutaneous GnRH agonist: RR 0.21, 95% CI 0.01 to 4.09; intranasal GnRH agonist: RR 0.41, 95% CI 0.02 to 8.84). Ultrasound antral follicular count (AFC) was not significantly different between groups (SMD 1.11, 95% CI 0.32 to 1.90). AUTHORS' CONCLUSIONS The use of GnRH agonists should be considered in women of reproductive age receiving chemotherapy. Intramuscular or subcutaneous GnRH analogues seem to be effective in protecting ovaries during chemotherapy and should be given before or during treatment, although no significant difference in pregnancy rates was seen.
Collapse
Affiliation(s)
- Hengxi Chen
- Department of Obstetrics and Gynecology, West China Second University Hospital, West China Women’s and Children’s Hospital,Chengdu, China.
| | | | | | | |
Collapse
|
47
|
Treves R, Grynberg M, Hesters L, Frydman R. Fertility Preservation in Women with Cancer: Importance of a Multidisciplinary Approach. WOMENS HEALTH 2011; 7:537-43. [DOI: 10.2217/whe.11.40] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
For all patients affected by a disease that could impair fertility before or during the reproductive lifespan, strategies to preserve their fertility and the ability to bear their own children is likely to be of utmost importance. While fertility preservation is a promising option, most of the technologies currently used are far from being well-established or are still experimental. Patients should be aware that no method guarantees success. Psychological and ethical impacts of fertility preservation are major concerns and should be included in the multidisciplinary approach to the patients.
Collapse
Affiliation(s)
- Rachel Treves
- Departments of Obstetrics & Gynecology & Reproductive Medicine, Université Paris XI, Institut National de la Santé et de la Recherche Médicale U782, Hôpital Antoine Béclère 157, rue de la Porte de Trivaux 92141 Clamart, France
| | - Michaël Grynberg
- Departments of Obstetrics & Gynecology & Reproductive Medicine, Université Paris XI, Institut National de la Santé et de la Recherche Médicale U782, Hôpital Antoine Béclère 157, rue de la Porte de Trivaux 92141 Clamart, France
| | - Laetitia Hesters
- Departments of Obstetrics & Gynecology & Reproductive Medicine, Université Paris XI, Institut National de la Santé et de la Recherche Médicale U782, Hôpital Antoine Béclère 157, rue de la Porte de Trivaux 92141 Clamart, France
| | - Rene Frydman
- Departments of Obstetrics & Gynecology & Reproductive Medicine, Université Paris XI, Institut National de la Santé et de la Recherche Médicale U782, Hôpital Antoine Béclère 157, rue de la Porte de Trivaux 92141 Clamart, France
| |
Collapse
|
48
|
Whitehead J, Toledo MG, Stern CJ. A pilot study to assess the use of the gonadotrophin antagonist cetrorelix in preserving ovarian function during chemotherapy. Aust N Z J Obstet Gynaecol 2011; 51:452-4. [PMID: 21810087 DOI: 10.1111/j.1479-828x.2011.01346.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cyclophosphamide treatment can cause premature ovarian failure. This pilot study evaluates the protective effect of the gonadotrophin releasing hormone (GnRH) antagonist, cetrorelix, on ovarian function, when used during cyclophosphamide chemotherapy in women aged 18-35. Primary outcomes measured were serum follicle stimulating hormone (FSH) and inhibin prior to and at 6 and 12 months after chemotherapy. Secondary outcomes were hormonal evidence of a suppressive effect and the side effect profile.
Collapse
|
49
|
Browne HN, Moon KS, Mumford SL, Schisterman EF, Decherney AH, Segars JH, Armstrong AY. Is anti-Müllerian hormone a marker of acute cyclophosphamide-induced ovarian follicular destruction in mice pretreated with cetrorelix? Fertil Steril 2011; 96:180-186.e2. [PMID: 21550044 DOI: 10.1016/j.fertnstert.2011.04.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2010] [Revised: 03/31/2011] [Accepted: 04/05/2011] [Indexed: 01/02/2023]
Abstract
OBJECTIVE To define whether anti-Müllerian hormone (AMH) may be a marker of acute cyclophosphamide (CTX)-induced germ cell destruction in mice pretreated with the GnRH antagonist, cetrorelix. DESIGN Controlled, experimental study. SETTING Research laboratory in a federal research facility. ANIMAL(S) Balb/c female mice (6 weeks old). INTERVENTION(S) Mice were treated with GnRH antagonist (cetrorelix) or saline for 15 days followed by 75 mg/kg or 100 mg/kg of CTX or saline control on day 9. MAIN OUTCOME MEASURE(S) Number of primordial follicles (PMF), DNA damage, AMH protein expression, and AMH serum levels. RESULT(S) Ovaries in mice pretreated with cetrorelix had significantly more PMFs and reduced DNA damage compared with those exposed to CTX alone. Immunohistochemical staining for AMH expression and serum AMH levels did not differ significantly between treatment groups. CONCLUSION(S) Cetrorelix protected PMFs and reduced DNA damage in follicles of mice treated with CTX, but AMH levels in tissue and serum did not correlate with germ cell destruction. Further research is needed to determine the mechanism responsible for the protective effects on PMF counts observed with cetrorelix.
Collapse
Affiliation(s)
- Hyacinth N Browne
- Program in Reproductive and Adult Endocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Modern combination chemotherapy and radiotherapy regimens have a substantial negative impact on reproduction. Premature ovarian failure and other poor reproductive outcomes subsequent to cancer therapies are being recognized. Furthermore, beside malignancies, treatment of certain precancerous and benign conditions such as myelodysplasia, aplastic anemia, and systemic lupus erythematosus may necessitate administration of high-dose chemotherapeutics with and without stem cell transplantation. Therefore, preservation of gonadal function and fertility has become one of the major quality of life issues for cancer survivors at reproductive ages. In this review, premature ovarian failure and other adverse reproductive outcomes in female patients who receive chemotherapy and radiation will be discussed and the options to preserve their fertility will be delineated. After completion of the educational activity, the reader will be better able to distinguish the experimental approaches to minimizing gonadotoxic therapy, and use the most effective treatment options.
Collapse
|