1
|
Seok J, Park HS, Cetin E, Ghasroldasht MM, Liakath FB, Al-Hendy A. The potent paracrine effect of umbilical cord mesenchymal stem cells mediates mitochondrial quality control to restore chemotherapy-induced damage in ovarian granulosa cells. Biomed Pharmacother 2024; 172:116263. [PMID: 38350369 DOI: 10.1016/j.biopha.2024.116263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/24/2024] [Accepted: 02/06/2024] [Indexed: 02/15/2024] Open
Abstract
The basic principle of chemotherapy is to attack cells with fast growth, and cancer cells are targeted by anticancer drugs because they have a faster growth rate than normal cells. High doses of anticancer drugs may cause an irreversible decline in reproductive capacity, and novel approaches for fertility preservation and/or restoration after anticancer treatment are urgently needed. Here, we provide important insights into the recovery of human reproductive cells damaged by chemotherapy. We performed a detailed screening of the cytokines of various human mesenchymal stem cells (hMSCs) to select superior MSCs. Also, we analyzed the Ovarian granulosa cell (OGC)-)-specific functions for restoring function, apoptosis, and mitochondrial functions to confirm the recovery mechanism in damaged OGCs. As a result, we demonstrated that conditioned media (CM) of Umbilical cord mesenchymal stem cells (UC-MSCs) could restore the functions of damaged OGCs primarily through antiapoptotic and antioxidant effects. Furthermore, CM changed the phenotype of damaged OGCs to an energetic status by restoring mitochondrial function and enhanced the mitochondrial metabolic activity decreased by chemotherapy. Finally, we demonstrated that the restoration of mitochondrial function in damaged OGCs was mediated through mitochondrial autophagy (mitophagy). Our findings offer new insights into the potential of stem cell-based therapy for fertility preservation and/or restoration in female cancer patients.
Collapse
Affiliation(s)
- Jin Seok
- Department of Obstetrics and Gynecology, University of Chicago, 5841 A. Maryland Ave, Chicago, IL 60637, USA
| | - Hang-Soo Park
- Department of Obstetrics and Gynecology, University of Chicago, 5841 A. Maryland Ave, Chicago, IL 60637, USA
| | - Esra Cetin
- Department of Obstetrics and Gynecology, Hurley Medical Center, Michigan State University, 1 Hurley Plaza, Flint, MI 48503, USA
| | | | - Farzana Begum Liakath
- Department of Obstetrics and Gynecology, University of Chicago, 5841 A. Maryland Ave, Chicago, IL 60637, USA
| | - Ayman Al-Hendy
- Department of Obstetrics and Gynecology, University of Chicago, 5841 A. Maryland Ave, Chicago, IL 60637, USA.
| |
Collapse
|
2
|
Himpe J, Lammerant S, Van den Bergh L, Lapeire L, De Roo C. The Impact of Systemic Oncological Treatments on the Fertility of Adolescents and Young Adults-A Systematic Review. Life (Basel) 2023; 13:life13051209. [PMID: 37240854 DOI: 10.3390/life13051209] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/11/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND Over the past decades, advancements in oncological treatments have led to major improvements in survival. Particularly for adolescents and young adults (AYAs), fertility is an important concern in cancer survivorship. The purpose of the review is to provide physicians with a practical overview of the current knowledge about the impact of systemic oncological treatments on the fertility of female and male AYAs. METHODS A systematic review was performed based on relevant articles obtained from 4 databases up until 31 December 2022. RESULTS The mechanisms of gonadotoxicity and the concurrent risk is described for the following categories: chemotherapy, targeted therapy and immunotherapy. For the category "chemotherapy", the specific effects and risks are listed for the different classes and individual chemotherapeutics. In the category "targeted therapy", a distinction was made between tyrosine kinase inhibitors (TKIs) and monoclonal antibodies. Information concerning immunotherapy is scarce. CONCLUSIONS The effects of chemotherapy on fertility are well investigated, but even in this category, results can be conflicting. Insufficient data are available on the fertility effects of targeted therapy and immunotherapy to draw definitive conclusions. More research is needed for these therapies and their evolving role in treating cancers in AYAs. It would be useful to include fertility endpoints in clinical trials that evaluate new and existing oncological treatments.
Collapse
Affiliation(s)
- Justine Himpe
- Department of Medical Oncology, Ghent University Hospital, 9000 Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Sander Lammerant
- Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Lore Van den Bergh
- Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Lore Lapeire
- Department of Medical Oncology, Ghent University Hospital, 9000 Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
- AYA Research Centre and Hub (ARCH), Ghent University, 9000 Ghent, Belgium
| | - Chloë De Roo
- AYA Research Centre and Hub (ARCH), Ghent University, 9000 Ghent, Belgium
- Department of Reproductive Medicine, Ghent University Hospital, 9000 Ghent, Belgium
| |
Collapse
|
3
|
Schuurman T, Song JY, Wolters V, van de Ven M, van Trommel N, Beerendonk I, Amant F, Lok C. Effects of chemotherapy on ovaries of pregnant mice. Arch Gynecol Obstet 2023; 307:1163-1176. [PMID: 36166083 DOI: 10.1007/s00404-022-06793-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 09/09/2022] [Indexed: 11/02/2022]
Abstract
PURPOSE It is unknown if future fertility is compromised by the administration of chemotherapy during pregnancy. The aim of this study was to identify if chemotherapy affects the maternal ovaries during pregnancy and whether these effects depend on type of chemotherapy and duration of exposure. METHODS Pregnant 8-week-old female BL6 mice were exposed to 6 different single chemotherapeutic agents (carboplatin, cisplatin, paclitaxel, epirubicin, doxorubicin, or cyclophosphamide) or saline at gestational day (GD) 13.5. The mice were sacrificed at GD 15.5 or GD 18.5. Ovaries were assessed by histopathology and immunohistochemistry. Follicle count was determined per follicle stage and per treatment modality. RESULTS Maternal ovarian damage was demonstrated by the presence of apoptosis and necrosis in preantral follicles. The extent of this damage depends upon type of chemotherapy and duration of exposure (2 or 5 days). After short exposure, 81% of ovaries showed histopathologic signs of damage compared to 36% after long exposure, which might suggest a transient effect. Loss of primordial follicles (PMFs) was observed after both short and long exposure, with a reduction of more than 70%. Evidence of DNA damage, as demonstrated by phospho-H2AX expression, was present in 23% (range 0-89%) of PMFs exposed to chemotherapy, but only in the short exposure group. Overall, the least damage was seen after administration of paclitaxel. CONCLUSION Despite physiological ovarian function suppression during gestation, chemotherapy-induced damage of the ovaries occurs in pregnant mouse models, potentially affecting future fertility.
Collapse
Affiliation(s)
- Teska Schuurman
- Department of Gynecologic Oncology, Netherlands Cancer Institute, Antoni Van Leeuwenhoek, Amsterdam, The Netherlands
| | - Ji-Ying Song
- Department of Experimental Animal Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Vera Wolters
- Department of Gynecologic Oncology, Netherlands Cancer Institute, Antoni Van Leeuwenhoek, Amsterdam, The Netherlands
| | - Marieke van de Ven
- Mouse Clinic for Cancer and Aging (MCCA), Preclinical Intervention Unit, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Nienke van Trommel
- Department of Gynecologic Oncology, Netherlands Cancer Institute, Antoni Van Leeuwenhoek, Amsterdam, The Netherlands
| | - Ina Beerendonk
- Department of Obstetrics and Gynecology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Frédéric Amant
- Department of Gynecologic Oncology, Netherlands Cancer Institute, Antoni Van Leeuwenhoek, Amsterdam, The Netherlands
- Department of Gynecologic Oncology, UZ Leuven, Leuven, Belgium
| | - Christianne Lok
- Department of Gynecologic Oncology, Netherlands Cancer Institute, Antoni Van Leeuwenhoek, Amsterdam, The Netherlands.
| |
Collapse
|
4
|
Ovarian Reserve Disorders, Can We Prevent Them? A Review. Int J Mol Sci 2022; 23:ijms232315426. [PMID: 36499748 PMCID: PMC9737352 DOI: 10.3390/ijms232315426] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
The ovarian reserve is finite and begins declining from its peak at mid-gestation until only residual follicles remain as women approach menopause. Reduced ovarian reserve, or its extreme form, premature ovarian insufficiency, stems from multiple factors, including developmental, genetic, environmental exposures, autoimmune disease, or medical/surgical treatment. In many cases, the cause remains unknown and resulting infertility is not ultimately addressed by assisted reproductive technologies. Deciphering the mechanisms that underlie disorders of ovarian reserve could improve the outcomes for patients struggling with infertility, but these disorders are diverse and can be categorized in multiple ways. In this review, we will explore the topic from a perspective that emphasizes the prevention or mitigation of ovarian damage. The most desirable mode of fertoprotection is primary prevention (intervening before ablative influence occurs), as identifying toxic influences and deciphering the mechanisms by which they exert their effect can reduce or eliminate exposure and damage. Secondary prevention in the form of screening is not recommended broadly. Nevertheless, in some instances where a known genetic background exists in discrete families, screening is advised. As part of prenatal care, screening panels include some genetic diseases that can lead to infertility or subfertility. In these patients, early diagnosis could enable fertility preservation or changes in family-building plans. Finally, Tertiary Prevention (managing disease post-diagnosis) is critical. Reduced ovarian reserve has a major influence on physiology beyond fertility, including delayed/absent puberty or premature menopause. In these instances, proper diagnosis and medical therapy can reduce adverse effects. Here, we elaborate on these modes of prevention as well as proposed mechanisms that underlie ovarian reserve disorders.
Collapse
|
5
|
Xiao Y, Peng Y, Zhang C, Liu W, Wang K, Li J. hucMSC-derived exosomes protect ovarian reserve and restore ovarian function in cisplatin treated mice. J Biomed Res 2022; 37:382-393. [PMID: 37198178 PMCID: PMC10541778 DOI: 10.7555/jbr.36.20220166] [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: 07/25/2022] [Revised: 08/28/2022] [Accepted: 09/04/2022] [Indexed: 11/17/2022] Open
Abstract
Anti-cancer therapy often causes premature ovarian insufficiency and infertility as the ovarian follicle reserve is extremely sensitive to chemotherapy drugs, such as cisplatin. Various fertility preservation methods have been explored for women, especially prepubertal girls undergoing radiotherapy and chemotherapy due to cancer. In recent years, mesenchymal stem cell-derived exosomes (MSC-exos) have been reported to play an important role in tissue repair and the treatment of various diseases. In the current study, we observed that human umbilical cord-derived MSC-exos (hucMSC-exos) after short-term culture improved follicular survival and development while receiving cisplatin treatment. Moreover, intravenous injection of hucMSC-exos improved ovarian function and ameliorated inflammatory environment within the ovary. The underlying mechanism of hucMSC-exos on fertility preservation was associated with the down-regulation of p53-related apoptosis and their anti-inflammatory function. Based on these findings, we propose that hucMSC-exos may be a potential approach to improve fertility in women diagnosed with cancer.
Collapse
Affiliation(s)
- Yue Xiao
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yue Peng
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Chi Zhang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Wei Liu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Kehan Wang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Jing Li
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|
6
|
Barberino RS, Lins TLBG, Monte APO, Silva RLS, Andrade KO, Campinho DSP, Palheta Junior RC, Smitz JEJ, Matos MHT. Epigallocatechin-3-gallate attenuates cyclophosphamide-induced damage in mouse ovarian tissue via suppressing inflammation, apoptosis, and expression of phosphorylated Akt, FOXO3a and rpS6. Reprod Toxicol 2022; 113:42-51. [PMID: 35981663 DOI: 10.1016/j.reprotox.2022.08.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 08/12/2022] [Accepted: 08/12/2022] [Indexed: 11/30/2022]
Abstract
This study was conducted to evaluate the protective effects of epigallocatechin-3-gallate (EGCG) against ovarian toxicity in cyclophosphamide-treated mice and to verify the possible involvement of phosphorylated Akt, FOXO3a and rpS6 in the EGCG actions. Mice received saline solution (i.p.; control) or a single dose of cyclophosphamide (200 mg/kg body weight, i.p.) or mice were pretreated with N-acetylcysteine (150 mg/kg body weight, i.p.; positive control) or with EGCG (5, 25 or 50 mg/kg body weight, i.p.) once daily for three days followed by injection with single dose of cyclophosphamide (200 mg/kg body weight, i.p.). Thereafter, the mice were euthanized, and the ovaries were harvested and destined to histological (follicular morphology and activation), immunohistochemistry (cleaved caspase-3 and TNF-α) and fluorescence (mitochondrial activity and GSH concentrations) analyses. Furthermore, we examined the participation of p-Akt, p-FOXO3a and p-rpS6 in the protective effects of EGCG in cyclophosphamide-induced ovarian damage by immunohistochemical staining. The results showed that pretreatment with N-acetylcysteine or EGCG at 25 and 50 mg/kg before cyclophosphamide administration preserved the normal follicular morphology, prevented primordial follicle loss, reduced atresia, inflammation, and mitochondrial damage, and increased GSH concentrations compared to the only cyclophosphamide treatment. Additionally, pretreatment with 25 mg/kg EGCG regulated phosphorylated Akt, FOXO3a and rpS6 after cyclophosphamide treatment. In conclusion, short-time pretreatment with 25 mg/kg EGCG can prevent follicle loss in cyclophosphamide-treated mice by reducing oxidative damage, inflammation, and apoptosis, and regulating of p-Akt, p-FOXO3a and p-rpS6.
Collapse
Affiliation(s)
- Ricássio S Barberino
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley - UNIVASF, Petrolina, Brazil.
| | - Thae Lanne B G Lins
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley - UNIVASF, Petrolina, Brazil
| | - Alane P O Monte
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley - UNIVASF, Petrolina, Brazil
| | - Regina Lucia S Silva
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley - UNIVASF, Petrolina, Brazil
| | - Kíscyla O Andrade
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley - UNIVASF, Petrolina, Brazil
| | - Daniela S P Campinho
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley - UNIVASF, Petrolina, Brazil
| | - Raimundo C Palheta Junior
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Federal University of São Francisco Valley - UNIVASF, Petrolina, Brazil
| | - Johan E J Smitz
- Follicle Biology Laboratory, Free University Brussels - VUB, Brussels, Belgium
| | - Maria Helena T Matos
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley - UNIVASF, Petrolina, Brazil
| |
Collapse
|
7
|
Barberino RS, Silva RLS, Palheta Junior RC, Smitz JEJ, Matos MHT. Protective Effects of Antioxidants on Cyclophosphamide-Induced Ovarian Toxicity. Biopreserv Biobank 2022; 21:121-141. [PMID: 35696235 DOI: 10.1089/bio.2021.0159] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The most common limitation of anticancer chemotherapy is the injury to normal cells. Cyclophosphamide, which is one of the most widely used alkylating agents, can cause premature ovarian insufficiency and infertility since the ovarian follicles are extremely sensitive to their effects. Although little information is available about the pathogenic mechanism of cyclophosphamide-induced ovarian damage, its toxicity is attributed to oxidative stress, inflammation, and apoptosis. The use of compounds with antioxidant and cytoprotective properties to protect ovarian function from deleterious effects during chemotherapy would be a significant advantage. Thus, this article reviews the mechanism by which cyclophosphamide exerts its toxic effects on the different cellular components of the ovary, and describes 24 cytoprotective compounds used to ameliorate cyclophosphamide-induced ovarian injury and their possible mechanisms of action. Understanding these mechanisms is essential for the development of efficient and targeted pharmacological complementary therapies that could protect and prolong female fertility.
Collapse
Affiliation(s)
- Ricássio S Barberino
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Department of Veterinary Medicine, Federal University of São Francisco Valley-UNIVASF, Petrolina, Brazil
| | - Regina Lucia S Silva
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Department of Veterinary Medicine, Federal University of São Francisco Valley-UNIVASF, Petrolina, Brazil
| | - Raimundo C Palheta Junior
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Federal University of São Francisco Valley-UNIVASF, Petrolina, Brazil
| | - Johan E J Smitz
- Follicle Biology Laboratory, Center for Reproductive Medicine, Free University Brussels-VUB, Brussels, Belgium
| | - Maria Helena T Matos
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Department of Veterinary Medicine, Federal University of São Francisco Valley-UNIVASF, Petrolina, Brazil
| |
Collapse
|
8
|
Schindler K. Inhibition of BIN2 extends reproductive lifespan. NATURE AGING 2021; 1:977-979. [PMID: 37118339 DOI: 10.1038/s43587-021-00132-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Affiliation(s)
- Karen Schindler
- Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA.
- Department of Genetics, Rutgers University, Piscataway, NJ, USA.
| |
Collapse
|
9
|
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: 32] [Impact Index Per Article: 10.7] [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
|
10
|
Sun B, Yeh J. Onco-fertility and personalized testing for potential for loss of ovarian reserve in patients undergoing chemotherapy: proposed next steps for development of genetic testing to predict changes in ovarian reserve. FERTILITY RESEARCH AND PRACTICE 2021; 7:13. [PMID: 34193292 PMCID: PMC8244159 DOI: 10.1186/s40738-021-00105-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 05/19/2021] [Indexed: 12/29/2022]
Abstract
Women of reproductive age undergoing chemotherapy face the risk of irreversible ovarian insufficiency. Current methods of ovarian reserve testing do not accurately predict future reproductive potential for patients undergoing chemotherapy. Genetic markers that more accurately predict the reproductive potential of each patient undergoing chemotherapy would be critical tools that would be useful for evidence-based fertility preservation counselling. To assess the possible approaches to take to develop personalized genetic testing for these patients, we review current literature regarding mechanisms of ovarian damage due to chemotherapy and genetic variants associated with both the damage mechanisms and primary ovarian insufficiency. The medical literature point to a number of genetic variants associated with mechanisms of ovarian damage and primary ovarian insufficiency. Those variants that appear at a higher frequency, with known pathways, may be considered as potential genetic markers for predictive ovarian reserve testing. We propose developing personalized testing of the potential for loss of ovarian function for patients with cancer, prior to chemotherapy treatment. There are advantages of using genetic markers complementary to the current ovarian reserve markers of AMH, antral follicle count and day 3 FSH as predictors of preservation of fertility after chemotherapy. Genetic markers will help identify upstream pathways leading to high risk of ovarian failure not detected by present clinical markers. Their predictive value is mechanism-based and will encourage research towards understanding the multiple pathways contributing to ovarian failure after chemotherapy.
Collapse
Affiliation(s)
- Bei Sun
- Sackler School of Medicine, New York State/American Program of Tel Aviv University, Tel Aviv University, Ramat Aviv 69978, Tel Aviv, Israel
| | - John Yeh
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics & Gynecology, University of Massachusetts Medical School, UMass Memorial Medical Center, 119 Belmont Street, Worcester, MA, 01605, USA.
| |
Collapse
|
11
|
Huang CC, Chou CH, Yang YS, Ho HN, Shun CT, Wen WF, Chen SU, Chen MJ. Metformin: a novel promising option for fertility preservation during cyclophosphamide-based chemotherapy. Mol Hum Reprod 2021; 27:gaaa084. [PMID: 33543290 PMCID: PMC8494485 DOI: 10.1093/molehr/gaaa084] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 11/22/2020] [Indexed: 12/19/2022] Open
Abstract
Cyclophosphamide (CP) could cause severe gonadotoxicity via imbalanced activation of primordial follicles through PI3K/AKT/mTOR activation. Whether metformin, a widely prescribed anti-diabetes agent with mTOR inhibitory effect, could preserve ovarian function against CP toxicity is unknown. Female C57BL/6 mice were randomized into seven groups (n = 11), including control, CP-alone, CP + metformin, CP + sirolimus or everolimus, metformin-alone and sirolimus-alone groups. The duration of pharmaceutical treatment was 4 weeks. CP treatment significantly impaired ovarian function and fertility in mice. CP + metformin treatment significantly attenuated the gonadotoxicity comparing to CP-alone treatment (primordial follicle count: 17.6 ± 4.2 versus 10.3 ± 2.7 follicles/high-power field; P = 0.027). CP + metformin treatment also tended to increase antral follicular count (5.4 ± 1.1 versus 2.5 ± 1.6 follicles/section), serum AMH levels (4.6 ± 1.2 versus 2.0 ± 0.8 ng/ml) and the litter size (4.2 ± 1.3 versus 1.5 ± 1.0 mice per pregnancy), compared with CP-alone group. Expression of phospho-mTOR and the number of TUNEL-positive granulosa cells increased after CP treatment and decreased in the CP + metformin groups, suggesting the mTOR inhibitory and anti-apoptotic effects of metformin. In in-vitro granulosa cell experiments, the anti-apoptotic effect of metformin was blocked after inhibiting p53 or p21 function, and the expression of p53 mRNA was blocked with AMPK inhibitor, suggesting that the anti-apoptotic effect was AMPK/p53/p21-mediated. In conclusion, concurrent metformin treatment during CP therapy could significantly preserve ovarian function and fertility and could be a promising novel fertility preserving agent during chemotherapy. The relatively acceptable cost and well-established long-term safety profiles of this old drug might prompt its further clinical application at a faster pace.
Collapse
Affiliation(s)
- Chu-Chun Huang
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei 100, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Chia-Hung Chou
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Yu-Shih Yang
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei 100, Taiwan
- Department of Obstetrics and Gynecology, Fu Jen Catholic University Hospital, New Taipei 243, Taiwan
| | - Hong-Nerng Ho
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei 100, Taiwan
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Taipei Medical University, Taipei 110, Taiwan
| | - Chia-Tung Shun
- Departments of Forensic Medicine and Pathology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Wen-Fen Wen
- Departments of Pathology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Shee-Uan Chen
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Mei-Jou Chen
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei 100, Taiwan
- Livia Shangyu Wan Scholar, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| |
Collapse
|
12
|
Cathcart-Rake EJ, Ruddy KJ, Bleyer A, Johnson RH. Breast Cancer in Adolescent and Young Adult Women Under the Age of 40 Years. JCO Oncol Pract 2021; 17:305-313. [PMID: 33449828 DOI: 10.1200/op.20.00793] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
In the United States, one in 196 women is diagnosed with breast cancer under the age of 40 years. Adolescents and young adults (AYAs), of age 15-39 years at diagnosis, experience a number of unique challenges when confronting breast cancer. The incidence of invasive breast cancer has increased among AYA women in the United States since 2004, and most of this change is due to an increase in young women diagnosed with distant disease. AYAs are more likely than older women to present with aggressive subtypes and advanced disease, and they often require systemic staging at diagnosis. Clinical trials should be considered whenever possible, particularly in AYAs with locally advanced or metastatic disease at diagnosis and those with disease progression or recurrence. A significant proportion of AYAs carry germline cancer predisposition mutations, which necessitates prompt genetic testing for all AYAs at diagnosis and may influence choice of local therapy. Suppression of ovarian function, as an adjunct to chemotherapy, may improve breast cancer survival in AYAs. To provide optimal care for AYAs with breast cancer, clinicians should engage multidisciplinary teams that offer fertility preservation, genetic counseling, physical and occupational therapy, nutrition, and psychosocial support, along with medical expertise in tailoring cancer-directed therapy and symptom management toward young women.
Collapse
Affiliation(s)
| | | | - Archie Bleyer
- St Charles Health System, Central Oregon Health and Science University, Portland, OR
| | | |
Collapse
|
13
|
Dolmans MM, Donnez J, Cacciottola L. Fertility Preservation: The Challenge of Freezing and Transplanting Ovarian Tissue. Trends Mol Med 2020; 27:777-791. [PMID: 33309205 DOI: 10.1016/j.molmed.2020.11.003] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/06/2020] [Accepted: 11/10/2020] [Indexed: 02/06/2023]
Abstract
Cancer treatments are increasingly effective, but can result in iatrogenic premature ovarian insufficiency. Ovarian tissue cryopreservation is the only option available to preserve fertility in prepubertal girls and young women who require immediate chemotherapy. Ovarian tissue transplantation has been shown to restore hormonal cycles and fertility, but a large proportion of the follicle reserve is lost as a consequence of exposure to hypoxia. Another crucial concern is the risk of reimplanting malignant cells together with the grafted tissue. In this review, the authors advance some challenging propositions, from prevention of chemotherapy-related gonadotoxicity to ovarian tissue cryopreservation and transplantation, including the artificial ovary approach.
Collapse
Affiliation(s)
- Marie-Madeleine Dolmans
- Gynecology Department, Cliniques universitaires St-Luc, Brussels, Belgium; Pôle de Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium.
| | - Jacques Donnez
- Prof. Em. Catholic University of Louvain, Brussels, Belgium; Société de Recherche pour l'Infertilité (SRI), Brussels, Belgium
| | - Luciana Cacciottola
- Pôle de Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
14
|
Salian SR, Uppangala S, Cheredath A, D’Souza F, Kalthur G, Nayak VC, Anderson RA, Adiga SK. Early prepubertal cyclophosphamide exposure in mice results in long-term loss of ovarian reserve, and impaired embryonic development and blastocyst quality. PLoS One 2020; 15:e0235140. [PMID: 32574203 PMCID: PMC7310698 DOI: 10.1371/journal.pone.0235140] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 06/09/2020] [Indexed: 01/11/2023] Open
Abstract
Background Due to improved treatment, there is an increasing focus on the reproductive potential of survivors of childhood cancer. Cytotoxic chemotherapy accelerates the decline in the number of primordial follicles within the mammalian ovary at all ages, but effects on the developmental potential of remaining oocytes following prepubertal cancer treatment are unclear. Objectives To investigate whether cyclophosphamide (CY) exposure in the prepubertal period in female mice influences ovarian function and the functional competence of oocytes in adulthood. Methods This study used Swiss albino mice as the experimental model. Female mice were treated with 200 mg/kg CY on either postnatal day 14 (CY14), 21 (CY21) or 28 (CY28) i.e at a prepubertal and 2 young postpubertal ages. At 14 weeks of life, ovarian function, functional competence of oocytes, and embryo quality were assessed. Results The number of primordial follicles decreased significantly in CY14 and CY21 groups compared to control (p < 0.01). The number of oocytes from superovulated was 8.5 ± 1.4, 24.1 ± 2.9 and 26.8 ± 2.1 in CY14, CY21 and CY28 respectively which was significantly lower than control (50.2 ± 3.2; p < 0.001). In vitro culture of CY14 embryos demonstrated only 55.4% blastocyst formation (p < 0.0001) and reduced ability of inner cell mass (ICM) to proliferate in vitro (p < 0.05) at 120 and 216 h post insemination respectively. On the other hand, ICM proliferation was unaltered in 2 young postpubertal ages. Conclusion Our results indicate long-term effects on the developmental competence of oocytes exposed to CY in early but not adult life. These data provide a mechanism whereby long-term fertility can be impaired after chemotherapy exposure, despite the continuing presence of follicles within the ovary, and support the need for fertility preservation in prepubertal girls before alkylating agent exposure.
Collapse
Affiliation(s)
- Sujith Raj Salian
- Department of Clinical Embryology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Shubhashree Uppangala
- Department of Clinical Embryology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Aswathi Cheredath
- Department of Clinical Embryology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Fiona D’Souza
- Department of Clinical Embryology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Guruprasad Kalthur
- Department of Clinical Embryology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Vinod C. Nayak
- Department of Forensic Medicine, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Richard A. Anderson
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| | - Satish Kumar Adiga
- Department of Clinical Embryology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
- * E-mail:
| |
Collapse
|
15
|
Spears N, Lopes F, Stefansdottir A, Rossi V, De Felici M, Anderson RA, Klinger FG. Ovarian damage from chemotherapy and current approaches to its protection. Hum Reprod Update 2020; 25:673-693. [PMID: 31600388 PMCID: PMC6847836 DOI: 10.1093/humupd/dmz027] [Citation(s) in RCA: 323] [Impact Index Per Article: 80.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 07/18/2019] [Accepted: 07/23/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Anti-cancer therapy is often a cause of premature ovarian insufficiency and infertility since the ovarian follicle reserve is extremely sensitive to the effects of chemotherapy and radiotherapy. While oocyte, embryo and ovarian cortex cryopreservation can help some women with cancer-induced infertility achieve pregnancy, the development of effective methods to protect ovarian function during chemotherapy would be a significant advantage. OBJECTIVE AND RATIONALE This paper critically discusses the different damaging effects of the most common chemotherapeutic compounds on the ovary, in particular, the ovarian follicles and the molecular pathways that lead to that damage. The mechanisms through which fertility-protective agents might prevent chemotherapy drug-induced follicle loss are then reviewed. SEARCH METHODS Articles published in English were searched on PubMed up to March 2019 using the following terms: ovary, fertility preservation, chemotherapy, follicle death, adjuvant therapy, cyclophosphamide, cisplatin, doxorubicin. Inclusion and exclusion criteria were applied to the analysis of the protective agents. OUTCOMES Recent studies reveal how chemotherapeutic drugs can affect the different cellular components of the ovary, causing rapid depletion of the ovarian follicular reserve. The three most commonly used drugs, cyclophosphamide, cisplatin and doxorubicin, cause premature ovarian insufficiency by inducing death and/or accelerated activation of primordial follicles and increased atresia of growing follicles. They also cause an increase in damage to blood vessels and the stromal compartment and increment inflammation. In the past 20 years, many compounds have been investigated as potential protective agents to counteract these adverse effects. The interactions of recently described fertility-protective agents with these damage pathways are discussed. WIDER IMPLICATIONS Understanding the mechanisms underlying the action of chemotherapy compounds on the various components of the ovary is essential for the development of efficient and targeted pharmacological therapies that could protect and prolong female fertility. While there are increasing preclinical investigations of potential fertility preserving adjuvants, there remains a lack of approaches that are being developed and tested clinically.
Collapse
Affiliation(s)
- N Spears
- Biomedical Sciences, University of Edinburgh, Edinburgh UK
| | - F Lopes
- Biomedical Sciences, University of Edinburgh, Edinburgh UK
| | | | - V Rossi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - M De Felici
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - R A Anderson
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh UK
| | - F G Klinger
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
16
|
Abstract
OBJECTIVE More than 5,000 premenopausal women are diagnosed with lung cancer annually in the United States. Limited data exist regarding the risk of treatment-related amenorrhea, a surrogate for infertility and early menopause, after systemic therapies for lung cancer. METHODS Premenopausal women diagnosed with lung cancer under age 50 were surveyed at diagnosis and annually thereafter about their menstrual status as a part of the Mayo Clinic Epidemiology and Genetics of Lung Cancer Research Program. Types of lung cancer-directed treatments were recorded, and frequencies of self-reported menopause at each survey were calculated. RESULTS A cohort of 182 premenopausal women were included in this study, with average age at lung cancer diagnosis 43 years (SD 6). Among the 85 patients who received chemotherapy, 64% self-reported that they had become menopausal within a year of diagnosis. Platinum salts were universally included in these chemotherapy regimens, and the majority of these women also received taxanes within 1 year of diagnosis. Only 15% of the 94 patients who did not receive systemic therapy within 1 year of diagnosis experienced self-reported menopause. Three patients received targeted therapy alone, two of whom remained premenopausal at the final qualifying survey, completed a median of 3 years after diagnosis. CONCLUSIONS Chemotherapy for lung cancer patients appears to increase risk of early loss of menses in survivors.
Collapse
|
17
|
Jeelani R, Chatzicharalampous C, Kohan-Ghadr HR, Awonuga A, Joshi N, Morris RT, Abu-Soud HM. Acrolein, a commonly found environmental toxin, causes oocyte mitochondrial dysfunction and negatively affects embryo development. Free Radic Res 2018; 52:929-938. [PMID: 29886754 DOI: 10.1080/10715762.2018.1487559] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Recent studies have revealed that acrolein, a commonly found toxin and a potent metabolite of cyclophosphamide (CTX), can cause deterioration of mouse oocyte quality through a mechanism involving the generation of reactive oxygen species (ROS). We extend these studies to evaluate the effects of acrolein, in varying concentrations, on the oocyte mitochondrial membrane and oocyte apoptosis and its effect on embryo development in vitro. Metaphase II mouse oocytes were exposed for 45 minutes to acrolein and CTX (10 & 25 µM) and mitochondrial dysfunction, a major source of ROS overproduction, was evaluated by the 5,5,6,6-tetrachloro-1,1,3,3-tetraethyl-β-benzimidazolylcarbocyanine iodide (JC-10) mitochondrial membrane potential assay. Treatment with acrolein led to mitochondrial membrane damage as well as induction of apoptosis compared to untreated control (p < 0.05). Similar results were obtained when oocytes were exposed to CTX (p < .05). Subsequently, the effect of acrolein exposure was evaluated by observing in vitro development of embryos after exposure. Acrolein treatment caused higher proportions of arrested and poor-quality embryos, evidenced by irregular cleavage, severe asymmetry of blastomeres, presence of large percentage of anuclear fragments, and dark granularity of the cytoplasm. Development at various durations in culture revealed that optimal embryo growth was significantly inhibited in a dose dependent manner, when compared to control (p < .05). A global model that links acrolein accumulation, generation of ROS, and mitochondrial dysfunction and their effect on oocyte and embryo quality is discussed further. Collectively, understanding the mechanism by which CTX and acrolein impact fertility is helpful in finding potential alternative or supplemental treatment options.
Collapse
Affiliation(s)
- Roohi Jeelani
- a Department of Obstetrics and Gynecology, the CS Mott Center for Human Growth and Development , Wayne State University School of Medicine , Detroit , Michigan , USA
| | - Charalampos Chatzicharalampous
- a Department of Obstetrics and Gynecology, the CS Mott Center for Human Growth and Development , Wayne State University School of Medicine , Detroit , Michigan , USA
| | - Hamid-Reza Kohan-Ghadr
- a Department of Obstetrics and Gynecology, the CS Mott Center for Human Growth and Development , Wayne State University School of Medicine , Detroit , Michigan , USA
| | - Awoniyi Awonuga
- a Department of Obstetrics and Gynecology, the CS Mott Center for Human Growth and Development , Wayne State University School of Medicine , Detroit , Michigan , USA
| | - Narendra Joshi
- b Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology , Wayne State University , Detroit , Michigan , USA
| | - Robert T Morris
- a Department of Obstetrics and Gynecology, the CS Mott Center for Human Growth and Development , Wayne State University School of Medicine , Detroit , Michigan , USA.,c Karmanos Cancer Institute , Detroit , Michigan , USA
| | - Husam M Abu-Soud
- a Department of Obstetrics and Gynecology, the CS Mott Center for Human Growth and Development , Wayne State University School of Medicine , Detroit , Michigan , USA.,d Department of Microbiology, Immunology and Biochemistry , Wayne State University School of Medicine, Detroit , Michigan , USA
| |
Collapse
|
18
|
Jeelani R, Khan SN, Shaeib F, Kohan-Ghadr HR, Aldhaheri SR, Najafi T, Thakur M, Morris R, Abu-Soud HM. Cyclophosphamide and acrolein induced oxidative stress leading to deterioration of metaphase II mouse oocyte quality. Free Radic Biol Med 2017; 110:11-18. [PMID: 28499912 PMCID: PMC6854673 DOI: 10.1016/j.freeradbiomed.2017.05.006] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 05/03/2017] [Accepted: 05/07/2017] [Indexed: 12/30/2022]
Abstract
Cyclophosphamide (CTX) is a chemotherapeutic agent widely used to treat ovarian, breast, and hematological cancers as well as autoimmune disorders. Such chemotherapy is associated with reproductive failure and premature ovarian insufficiency. The mechanism by which CTX and/or its main metabolite, acrolein, affect female fertility remains unclear, but it is thought to be caused by an overproduction of reactive oxygen species (ROS). Here, we investigated the effect of CTX on metaphase II mouse oocytes obtained from treated animals (120mg/kg, 24h of single treatment), and oocytes directly exposed to increasing concentrations of CTX and acrolein (n=480; 0, 5, 10, 25, 50, and 100μM) with and without cumulus cells (CCs) for 45min which correlates to the time of maximum peak plasma concentrations after administration. Oocytes were fixed and subjected to indirect immunofluorescence and were scored based on microtubule spindle structure (MT) and chromosomal alignment (CH). Generation of ROS was evaluated using the Cellular Reactive Oxygen Species Detection Assay Kit. Deterioration of oocyte quality was noted when oocytes were obtained from CTX treated mice along with CTX and acrolein treated oocytes in a dose-dependent manner as shown by an increase in poor scores. Acrolein had an impact at a significantly lower level as compared to CTX, plateau at 10μM versus 50μM, respectively. These variation is are associated with the higher amount of ROS generated with acrolein exposure as compared to CTX (p<0.05). Utilization of antioxidant therapy and acrolein scavengers may mitigate the damaging effects of these compounds and help women undergoing such treatment.
Collapse
Affiliation(s)
- Roohi Jeelani
- Department of Obstetrics and Gynecology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Sana N Khan
- Department of Obstetrics and Gynecology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Faten Shaeib
- Department of Obstetrics and Gynecology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Hamid-Reza Kohan-Ghadr
- Department of Obstetrics and Gynecology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Sarah R Aldhaheri
- Department of Obstetrics and Gynecology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Tohid Najafi
- Department of Obstetrics and Gynecology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Mili Thakur
- Department of Obstetrics and Gynecology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI 48201, USA; Division of Genetic, Genomic and Metabolic Disorders, Department of Pediatrics and Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Robert Morris
- Department of Obstetrics and Gynecology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI 48201, USA; Karmanos Cancer Institute, Detroit, MI 48201, USA
| | - Husam M Abu-Soud
- Department of Obstetrics and Gynecology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI 48201, USA; Department of Biochemistry and Molecular Biology, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| |
Collapse
|
19
|
Kim IR, Kim SH, Ok ON, Kim SH, Lee S, Choi E, Kim SJ, Yoon DH, Lee MH. Sexual problems in male vs. female non-Hodgkin lymphoma survivors: prevalence, correlates, and associations with health-related quality of life. Ann Hematol 2017; 96:739-747. [DOI: 10.1007/s00277-017-2940-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 01/31/2017] [Indexed: 01/17/2023]
|
20
|
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: 7.1] [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
|
21
|
Brunner HI, Bishnoi A, Barron AC, Houk LJ, Ware A, Farhey Y, Mongey AB, Strife CF, Graham TB, Passo MH. Disease outcomes and ovarian function of childhood-onset systemic lupus erythematosus. Lupus 2016; 15:198-206. [PMID: 16686258 DOI: 10.1191/0961203306lu2291oa] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The objective of this study was to determine the medical outcomes including the ovarian function childhood-onset SLE (cSLE). The medical records of all patients diagnosed with cSLE in the Greater Cincinnati area between 1981 and 2002 were reviewed. Patient interviews were performed to obtain additional information on current medication regimens, disease activity [SLE Disease Activity Index (SLEDAI-2k)], and damage [Systemic Lupus International Collaborating Clinics/American College of Rheumatology Damage Index (SDI)]. The occurence of premature ovarian failure (POF) and reduction of the ovarian reserve was assessed by timed gonadotropin levels. There were 77 patients (F: M = 70: 7, 53% Caucasian, 45% African-American and 2% Asian) with a mean age at diagnosis of 14.6 years. Nine patients died (88.3% survival) during the mean follow-up of 7.1 years (standard deviation [SD] 5.6) and 88% of the patients continued to have active disease (SLEDAI-2k mean/SD: 6.6/6.7), with 42% of them having disease damage (SDI mean/SD: 1.62/2.1); Non-Caucasian patients had higher disease activity (mean SLEDAI-2k: 10 versus 3.4; P < 0.0001) and more disease damage (mean SDI: 2.1 versus 1.2; P < 0.02) than Caucasian patients. Cyclophosphamide was given to 47% of the patients during the course of their disease and associated with the presence of significantly reduced ovarian reserve (RR = 2.8; 95% CI: 1.7-4.8; P = 0.026). Patient mortality and disease damage with cSLE continue to be high. Although overt POF with cyclophosphamide exposure is rare, it is a risk factor for significantly decreased ovarian reserve cSLE.
Collapse
Affiliation(s)
- H I Brunner
- Cincinnati Children's Hospital Medical Center, University of Cincinnati, William Rowe Division of Rheumatology, OH 45229, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Chan JL, Johnson LNC, Efymow BL, Sammel MD, Gracia CR. Outcomes of ovarian stimulation after treatment with chemotherapy. J Assist Reprod Genet 2015; 32:1537-45. [PMID: 26400507 PMCID: PMC4615911 DOI: 10.1007/s10815-015-0575-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 09/07/2015] [Indexed: 11/28/2022] Open
Abstract
PURPOSE Chemotherapeutic agents have a known gonadotoxic effect; however, it is difficult to predict the impact they may have on ovarian stimulation. The objective of this study was to evaluate response to ovarian stimulation in patients exposed to chemotherapy compared with patients who were chemotherapy-naïve. METHODS A retrospective cohort study of 130 patients with cancer or autoimmune disease was performed. Demographics, ovarian reserve, ovarian response and stimulation parameters, and oocyte data were compared between patients who were pre- and post-chemotherapy. Logistic regression modeling was performed to identify risk factors for cancellation and low oocyte yield, adjusting for confounders as appropriate. RESULTS Antral follicle count (AFC) was significantly lower in post-chemo patients (9 vs. 17, p < 0.001). Post-chemotherapy patients were more likely to be cancelled during stimulation (23 vs. 4 %, p = 0.003). Among those that went to retrieval, there was no difference in total number of oocytes (10 vs. 10, p = 0.31) or mature oocytes retrieved (8 vs. 8, p = 0.38), despite higher starting (300 vs. 450 IU, p < 0.001) and total gonadotropin (3075 vs. 4612.5 IU, p = 0.008) doses in post-chemotherapy patients. Low AFC (≤6) was associated with cycle cancellation (OR 7.7, 95 % CI 1.8-33.2) and low oocyte yield (<6) (OR 5.4, 95 % CI 1.6-17.7). CONCLUSIONS Patients post-chemotherapy have lower AFC compared with the chemotherapy-naïve and have higher cancellation rates. Among those who underwent oocyte retrieval, oocyte yield was similar in both groups. Low AFC was most strongly associated with cycle cancellation and oocyte yield. Post-chemotherapy patients had higher rates of cycle cancellation but did equally well as pre-chemotherapy patients if they reached retrieval.
Collapse
Affiliation(s)
- Jessica L Chan
- Department of Obstetrics and Gynecology, University of Pennsylvania, 3701 Market Street, Suite 800, Philadelphia, PA, 19104, USA.
| | - Lauren N C Johnson
- Department of Obstetrics and Gynecology, University of Pennsylvania, 3701 Market Street, Suite 800, Philadelphia, PA, 19104, USA
| | - Brenda L Efymow
- Department of Obstetrics and Gynecology, University of Pennsylvania, 3701 Market Street, Suite 800, Philadelphia, PA, 19104, USA
| | - Mary D Sammel
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, 423 Guardian Drive, 605 Blockley Hall, Philadelphia, PA, 19104, USA
| | - Clarisa R Gracia
- Department of Obstetrics and Gynecology, University of Pennsylvania, 3701 Market Street, Suite 800, Philadelphia, PA, 19104, USA
| |
Collapse
|
23
|
Gui T, Yuan G, Shen K, Cao D, Yang J, Wu M, Lang J. Protective effect of gonadotropin-releasing hormone analog on the ovarian reserve in rats receiving cyclophosphamide treatment. Onco Targets Ther 2015; 8:661-7. [PMID: 25848306 PMCID: PMC4376258 DOI: 10.2147/ott.s78729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Objective The aim of the study reported here was to investigate the protective effect of gonadotropin-releasing hormone analog (GnRHa) against cyclophosphamide (CTX)-induced gonadotoxicity. Methods Eighty Fischer 344 rats were divided randomly into four groups (20 per group). One group received normal saline, one GnRHa, one CTX, and one GnRHa+CTX. Several parameters were used to observe the ovarian reserve, including ovary weight, follicle number and diameter, concentrations of estradiol (E2) and follicle-stimulating hormone (FSH), and expressions of sex hormone receptors. Results When treatment was finished, the number of small follicles in the GnRHa+CTX group was significantly higher than in the CTX-alone group. Thirty days after treatment, the ovary weight, percentage of small follicles, mean follicular diameter, and serum concentrations of E2 and FSH in the GnRHa+CTX group all recovered, approaching normal levels. Sex hormone receptors did not show significant differences between the four groups. Conclusion Combination treatment with GnRHa could prevent CTX-induced damage to ovarian reserve.
Collapse
Affiliation(s)
- Ting Gui
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Beijing, People's Republic of China
| | - Guangwen Yuan
- Department of Gynecologic Oncology, Cancer Hospital, Peking Union Medical College, Chinese Academy of Medical Science, Beijing, People's Republic of China
| | - Keng Shen
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Beijing, People's Republic of China
| | - Dongyan Cao
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Beijing, People's Republic of China
| | - Jiaxin Yang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Beijing, People's Republic of China
| | - Ming Wu
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Beijing, People's Republic of China
| | - Jinghe Lang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Beijing, People's Republic of China
| |
Collapse
|
24
|
Roti Roti EC, Leisman SK, Abbott DH, Salih SM. Acute doxorubicin insult in the mouse ovary is cell- and follicle-type dependent. PLoS One 2012; 7:e42293. [PMID: 22876313 PMCID: PMC3410926 DOI: 10.1371/journal.pone.0042293] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 07/06/2012] [Indexed: 01/20/2023] Open
Abstract
Primary ovarian insufficiency (POI) is one of the many unintended consequences of chemotherapy faced by the growing number of female cancer survivors. While ovarian repercussions of chemotherapy have long been recognized, the acute insult phase and primary sites of damage are not well-studied, hampering efforts to design effective intervention therapies to protect the ovary. Utilizing doxorubicin (DXR) as a model chemotherapy agent, we defined the acute timeline for drug accumulation, induced DNA damage, and subsequent cellular and follicular demise in the mouse ovary. DXR accumulated first in the core ovarian stroma cells, then redistributed outwards into the cortex and follicles in a time-dependent manner, without further increase in total ovarian drug levels after four hours post-injection. Consistent with early drug accumulation and intimate interactions with the blood supply, stroma cell-enriched populations exhibited an earlier DNA damage response (measurable at 2 hours) than granulosa cells (measurable at 4 hours), as quantified by the comet assay. Granulosa cell-enriched populations were more sensitive however, responding with greater levels of DNA damage. The oocyte DNA damage response was delayed, and not measurable above background until 10-12 hours post-DXR injection. By 8 hours post-DXR injection and prior to the oocyte DNA damage response, the number of primary, secondary, and antral follicles exhibiting TUNEL (terminal deoxynucleotidyl transferase dUTP nick end labeling)-positive granulosa cells plateaued, indicating late-stage apoptosis and suggesting damage to the oocytes is subsequent to somatic cell failure. Primordial follicles accumulate significant DXR by 4 hours post-injection, but do not exhibit TUNEL-positive granulosa cells until 48 hours post-injection, indicating delayed demise. Taken together, the data suggest effective intervention therapies designed to protect the ovary from chemotherapy accumulation and induced insult in the ovary must act almost immediately to prevent acute insult as significant damage was seen in stroma cells within the first two hours.
Collapse
Affiliation(s)
- Elon C. Roti Roti
- University of Wisconsin-Madison, Department of Obstetrics and Gynecology, Reproductive Endocrinology and Infertility Division, Madison, Wisconsin, United States of America
| | - Scott K. Leisman
- University of Wisconsin-Madison, Department of Obstetrics and Gynecology, Reproductive Endocrinology and Infertility Division, Madison, Wisconsin, United States of America
| | - David H. Abbott
- University of Wisconsin-Madison, Department of Obstetrics and Gynecology, Reproductive Endocrinology and Infertility Division, Madison, Wisconsin, United States of America
| | - Sana M. Salih
- University of Wisconsin-Madison, Department of Obstetrics and Gynecology, Reproductive Endocrinology and Infertility Division, Madison, Wisconsin, United States of America
| |
Collapse
|
25
|
Low JJ, Ilancheran A, Ng JS. Malignant ovarian germ-cell tumours. Best Pract Res Clin Obstet Gynaecol 2012; 26:347-55. [DOI: 10.1016/j.bpobgyn.2012.01.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Accepted: 01/10/2012] [Indexed: 11/26/2022]
|
26
|
Abstract
At birth, the mammalian ovary contains a finite number of primordial follicles, which once depleted, cannot be replaced. Xenobiotic exposures can destroy primordial follicles resulting in premature ovarian failure and, consequently, early entry into menopause. A number of chemical classes can induce premature ovarian failure, including environmental, chemotherapeutic and industrial exposures. While our knowledge on the mechanistic events that occur in the ovary with chemical exposures is increasing, our understanding of the ovary's capacity to metabolize such compounds is less established. This review will focus on three chemicals for which information on ovarian metabolism is known: trichloroethylene, 7,12-dimethylbenz[a]anthracene and 4-vinylcyclohexene. The current state of understanding of ovarian bioactivation and detoxification processes for each will be described.
Collapse
|
27
|
Arden-Close E, Eiser C, Pacey A. Sexual functioning in male survivors of lymphoma: a systematic review (CME). J Sex Med 2011; 8:1833-41. [PMID: 21324087 DOI: 10.1111/j.1743-6109.2011.02209.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION The lymphomas (Hodgkin's lymphoma [HL] and non-Hodgkin's lymphoma [NHL]) are among the most common cancers affecting men under 45 years. Survival rates are now excellent, but treatment is associated with a number of side effects including sexual dysfunction with potential implications for compromised quality of life (QoL). AIMS To address the (i) prevalence of sexual dysfunction among lymphoma survivors relative to the general population, survivors of other cancers, and in survivors of HL and NHL; and (ii) relationships between sexual functioning and disease and treatment, demographic, and psychological variables. METHODS Inclusion criteria were quantitative studies that focused on adult male survivors of lymphoma and included a comparison group and presented results separately for HL and NHL. Standardized systematic searches were used. Information about design, sample size, age, time since diagnosis, type of treatment, comparison group, measures, and findings were extracted from eligible studies. RESULTS Ten articles met the inclusion criteria, of which, nine included patients with HL only, and one included patients with HL or NHL. Sexual function was compromised relative to the general population, better than testicular cancer survivors, and worse than leukemia survivors. Depression was consistently associated with sexual dysfunction. There was evidence that chemotherapy, relapse, reduced testosterone levels, older age at survey, and worse physical QoL were associated with worse sexual function. CONCLUSIONS Conclusions are limited by methodological issues including lack of utilization of standardized measures of sexual function and longitudinal research. Even so, there is evidence of sexual dysfunction among lymphoma survivors. Clinicians need to be sensitive to these issues. Future longitudinal work is necessary to determine the likelihood of recovery.
Collapse
Affiliation(s)
- Emily Arden-Close
- University of Sheffield-Department of Psychology, Sheffield, UK University of Sheffield-School of Medicine and Biomedical Sciences, Academic Unit of Reproductive and Developmental Medicine, Sheffield, UK
| | | | | |
Collapse
|
28
|
Abstract
The treatment of acute leukemia is usually similar in women and men. The outcome is also generally the same. However, diagnosis in women poses additional challenges in clinical practice such as leukemia following breast or ovarian cancers, prevention of abnormal uterine bleeding in premenopausal females, treatment during pregnancy related-problems in long-term survivors. All these special issues are addressed in this review of the literature.
Collapse
|
29
|
Su HI, Sammel MD, Velders L, Horn M, Stankiewicz C, Matro J, Gracia CR, Green J, DeMichele A. Association of cyclophosphamide drug-metabolizing enzyme polymorphisms and chemotherapy-related ovarian failure in breast cancer survivors. Fertil Steril 2009; 94:645-54. [PMID: 19376514 DOI: 10.1016/j.fertnstert.2009.03.034] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2008] [Revised: 03/04/2009] [Accepted: 03/04/2009] [Indexed: 10/20/2022]
Abstract
OBJECTIVE To determine if genetic variation in chemotherapy metabolism are associated with risk of ovarian failure in breast cancer patients after adjuvant chemotherapy. DESIGN Prospective cohort study. SETTING Comprehensive cancer center. PATIENT(S) Early-stage breast cancer patients who were premenopausal at cancer diagnosis and treatment. INTERVENTION(S) None. MAIN OUTCOMES MEASURE(S) Chemotherapy-related ovarian failure (CROF). RESULT(S) A total of 127 breast cancer subjects who were premenopausal at cancer diagnosis and underwent cyclophosphamide-based chemotherapy were genotyped for nine single-nucleotide polymorphisms (SNPs) in enzymes involved in cyclophosphamide activation (CYP3A4, CYP2B6, CYP3A5) and detoxification (GSTA1, GSTM1, GSTP1, GSTT1). Median age at chemotherapy was 43.2 years. Median follow-up after chemotherapy was 5.2 years. For the entire cohort, there was no significant association between CROF and SNPs. However, the association between CROF and SNPs was modified by age at chemotherapy. In subjects younger than 45 years old at chemotherapy, CYP3A4 *1B variants had significantly longer time to CROF than CYP3A4 *1A homozygotes in an adjusted multivariable Cox model. Age and tamoxifen use were also independently associated with CROF. CONCLUSION(S) A common SNP in a cyclophosphamide drug-metabolizing enzyme appears to be related to ovarian failure after cyclophosphamide-based chemotherapy in young women with breast cancer. Larger prospective studies to validate these results should be directed toward women younger than 45 years of age at chemotherapy.
Collapse
Affiliation(s)
- H Irene Su
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Pennsylvania School of Medicine, Philadelphia, PA 19146, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Stroud JS, Mutch D, Rader J, Powell M, Thaker PH, Grigsby PW. Effects of cancer treatment on ovarian function. Fertil Steril 2008; 92:417-27. [PMID: 18774559 DOI: 10.1016/j.fertnstert.2008.07.1714] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2008] [Revised: 06/23/2008] [Accepted: 07/09/2008] [Indexed: 12/16/2022]
Abstract
Causes of primary ovarian failure are reviewed, focusing specifically on cancer treatment-related modalities. Strategies and future directions for protection of the ovaries during cancer therapy, including ovarian transposition, and conformal radiation techniques are presented.
Collapse
Affiliation(s)
- Jaymeson S Stroud
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | | | | | | | | | | |
Collapse
|
31
|
Gershenson DM, Miller AM, Champion VL, Monahan PO, Zhao Q, Cella D, Williams SD. Reproductive and sexual function after platinum-based chemotherapy in long-term ovarian germ cell tumor survivors: a Gynecologic Oncology Group Study. J Clin Oncol 2007; 25:2792-7. [PMID: 17602084 DOI: 10.1200/jco.2006.08.4590] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To compare malignant ovarian germ cell tumor survivors with a matched control group of females on menstrual and reproductive outcomes, sexual functioning, and dyadic adjustment. PATIENTS AND METHODS Eligible patients met the following criteria: (1) history of malignant ovarian germ cell tumor; (2) treatment with surgery plus platinum-based chemotherapy; (3) age at least 18 years and continuously disease-free with minimum follow-up of 2 years; (4) capability of completing questionnaire and telephone interview; and (5) completion of written informed consent. The control group was drawn from acquaintances recommended by survivors and matched for age, race, and education. Scales with established reliability and validity were used to measure quality-of-life concepts of sexual functioning and social networks. RESULTS One hundred thirty-two survivors and 137 controls completed the study. Of 132 survivors, 71 (53.8%) had fertility-sparing surgery. Of fertile survivors, 62 (87.3%) reported still having menstrual periods. Twenty-four survivors reported 37 offspring after cancer treatment. Compared with controls, survivors had significantly greater reproductive concerns (P < .0001), less sexual pleasure (P = .003), and lower scores on the total Sexual Activity Scale Score (P = .001). However, survivors had better dyadic consensus (P = .004), dyadic satisfaction (P = .005), and dyadic cohesion (P = .014). CONCLUSION Women who had fertility-sparing surgery were very likely to retain menstrual function and fertility after chemotherapy. Although there is some increase in gynecologic symptoms and diminution in sexual pleasure, survivors tended to have stronger, more positive relationships with significant others.
Collapse
Affiliation(s)
- David M Gershenson
- Department of Gynecologic Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77230-1439, USA.
| | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
Systemic lupus erythematosus (SLE) is a prototypical autoimmune disease, characterised by flares of rampant inflammation that can threaten, in an unpredictable manner, almost any organ in the body. Current standard of care is largely empiric, involving the use of corticosteroids and toxic immune suppressive agents that are widely acknowledged to have unacceptable side effects for long-term use. Recently, there have been significant advances in understanding the nature of some fundamental immune imbalances underlying the complicated clinical manifestations of SLE. Nevertheless attempts to develop and test more targeted, and potentially safer immune-modulating drugs for lupus have encountered significant obstacles, due to the lack of validated biological markers for disease flare and remission, and difficulties in the clinical assessment of the heterogeneous patients. In support of renewed interest in drug development for lupus, large collaborative groups have formed, and efforts are underway to develop objective biomarkers for SLE as well as to improve the standardisation and reproducibility of clinical outcome measures in multi-centre trials.
Collapse
Affiliation(s)
- Joan T Merrill
- Oklahoma Medical Research Foundation, Clinical Pharmacology Research Program, 825 Northeast 13 St., Oklahoma City, OK 73104, USA.
| |
Collapse
|
33
|
Merrill JT, Erkan D, Buyon JP. Challenges in bringing the bench to bedside in drug development for sle. Nat Rev Drug Discov 2004; 3:1036-46. [PMID: 15573102 DOI: 10.1038/nrd1577] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
It is now widely accepted that the current standard of care for systemic lupus erythematosus (SLE) patients is inadequate. There has not been a new medication approved for this disease in thirty years. Attempts to develop and test new drugs have been ongoing since the mid-1990s, but have encountered formidable obstacles. Current models for lupus pathogenesis have provided a theoretical framework for understanding how heterogeneous genetic defects might combine in various ways to increase susceptibility to SLE in different individuals, and could have important implications for new drug development. With the current burst of drug discovery and increased public awareness of SLE, the impetus to overcome these obstacles has never been greater.
Collapse
Affiliation(s)
- Joan T Merrill
- Clinical Pharmacology Research Program, Oklahoma Medical Research Foundation, 825 Northeast 13th Street Oklahoma City, Oklahoma 73104, USA
| | | | | |
Collapse
|
34
|
Tilly JL, Kolesnick RN. Realizing the promise of apoptosis-based therapies: separating the living from the clinically undead. Cell Death Differ 2003; 10:493-5. [PMID: 12728247 DOI: 10.1038/sj.cdd.4401217] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
35
|
Minton SE, Munster PN. Chemotherapy-induced amenorrhea and fertility in women undergoing adjuvant treatment for breast cancer. Cancer Control 2002; 9:466-72. [PMID: 12514564 DOI: 10.1177/107327480200900603] [Citation(s) in RCA: 123] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The majority of women diagnosed with early-stage breast cancer have an excellent long-term prognosis, but many will undergo temporary or permanent chemotherapy-induced amenorrhea. METHODS While breast cancer is more common in older women, about 1 in 200 women under the age of 40 is at risk to develop breast cancer. Many of these women benefit from chemotherapy but are afraid to risk the opportunity to bear children. The authors review the current studies on the impact of adjuvant chemotherapy on amenorrhea and fertility in women with breast cancer. RESULTS The likelihood of amenorrhea is based on the specific adjuvant chemotherapy regimen administered and the age of the patient. Future childbirth is a viable option for women treated for breast cancer at an early stage. While the use of tamoxifen as a hormonal therapy in premenopausal breast cancer is now the standard of care, no conclusive data confirm the benefit of GnRH agonists in adjuvant therapy after treatment with chemotherapy followed by tamoxifen. CONCLUSIONS As more women over the age of 35 consider pregnancy, fertility issues are becoming important areas of investigation for the adjuvant treatment of breast cancer. Whether chemotherapy-induced amenorrhea has a prognostic effect remains unclear, and further studies are warranted.
Collapse
Affiliation(s)
- Susan E Minton
- Comprehensive Breast Cancer Program, H Lee Moffitt Cancer Center Research Institute, Tampa, Florida 33612, USA.
| | | |
Collapse
|
36
|
Kwon JS, Case AM. Effects of cancer treatment on reproduction and fertility. JOURNAL OF OBSTETRICS AND GYNAECOLOGY CANADA 2002; 24:619-27. [PMID: 12196840 DOI: 10.1016/s1701-2163(16)30192-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE (1) To review the effects of cancer treatment on reproduction and fertility in both women and men and (2) to review current and new techniques that could be used to preserve fertility and hormone production in women who have undergone cancer treatment. DATA SOURCES Entries to MEDLINE and CANCERLIT through to October 2001 were searched for evidence relevant to this article. METHODS OF STUDY SELECTION This document is primarily based on large retrospective cohort series and case reports, as no randomized trials were available. TABULATION, INTEGRATION, AND RESULTS The first part of this review article focuses on the effects of radiation and chemotherapy on reproductive function and innovative medical and surgical techniques employed to minimize these effects. The second part of this article describes conservative methods of managing cervical, endometrial, and ovarian cancer for women who wish to preserve their fertility. CONCLUSIONS Over the last decade, there has been a trend toward trying to preserve reproductive function in young patients with malignancy. Information acquired over the last century on the reproductive effects of radiation and chemo-therapy has enabled us to tailor our treatment of various malignancies affecting young patients. As we learn more about the biology and clinical behaviour of gynaecologic malignancies, we are able to modify surgical management to preserve fertility in select cases. All of these trends are in keeping with our goal to optimize quality of life in cancer care without compromising safety or survival.
Collapse
Affiliation(s)
- Janice S Kwon
- Department of Obstetrics and Gynecology, University of Western Ontario, London, ON, Canada
| | | |
Collapse
|
37
|
Blumenfeld Z. Preservation of fertility and ovarian function and minimalization of chemotherapy associated gonadotoxicity and premature ovarian failure: the role of inhibin-A and -B as markers. Mol Cell Endocrinol 2002; 187:93-105. [PMID: 11988316 DOI: 10.1016/s0303-7207(01)00712-2] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
BACKGROUND Following the improved long term survival in young women with lymphoma and leukemia undergoing chemotherapy, the preservation of future fertility has been the focus of recent interest. The investigational endeavors of ovarian cryopreservation awaits the clinical experience of in-vitro maturation of thawed primordial follicles, their in-vitro fertilization and ET. Although promising, this experience is not available yet. Moreover, the risk of possible reimplantation of malignant stem cells with the thawed cryopreserved ovary has been raised, following experimental animal observations. Therefore, until these innovative endeavors prove successful, and in parallel to them we attempted to minimize the gonadotoxic effect of chemotherapy by the co-treatment with a GnRH agonistic analogue to induce a temporary prepubertal milieu. Whereas, inhibin-B concentrations in serum may reflect the ovarian granulosa cell compartment, inhibin-A reflects luteal function. The immunoreactive inhibin-A and -B in these patients, before, during, and following the gonadotoxic chemotherapy were measured. METHODS A prospective clinical protocol was undertaken in 55 women with lymphoma, aged 15-40 years, ten with leukemia and eight undergoing chemotherapeutic treatments for non malignant diseases such as systemic lupus erythematosus (SLE) or other autoimmune diseases. A monthly injection of depot D-TRP6-GnRH-a was administered from before starting the chemotherapy until its conclusion, up to a maximum of 6 months. Hormonal profile (FSH, LH, E2, T, P4, IGF-1, IGF-BP3, and PRL) was taken before starting the GnRH-a/chemotherapy co-treatment, and monthly thereafter, until resuming spontaneous ovulation. This group was compared with a control group of 55 women who have been treated with similar chemotherapy. Inhibin-A and -B immunoactivity was measured by an ELISA commercial kit (Serotec). RESULTS Whereas, all but three (40- and 36-year-old) of the surviving patients with GnRH-a/chemotherapy co-treatment group resumed spontaneous ovulation and menses within 12 months, less than half of the patients in the control group (chemotherapy without GnRH-a co-treatment) resumed ovarian function and regular cyclic activity (P<0.05). The remaining 56% experienced premature ovarian failure (POF). Temporary increased FSH concentrations were experienced by about a third of the patients resuming cyclic ovarian function, suggesting a reversible ovarian damage in a larger proportion of women than those experiencing POF. Inhibin-A and -B decreased during the GnRH-a/chemotherapy co-treatment but increased to normal levels in patients who resumed regular ovarian cyclicity, and/or spontaneously conceived, as compared with low levels in those who developed POF. CONCLUSIONS If these preliminary data are consistent in a larger group of patients, inhibin-A or -B concentrations may serve as prognostic factors for predicting the resumption of ovarian function, in addition to the levels of FSH, LH and E2. The GnRH-a co-treatment should be considered in every woman in the reproductive age receiving chemotherapy, in addition to ART, and to the investigational attempts of ovarian cryopreservation for future in-vitro maturation, autotransplantation, or xenotransplantation.
Collapse
Affiliation(s)
- Z Blumenfeld
- Reproductive Endocrinology and Infertility Section, Department of Obstetrics and Gynecology, Rambam Medical Center, Technion-Faculty of Medicine, Technion-Israel Institute of Technology, 31096, Haifa, Israel.
| |
Collapse
|
38
|
Daikh DI, Wofsy D. Cutting edge: reversal of murine lupus nephritis with CTLA4Ig and cyclophosphamide. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 166:2913-6. [PMID: 11207238 DOI: 10.4049/jimmunol.166.5.2913] [Citation(s) in RCA: 170] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Cyclophosphamide (CTX) prevents progression of nephritis and prolongs survival in (NZB x NZW)F(1) (B/W) mice and is used to treat humans with lupus nephritis. To compare the efficacy of CTLA4Ig with CTX and determine whether there is an incremental benefit to combining CTLA4Ig with CTX, we treated B/W mice with CTX, CTLA4Ig, or both agents. In mice with mild renal disease, treatment delayed the onset of proteinuria and prolonged survival in all groups. In mice with advanced renal disease, treatment with both agents reduced proteinuria in 71% of mice, whereas mice treated with either agent alone had no such improvement. Survival was also markedly improved among mice treated with both agents. Thus, combination treatment with CTX and CTLA4Ig is more effective than either agent alone in reducing renal disease and prolonging survival of B/W mice with advanced nephritis. This striking reversal of proteinuria is unprecedented in animal models of SLE.
Collapse
Affiliation(s)
- D I Daikh
- Department of Medicine, Department of Veterans Affairs Medical Center and the University of California, San Francisco, CA 94121, USA.
| | | |
Collapse
|
39
|
Irving JA, McFarland DF, Stuart DS, Gilks CB. Mitotic arrest of endometrial epithelium after paclitaxel therapy for breast cancer. Int J Gynecol Pathol 2000; 19:395-7. [PMID: 11109173 DOI: 10.1097/00004347-200010000-00018] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
We report the histopathologic findings in endometrial curettings from a 31-year-old woman with dysfunctional uterine bleeding who had received paclitaxel therapy for breast carcinoma. Paclitaxel, a member of the taxane family of antineoplastic agents that is used in the treatment of breast carcinoma, ovarian carcinoma, and non-small cell lung carcinoma, acts by the simultaneous promotion of tubulin assembly into microtubules and inhibition of microtubule disassembly. The curettings in this case showed fragmented menstrual phase endometrium with striking numbers of mitotic figures. Cell divisions were arrested in metaphase. Glandular epithelial cells showed strong immunoreactivity for bcl-2 and MIB-1. We attribute this marked morphologic effect to paclitaxel-induced mitotic arrest of the endometrium.
Collapse
Affiliation(s)
- J A Irving
- Department of Pathology, University of British Columbia, Vancouver, Canada
| | | | | | | |
Collapse
|
40
|
Low JJH, Perrin LC, Crandon AJ, Hacker NF. Conservative surgery to preserve ovarian function in patients with malignant ovarian germ cell tumors. Cancer 2000. [DOI: 10.1002/1097-0142(20000715)89:2<391::aid-cncr26>3.0.co;2-v] [Citation(s) in RCA: 161] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
41
|
Kanazawa K, Suzuki T, Sakumoto K. Treatment of malignant ovarian germ cell tumors with preservation of fertility: reproductive performance after persistent remission. Am J Clin Oncol 2000; 23:244-8. [PMID: 10857886 DOI: 10.1097/00000421-200006000-00007] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
To describe our experience with malignant ovarian germ cell tumors with special reference to reproductive performance after remission, medical records of 31 patients were reviewed. The mean age at diagnosis was 18.6 years. Tumor by stage was I in 16 cases, II in 5, III in 5, IV in 3, and recurrence in 2. Histology was dysgerminoma in 7 cases, yolk sac tumor in 10, immature teratoma in 7, choriocarcinoma in 1, and mixed-type tumor in 6. Conservative surgery for fertility preservation was performed in 21 cases. Postoperative chemotherapy was given to all cases except two with stage Ia dysgerminoma. Of 31 cases, 4 including one fertility-preserved case died of disease. The other 27 cases including 20 fertility-preserved cases were successfully treated. Twenty-five cases (92.6%) have been followed longer than 60 months and 13 cases (48.1%) longer than 120 months. By the last follow-up, 8 of the 20 fertility-preserved cases delivered a total of 9 normal babies. Of the remaining 12 nonpregnant cases, 3 married, 9 have had regular menses, and 3 have had menstrual problems. Two of the latter three cases have been in hypergonadotropic anovulatory cycles. One patient has been diagnosed with tubal infertility caused by peritubal adhesion. Thus, management of the disease with fertility preservation is safe and the majority of patients can attain or retain normal ovarian function and reproductive potential.
Collapse
Affiliation(s)
- K Kanazawa
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of the Ryukyus, Okinawa, Japan
| | | | | |
Collapse
|
42
|
Abstract
Remarkable advances have been made in the treatment of cancers that afflict patients of the reproductive age. Many survivors must now face the effects on gonadal function and have concerns about reproductive capacity. The sequelae of different modalities of cancer therapy specifically addressing surgery, chemotherapy, and radiotherapy on reproductive system are reviewed. Assisted reproductive technologies, prenatal diagnosis methods, and contraception counseling are briefly summarized in conclusion.
Collapse
Affiliation(s)
- M S Shahin
- Department of Obstetrics and Gynecology, University of Iowa Hospitals and Clinics, Iowa City, USA
| | | |
Collapse
|
43
|
Yeung SC, Chiu AC, Vassilopoulou-Sellin R, Gagel RF. The endocrine effects of nonhormonal antineoplastic therapy. Endocr Rev 1998; 19:144-72. [PMID: 9570035 DOI: 10.1210/edrv.19.2.0328] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- S C Yeung
- Joint Baylor College of Medicine-The University of Texas M. D. Anderson Cancer Center Endocrinology Fellowship Program, Houston 77030, USA
| | | | | | | |
Collapse
|
44
|
Headley JA, Theriault RL, LeBlanc AD, Vassilopoulou-Sellin R, Hortobagyi GN. Pilot study of bone mineral density in breast cancer patients treated with adjuvant chemotherapy. Cancer Invest 1998; 16:6-11. [PMID: 9474245 DOI: 10.3109/07357909809039747] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The objective of this cross-sectional study was to determine lumbar spine bone mineral density (BMD) in breast cancer patients previously treated with adjuvant chemotherapy. Sixteen of 27 patients who received adjuvant chemotherapy became permanently amenorrheic as a result of chemotherapy. BMD was measured at the lumbar spine using dual energy X-ray absorptiometry (DEXA). Chemotherapy drugs and dosages along with a history of risk factors for reduced bone density including activity level, tobacco and/or alcohol use, metabolic bone disease, family history, and hormone exposure were identified. Results showed that women who became permanently amenorrheic as a result of chemotherapy had BMD 14% lower than women who maintained menses after chemotherapy. Chemotherapy-treated women who maintained ovarian function had normal BMD. This study suggests that women who have premature menopause as a result of chemotherapy for breast cancer are at increased risk of bone loss and may be at risk for early development of osteoporosis. Women who maintain menses do not appear to be at risk for accelerated trabecular bone loss.
Collapse
Affiliation(s)
- J A Headley
- University of Texas Health Science Center-Houston School of Nursing, UT M.D. Anderson Cancer Center, Baylor College of Medicine, USA
| | | | | | | | | |
Collapse
|
45
|
Abstract
Infertility represents one of the main remote sequelae of cytotoxic chemotherapy given for various malignant diseases. The impairment of gonadal function after cytotoxic chemotherapy is more frequent in the male than in the female. Because dividing cells are more sensitive to the cytotoxic effects of alkylating agents than are cells at rest, it has been hypothesized that inhibition of the pituitary-gonadal axis by gonadotropin-releasing hormone (GnRH) agonists would render the germinal epithelium less susceptible to the cytotoxic effects of chemotherapy. This hypothesis has not been thoroughly clinically tested until recently, although several investigators have demonstrated that GnRH-agonistic analogues (GnRH-a) inhibit chemotherapy-induced ovarian follicular depletion in the rat and Rhesus monkeys. Based on this rationale, we have undertaken a prospective evaluation to determine whether GnRH-a administration during combination chemotherapy for Hodgkin's and non-Hodgkin's lymphoma could prevent posttreatment ovarian damage in women by inducing a temporary prepubertal hormonal milieu. While over 93% of the surviving patients in the GnRH-a and chemotherapy group resumed spontaneous ovulation and menses, less than 40% of the women in the control group of chemotherapy without the GnRH-a cotreatment resumed normal ovarian cyclic activity. More than 60% of the women experienced premature ovarian failure (POF) in the chemotherapy alone group. Our preliminary results suggest that GnRH-a cotreatment protects against POF during cytotoxic chemotherapy. The GnRH-a and chemotherapy cotreatment may be also suggested for young women treated by cyclophosphamide pulse therapy or other gonadotoxic treatments for systemic lupus erythematosus, organ transplantation and other autoimmune diseases. The technology of cryopreservation of human ova for future fertility in these patients awaits clinical validation and substantiation. This review discusses possibilities to prevent gonadal damage induced by cytotoxic therapy and presents the clinical data currently available.
Collapse
Affiliation(s)
- Z Blumenfeld
- Department of Obstetrics and Gynecology, Rambam Medical Center, The Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.
| | | |
Collapse
|
46
|
Goldwasser F, Pico JL, Cerrina J, Fernandez H, Pons JC, Cosset JM, Hayat M. Successful chemotherapy including epirubicin in a pregnant non-Hodgkin's lymphoma patient. Leuk Lymphoma 1995; 20:173-6. [PMID: 8750642 DOI: 10.3109/10428199509054772] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
We report a case of non-Hodgkin's lymphoma (NHL) successfully treated with combination chemotherapy during pregnancy. The histological diagnosis was large-cell B-type NHL. Four courses of chemotherapy with epirubicin, vincristine and prednisolone were given before delivery. The patient is in complete remission and her baby, now four years old, has developed normally. To our knowledge, this is the first reported case of epirubicin administration during pregnancy and in which chemotherapy was given safely to NHL patients during the second and third trimester of pregnancy.
Collapse
|
47
|
Apperley JF, Reddy N. Mechanism and management of treatment-related gonadal failure in recipients of high dose chemoradiotherapy. Blood Rev 1995; 9:93-116. [PMID: 7580395 DOI: 10.1016/s0268-960x(95)90030-6] [Citation(s) in RCA: 103] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
For the last two decades, the use of bone marrow transplantation for leukaemia, and most recently for lymphomas and other cancers, has increased dramatically and all of the systemic chemotherapy or radiotherapy used in these patients has a potential for causing infertility. This review describes the normal reproductive function of relevance to patients at risk of chemotherapy- or radiotherapy-induced infertility. The mechanism by which fertility may be affected by these agents is also described and therapeutic strategies for reducing or preventing these problems are outlined. The treatment of men and women who are infertile after chemotherapy or radiotherapy is addressed, as are the ethical and legal aspects of assisted fertilization.
Collapse
Affiliation(s)
- J F Apperley
- Department of Haematology, Royal Postgraduate Medical School, Hammersmith Hospital, London, UK
| | | |
Collapse
|
48
|
Grigsby PW, Russell A, Bruner D, Eifel P, Koh WJ, Spanos W, Stetz J, Stitt JA, Sullivan J. Late injury of cancer therapy on the female reproductive tract. Int J Radiat Oncol Biol Phys 1995; 31:1281-99. [PMID: 7713788 DOI: 10.1016/0360-3016(94)00426-l] [Citation(s) in RCA: 93] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The purpose of this article is to review the late effects of cancer therapy on the female reproductive tract. The anatomic sites detailed are the vulva, vagina, cervix, uterus, fallopian tubes, and ovaries. The available pathophysiology is discussed. Clinical syndromes are presented. Tolerance doses of irradiation for late effects are rarely presented in the literature and are reviewed where available. Management strategies for surgical, radiotherapeutic, and chemotherapeutic late effects are discussed. Endpoints for evaluation of therapeutic late effects have been formulated utilizing the symptoms, objective, management, and analytic (SOMA) format. Late effects on the female reproductive tract from cancer therapy should be recognized and managed appropriately. A grading system for these effects is presented. Endpoints for late effects and tolls for the evaluation need to be further developed.
Collapse
Affiliation(s)
- P W Grigsby
- Washington University School of Medicine, St. Louis, MO, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Wang CL, Wang F, Bosco JJ. Ovarian failure in oral cyclophosphamide treatment for systemic lupus erythematosus. Lupus 1995; 4:11-4. [PMID: 7767332 DOI: 10.1177/096120339500400103] [Citation(s) in RCA: 73] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Ninety-two women with systemic lupus erythematosus treated with oral cyclophosphamide were studied to ascertain the prevalence and the factors associated with ovarian dysfunction. Menstrual disturbance during treatment occurred in 55% of patients: 36% had amenorrhoea and 19% had oligomenorrhoea. Sustained oligomenorrhoea occurred in 12% patients. Permanent amenorrhoea (> 12 months) after cessation of oral cyclophosphamide occurred in 27% of patients. Hormonal studies in these patients were consistent with ovarian failure. Older age at initiation of treatment and high cumulative dose of cyclophosphamide were found to be associated with this outcome. There was a trend towards linear relationship between the age of initiation of cyclophosphamide therapy and frequency of amenorrhoea. A statistically significant association between amenorrhoea and cumulative dose of cyclophosphamide after adjustment for age was found whereas no such association was linked to the duration of treatment. Fourteen of the 23 women who wished to become pregnant after cessation of treatment conceived resulting in 20 live births and two abortions.
Collapse
Affiliation(s)
- C L Wang
- Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur
| | | | | |
Collapse
|
50
|
Busch DB. Radiation and chemotherapy injury: pathophysiology, diagnosis, and treatment. Crit Rev Oncol Hematol 1993; 15:49-89. [PMID: 8240706 DOI: 10.1016/1040-8428(93)90020-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The text in general is not meant to represent the participants' entire presentations. The lecture presenters in general are not responsible for the summaries, and cannot necessarily be assumed to agree with all that is stated, but they deserve credit for providing the lecture and handout material on which the summaries are based, and in most cases have contributed far more to the summaries than I have.
Collapse
Affiliation(s)
- D B Busch
- Department of Environmental and Toxicologic Pathology, Armed Forces Institute of Pathology, Washington, D.C. 20306-6000
| |
Collapse
|