1
|
Valsamakis G, Valtetsiotis K, Charmandari E, Lambrinoudaki I, Vlahos NF. GnRH Analogues as a Co-Treatment to Therapy in Women of Reproductive Age with Cancer and Fertility Preservation. Int J Mol Sci 2022; 23:2287. [PMID: 35216409 PMCID: PMC8875398 DOI: 10.3390/ijms23042287] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/10/2022] [Accepted: 02/16/2022] [Indexed: 11/17/2022] Open
Abstract
In this review, we analyzed existing literature regarding the use of Gonadotropin-releasing Hormone (GnRH) analogues (agonists, antagonists) as a co-treatment to chemotherapy and radiotherapy. There is a growing interest in their application as a prophylaxis to gonadotoxicity caused by chemotherapy and/or radiotherapy due to their ovarian suppressive effects, making them a potential option to treat infertility caused by such chemotherapy and/or radiotherapy. They could be used in conjunction with other fertility preservation options to synergistically maximize their effects. GnRH analogues may be a valuable prophylactic agent against chemotherapeutic infertility by inhibiting rapid cellular turnover on growing follicles that contain types of cells unintentionally targeted during anti-cancer treatments. These could create a prepubertal-like effect in adult women, limiting the gonadotoxicity to the lower levels that young girls have. The use of GnRH agonists was found to be effective in hematological and breast cancer treatment whereas for ovarian endometrial and cervical cancers the evidence is still limited. Studies on GnRH antagonists, as well as the combination of both agonists and antagonists, were limited. GnRH antagonists have a similar protective effect to that of agonists as they preserve or at least alleviate the follicle degradation during chemo-radiation treatment. Their use may be preferred in cases where treatment is imminent (as their effects are almost immediate) and whenever the GnRH agonist-induced flare-up effect may be contra-indicated. The combination treatment of agonists and antagonists has primarily been studied in animal models so far, especially rats. Factors that may play a role in determining their efficacy as a chemoprotective agent that limits gonadal damage, include the type and stage of cancer, the use of alkylating agents, age of patient and prior ovarian reserve. The data for the use of GnRH antagonist alone or in combination with GnRH agonist is still very limited. Moreover, studies evaluating the impact of this treatment on the ovarian reserve as measured by Anti-Müllerian Hormone (AMH) levels are still sparse. Further studies with strict criteria regarding ovarian reserve and fertility outcomes are needed to confirm or reject their role as a gonadal protecting agent during chemo-radiation treatments.
Collapse
Affiliation(s)
- Georgios Valsamakis
- Second University Department of Obstetrics and Gynecology, Aretaieion University Hospital, Athens Medical School, Ethnikon and Kapodistriakon University of Athens, 115 28 Athens, Greece; (K.V.); (I.L.); (N.F.V.)
| | - Konstantinos Valtetsiotis
- Second University Department of Obstetrics and Gynecology, Aretaieion University Hospital, Athens Medical School, Ethnikon and Kapodistriakon University of Athens, 115 28 Athens, Greece; (K.V.); (I.L.); (N.F.V.)
| | - Evangelia Charmandari
- First University Department of Paediatrics, Aghia Sophia Childrens Hospital, Athens Medical School, Ethnikon and Kapodistriakon University of Athens, 152 33 Athens, Greece;
| | - Irene Lambrinoudaki
- Second University Department of Obstetrics and Gynecology, Aretaieion University Hospital, Athens Medical School, Ethnikon and Kapodistriakon University of Athens, 115 28 Athens, Greece; (K.V.); (I.L.); (N.F.V.)
| | - Nikolaos F. Vlahos
- Second University Department of Obstetrics and Gynecology, Aretaieion University Hospital, Athens Medical School, Ethnikon and Kapodistriakon University of Athens, 115 28 Athens, Greece; (K.V.); (I.L.); (N.F.V.)
| |
Collapse
|
2
|
Mangiardi-Veltin M, Sebbag C, Rousset-Jablonski C, Ray-Coquard I, Berkach C, Laot L, Wang Y, Abdennebi I, Labrosse J, Sautter C, Toussaint A, Sablone L, Laas E, Khallouch S, Coussy F, Santulli P, Chapron C, Bobrie A, Jacot W, Sella N, Dumas E, Sénéchal-Davin C, Espie M, Giacchetti S, Maitrot L, Plu-Bureau G, Coutant C, Guerin J, Asselain B, Fumoleau P, Rodrigues M, Decanter C, Mailliez A, Delrieu L, Lemoine A, Jouannaud C, Houdre D, Reyal F, Hamy AS. Pregnancy, fertility concerns, and fertility preservation procedures in French breast cancer survivors in the FEERIC national study (on behalf of the Seintinelles research network). Reprod Biomed Online 2022; 44:1031-1044. [DOI: 10.1016/j.rbmo.2021.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 12/06/2021] [Accepted: 12/22/2021] [Indexed: 10/19/2022]
|
3
|
Wang HQ, Wang WH, Chen CZ, Guo HX, Jiang H, Yuan B, Zhang JB. Regulation of FSH Synthesis by Differentially Expressed miR-488 in Anterior Adenohypophyseal Cells. Animals (Basel) 2021; 11:ani11113262. [PMID: 34827994 PMCID: PMC8614264 DOI: 10.3390/ani11113262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/27/2021] [Accepted: 11/12/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary GnRH and FSH play an important regulatory role in the reproductive activities of mammals. At present, many artificially synthesized GnRH analogues have been used in the regulation of cattle reproduction and the clinical treatment of various reproductive diseases. This study explored the potential mechanism of miR-488 in GnRH regulation of FSH synthesis and secretion and provides a theoretical basis for the application of GnRH analogue in cattle artificial breeding. We hope to provide a research foundation for improving the processing procedures of cattle estrus control and the domestic application of hormone products. Abstract Gonadotropin-releasing hormone (GnRH), which is synthesized and released by the hypothalamus, promotes the synthesis and secretion of follicle-stimulating hormone (FSH), thereby regulating the growth and reproduction of animals. GnRH analogues have been widely used in livestock production. MiRNAs, which are endogenous non-coding RNAs, have been found to play important roles in hormone regulation and other physiological processes in recent years. However, the roles of miRNAs in GnRH-mediated regulation of FSH secretion have rarely been studied. Herein, we treated bovine anterior adenohypophyseal cells with an exogenous GnRH analogue and found that miR-488 was differentially expressed. Through a combination of TargetScan prediction and dual luciferase reporter analysis, miR-488 was confirmed to be able to target the FSHB gene. Based on this finding, we verified the expression of Fshβ and Lhβ mRNA in the rat adenohypophysis before and after exogenous GnRH treatment in vivo and in vitro. Experiments on rat anterior adenohypophyseal cells showed that overexpression of miR-488 significantly inhibited Fshβ expression and FSH synthesis, while knockdown of miR-488 had the opposite effects. Our results demonstrate that GnRH relies on miR-488 to regulate FSH synthesis, providing additional useful evidence for the significance of miRNAs in the regulation of animal reproduction.
Collapse
Affiliation(s)
| | | | | | | | | | - Bao Yuan
- Correspondence: (B.Y.); (J.-B.Z.); Tel.: +86-431-8783-6536 (B.Y.); +86-431-8783-6551 (J.-B.Z.)
| | - Jia-Bao Zhang
- Correspondence: (B.Y.); (J.-B.Z.); Tel.: +86-431-8783-6536 (B.Y.); +86-431-8783-6551 (J.-B.Z.)
| |
Collapse
|
4
|
Rodgers RJ, Abbott JA, Walters KA, Ledger WL. Translational Physiology of Anti-Müllerian Hormone: Clinical Applications in Female Fertility Preservation and Cancer Treatment. Front Endocrinol (Lausanne) 2021; 12:689532. [PMID: 34557157 PMCID: PMC8454407 DOI: 10.3389/fendo.2021.689532] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 08/09/2021] [Indexed: 12/21/2022] Open
Abstract
Background Whilst the ability of AMH to induce the regression of the Müllerian ducts in the male fetus is well appreciated, AMH has additional biological actions in relation to steroid biosynthesis and ovarian follicle dynamics. An understanding of the physiology of AMH illuminates the potential therapeutic utility of AMH to protect the ovarian reserve during chemotherapy and in the treatment of female malignancies. The translation of the biological actions of AMH into clinical applications is an emerging focus of research, with promising preliminary results. Objective and Rationale Studies indicate AMH restrains primordial follicle development, thus administration of AMH during chemotherapy may protect the ovarian reserve by preventing the mass activation of primordial follicles. As AMH induces regression of tissues expressing the AMH receptor (AMHRII), administration of AMH may inhibit growth of malignancies expressing AMHR II. This review evaluates the biological actions of AMH in females and appraises human clinical applications. Search Methods A comprehensive search of the Medline and EMBASE databases seeking articles related to the physiological functions and therapeutic applications of AMH was conducted in July 2021. The search was limited to studies published in English. Outcomes AMH regulates primordial follicle recruitment and moderates sex steroid production through the inhibition of transcription of enzymes in the steroid biosynthetic pathway, primarily aromatase and 17α-hydroxylase/17,20-lyase. Preliminary data indicates that administration of AMH to mice during chemotherapy conveys a degree of protection to the ovarian reserve. Administration of AMH at the time of ovarian tissue grafting has the potential to restrain uncontrolled primordial follicle growth during revascularization. Numerous studies demonstrate AMH induced regression of AMHR II expressing malignancies. As this action occurs via a different mechanism to traditional chemotherapeutic agents, AMH has the capacity to inhibit proliferation of chemo-resistant ovarian cancer cells and cancer stem cells. Wider Implications To date, AMH has not been administered to humans. Data identified in this review suggests administration of AMH would be safe and well tolerated. Administration of AMH during chemotherapy may provide a synchronistic benefit to women with an AMHR II expressing malignancy, protecting the ovarian reserve whilst the cancer is treated by dual mechanisms.
Collapse
Affiliation(s)
- Rachael Jean Rodgers
- School of Women’s and Children’s Health, University of New South Wales, Sydney, NSW, Australia
| | | | | | | |
Collapse
|
5
|
Günther V, Alkatout I, Maass N, von Otte S. Fertilitätserhalt bei Patienten in der Dermatoonkologie – Eine aktuelle Übersicht. AKTUELLE DERMATOLOGIE 2021. [DOI: 10.1055/a-1426-2908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
ZusammenfassungBei vielen Patienten, die an einem Malignom erkranken, ist die Familienplanung noch nicht abgeschlossen, sodass für den Erhalt des fertilen Potenzials Maßnahmen der Fertilitätsprotektion sinnvoll sind. Durch eine Polychemotherapie, unabhängig ob im neoadjuvanten oder adjuvanten Setting, Molekular- oder Immuntherapien kann es zu einer irreversiblen Schädigung der Follikel bzw. Spermatogenese kommen, was u. U. zu einer permanenten Infertilität führen kann. Abhängig von der verwendeten Therapie und der altersabhängigen Ovarialreserve der Frau muss das gonadotoxische Risiko als niedrig, mittel oder hoch eingeschätzt werden. Möglichkeiten des Fertilitäserhalts sind: a) die Kryokonservierung von fertilisierten oder unfertilisierten Oozyten. Hierbei werden nach ovarieller Hyperstimulation reife Oozyten mittels transvaginaler Follikelaspiration gewonnen und im Anschluss entweder unfertilisiert oder nach erfolgter IVF- oder ICSI-Behandlung kryokonserviert. Bei b) der Kryokonservierung von Ovarialgewebe wird mithilfe eines laparoskopischen Eingriffs etwa 50 % des Ovarkortex eines Ovars reseziert und kryokonserviert. Die Verwendung von c) GnRH-Agonisten als medikamentöse Therapieoption unternimmt den Versuch einer endokrinen Ovarialsuppression, um Oozyten, Granulosa- und Thekazellen vor dem zytotoxischen Einfluss der jeweiligen Therapie zu schützen. Bei männlichen Patienten können Spermien vor Therapiebeginn kryokonserviert werden.
Collapse
Affiliation(s)
- V. Günther
- Klinik für Gynäkologie und Geburtshilfe, Universitätsklinikum Schleswig-Holstein, Campus Kiel
- Universitäres Kinderwunschzentrum, MVZ, Universitätsklinikum Schleswig-Holstein, Campus Kiel
| | - I. Alkatout
- Klinik für Gynäkologie und Geburtshilfe, Universitätsklinikum Schleswig-Holstein, Campus Kiel
| | - N. Maass
- Klinik für Gynäkologie und Geburtshilfe, Universitätsklinikum Schleswig-Holstein, Campus Kiel
| | - S. von Otte
- Universitäres Kinderwunschzentrum, MVZ, Universitätsklinikum Schleswig-Holstein, Campus Kiel
| |
Collapse
|
6
|
Choi JH, Seok J, Lim SM, Kim TH, Kim GJ. Microenvironmental changes induced by placenta-derived mesenchymal stem cells restore ovarian function in ovariectomized rats via activation of the PI3K-FOXO3 pathway. Stem Cell Res Ther 2020; 11:486. [PMID: 33198818 PMCID: PMC7667861 DOI: 10.1186/s13287-020-02002-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 10/29/2020] [Indexed: 12/23/2022] Open
Abstract
Background Translational studies have explored the therapeutic potential and feasibility of mesenchymal stem cells (MSCs) in several degenerative diseases; however, mechanistic studies of the function of these cells have been insufficient. As ovarian failure causes anovulation as well as ovarian steroid hormonal imbalances, the specific aims of this study were to analyze the therapeutic role of placenta-derived MSCs (PD-MSCs) in an ovarian failure ovariectomy (OVX) rat model and evaluate whether PD-MSC transplantation (Tx) improved folliculogenesis and oocyte maturation in the injured ovary through PI3K/Akt and FOXO signaling. Methods Blood and ovary tissue were collected and analyzed after various PD-MSC Tx treatments in an ovariectomized rat model. Changes in the expression of folliculogenesis- and ovary regeneration-related genes induced by PD-MSC treatments were analyzed by qRT-PCR, Western blotting, and histological analysis. Results The levels of hormones related to ovary function were significantly increased in the PD-MSC Tx groups compared with those in the nontransplantation group (NTx). The follicle numbers in the ovarian tissues were increased along with the increased expression of genes related to folliculogenesis in the PD-MSC Tx groups compared with the NTx groups. Furthermore, Tx PD-MSCs induced follicle maturation by increasing the phosphorylation of GSK3 beta and FOXO3 (p < 0.05) and shifting the balance of growth and apoptosis in oocytes. Conclusions Taken together, these results show that PD-MSC Tx can restore ovarian function and induce ovarian folliculogenesis via the PI3K/Akt and FOXO signaling pathway.
Collapse
Affiliation(s)
- Jong Ho Choi
- Department of Oral Pathology, College of Dentistry, Gangneung-Wonju National University, Gangneung-si, 25457, Republic of Korea
| | - Jin Seok
- Department of Biomedical Science, CHA University, 689, Sampyeong-dong, Bundang-gu, Seongnam-si, 13488, Gyeonggi-do, Republic of Korea
| | - Seung Mook Lim
- Department of Biomedical Science, CHA University, 689, Sampyeong-dong, Bundang-gu, Seongnam-si, 13488, Gyeonggi-do, Republic of Korea
| | - Tae Hee Kim
- Department of Obstetrics and Gynecology, Soonchunhyang University College of Medicine Hospital, Asan, 14584, Gyoenggi-do, Republic of Korea
| | - Gi Jin Kim
- Department of Biomedical Science, CHA University, 689, Sampyeong-dong, Bundang-gu, Seongnam-si, 13488, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
7
|
Cho HW, Lee S, Min KJ, Hong JH, Song JY, Lee JK, Lee NW, Kim T. Advances in the Treatment and Prevention of Chemotherapy-Induced Ovarian Toxicity. Int J Mol Sci 2020; 21:E7792. [PMID: 33096794 PMCID: PMC7589665 DOI: 10.3390/ijms21207792] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 10/20/2020] [Indexed: 12/24/2022] Open
Abstract
Due to improvements in chemotherapeutic agents, cancer treatment efficacy and cancer patient survival rates have greatly improved, but unfortunately gonadal damage remains a major complication. Gonadotoxic chemotherapy, including alkylating agents during reproductive age, can lead to iatrogenic premature ovarian insufficiency (POI), and loss of fertility. In recent years, the demand for fertility preservation has increased dramatically among female cancer patients. Currently, embryo and oocyte cryopreservation are the only established options for fertility preservation in women. However, there is growing evidence for other experimental techniques including ovarian tissue cryopreservation, oocyte in vitro maturation, artificial ovaries, stem cell technologies, and ovarian suppression. To prevent fertility loss in women with cancer, individualized fertility preservation options including established and experimental techniques that take into consideration the patient's age, marital status, chemotherapy regimen, and the possibility of treatment delay should be provided. In addition, effective multidisciplinary oncofertility strategies that involve a highly skilled and experienced oncofertility team consisting of medical oncologists, gynecologists, reproductive biologists, surgical oncologists, patient care coordinators, and research scientists are necessary to provide cancer patients with high-quality care.
Collapse
Affiliation(s)
| | - Sanghoon Lee
- Department of Obstetrics and Gynecology, Korea University College of Medicine, Seoul 02841, Korea; (H.-W.C.); (K.-J.M.); (J.H.H.); (J.Y.S.); (J.K.L.); (N.W.L.); (T.K.)
| | | | | | | | | | | | | |
Collapse
|
8
|
Gini G, Annibali O, Lupasco D, Bocci C, Tomarchio V, Sampaolo M, Trappolini S, Tafuri MA, Cacciagiù S, Ciccarone M, Barucca A, Sarlo C, Vincenzi B, Avvisati G, Leoni P, Olivieri A. Gonadal Function Recovery and Fertility in Women Treated with Chemo- and/or Radiotherapy for Hodgkin's and Non-Hodgkin Lymphoma. Chemotherapy 2019; 64:36-41. [PMID: 31117081 DOI: 10.1159/000499535] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 03/12/2019] [Indexed: 11/19/2022]
Abstract
BACKGROUND Fertility and gonadal function represent one of the most important aspects for long-term lymphoma survivors. AIMS The aim of our study was to determine possible risk factors, such as age at treatment, chemotherapeutic regimen, protection with oral contraceptives (OCs), and gonadotropin-releasing hormone (GnRH) analogues in female patients treated for Hodgkin's lymphoma (HL) or non-Hodgkin lymphoma (NHL) at a reproductive age. METHODS Patients between the age of 16 and 50 years at the time of HL or NHL diagnosis were selected. Eligible patients were requested to respond to a questionnaire by phone interview about fertility, menstrual status, sexual aspects, and treatment with OCs or GnRH analogues during chemotherapy. RESULTS The resumption of menstrual activity was associated with the use of the OCs and GnRH analogues during chemotherapy (p = 0.008 and 0.034, respectively). At univariate analysis, the use of OCs during chemotherapy was associated with a lower risk of amenorrhea (prevalence ratio [PR] = 0.37; 95% CI 0.17-0.82). A higher age at the time of treatment correlated positively with therapy-induced amenorrhea, with a difference of 12.8 years between the mean age at diagnosis of the women with therapy-induced amenorrhea and those who resumed their menses. Amenorrhea was significantly higher in women receiving R-CHOP than in women treated with ABVD (PR = 6.00; 95% CI 2.32-15.54). Moreover, NHL had an infertility PR of 1.51 (95% CI 0.86-2.45) at multivariate analysis compared to HL. CONCLUSIONS This study suggests a possible role of pharmacological prophylaxis with OCs and GnRH analogues.
Collapse
Affiliation(s)
- Guido Gini
- Unit of Hematology, AUO Ospedali Riuniti, Università Politecnica delle Marche, Ancona, Italy
| | - Ombretta Annibali
- Unit of Hematology and Stem Cell Transplantation, Campus Bio-Medico University, Rome, Italy,
| | - Diana Lupasco
- Unit of Hematology, AUO Ospedali Riuniti, Università Politecnica delle Marche, Ancona, Italy
| | - Caterina Bocci
- Unit of Hematology, AUO Ospedali Riuniti, Università Politecnica delle Marche, Ancona, Italy
| | - Valeria Tomarchio
- Unit of Hematology and Stem Cell Transplantation, Campus Bio-Medico University, Rome, Italy
| | - Michela Sampaolo
- Unit of Hematology, AUO Ospedali Riuniti, Università Politecnica delle Marche, Ancona, Italy
| | - Silvia Trappolini
- Unit of Hematology, AUO Ospedali Riuniti, Università Politecnica delle Marche, Ancona, Italy
| | | | - Sonia Cacciagiù
- Unit of Hematology, AUO Ospedali Riuniti, Università Politecnica delle Marche, Ancona, Italy
| | - Mariavita Ciccarone
- Unit of Gynecology, Ospedale San Carlo di Nancy, Rome, Italy.,Associazione Gemme Dormienti ONLUS, Rome, Italy
| | - Alessandra Barucca
- Unit of Hematology, AUO Ospedali Riuniti, Università Politecnica delle Marche, Ancona, Italy
| | - Chiara Sarlo
- Unit of Hematology and Stem Cell Transplantation, Campus Bio-Medico University, Rome, Italy
| | - Bruno Vincenzi
- Medical Oncology, University Campus Bio Medico, Rome, Italy
| | - Giuseppe Avvisati
- Unit of Hematology and Stem Cell Transplantation, Campus Bio-Medico University, Rome, Italy
| | - Pietro Leoni
- Unit of Hematology, AUO Ospedali Riuniti, Università Politecnica delle Marche, Ancona, Italy
| | - Attilio Olivieri
- Unit of Hematology, AUO Ospedali Riuniti, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
9
|
Shah NM, Scott DM, Kandagatla P, Moravek MB, Cobain EF, Burness ML, Jeruss JS. Young Women with Breast Cancer: Fertility Preservation Options and Management of Pregnancy-Associated Breast Cancer. Ann Surg Oncol 2019; 26:1214-1224. [PMID: 30680478 PMCID: PMC6458084 DOI: 10.1245/s10434-019-07156-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Breast cancer is the most common malignancy diagnosed in women of childbearing age. A breast cancer diagnosis in this young patient population can be uniquely complex to navigate when considering the potential impact of fertility loss associated with specific gonadotoxic therapies. Another unique challenge for young breast cancer patients is pregnancy-associated breast cancer (PABC), which occurs in approximately 1 of every 3000 pregnancies. Pregnancy adds a layer of complexity to breast cancer treatment planning as many therapies can affect the developing fetus. These two clinical challenges require nuanced multidisciplinary approaches to facilitate optimal treatment outcomes. We sought to review and summarize the management strategy options for both fertility preservation and PABC. METHODS A guideline and literature review was performed for fertility preservation, young patients with breast cancer, and pregnancy-associated breast cancer. RESULTS Fertility preservation options, both established and experimental, are detailed. Suggested clinical practice guidelines for PABC are also presented, which delineate breast cancer treatment recommendations based on pregnancy trimester. CONCLUSION A multidisciplinary approach to patient care, including oncologists and early referral to reproductive specialists, can provide young breast cancer patients with options for fertility preservation. Under the guidance of a multidisciplinary treatment team, PABC can also be diagnosed and treated to permit the best possible outcomes for the mother and the developing fetus.
Collapse
Affiliation(s)
- Nikita M Shah
- Division of Surgical Oncology, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Dana M Scott
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
| | - Pridvi Kandagatla
- Division of Surgical Oncology, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Department of Surgery, Henry Ford Health System/Wayne State University, Detroit, MI, USA
| | - Molly B Moravek
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
| | - Erin F Cobain
- Division of Medical Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Monika L Burness
- Division of Medical Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Jacqueline S Jeruss
- Division of Surgical Oncology, Department of Surgery, University of Michigan, Ann Arbor, MI, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
10
|
Lambertini M, Richard F, Nguyen B, Viglietti G, Villarreal-Garza C. Ovarian Function and Fertility Preservation in Breast Cancer: Should Gonadotropin-Releasing Hormone Agonist be administered to All Premenopausal Patients Receiving Chemotherapy? CLINICAL MEDICINE INSIGHTS. REPRODUCTIVE HEALTH 2019; 13:1179558119828393. [PMID: 30886529 PMCID: PMC6410390 DOI: 10.1177/1179558119828393] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 01/10/2019] [Indexed: 11/15/2022]
Abstract
Chemotherapy-induced premature ovarian insufficiency (POI) is one of the potential drawbacks of chemotherapy use of particular concern for newly diagnosed premenopausal breast cancer patients. Temporary ovarian suppression obtained pharmacologically with the administration of a gonadotropin-releasing hormone agonist (GnRHa) during chemotherapy has been specifically developed as a method to counteract chemotherapy-induced gonadotoxicity with the main goal of diminishing the risk of POI. In recent years, important clinical evidence has become available on the efficacy and safety of this strategy that should now be considered a standard option for ovarian function preservation in premenopausal breast cancer patients, including women who are not interested in conceiving after treatment or that would not be candidates for fertility preservation strategies because of their age. Nevertheless, in women interested in fertility preservation, this is not an alternative to gamete cryopreservation, which remains as the first option to be offered. In this setting, temporary ovarian suppression with GnRHa during chemotherapy should be also proposed following gamete cryopreservation or to women who have no access, refuse, or have contraindications to surgical fertility preservation techniques. In this article, we present an overview about the role of temporary ovarian suppression with GnRHa during chemotherapy in breast cancer patients by addressing the available clinical evidence with the aim of identifying both the best candidates for the use of this strategy and the still existing gray zones requiring further investigation.
Collapse
Affiliation(s)
- Matteo Lambertini
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, Ospedale Policlinico San Martino, Genova, Italy.,Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - François Richard
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles (U.L.B), Brussels, Belgium
| | - Bastien Nguyen
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles (U.L.B), Brussels, Belgium
| | - Giulia Viglietti
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles (U.L.B), Brussels, Belgium
| | - Cynthia Villarreal-Garza
- Centro de Cancer de Mama del Hospital Zambrano Hellion, Tecnologico de Monterrey, Nuevo Leon, Mexico.,Departamento de Investigacion y de Tumores Mamarios del Instituto Nacional de Cancerologia, Mexico City, Mexico
| |
Collapse
|
11
|
Chen H, Xiao L, Li J, Cui L, Huang W. Adjuvant gonadotropin-releasing hormone analogues for the prevention of chemotherapy-induced premature ovarian failure in premenopausal women. Cochrane Database Syst Rev 2019; 3:CD008018. [PMID: 30827035 PMCID: PMC6397718 DOI: 10.1002/14651858.cd008018.pub3] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND This is an update of the original review published in the Cochrane Database of Systematic Reviews 2011, Issue 11, and updated in 2015, Issue 4.Chemotherapy has significantly improved prognosis for women with malignant and some non-malignant conditions. This treatment, however, is associated with ovarian toxicity. The use of gonadotropin-releasing hormone (GnRH) analogues, both agonists and antagonists, may have a protective effect on the ovaries. The primary mechanism of action of GnRH analogues is to suppress the gonadotropin levels to simulate pre-pubertal hormonal milieu and subsequently prevent primordial follicles from maturation and therefore decrease the number of follicles that are more vulnerable to chemotherapy. OBJECTIVES To assess the efficacy and safety of GnRH analogues given before or in parallel to chemotherapy to prevent chemotherapy-related ovarian damage in premenopausal women with malignant or non-malignant conditions. SEARCH METHODS The search was run for the original review in July 2011, and for the first update in July 2014. For this update we searched the following databases in November 2018: the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, Embase, and the Chinese Biomedicine Database (CBM). SELECTION CRITERIA Randomised controlled trials (RCTs), in all languages, which examined the effect of GnRH analogues for chemotherapy-induced ovarian failure in premenopausal women, were eligible for inclusion in the review. DATA COLLECTION AND ANALYSIS Two review authors independently extracted data and assessed trial quality using the Cochrane 'Risk of bias' tool. We analysed binary data using risk ratios (RRs) with 95% confidence intervals (CI) and for continuous data, we used the standardized mean difference (SMD) to combine trials. We applied the random-effects model in our analyses. We used the GRADE approach to produce a 'Summary of findings' table for our main outcomes of interest. MAIN RESULTS We included 12 RCTs involving 1369 women between the ages of 12 and 51.1 years. Participants were diagnosed with breast malignancy, ovarian malignancy, or Hodgkin's lymphoma, and most of them received alkylating, or platinum complexes, based chemotherapy. The included studies were funded by a university (n = 1), research centres (n = 4), and pharmaceutical companies (n = 1). Trials were at high or unclear risk of bias.Comparison 1: GnRH agonist plus chemotherapy versus chemotherapy aloneThe incidence of menstruation recovery or maintenance was 178 of 239 (74.5%) in the GnRH agonist group and 110 of 221 (50.0%) in the control group during a follow-up period no longer than 12 months (RR 1.60, 95% CI 1.14 to 2.24; 5 studies, 460 participants; I2 = 79%; low-certainty evidence), with an overall effect favouring treatment with GnRH agonist (P = 0.006). However, we observed no difference during a follow-up period longer than 12 months between these two groups (P = 0.24). In the GnRH agonist group, 326 of 447 participants had menstruation recovery or maintenance (72.9%) in comparison to the control group, in which 276 of 422 participants had menstruation recovery or maintenance (65.4%) during a follow-up period longer than 12 months (RR 1.08, 95% CI 0.95 to 1.22; 8 studies, 869 participants; I2 = 56%; low-certainty evidence).The incidence of premature ovarian failure was 43 of 401 (10.7%) in the GnRH agonist group and 96 of 379 (25.3%) in the control group (RR 0.44, 95% CI 0.31 to 0.61; 4 studies, 780 participants; I2 = 0%; moderate-certainty evidence), with an overall effect favouring treatment with GnRH agonist (P < 0.00001).The incidence of pregnancy was 32 of 356 (9.0%) in the GnRH agonist group and 22 of 347 (6.3%) in the control group (RR 1.59, 95% CI 0.93 to 2.70; 7 studies, 703 participants; I2 = 0%; low-certainty evidence), with no difference between groups (P = 0.09). However, we are cautious about this conclusion because there were insufficient data about whether the participants intended to become pregnant.The incidence of ovulation was 29 of 47 (61.7%) in the GnRH agonist group and 12 of 48 (25.0%) in the control group (RR 2.47, 95% CI 1.43 to 4.26; 2 studies, 95 participants; I2 = 0%; low-certainty evidence) with an overall effect favouring treatment with GnRH (P = 0.001).The most common adverse effects of GnRH analogues included hot flushes, vaginal dryness, urogenital symptoms, and mood swings. The pooled analysis of safety data showed no difference in adverse effects between GnRH agonist group and control group.Comparison 2: GnRH agonist-antagonist cotreatment plus chemotherapy versus chemotherapy aloneOnly one RCT discussed GnRH agonist-antagonist cotreatment. The limited evidence showed the incidence of menstruation recovery or maintenance was 20 of 25 (80%) in both cotreatment group and control group during a 12-month follow-up period (RR 1.00, 95% CI 0.76 to 1.32; 50 participants; very low-certainty evidence), with no difference between groups (P = 1.00). In the cotreatment group, 13 of 25 participants had menstruation recovery or maintenance (52.0%) in comparison to the control group, in which 14 of 25 participants had menstruation recovery or maintenance (56.0%) during a follow-up period longer than 12 months (RR 0.93, 95% CI 0.56 to 1.55; 50 participants; very low-certainty evidence), with no difference between groups (P = 0.78). The incidence of pregnancy was 1 of 25 (4.0%) in the cotreatment group and 0 of 25 (0%) in the control group (RR 3.00, 95% CI 0.13 to 70.30; 50 participants; very low-certainty evidence), with no difference between groups (P = 0.49). AUTHORS' CONCLUSIONS GnRH agonist appears to be effective in protecting the ovaries during chemotherapy, in terms of maintenance and resumption of menstruation, treatment-related premature ovarian failure and ovulation. Evidence for protection of fertility was insufficient and needs further investigation. Evidence was also insufficient to assess the effect of GnRH agonist and GnRH antagonist cotreatment on ovarian protection against chemotherapy. The included studies differed in some important aspects of design, and most of these studies had no age-determined subgroup analysis. Large and well-designed RCTs with longer follow-up duration should be conducted to clarify the effects of GnRH analogues in preventing chemotherapy-induced ovarian failure, especially on different age groups or different chemotherapy regimens. Furthermore, studies should address the effects on pregnancy rates and anti-tumour therapy.
Collapse
Affiliation(s)
- Hengxi Chen
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, No. 17, Section Three, Ren Min Nan Lu Avenue, Chengdu, Sichuan, China, 610041
| | | | | | | | | |
Collapse
|
12
|
Kim H, Kim H, Ku SY. Fertility preservation in pediatric and young adult female cancer patients. Ann Pediatr Endocrinol Metab 2018; 23:70-74. [PMID: 29969877 PMCID: PMC6057020 DOI: 10.6065/apem.2018.23.2.70] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 06/14/2018] [Indexed: 01/08/2023] Open
Abstract
As the 5-year survival rate increases up to 80% in pediatric cancer patients, the number of women patients with reduced gonadal function by chemotherapy and radiotherapy increases. The gonadal toxicity of pediatric patients varies highly according to the chemotherapeutic agent and the type of radiotherapy. Although American Society of Clinical Oncology published the guideline for fertility preservation, additional scientific and ethical concerns should be considered for clinical practice. In addition, only the experimental method can be applied for the prepubertal patients in contrast to the postpubertal patients. In this review, we will discuss some options for preserving fertility among women's quality of life issues.
Collapse
Affiliation(s)
- Hashin Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Hoon Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Seung-Yup Ku
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
13
|
Hickman LC, Llarena NC, Valentine LN, Liu X, Falcone T. Preservation of gonadal function in women undergoing chemotherapy: a systematic review and meta-analysis of the potential role for gonadotropin-releasing hormone agonists. J Assist Reprod Genet 2018; 35:571-581. [PMID: 29470701 PMCID: PMC5949114 DOI: 10.1007/s10815-018-1128-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 01/23/2018] [Indexed: 01/15/2023] Open
Abstract
PURPOSE To evaluate the available randomized controlled trials (RCTs) in the literature investigating the use of gonadotropin-releasing hormone agonist (GnRHa) co-treatment for ovarian preservation in women receiving chemotherapy. METHODS A systematic review of the literature was performed from 1960 through 2017 to identify relevant RCTs. Included patients had lymphoma, ovarian cancer, or breast cancer. The primary outcome was the proportion of women who retained ovarian function after chemotherapy. Extracted data points included study design, patient characteristics, and proportion of women who developed premature ovarian failure (POF). A risk of bias assessment was performed according to the criteria outlined in the Cochrane Handbook for Systematic Reviews of Interventions. The pooled odds ratio was calculated, and outcomes of individual studies were compared using the random-effects model with the inverse-variance method and the DerSimonian-Laird estimator. RESULTS Twenty-nine RCTs were identified, and 10 met criteria for inclusion in the meta-analysis. An analysis of patients who did not develop POF after chemotherapy revealed eight studies supporting the use of GnRHa (OR 1.83; 95% CI 1.34-2.49). The duration of benefit of GnRHa is unclear. An analysis of three studies with outcome data at 2 years revealed a non-significant OR of 0.53 (95% CI 0.22-1.30) for the preservation of ovarian function with GnRHa treatment. CONCLUSION GnRHa may have a protective effect against the development of POF after gonadotoxic chemotherapy; however, the duration of benefit is unclear and requires further study.
Collapse
Affiliation(s)
- Lisa C Hickman
- Department of Obstetrics/Gynecology, Cleveland Clinic Foundation, 9500 Euclid Avenue, A81, Cleveland, OH, 44195, USA
| | - Natalia C Llarena
- Department of Obstetrics/Gynecology, Cleveland Clinic Foundation, 9500 Euclid Avenue, A81, Cleveland, OH, 44195, USA
| | - Lindsey N Valentine
- Department of Obstetrics/Gynecology, Cleveland Clinic Foundation, 9500 Euclid Avenue, A81, Cleveland, OH, 44195, USA
| | - Xiaobo Liu
- Department of Quantitative Health Science, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Tommaso Falcone
- Department of Obstetrics/Gynecology, Cleveland Clinic Foundation, 9500 Euclid Avenue, A81, Cleveland, OH, 44195, USA.
| |
Collapse
|
14
|
Tang M, Webber K. Fertility and pregnancy in cancer survivors. Obstet Med 2018; 11:110-115. [PMID: 30214475 DOI: 10.1177/1753495x18757816] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 01/10/2018] [Indexed: 02/05/2023] Open
Abstract
Cancer survivors are increasing as improvements in cancer diagnosis and treatment translate to improved outcomes. As cancer survivors in their reproductive years contemplate pregnancy, it is important to understand the impact of cancer and its treatment on fertility and pregnancy outcomes. Cancer treatments such as chemotherapy and radiotherapy can affect patients' fertility, and strategies are available to help preserve the future fertility of survivors. The potential impact of previous cancer diagnoses and treatments on pregnancy and maternal and fetal outcomes needs to be assessed and discussed with survivors, with support from materno-fetal medicine specialists and high-risk antenatal services as needed.
Collapse
Affiliation(s)
- Monica Tang
- Department of Medical Oncology, Prince of Wales Hospital, Randwick, NSW, Australia
| | - Kate Webber
- Department of Medical Oncology, Prince of Wales Hospital, Randwick, NSW, Australia
| |
Collapse
|
15
|
Karunakaran P, Malhotra P, Lad D. Fertility Management for the Hemato-Oncologist. Indian J Hematol Blood Transfus 2018; 34:13-18. [PMID: 29398794 DOI: 10.1007/s12288-017-0887-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 10/03/2017] [Indexed: 11/24/2022] Open
Abstract
For cancer survivors, social parenthood buffers distress and improves quality of life. It is important that physicians dealing with treatment of malignancies are aware of this patient perspective. This review deals with risks of infertility with hematological malignancies and chemotherapy, modes of fertility preservation interventions, time and barriers to referral to specialists offering these interventions. This review aims to guide the hemato-oncologist to make an informed decision with the patient and the partner about fertility preservation at the right time.
Collapse
Affiliation(s)
- Parathan Karunakaran
- Clinical Hematology, Blood and Marrow Transplantation Unit, #28, Department of Internal Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Pankaj Malhotra
- Clinical Hematology, Blood and Marrow Transplantation Unit, #28, Department of Internal Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Deepesh Lad
- Clinical Hematology, Blood and Marrow Transplantation Unit, #28, Department of Internal Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
16
|
Senra JC, Roque M, Talim MCT, Reis FM, Tavares RLC. Gonadotropin-releasing hormone agonists for ovarian protection during cancer chemotherapy: systematic review and meta-analysis. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2018; 51:77-86. [PMID: 29055060 DOI: 10.1002/uog.18934] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 10/13/2017] [Accepted: 10/16/2017] [Indexed: 06/07/2023]
Abstract
OBJECTIVE To evaluate the effectiveness of gonadotropin-releasing hormone agonist (GnRHa) administration before and/or during cancer chemotherapy for the protection of ovarian reserve in premenopausal women without prior diagnosis of infertility. METHODS This was a systematic review and meta-analysis of randomized controlled trials (RCTs) comparing administration of GnRHa before and/or during chemotherapy vs chemotherapy alone. Eligible participants were premenopausal women at any stage of cancer, without previous diagnosis of infertility. An electronic database search in MEDLINE, CENTRAL, LILACS and ClinicalTrials.gov was performed. After selecting eligible studies, the relative risk (RR) was assessed for primary ovarian insufficiency (POI)/amenorrhea and for spontaneous pregnancy after completion of treatment. RESULTS Thirteen RCTs comparing concurrent use of GnRHa and chemotherapy (609 participants) with chemotherapy alone (599 participants) were eligible for meta-analysis. All trials were open-label and patients had been treated for breast cancer (n = 1099) or lymphoma (n = 109). GnRHa had a significant benefit on the risk of POI/amenorrhea (RR, 0.60; 95% CI, 0.45-0.79), which persisted in subgroup analysis for breast cancer (RR, 0.57; 95% CI, 0.43-0.77) but not for lymphoma patients (RR, 0.70; 95% CI, 0.20-2.47). The rate of spontaneous pregnancy after completion of treatment was higher in women receiving GnRHa plus chemotherapy compared with those receiving chemotherapy alone (RR, 1.43; 95% CI, 1.01-2.02). Overall, the quality of evidence was low due to the unclear risk of bias, short follow-up and lack of objective assessment of ovarian function and reserve. CONCLUSIONS Evidence, albeit of low quality, supports the use of GnRHa before and/or during chemotherapy to reduce the risk of POI and increase the probability of spontaneous pregnancy in the short term. Further high quality RCTs with more accurate assessment of ovarian reserve are needed to support definitive recommendations for clinical practice. Copyright © 2017 ISUOG. Published by John Wiley & Sons Ltd.
Collapse
Affiliation(s)
- J C Senra
- Division of Human Reproduction, Hospital das Clínicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - M Roque
- ORIGEN - Center for Reproductive Medicine, Rio de Janeiro, Brazil
| | - M C T Talim
- Division of Human Reproduction, Hospital das Clínicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - F M Reis
- Division of Human Reproduction, Hospital das Clínicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - R L C Tavares
- Division of Human Reproduction, Hospital das Clínicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
17
|
Lee DY, Kim SK, Kim M, Hwang KJ, Kim SH. Fertility preservation for patients with hematologic malignancies: The Korean Society for Fertility Preservation clinical guidelines. Clin Exp Reprod Med 2017; 44:187-192. [PMID: 29376015 PMCID: PMC5783915 DOI: 10.5653/cerm.2017.44.4.187] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 03/14/2017] [Accepted: 03/28/2017] [Indexed: 12/16/2022] Open
Abstract
Although the survival rate of hematologic malignancies in young patients is very high, cytotoxic therapies such as chemotherapy and total body irradiation therapy can significantly reduce a patient's reproductive capacity and cause irreversible infertility. Early ovarian failure also commonly occurs following additional cancer treatment, bone marrow transplantation, or autologous transplantation. Because the risk of early ovarian failure depends on the patient's circumstances, patients with a hematologic malignancy must consult health professionals regarding fertility preservation before undergoing treatments that can potentially damage their ovaries. While it is widely known that early menopause commonly occurs following breast cancer treatment, there is a lack of reliable study results regarding fertility preservation during hematologic malignancy treatment. Therefore, an in-depth discussion between patients and health professionals about the pros and cons of the various options for fertility preservation is necessary. In this study, we review germ cell toxicity, which occurs during the treatment of hematologic malignancies, and propose guidelines for fertility preservation in younger patients with hematologic malignancies.
Collapse
Affiliation(s)
- Dong-Yun Lee
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seul Ki Kim
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Miran Kim
- Department of Obstetrics and Gynecology, Ajou University School of Medicine, Suwon, Korea
| | - Kyung Joo Hwang
- Department of Obstetrics and Gynecology, Ajou University School of Medicine, Suwon, Korea
| | - Seok Hyun Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
18
|
Weterings MAJ, Glanville E, van Eekelen R, Den Hartog JE, Farquhar C. Interventions for fertility preservation in women with cancer undergoing chemotherapy. Hippokratia 2017. [DOI: 10.1002/14651858.cd012891] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Maria AJ Weterings
- Maastrict University Medical Center; P. Debyelaan 25 Maastricht Limburg Netherlands 6229 HX
| | | | - Rik van Eekelen
- Academic Medical Center, Amsterdam; Centre for Reproductive Medicine; Meibergdreef 9 Amsterdam Netherlands 1105 AZ
| | - Janneke Eva Den Hartog
- Maastricht University Medical Centre; Department of Gynecology and Obstetrics; P. Debyelaan 25 Maastricht Netherlands 6229 HX
| | - Cindy Farquhar
- University of Auckland; Department of Obstetrics and Gynaecology; FMHS Park Road Grafton Auckland New Zealand 1003
| |
Collapse
|
19
|
Srikanthan A, Amir E, Bedard P, Giuliani M, Hodgson D, Laframboise S, Prica A, Yee K, Greenblatt E, Lewin J, Gupta A. Fertility preservation in post-pubescent female cancer patients: A practical guideline for clinicians. Mol Clin Oncol 2017; 8:153-158. [PMID: 29387409 DOI: 10.3892/mco.2017.1486] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 10/30/2017] [Indexed: 11/06/2022] Open
Abstract
Increasing accessibility of fertility preservation (FP) options has permitted women to retain fertility following anticancer therapies. Several published guidelines have made recommendations for FP however their implementation into practice is currently unknown. In this review, we aim to provide oncology clinicians practical information about FP options for post-pubescent female cancer patients and recommendations for care delivery in order to answer preliminary questions and help triage whether FP referral is appropriate. Herein, we present a resource for oncology providers to guide them with FP discussions. Key points that are discussed in this critical review include: i) All cancer patients beginning a new plan of care should be informed of potential fertility risk. ii) If a woman requests further information on FP interventions, referral to a FP clinic should be made. iii) Given the evolving technologies in this area, patients should be informed of those which are proven and unproven, with oocyte and embryo preservation recognized as standard practice. iv) Random start (independent of menstrual cycle day) techniques are available to minimize oncologic treatment delays. v) Specific protocols for ovarian stimulation may be center-specific. vi) There is unlikely an increased cancer recurrence risk as a result of stimulation protocols in women with hormone-sensitive cancers. vii) Lastly, given the absence of consensus in the literature, routine use of GnRH analogs is not recommended for all cancer patients, however may be considered in select cases, such as high-risk women in whom definitive FP is not possible or feasible.
Collapse
Affiliation(s)
- Amirrtha Srikanthan
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada
| | - Eitan Amir
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada
| | - Philippe Bedard
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada
| | - Meredith Giuliani
- Department of Radiation Oncology, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada
| | - David Hodgson
- Department of Radiation Oncology, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada
| | - Stephanie Laframboise
- Department of Gynecologic Oncology, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada
| | - Anca Prica
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada
| | - Karen Yee
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada
| | - Ellen Greenblatt
- Mount Sinai Centre for Fertility and Reproductive Health, Mount Sinai Hospital, Toronto, ON M5T 2Z5, Canada
| | - Jeremy Lewin
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada
| | - Abha Gupta
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada.,Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| |
Collapse
|
20
|
Cima LN, Colita A, Fica S. Perspectives on the co-treatment with GnRHa in female patients undergoing hematopoietic stem cell transplantation. Endocr Connect 2017; 6:R162-R170. [PMID: 28947558 PMCID: PMC5655684 DOI: 10.1530/ec-17-0246] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 09/25/2017] [Indexed: 11/08/2022]
Abstract
Outcomes after hematopoietic stem cell transplantation (HSCT) for patients with both malignant and nonmalignant diseases have improved significantly in recent years. However, the endocrine system is highly susceptible to damage by the high-dose chemotherapy and/or irradiation used in the conditioning regimen before HSCT. Ovarian failure and subsequent infertility are frequent complications that long-term HSCT survivors and their partners face with a negative impact on their QoL. Several meta-analyses of randomized clinical trials showed that gonadotropin-releasing hormone agonist (GnRHa) administration in advance of starting standard chemotherapy decreases the risk of gonadal dysfunction and infertility in cancer patients, but GnRHa use for ovarian protection in HSCT patients is not fully determined. In this review, we are discussing the potential preservation of ovarian function and fertility in pubertal girls/premenopausal women who undergo HSCT using GnRHa in parallel with conditioning chemotherapy, focusing on the current data available and making some special remarks regarding the use of GnRHa.
Collapse
Affiliation(s)
| | - Anca Colita
- Carol Davila University of Medicine and PharmacyBucharest, Romania
- Hematology DepartmentFundeni Hospital, Bucharest, Romania
| | - Simona Fica
- Carol Davila University of Medicine and PharmacyBucharest, Romania
- Endocrine DepartmentElias Hospital, Bucharest, Romania
| |
Collapse
|
21
|
Guenther V, Alkatout I, Junkers W, Bauerschlag D, Maass N, von Otte S. Fertility Preservation in Female Patients with Breast Cancer - a Current Overview. Geburtshilfe Frauenheilkd 2017; 77:1088-1094. [PMID: 29093602 PMCID: PMC5658233 DOI: 10.1055/s-0043-119543] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 09/08/2017] [Accepted: 09/11/2017] [Indexed: 12/12/2022] Open
Abstract
Many premenopausal patients who develop breast cancer have not yet completed their family planning, so measures of fertility protection to preserve their fertile potential would be beneficial. Polychemotherapy causes irreversible damage to the ovarian follicles – irrespective of whether in a neoadjuvant or adjuvant setting – and this can sometimes result in permanent infertility. Depending on which cytostatic agents are used and on the age-related ovarian reserve of the woman, gonadotoxic risk must be classified as low, moderate or high. Options of fertility preservation include: a) cryopreservation of fertilised or unfertilised oocytes. After ovarian hyperstimulation, mature oocytes are retrieved by transvaginal follicle aspiration, after which they are cryopreserved, either unfertilised or on completion of IVF or ICSI treatment. During b) cryopreservation of ovarian tissue, about 50% of the ovarian cortex of one ovary is resected with the aid of a laparoscopic procedure and cryopreserved. The application of c) GnRH agonists as a medicinal therapy option is an attempt at endocrine ovarian suppression in order to protect oocytes, granulosa cells and theca cells from the cytotoxic effect of chemotherapy.
Collapse
Affiliation(s)
- Veronika Guenther
- Department of Gynaecology and Obstetrics, UKSH Campus Kiel, Kiel, Germany
| | - Ibrahim Alkatout
- Department of Gynaecology and Obstetrics, UKSH Campus Kiel, Kiel, Germany
| | - Wiebe Junkers
- University Fertility Centre, Medical Care Centre, University Medical Centre Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Dirk Bauerschlag
- Department of Gynaecology and Obstetrics, UKSH Campus Kiel, Kiel, Germany
| | - Nicolai Maass
- Department of Gynaecology and Obstetrics, UKSH Campus Kiel, Kiel, Germany
| | - Soeren von Otte
- University Fertility Centre, Medical Care Centre, University Medical Centre Schleswig-Holstein, Campus Kiel, Kiel, Germany
| |
Collapse
|
22
|
Abstract
Early menopause or premature ovarian insufficiency (POI) is a common cause of infertility in women and affects about one per cent of young women. This disorder has significant psychological sequelae and major health implications. Its relevance has increased in recent years due to the fact that age of motherhood is being delayed in developed countries, with the risk of having either primary ovarian insufficiency or less possibilities of pregnancy. The main characteristics are absence of ovulation, amenorrhoea and high levels of serum gonadothropins (hypergonadotropic hypogonadism). Although the aetiology remains uncertain in most cases, several rare specific causes have been elucidated. Potential causes for POI are iatrogenic (ovarian surgery, radiotherapy or chemotherapy), environmental factors, viral infections, metabolic and autoinmune diseases, and genetic alterations. Because of the association with other autoimmune diseases, close follow up is recommended in patients with POI. The traditional indicators to evaluate ovarian ageing are age, serum hormonal levels, anti-Mullerian hormone, antral follicle count, and ultrasonography of ovaries. Hormone replacement therapy remains the mainstay of treatment, and the best chance of achieving a pregnancy is through oocyte donation. This article aims to present an overview of potential causes, clinical manifestations, and treatment options of POI.
Collapse
Affiliation(s)
- Claudio Hernández-Angeles
- Gynecoobstetrics number 4 "Luis Castelazo Ayala" Hospital, Mexican Social Security Institute, Mexico
| | - Camil Castelo-Branco
- Clínic Institute of Gynecology, Obstetrics and Neonatology, Hospital Clínic- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Faculty of Medicine-University of Barcelona, Barcelona, Spain
| |
Collapse
|
23
|
Garrido-Oyarzún MF, Castelo-Branco C. Controversies over the use of GnRH agonists for reduction of chemotherapy-induced gonadotoxicity. Climacteric 2016; 19:522-525. [PMID: 27644002 DOI: 10.1080/13697137.2016.1225713] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The increase in cancer incidence in younger people and the significant improvement in long and permanent remission have brought concern about their reproductive future and quality of life. Up to two-thirds of adult female patients undergoing chemotherapy for malignancies eventually develop premature ovarian failure. This condition is related to many complaints including vasomotor symptoms, osteoporosis, increased risk of cardiovascular diseases, sexual dysfunction, and infertility. Therefore, protection against iatrogenic infertility and loss of endocrine ovarian function caused by chemotherapy is currently of high priority. Several options have been used for preserving ovarian function. Established methods include cryopreservation of embryos and/or ova, and ovarian transposition, while others such as ovarian tissue preservation are new, yet promising treatments for fertility preservation. The administration of gonadotropin releasing hormone (GnRH) agonistic analogs (GnRH-a) is still considered experimental. However, the recent evidence is strong to recommend the use of GnRH-a co-treatment during chemotherapy in young women with cancer to protect ovarian function, with promising results regarding fertility preservation. As the use of GnRH-a is non-invasive, highly available and without impact on cancer treatment outcomes, it should be offered to all young female cancer patients to preserve their ovarian function.
Collapse
Affiliation(s)
- M F Garrido-Oyarzún
- a Clínica Universidad de los Andes, Department of Obstetrics & Gynecology and Reproductive Biology, Faculty of Medicine , Universidad de los Andes , Santiago , Chile
| | - C Castelo-Branco
- b Clinic Institute of Gynecology, Obstetrics and Neonatology , Hospital Clinic-Institut d'Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona , Barcelona , Spain
| |
Collapse
|
24
|
Zavras N, Siristatidis C, Siatelis A, Koumarianou A. Fertility Risk Assessment and Preservation in Male and Female Prepubertal and Adolescent Cancer Patients. CLINICAL MEDICINE INSIGHTS-ONCOLOGY 2016; 10:49-57. [PMID: 27398041 PMCID: PMC4927042 DOI: 10.4137/cmo.s32811] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 05/22/2016] [Accepted: 05/26/2016] [Indexed: 12/15/2022]
Abstract
Cancer represents the second cause of death in prepubertal children and adolescents, although it is currently associated with an overall survival rate of 80%–85%. The annual incidence rate is 186.6 per 1 million children and adolescents aged up to 19 years. Both disease and treatment options are associated with life-altering, long-term effects that require monitoring. Infertility is a common issue, and as such, fertility preservation represents an essential part in the management of young patients with cancer who are at risk of premature gonadal failure. This review deals with the up-to-date available data on fertility risk assessment and preservation strategies that should be addressed prior to antineoplastic therapy in this vulnerable subgroup of cancer patients.
Collapse
Affiliation(s)
- Nikolaos Zavras
- Unit of Pediatric and Adolescent Surgery, Third Department of Surgery, Attikon Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Charalampos Siristatidis
- Assisted Reproduction Unit, Third Department of Obstetrics and Gynecology, Attikon Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Argyris Siatelis
- Urology Department, Attikon Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Anna Koumarianou
- Consultant in Medical Oncology, Hematology-Oncology Unit, Fourth Department of Internal Medicine, Attikon Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
25
|
Munhoz RR, Pereira AAL, Sasse AD, Hoff PM, Traina TA, Hudis CA, Marques RJ. Gonadotropin-Releasing Hormone Agonists for Ovarian Function Preservation in Premenopausal Women Undergoing Chemotherapy for Early-Stage Breast Cancer: A Systematic Review and Meta-analysis. JAMA Oncol 2016; 2:65-73. [PMID: 26426573 DOI: 10.1001/jamaoncol.2015.3251] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
IMPORTANCE Chemotherapy may result in a detrimental effect on ovarian function and fertility in premenopausal women undergoing treatment for early-stage breast cancer (EBC). To minimize risk of harm to ovarian function and fertility for patients in this setting, careful considerations should be made. Gonadotropin-releasing hormone agonists (GnRHa) have been suggested as an alternative to prevent the loss of ovarian function due to exposure to cytotoxic agents, but GnRHa use for ovarian protection in EBC patients is not fully resolved. OBJECTIVE To determine the effectiveness of GnRHa administered concurrently with chemotherapy for ovarian function preservation. DATA SOURCES PubMed, SCOPUS, and Cochrane databases were searched for studies published between January 1975 and March 2015. The abstracts of the American Society of Clinical Oncology Annual Meeting between 1995 and 2014 and the San Antonio Breast Cancer Symposium between 2009 and 2014 were searched as well. STUDY SELECTION Prospective, randomized, clinical trials addressing the role of ovarian suppression with GnRHa in preventing early ovarian dysfunction in premenopausal women undergoing treatment for EBC were selected. DATA EXTRACTION AND SYNTHESIS Data extraction was performed independently by 2 authors. The methodology and the risk of bias were assessment based on the description of randomization method, withdrawals, and blinding process. MAIN OUTCOMES AND MEASURES Rate of resumption of regular menses after a minimal follow-up period of 6 months following chemotherapy was used as a surrogate to assess the incidence of ovarian dysfunction. Additional secondary outcomes included hormone levels and number of pregnancies. Risk ratio estimates were calculated based on the number of evaluable patients. Analyses were conducted using a random effect model. RESULTS Seven studies were included in this analysis, totaling 1047 randomized patients and 856 evaluable patients. The use of GnRHa was associated with a higher rate of recovery of regular menses after 6 months (odds ratio [OR], 2.41; 95% CI, 1.40-4.15; P = .002) and at least 12 months (OR, 1.85; 95% CI, 1.33-2.59; P < .001) following the last chemotherapy cycle. The use of GnRHa was also associated with a higher number of pregnancies (OR, 1.85; 95% CI, 1.02-3.36; P = .04), although this outcome was not uniformly reported and fertility or rate of pregnancy was not the primary outcome in any of the trials. CONCLUSIONS AND RELEVANCE Gonadotropin-releasing hormone agonists given with chemotherapy was associated with increased rates of recovery of regular menses in this meta-analysis. Evidence was insufficient to assess outcomes related to GnRHa and ovarian function and fertility and needs further investigation.
Collapse
Affiliation(s)
- Rodrigo R Munhoz
- Hospital Sírio Libanês, São Paulo, Brazil2Instituto do Cancer do Estado de São Paulo, Universidade de São Paulo, São Paulo, Brazil
| | - Allan A L Pereira
- Hospital Sírio Libanês, São Paulo, Brazil2Instituto do Cancer do Estado de São Paulo, Universidade de São Paulo, São Paulo, Brazil
| | | | - Paulo M Hoff
- Hospital Sírio Libanês, São Paulo, Brazil2Instituto do Cancer do Estado de São Paulo, Universidade de São Paulo, São Paulo, Brazil
| | - Tiffany A Traina
- Breast Cancer Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Clifford A Hudis
- Breast Cancer Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | | |
Collapse
|
26
|
Sonigo C, Simon C, Boubaya M, Benoit A, Sifer C, Sermondade N, Grynberg M. What threshold values of antral follicle count and serum AMH levels should be considered for oocyte cryopreservation after in vitro maturation? Hum Reprod 2016; 31:1493-500. [PMID: 27165625 DOI: 10.1093/humrep/dew102] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 04/12/2016] [Indexed: 11/12/2022] Open
Abstract
STUDY QUESTION What threshold values of ultrasonographic antral follicle count (AFC) and serum anti-Müllerian hormone (AMH) levels should be considered for ensuring the cryopreservation of sufficient number of in vitro matured (IVM) oocytes, in cancer patients seeking fertility preservation (FP)? SUMMARY ANSWER AFC and serum AMH values >20 follicles and 3.7 ng/ml, respectively, are required for obtaining at least 10 IVM oocytes for cryopreservation. WHAT IS KNOWN ALREADY IVM of cumulus oocyte complexes (COCs) followed by oocyte cryopreservation has emerged recently as an option for urgent FP. Recent data have reported that, in healthy patients, 8-20 cryopreserved oocytes after ovarian stimulation would maximize the chance of obtaining a live birth. Although both AFC and AMH have been reported as predictive factors of IVM success in infertile patients with polycystic ovary syndrome (PCOS), there is a dramatic lack of data regarding the values of these parameters in oncological patients as candidates for FP. STUDY DESIGN, SIZE, DURATION From January 2009 to April 2015, we prospectively studied 340 cancer patients, aged 18-41 years, as candidates for oocyte cryopreservation following IVM. PARTICIPANTS/MATERIALS, SETTING, METHODS All patients had AFC and AMH measurements, 48-72 h before oocyte retrieval, regardless of the phase of the cycle. COCs were recovered under ultrasound guidance 36 h after hCG priming. Logistic regression allowed the determination of threshold values of AFC and AMH, for obtaining at least 8, 10 or 15 matures oocytes frozen after the IVM procedure. Similar analyses were performed for a final number of mature oocytes ≤2. MAIN RESULTS AND THE ROLE OF CHANCE Among the 340 cancer patients included, 300 were diagnosed with breast cancers, 14 had hematological malignancies and 26 underwent the procedure for others indications. Overall, the mean age of the population was 31.8 ± 4.5 years. Mean AFC and serum AMH levels were 21.7 ± 13.3 follicles and 4.4 ± 3.8 ng/ml, respectively. IVM was performed in equal proportions during the follicular or luteal phase of the cycle (49 and 51%, respectively). Statistical analysis showed that AFC and AMH values above 28 follicles and 3.9 ng/ml, 20 follicles and 3.7 ng/ml and 19 follicles and 3.5 ng/ml are required, respectively, for obtaining at least 15, 10 or 8 frozen IVM oocytes with a sensitivity ranging from 0.82 to 0.90. On the contrary, ≤2 IVM oocytes were cryopreserved when AFC and AMH were <19 follicles and 3.0 ng/ml, respectively. LIMITATIONS, REASONS FOR CAUTION Although the potential of cryopreserved IVM oocytes from cancer patients remains unknown, data obtained from infertile PCOS women have shown a dramatically reduced competence of these oocytes when compared with that of oocytes recovered after ovarian stimulation. As a consequence, the optimal number of IVM oocytes frozen in candidates for FP is currently unpredictable. WIDER IMPLICATIONS OF THE FINDINGS Cryopreservation of oocytes after IVM should be considered in the FP strategy when ovarian stimulation is unfeasible, in particular when markers of the follicular ovarian status are at a relatively high range. Further investigation is needed to objectively assess the real potential of these IVM oocytes after cryopreservation. Therefore, even when a good COCs yield is expected, we should systematically encourage IVM in combination with ovarian tissue cryopreservation. STUDY FUNDING/COMPETING INTERESTS No external funding was obtained for the present study. The authors have no conflict of interest to declare. TRIAL REGISTRATION NUMBER Not applicable.
Collapse
Affiliation(s)
- C Sonigo
- AP-HP, Service de Médecine de la Reproduction et Préservation de la Fertilité, Hôpital Jean Verdier, Avenue du 14 Juillet, Bondy 93143, France INSERM, U1185, Université Paris-Sud, Le Kremlin-Bicêtre 94270, France Université Paris XIII, Bobigny 93000, France
| | - C Simon
- AP-HP, Service de Médecine de la Reproduction et Préservation de la Fertilité, Hôpital Jean Verdier, Avenue du 14 Juillet, Bondy 93143, France
| | - M Boubaya
- Unité de Recherche Clinique, Hôpital Avicenne, Bobigny 93009, France
| | - A Benoit
- AP-HP, Service de Médecine de la Reproduction et Préservation de la Fertilité, Hôpital Jean Verdier, Avenue du 14 Juillet, Bondy 93143, France
| | - C Sifer
- AP-HP, Service de Cytogénétique et Biologie de la Reproduction, Hôpital Jean Verdier, Avenue du 14 Juillet, Bondy 93143, France
| | - N Sermondade
- AP-HP, Service de Cytogénétique et Biologie de la Reproduction, Hôpital Jean Verdier, Avenue du 14 Juillet, Bondy 93143, France
| | - M Grynberg
- AP-HP, Service de Médecine de la Reproduction et Préservation de la Fertilité, Hôpital Jean Verdier, Avenue du 14 Juillet, Bondy 93143, France Université Paris XIII, Bobigny 93000, France INSERM, U1133, Université Paris-Diderot, Paris 75013, France
| |
Collapse
|
27
|
Lambertini M, Del Mastro L, Pescio MC, Andersen CY, Azim HA, Peccatori FA, Costa M, Revelli A, Salvagno F, Gennari A, Ubaldi FM, La Sala GB, De Stefano C, Wallace WH, Partridge AH, Anserini P. Cancer and fertility preservation: international recommendations from an expert meeting. BMC Med 2016; 14:1. [PMID: 26728489 PMCID: PMC4700580 DOI: 10.1186/s12916-015-0545-7] [Citation(s) in RCA: 353] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 12/16/2015] [Indexed: 12/28/2022] Open
Abstract
In the last years, thanks to the improvement in the prognosis of cancer patients, a growing attention has been given to the fertility issues. International guidelines on fertility preservation in cancer patients recommend that physicians discuss, as early as possible, with all patients of reproductive age their risk of infertility from the disease and/or treatment and their interest in having children after cancer, and help with informed fertility preservation decisions. As recommended by the American Society of Clinical Oncology and the European Society for Medical Oncology, sperm cryopreservation and embryo/oocyte cryopreservation are standard strategies for fertility preservations in male and female patients, respectively; other strategies (e.g. pharmacological protection of the gonads and gonadal tissue cryopreservation) are considered experimental techniques. However, since then, new data have become available, and several issues in this field are still controversial and should be addressed by both patients and their treating physicians.In April 2015, physicians with expertise in the field of fertility preservation in cancer patients from several European countries were invited in Genova (Italy) to participate in a workshop on the topic of "cancer and fertility preservation". A total of ten controversial issues were discussed at the conference. Experts were asked to present an up-to-date review of the literature published on these topics and the presentation of own unpublished data was encouraged. On the basis of the data presented, as well as the expertise of the invited speakers, a total of ten recommendations were discussed and prepared with the aim to help physicians in counseling their young patients interested in fertility preservation.Although there is a great interest in this field, due to the lack of large prospective cohort studies and randomized trials on these topics, the level of evidence is not higher than 3 for most of the recommendations highlighting the need of further research efforts in many areas of this field. The participation to the ongoing registries and prospective studies is crucial to acquire more robust information in order to provide evidence-based recommendations.
Collapse
Affiliation(s)
- Matteo Lambertini
- Department of Medical Oncology, U.O. Oncologia Medica 2, IRCCS AOU San Martino - IST, Genoa, Italy.
| | - Lucia Del Mastro
- Department of Medical Oncology, U.O. Sviluppo Terapie Innovative, IRCCS AOU San Martino - IST, Genoa, Italy
| | - Maria C Pescio
- Physiopathology of Human Reproduction, IRCCS AOU San Martino - IST, Genoa, Italy
| | - Claus Y Andersen
- Laboratory of Reproductive Biology, Section 5712, Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Hatem A Azim
- BrEAST Data Centre, Department of Medicine, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Fedro A Peccatori
- Fertility and Procreation Unit, Gynecologic Oncology Department, European Institute of Oncology, Milan, Italy
| | - Mauro Costa
- Reproductive Medicine Department, International Evangelic Hospital, Genoa, Italy
| | - Alberto Revelli
- Physiopathology of Reproduction and In Vitro Fertilization Unit, S. Anna Hospital, University of Turin, Turin, Italy
| | - Francesca Salvagno
- Physiopathology of Reproduction and In Vitro Fertilization Unit, S. Anna Hospital, University of Turin, Turin, Italy
| | | | - Filippo M Ubaldi
- GENERA Centre for Reproductive Medicine, Clinica Valle Giulia, Rome, Italy
| | - Giovanni B La Sala
- Obstetric and Gynecology Department, Azienda Ospedaliera Arcispedale S. Maria Nuova-IRCCS, University of Modena and Reggio Emilia, Reggio Emilia, Italy
| | - Cristofaro De Stefano
- Children and Women Health Department, Physiopathology of Human Reproduction Unit, "San Giuseppe Moscati" Hospital, Avellino, Italy
| | - W Hamish Wallace
- Department of Haematology/Oncology, Royal Hospital for Sick Children, and Department of Child Life and Health, University of Edinburgh, Edinburgh, UK
| | - Ann H Partridge
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Paola Anserini
- Physiopathology of Human Reproduction, IRCCS AOU San Martino - IST, Genoa, Italy
| |
Collapse
|
28
|
|
29
|
Abstract
Fertility preservation is the process by which either oocytes (eggs) or sperm undergo an intervention to preserve their use for future attempts at conception. Consideration of fertility preservation in the pediatric and adolescent population is important, as future childbearing is usually a central life goal. For postpubertal girls, both oocyte and embryo cryopreservation are standard of care and for postpubertal boys, sperm cryopreservation continues to be recommended. Although all the risks are unknown, it appears that fertility preservation in most cases does not worsen prognosis, allows for the birth of healthy children, and does not increase the chance of recurrence.
Collapse
Affiliation(s)
- Stephanie J Estes
- Donor Oocyte Program, Robotic Surgical Services, Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Pennsylvania State University, College of Medicine, Hershey Medical Center, Mail Code H103, 500 University Drive, Hershey, PA 17033-0850, USA.
| |
Collapse
|
30
|
Blumenfeld Z, Zur H, Dann EJ. Gonadotropin-Releasing Hormone Agonist Cotreatment During Chemotherapy May Increase Pregnancy Rate in Survivors. Oncologist 2015; 20:1283-9. [PMID: 26463871 DOI: 10.1634/theoncologist.2015-0223] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 08/13/2015] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The use of gonadotropin-releasing hormone analogs (GnRHas) for fertility preservation is not unequivocally accepted. It is controversial whether GnRHa can increase the pregnancy rate in survivors. PATIENTS AND METHODS This is a retrospective cohort study. Every patient referred for fertility preservation was offered cryopreservation of embryos, ova, and ovarian tissue and GnRHa. The patients were consecutively included. The primary outcome was spontaneous pregnancies. The secondary outcome was cyclic ovarian function (COF) versus premature ovarian failure (POF). These outcomes were assessed 2 years or more after chemotherapy. RESULTS We compared 286 patients who received gonadotropin-releasing hormone agonist (GnRHa) with chemotherapy with 188 patients who were treated with chemotherapy alone. Ovarian function could be determined in 217 patients. Overall, 87% (127 of 146) of the patients in the GnRHa group retained COF and 13% (19 of 146) suffered POF, whereas in the control group, 49% (35 of 71) experienced COF and 51% (36 of 71) suffered POF (p = .0001). The odds ratio (OR) for preserving COF was 6.87 for the patients who received GnRHa (95% confidence interval [CI] 3.4-13.4). Overall 60% (112 of 188) of the survivors conceived: 69.3% (84 of 122) of the patients in the GnRHa group compared with 42.4% (28 of 66) in the control group (p = .006). In the GnRHa group, 123 healthy newborns were delivered, versus 40 in the controls. Spontaneous pregnancies occurred in 65.6% (80 of 122) of the survivors in the GnRHa group versus 37.9% (25 of 66) in the control group (p = .0004, OR 3.12, 95% CI 1.7-5.8). CONCLUSION Adding GnRHa to chemotherapy significantly increases the OR for spontaneous conception, in addition to COF. It is suggested that GnRHa cotreatment should be added before and during gonadotoxic chemotherapy. IMPLICATIONS FOR PRACTICE The use of gonadotropin-releasing hormone analogs (GnRHa) for fertility preservation is not unequivocally accepted and is even controversial. This study compared 286 patients who received GnRHa with chemotherapy with 188 patients who were treated with chemotherapy alone. Ovarian function could be determined in 217 patients. The odds ratio for preserving cyclic ovarian function was 6.87 for the patients who received GnRHa. Furthermore, the total and spontaneous pregnancy rate was significantly higher for those who received the agonist (p = .006). Adding GnRHa to chemotherapy significantly increased the odds ratio for spontaneous conception, in addition to preserving regular ovarian function. It is suggested that GnRHa cotreatment should be administered to young women in conjunction with gonadotoxic chemotherapy.
Collapse
Affiliation(s)
- Zeev Blumenfeld
- Department of Gynecology and Obstetrics, Rambam Health Care Campus and Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel Department of Reproductive Endocrinology, Rambam Health Care Campus and Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Hilli Zur
- Department of Reproductive Endocrinology, Rambam Health Care Campus and Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Eldad J Dann
- Department of Hematology, Rambam Health Care Campus and Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
31
|
Biglia N, Torrisi R, D'Alonzo M, Codacci Pisanelli G, Rota S, Peccatori FA. Attitudes on fertility issues in breast cancer patients: an Italian survey. Gynecol Endocrinol 2015; 31:458-64. [PMID: 25982361 DOI: 10.3109/09513590.2014.1003293] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Fertility issues should be discussed with young women before the start of any anticancer treatment. The study is aimed to investigate the attitude on fertility among Italian oncologists and breast surgeons dealing with BCa, and to report the consensus achieved on specific statements. METHODS One hundred and sixty-two panelists anonymously expressed an opinion through a web-based platform on 19 statements based on the Delphi method. RESULTS Ninety-one percent of oncologists considered important to discuss with patients about fertility issues and 83% believed estrogens could stimulate the growth of hidden cancer cells in ER(+) tumors. Difficulties in accessing fertility preservation procedures were mainly due to patients' reluctance, but also to lack of coordination with the assisted reproduction specialists. No full consensus was reached on the prognostic role of pregnancy after BCa. Fifty-four percent of oncologists declared that pregnancy does not affect oncologic prognosis. Treatment with GnRHa during chemotherapy was considered the only mean for preserving ovarian function. CONCLUSIONS Fertility preservation in BCa patients is a well-accepted practice among Italian oncologists. A poor knowledge of this specific issue emerged from the survey, even if a certain degree of agreement was observed on most fertility-related issues.
Collapse
Affiliation(s)
- Nicoletta Biglia
- Department of Gynaecology and Obstetrics, University of Turin , Turin , Italy
| | | | | | | | | | | |
Collapse
|
32
|
Zhu SF, Hu HB, Xu HY, Fu XF, Peng DX, Su WY, He YL. Human umbilical cord mesenchymal stem cell transplantation restores damaged ovaries. J Cell Mol Med 2015; 19:2108-17. [PMID: 25922900 PMCID: PMC4568915 DOI: 10.1111/jcmm.12571] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 01/29/2015] [Indexed: 12/20/2022] Open
Abstract
Ovarian injury because of chemotherapy can decrease the levels of sexual hormones and potentia generandi of patients, thereby greatly reducing quality of life. The goal of this study was to investigate which transplantation method for human umbilical cord mesenchymal stem cells (HUMSCs) can recover ovarian function that has been damaged by chemotherapy. A rat model of ovarian injury was established using an intraperitoneal injection of cyclophosphamide. Membrane-labelled HUMSCs were subsequently injected directly into ovary tissue or tail vein. The distribution of fluorescently labelled HUMSCs, estrous cycle, sexual hormone levels, and potentia generandi of treated and control rats were then examined. HUMSCs injected into the ovary only distributed to the ovary and uterus, while HUMSCs injected via tail vein were detected in the ovary, uterus, kidney, liver and lung. The estrous cycle, levels of sex hormones and potentia generandi of the treated rats were also recovered to a certain degree. Moreover, in some transplanted rats, fertility was restored and their offspring developed normally. While ovary injection could recover ovarian function faster, both methods produced similar results in the later stages of observation. Therefore, our results suggest that transplantation of HUMSCs by tail vein injection represents a minimally invasive and effective treatment method for ovarian injury.
Collapse
Affiliation(s)
- Shao-Fang Zhu
- Medical College of Shaoguan University, Shaoguan, GuangDong, China.,Department of Obstetrics and Gynaecology, Zhujiang Hospital, Southern Medical University, Guangzhou, GuangDong, China
| | - Hong-Bo Hu
- Department of Obstetrics and Gynaecology, The Yuebei People's Hospital of Shaoguan, Shaoguan, GuangDong, China
| | - Hong-Yan Xu
- Department of Obstetrics and Gynaecology, The Yuebei People's Hospital of Shaoguan, Shaoguan, GuangDong, China
| | - Xia-Fei Fu
- Department of Obstetrics and Gynaecology, Zhujiang Hospital, Southern Medical University, Guangzhou, GuangDong, China
| | - Dong-Xian Peng
- Department of Obstetrics and Gynaecology, Zhujiang Hospital, Southern Medical University, Guangzhou, GuangDong, China
| | - Wei-Yan Su
- Medical College of Shaoguan University, Shaoguan, GuangDong, China
| | - Yuan-Li He
- Department of Obstetrics and Gynaecology, Zhujiang Hospital, Southern Medical University, Guangzhou, GuangDong, China
| |
Collapse
|
33
|
Mahajan N. Fertility preservation in female cancer patients: An overview. J Hum Reprod Sci 2015; 8:3-13. [PMID: 25838742 PMCID: PMC4381379 DOI: 10.4103/0974-1208.153119] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 02/06/2015] [Accepted: 01/29/2015] [Indexed: 12/20/2022] Open
Abstract
Fertility preservation is becoming increasingly important to improve the quality of life in cancer survivors. Despite guidelines suggesting that discussion of fertility preservation should be done prior to starting cancer therapies, there is a lack of implementation in this area. A number of techniques are available for fertility preservation, and they can be used individually or together in the same patient to maximize efficiency. Oocyte and embryo cryopreservation are now established techniques but have their limitations. Ovarian tissue cryopreservation though considered experimental at present, has a wider clinical application and the advantage of keeping the fertility window open for a longer time. Both chemotherapy and radiotherapy have a major impact on reproductive potential and fertility preservation procedures should be carried out prior to these treatments. The need for fertility preservation has to be weighed against morbidity and mortality associated with cancer. There is thus a need for a multidisciplinary collaboration between oncologists and reproductive specialists to improve awareness and availability.
Collapse
Affiliation(s)
- Nalini Mahajan
- Department of Reproductive Medicine, Nova IVI Fertilit, New Delhi, India
| |
Collapse
|
34
|
Huser M, Smardova L, Janku P, Crha I, Zakova J, Stourac P, Jarkovsky J, Mayer J, Ventruba P. Fertility status of Hodgkin lymphoma patients treated with chemotherapy and adjuvant gonadotropin-releasing hormone analogues. J Assist Reprod Genet 2015; 32:1187-93. [PMID: 25724588 DOI: 10.1007/s10815-015-0452-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 02/19/2015] [Indexed: 11/29/2022] Open
Abstract
PURPOSE Aim of this prospective observational study was to analyze fertility status of Hodgkin lymphoma (HL) patients treated with different types of chemotherapy while receiving GnRH analogues to preserve ovarian function. METHODS Fertility status was assessed among 108 females in reproductive age treated by curative chemotherapy for freshly diagnosed HL between 2005 and 2010 in university-based tertiary fertility and oncology center. All patients received GnRH analogues during chemotherapy to preserve their ovarian function. Their reproductive functions were assessed by follicle-stimulating hormone (FSH) measurement and pregnancy achievement. Ovarian function was determined separately in three groups with increasing gonadotoxicity of chemotherapy. RESULTS One year following the treatment, normal ovarian function was found in 89 (82.4%) of patients. Two years after chemotherapy, 98 (90.7%) of patients retained their ovarian function, and 23 (21.3%) achieved clinical pregnancy during the follow-up period. Average FSH after chemotherapy was 11.6 ± 17.9 IU/l 1 year after the treatment resp. 9.0 ± 13.8 at the 2 years interval. There were significantly more patients with chemotherapy induced diminished ovarian reserve (chDOR) among the group receiving escalated BEACOPP chemotherapy in comparison with the other types of treatment (58.1% vs. 87.9% resp. 95.5%). CONCLUSION The rate of chDOR is significantly higher after EB poly-chemotherapy and there is no tendency for improvement in time. The 2 + 2 chemotherapy with GnRH-a required for more advanced HL retained ovarian function significantly better after 2 years. Another important advantage of GnRH-a co-treatment is the excellent control of patient's menstrual cycle.
Collapse
Affiliation(s)
- M Huser
- Department of Obstetrics and Gynecology, Brno University Hospital and Masaryk University Medical School, Jihlavska 20, 625 00, Brno, Czech Republic,
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
GNRH agonists and antagonists in rescue for cyclophosphamide-induced ovarian damage: friend or foe? Arch Gynecol Obstet 2014; 291:1403-10. [DOI: 10.1007/s00404-014-3564-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 11/25/2014] [Indexed: 10/24/2022]
|
36
|
Tan SJ, Lee LJ, Tzeng CR, Wang CW, Hsu MI, Chen CH. Targeted anti-apoptosis activity for ovarian protection against chemotherapy-induced ovarian gonadotoxicity. Reprod Biomed Online 2014; 29:612-20. [DOI: 10.1016/j.rbmo.2014.07.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Revised: 07/05/2014] [Accepted: 07/08/2014] [Indexed: 11/28/2022]
|
37
|
De Pinho JC, Sauer MV. Infertility and ART after transplantation. Best Pract Res Clin Obstet Gynaecol 2014; 28:1235-50. [DOI: 10.1016/j.bpobgyn.2014.07.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 07/14/2014] [Accepted: 07/18/2014] [Indexed: 11/27/2022]
|
38
|
Wilkosz P, Greggains GD, Tanbo TG, Fedorcsak P. Female reproductive decline is determined by remaining ovarian reserve and age. PLoS One 2014; 9:e108343. [PMID: 25310678 PMCID: PMC4195570 DOI: 10.1371/journal.pone.0108343] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 08/29/2014] [Indexed: 01/28/2023] Open
Abstract
The early decline and loss of female fertility in humans and other species represents an evolutionary paradox. Despite being born with a vast stock of oocytes, females encounter an exhaustion of ovarian reserve and sterility half way through their natural lives. Female reproductive ageing has been proposed to proceed as an ongoing decline in ovarian reserve, determined by remaining ovarian follicle number. However, despite extensive modelling, the respective contributions of intra-, inter-, and extra-ovarian signalling have not been fully characterised. It remains unclear whether reproductive ageing progresses simply as a pre-determined function of remaining ovarian follicles, or as an age-dependent process in humans. Here, we have analysed ovarian response to hormonal stimulation in women who have undergone surgical removal of a single ovary, in order to investigate the relative contributions of intra-, inter, and extra-ovarian signalling on reproductive ageing. Our data show that in unilaterally oophorectomised women, ovarian response to follicle stimulating hormone (FSH) declines beyond levels predicted by a total ovarian follicle pool model of reproductive ageing. Maintenance of ovarian function later in reproductive life, despite the removal of half of the total ovarian reserve, suggests a role for an extra-ovarian age-dependent regulation of reproductive decline. This highlights the need for further work to identify signalling factors that communicate age-related signals between the soma and the germline.
Collapse
Affiliation(s)
- Pawel Wilkosz
- Section for Reproductive Medicine, Department of Gynecology, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- * E-mail:
| | - Gareth D. Greggains
- Section for Reproductive Medicine, Department of Gynecology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Tom G. Tanbo
- Section for Reproductive Medicine, Department of Gynecology, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Peter Fedorcsak
- Section for Reproductive Medicine, Department of Gynecology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| |
Collapse
|
39
|
Affiliation(s)
- Zeev Blumenfeld
- Department of Obstetrics and Gynecology, Reproductive Endocrinology, Rambam Health Care Campus, The Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
40
|
Kumar P, Sharma A. Gonadotropin-releasing hormone analogs: Understanding advantages and limitations. J Hum Reprod Sci 2014; 7:170-4. [PMID: 25395741 PMCID: PMC4229791 DOI: 10.4103/0974-1208.142476] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 09/23/2014] [Accepted: 09/19/2014] [Indexed: 12/05/2022] Open
Abstract
Pituitary stimulation with pulsatile gonadotropin-releasing hormone (GnRH) analogs induces both follicle-stimulating hormone (FSH) and luteinizing hormone (LH). Pituitary gonadotropin secretions are blocked upon desensitization when a continuous GnRH stimulus is provided by means of an agonist or when the pituitary receptors are occupied with a competitive antagonist. GnRH antagonists were not available originally; therefore, prolonged daily injections of agonist with its desensitizing effect were used. Today, single- and multiple-dose injectable antagonists are also available to block the LH surge and thus to cause desensitization. This review provides an overview of the use of GnRH analogs which is potent therapeutic agents that are considerably useful in a variety of clinical indications from the past to the future with some limitations. These indications include management of endometriosis, uterine leiomyomas, hirsutism, dysfunctional uterine bleeding, premenstrual syndrome, assisted reproduction, and some hormone-dependent tumours, other than ovulation induction.
Collapse
Affiliation(s)
- Pratap Kumar
- Department of Obstetrics and Gynecology, Kasturba Medical College, Manipal University, Manipal, Karnataka, India
| | - Alok Sharma
- Department of Obstetrician and Gynecologist, Deen Dayal Upadhyaya Hospital, Shimla, Himachal Pradesh, India
| |
Collapse
|
41
|
Fernbach A, Lockart B, Armus CL, Bashore LM, Levine J, Kroon L, Sylvain G, Rodgers C. Evidence-Based Recommendations for Fertility Preservation Options for Inclusion in Treatment Protocols for Pediatric and Adolescent Patients Diagnosed With Cancer. J Pediatr Oncol Nurs 2014; 31:211-222. [PMID: 24799444 PMCID: PMC5213740 DOI: 10.1177/1043454214532025] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
As survival rates improve for pediatric cancers, increased attention has been paid to late effects of cancer therapy, in particular, infertility. Fertility preservation options are available for pre- and postpubertal cancer patients; however, many providers lack knowledge regarding options. The aim of this article is to provide a comprehensive synthesis of current evidence and recommendations regarding fertility preservation options for children, adolescents, and young adults undergoing cancer treatment. A systematic search was performed to identify fertility preservation evidence. Fifty-three studies and 4 clinical guidelines were used for the review. Final recommendations consisted of 2 strong and 1 weak recommendation for both female and male fertility preservation options. The treatment team should be knowledgeable about fertility preservation so that they can educate patients and families about available fertility preservation options. It is important to consider and discuss all available fertility options with patients at the time of diagnosis.
Collapse
Affiliation(s)
| | | | - Cheryl L Armus
- MACC Fund Center for Cancer and Blood Disorders, Milwaukee, WI, USA
| | - Lisa M Bashore
- Life After Cancer Program, Cook Children's Medical Center, Fort Worth, TX, USA
| | | | - Leah Kroon
- Seattle Children's Hospital, Seattle, WA, USA
| | | | | |
Collapse
|
42
|
Abstract
Menstrual suppression to provide relief of menstrual-related symptoms or to manage medical conditions associated with menstrual morbidity or menstrual exacerbation has been used clinically since the development of steroid hormonal therapies. Options range from the extended or continuous use of combined hormonal oral contraceptives, to the use of combined hormonal patches and rings, progestins given in a variety of formulations from intramuscular injection to oral therapies to intrauterine devices, and other agents such as gonadotropin-releasing hormone (GnRH) antagonists. The agents used for menstrual suppression have variable rates of success in inducing amenorrhea, but typically have increasing rates of amenorrhea over time. Therapy may be limited by side effects, most commonly irregular, unscheduled bleeding. These therapies can benefit women's quality of life, and by stabilizing the hormonal milieu, potentially improve the course of underlying medical conditions such as diabetes or a seizure disorder. This review addresses situations in which menstrual suppression may be of benefit, and lists options which have been successful in inducing medical amenorrhea.
Collapse
Affiliation(s)
- Paula Adams Hillard
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
43
|
Sacco JJ, Cliff J, Green JA. Chemotherapy for gynaecological malignancies and fertility preservation. World J Obstet Gynecol 2014; 3:54-60. [DOI: 10.5317/wjog.v3.i2.54] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 05/31/2013] [Accepted: 08/06/2013] [Indexed: 02/05/2023] Open
Abstract
Infertility is an increasingly important issue for patients surviving cancer. Significant improvements in cancer management have led to greater numbers of patients living healthy and fulfilling lives for many years after a diagnosis of cancer, and the ability to bear children is a major component of well-being. Infertility is particularly challenging in gynaecological cancer, where multiple treatment modalities are often employed. Surgery may involve the removal of reproductive organs and subsequent chemotherapy may also lead to infertility. Mitigation of this through the use of cryopreservation of embryos, oocytes or ovarian tissue before chemotherapy may enable subsequent pregnancy in the patient or a surrogate mother. Suppression of ovarian function during chemotherapy is less well established, but promises a reduction in infertility without the risks associated with surgery. Similarly, evolving chemotherapy regimens with replacement of alkylating agents will reduce the incidence of infertility. With a combination of these techniques, an increasing proportion of patients may be able to conceive after completion of treatment, and there is no evidence of an increase in congenital abnormalities. This review discusses chemotherapy-induced infertility, interventions and success rates, and demonstrates that individualisation of management is required for optimum outcome.
Collapse
|
44
|
Harp D, Chowdhury I, Garcia-Barrio M, Welch C, Matthews R, Thompson W. Maintaining the reproductive potential of cancer patients during cancer treatment. Front Biosci (Schol Ed) 2014; 6:39-49. [PMID: 24389259 PMCID: PMC6178803 DOI: 10.2741/s412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cancer therapies are known to alter the reproductive potential in cancer patients. Due to improved survival rates in cancer patients of reproductive age, considerations of the long-term effects of cancer therapy have become more significant. Oncofertility is a new discipline in medicine that deals with maintaining the reproductive potential of cancer patients while they are receiving gonadotoxic cancer treatment. The purpose of this review is to explore how cancer treatment impairs reproductive functioning and present the current options for preservation of fertility in women. All patients with reproductive potential should be made aware of the possible treatment-related infertility and be offered appropriate fertility preservation options before cancer treatment is instituted. The hope is that, in the future, mechanism(s) can be developed to preserve immature germ cells in the ovary, so that they can be used for fertilization in vivo or in vitro.
Collapse
Affiliation(s)
- Djana Harp
- Department of Obstetrics and Gynecology
- Reproductive Science Research Program
| | - Indrajit Chowdhury
- Department of Obstetrics and Gynecology
- Reproductive Science Research Program
| | - Minerva Garcia-Barrio
- Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, Georgia 30310
| | | | - Roland Matthews
- Department of Obstetrics and Gynecology
- Reproductive Science Research Program
| | - Winston Thompson
- Department of Obstetrics and Gynecology
- Reproductive Science Research Program
| |
Collapse
|
45
|
Thomin A, Torre A, Daraï É, Chabbert-Buffet N. [Role of GnRH agonists in preserving female fertility]. ACTA ACUST UNITED AC 2013; 43:267-74. [PMID: 24321862 DOI: 10.1016/j.jgyn.2013.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 10/30/2013] [Accepted: 11/06/2013] [Indexed: 11/18/2022]
Abstract
The impact of cancer treatment on ovarian function and fertility has been known since the 70s. Preservation of fertility is now an important focus of care for patients of reproductive age with cancer. The beneficial role of GnRH agonists in fertility preservation is controversial since the early 2000s. Recent randomized studies come to overturn this role. The POEMS multicenter randomized trial with long-term follow-up is ongoing and will provide results that could help clarify the current uncertain indication of these compounds in this context.
Collapse
Affiliation(s)
- A Thomin
- Service de gynécologie obstétrique, médecine de la reproduction, hôpital Tenon, AP-HP, 4, rue de la Chine, 75020 Paris, France.
| | - A Torre
- Service de gynécologie obstétrique, médecine de la reproduction, centre hospitalier intercommunal Poissy, 78300 Saint-Germain-en-Laye, France
| | - É Daraï
- Service de gynécologie obstétrique, médecine de la reproduction, hôpital Tenon, AP-HP, 4, rue de la Chine, 75020 Paris, France
| | - N Chabbert-Buffet
- Service de gynécologie obstétrique, médecine de la reproduction, hôpital Tenon, AP-HP, 4, rue de la Chine, 75020 Paris, France
| |
Collapse
|
46
|
Gonadotropin-releasing hormone analogues for the prevention of chemotherapy-induced premature ovarian failure in cancer women: systematic review and meta-analysis of randomized trials. Cancer Treat Rev 2013; 40:675-83. [PMID: 24360817 DOI: 10.1016/j.ctrv.2013.12.001] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 11/27/2013] [Accepted: 12/02/2013] [Indexed: 12/22/2022]
Abstract
BACKGROUND The role of temporary ovarian suppression with gonadotropin-releasing hormone analogues (GnRHa) in the prevention of chemotherapy-induced premature ovarian failure (POF) is still controversial. We conducted a systematic review and meta-analysis of randomized trials evaluating the efficacy of GnRHa, given before and during chemotherapy, in the prevention of POF in premenopausal cancer patients. METHODS Studies were retrieved by searching PubMed, Web of Knowledge database and the proceedings of major conferences. We calculated Odds Ratios (OR) and 95% confidence intervals (CIs) for POF from each trial and obtained pooled estimates through the random effects model as suggested by DerSimonian and Laird. RESULTS Nine studies were included in the meta-analysis with 225 events of POF occurring in 765 analyzed patients. The pooled OR estimate indicates a highly significant reduction in the risk of POF (OR=0.43; 95% CI: 0.22-0.84; p=0.013) in patients receiving GnRHa. There was statistically significant heterogeneity among studies (I(2)=55.8%; p=0.012). There was no evidence of publication bias. Subgroups analyses showed that the protective effect of GnRHa against POF was similar in subgroups of patients defined by age and timing of POF assessment, while it was present in breast cancer but unclear in ovarian cancer and lymphoma patients. CONCLUSIONS Our pooled analysis of randomized studies shows that the temporary ovarian suppression induced by GnRHa significantly reduces the risk of chemotherapy-induced POF in young cancer patients.
Collapse
|
47
|
Ronn R, Holzer HEG. Oncofertility in Canada: gonadal protection and fertility-sparing strategies. ACTA ACUST UNITED AC 2013; 20:e602-7. [PMID: 24311962 DOI: 10.3747/co.20.1359] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Cancer can be a devastating diagnosis. In particular, malignancy and its indicated treatments have profoundly negative effects on the fertility of young cancer patients. Oncofertility has emerged as a new interdisciplinary field to address the issue of gonadotoxicity associated with cancer therapies and to facilitate fertility preservation. In Canada, fertility issues are often inadequately addressed despite the availability of resources. The goal of this four-part series is to facilitate systemic improvements in fertility preservation for adolescent and young adult Canadians with a new diagnosis of cancer. METHODS Here, we review the fertility preservation measures currently available. Medical and surgical strategies are both outlined. RESULTS Fertility-preserving strategies and gonadal protection have demonstrated variable success in a number of approaches. The value of hormone suppression is still in question for women. Progestins for endometrial cancer and alternative chemotherapies are other medical approaches. Gonadal shielding and protective surgical approaches have also been attempted. CONCLUSIONS The techniques discussed here may be selectively considered and integrated into patient care in an attempt to preserve future fertility before initiating cancer treatment.
Collapse
Affiliation(s)
- R Ronn
- Department of Obstetrics and Gynecology, Queen's University, Kingston, ON
| | | |
Collapse
|
48
|
Zhang Y, Xiao Z, Wang Y, Luo S, Li X, Li S. Gonadotropin-releasing hormone for preservation of ovarian function during chemotherapy in lymphoma patients of reproductive age: a summary based on 434 patients. PLoS One 2013; 8:e80444. [PMID: 24312222 PMCID: PMC3842920 DOI: 10.1371/journal.pone.0080444] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 10/02/2013] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Gonadotropin-releasing hormone agonists (GnRHa) might play a role in preserving ovarian function in lymphoma patients by inhibiting chemotherapy-induced ovarian follicular damage. However, studies of its clinical efficacy have reported conflicting results. METHOD We conducted a meta-analysis to determine the effect of the preservation of ovarian function by administering GnRHa in young patients with lymphoma undergoing chemotherapy. Seven studies were identified that met inclusion criteria and comprised 434 patients assigned to GnRHa combined chemotherapy or chemotherapy alone. RESULTS The incidence of women with premature ovarian failure (POF) demonstrated a statistically significant difference in favor of the use of GnRHa (OR=0.32, 95% CI 0.13-0.77). In addition, the final level of FSH in the GnRH group was significantly lower than control group. (MD= -11.73, 95% CI,-22.25- -1.20), and the final level of AMH in the GnRH group was significantly higher than control group (MD=0.80; 95% CI, 0.61-0.98). However, there was no statistically significant difference between treatment and the control groups in the incidence of a spontaneous pregnancy (OR=1.11; 95% CI, 0.55-2.26). CONCLUSION This meta-analysis suggests that GnRHa may be effective in protecting ovarian function during chemotherapy in lymphoma patients. More well-designed prospective studies are needed to carry out for further understanding of this topic.
Collapse
Affiliation(s)
- Yaoyao Zhang
- Reproductive Medical Center, Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P. R. China
| | - Zhun Xiao
- Reproductive Medical Center, Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P. R. China
| | - Yan Wang
- Reproductive Medical Center, Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P. R. China
| | - Shan Luo
- Reproductive Medical Center, Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P. R. China
| | - Xiaohong Li
- Reproductive Medical Center, Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P. R. China
| | - Shangwei Li
- Reproductive Medical Center, Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P. R. China
- * E-mail:
| |
Collapse
|
49
|
Utility of GnRH-agonists for Fertility Preservation in Women With Operable Breast Cancer: Is It Protective? CURRENT BREAST CANCER REPORTS 2013; 5:302-308. [PMID: 24707345 DOI: 10.1007/s12609-013-0123-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Breast cancer is the most common type of malignancy in reproductive-age women. Breast cancer chemotherapy is associated with premature ovarian failure, infertility and negative psychosocial effects related to these reproductive changes. As a result of this, fertility preservation becomes highly critical in this group of women. Besides the fertility preservation methods that utilize assisted reproductive technologies such as embryo, oocyte, and ovarian tissue cryopreservation, another suggested strategy for fertility preservation is suppression of ovarian ovulatory function by gonadotropin-releasing hormone agonist (GnRHa) administration before and during chemotherapy. However, both the efficacy and safety of GnRH agonists for prevention of ovarian damage are unproven and the preponderance of evidence indicates that this is an ineffective strategy. This review details the most recent information and studies on this controversial topic.
Collapse
|
50
|
Resetkova N, Hayashi M, Kolp LA, Christianson MS. Fertility Preservation for Prepubertal Girls: Update and Current Challenges. CURRENT OBSTETRICS AND GYNECOLOGY REPORTS 2013; 2:218-225. [PMID: 25110617 DOI: 10.1007/s13669-013-0060-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
With increasing rates of diagnosis of childhood cancers and the evolution of more effective treatment options resulting in prolonged life spans, fertility preservation counseling is an integral component of the discussion at the time of diagnosis of childhood cancers. The primary fertility preservation option that exists for prepubertal girls is ovarian tissue cryopreservation. Although ovarian tissue cryopreservation is still considered to be experimental in nature, live births have resulted from orthotopic tissue transplantation. Fertility preservation should be offered to all prepubertal girls at high-risk for premature ovarian failure as a result of gonadotoxic treatment. Ethical and legal questions surrounding these issues must be considered as more and more pediatric patients pursue fertility preservation.
Collapse
Affiliation(s)
- Nina Resetkova
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, 600 N. Wolfe St., Phipps 279, Baltimore, MD 21287, USA
| | - Masanori Hayashi
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, 1650 Orleans St., CRB1, Room 254, Baltimore, MD 21231, USA
| | - Lisa A Kolp
- Department of Gynecology and Obstetrics, Division of Reproductive Endocrinology, Johns Hopkins University School of Medicine, 10751 Falls Road, Suite #280, Lutherville, MD 20193, USA
| | - Mindy S Christianson
- Department of Gynecology and Obstetrics, Division of Reproductive Endocrinology, Johns Hopkins University School of Medicine, 10751 Falls Road, Suite #280, Lutherville, MD 20193, USA
| |
Collapse
|