1
|
Annibali O, Castellino A, Cenfra N, Ciccarone M, Rotondo F, Minoia C, Petrucci L, Gini G, Rusconi C, Bozzoli V, Nicolosi M, Margiotta G, Meli E, Cocito F, Bigliardi S, Ciavarella S, Tesei C, Zaja F. Fertility preservation in lymphoma patients treated with immunochemotherapy regimens with or without radiotherapy: results of a retrospective multicenter Italian study (Ferty care). Leuk Lymphoma 2024; 65:1448-1455. [PMID: 38847543 DOI: 10.1080/10428194.2024.2361358] [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: 02/19/2024] [Revised: 05/05/2024] [Accepted: 05/24/2024] [Indexed: 10/02/2024]
Abstract
This is a retrospective, multicentric study, aimed to describe the real-life application of fertility preservation methods during treatment in female lymphoma patients, aged 18-40 years old, diagnosed between Oct 1st/2010 and May 31st/2018. Among 414 women included, median age was 28 years old, histologies were: HL 74%, PMBCL 13%, DLBCL 10%, others 3%. First line treatments were: ABVD in 295 (71%), R-CHOP like in 102 (25%), higher intensity regimens in 17 (4%) cases. Fertility preservation strategies were: GnRHa in 315 (78%), Oral Contraceptive in 41 (10%), oocytes and ovarian tissue cryopreservation in 55 and 42 patients, respectively. After therapy, we observed a restored regular period in 293 (70%) and premature ovarian failure (POF) in 33 (8%), Furthermore we recorded 43 pregnancies, all spontaneous with 5 years median follow-up. Median age at diagnosis and number of lines of treatment correlate with higher rate of amenorrhea, risk of POF and menopause (p < 0.001).
Collapse
Affiliation(s)
- Ombretta Annibali
- Unit of Hematology, Stem cell Transplantation, Fondazione Policlinico Universitario Campus Bio Medico, Roma, Italy
| | | | - Natalia Cenfra
- Unit of Hematology, Santa Maria Goretti Hospital, Latina, Italy
| | - Mariavita Ciccarone
- Gemme Dormienti Onlus Association, Roma, Italy
- San Carlo di Nancy Hospital, Rome, Italy
| | | | - Carla Minoia
- IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Luigi Petrucci
- Policlinico Umberto I Hospital and University, Roma, Italy
| | - Guido Gini
- Ospedali Riuniti, Università Politecnica delle Marche Hospital and University, Ancona, Italy
| | - Chiara Rusconi
- Fondazione IRCCS Istituto nazionale dei tumori, Milano, Italy
| | | | - Maura Nicolosi
- Hospital and University Maggiore della Carità, Novara, Italy
| | | | - Erika Meli
- ASSt Grande Niguarda Hospital, Milano, Italy
| | | | - Sara Bigliardi
- UOSD di oncologia Area Sud sede di Sassuolo, AUSL Modena Osedale di Sassuolo, Modena, Italy
| | | | - Cristiano Tesei
- Department of Hematology, University of Rome Tor Vergata, Rome, Italy
| | | |
Collapse
|
2
|
Chitoran E, Rotaru V, Mitroiu MN, Durdu CE, Bohiltea RE, Ionescu SO, Gelal A, Cirimbei C, Alecu M, Simion L. Navigating Fertility Preservation Options in Gynecological Cancers: A Comprehensive Review. Cancers (Basel) 2024; 16:2214. [PMID: 38927920 PMCID: PMC11201795 DOI: 10.3390/cancers16122214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 06/08/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
(1) Background: Currently, an increasing number of women postpone pregnancy beyond the age of 35. Gynecological cancers affect a significant proportion of women of reproductive age, necessitating the development of fertility preservation methods to fulfill family planning. Consequently, providing treatment options that preserve fertility in women diagnosed with gynecological cancers has become a crucial component of care for survivors. (2) Methods: We conducted an extensive search of relevant scientific publications in PubMed and Embase databases and performed a narrative review, including high-quality peer-reviewed research on fertility after being treated for gynecologic cancers, reporting pregnancy rates, birth rates, and pregnancy outcomes in cancer survivors as well as therapeutic options which partially preserve fertility and methods for obtaining a pregnancy in survivors. (3) Discussion: The medicine practiced today is focused on both treating the neoplasm and preserving the quality of life of the patients, with fertility preservation being an important element of this quality. This leads to an improved quality of life, allowing these women to become mothers even in the seemingly adverse circumstances posed by such a pathology. However, although there are guidelines on female fertility preservation in the context of neoplasms, an analysis shows that physicians do not routinely consider it and do not discuss these options with their patients. (4) Conclusions: Advancements in medicine have led to a better understanding and management of gynecological neoplasms, resulting in increased survival rates. Once the battle against these neoplasms is won, the issue of preserving the quality of life for these women arises, with fertility preservation being an important aspect for women who have not yet fulfilled their family planning desires at the time of diagnosis. It is important for patients to be informed about the available options for fertility preservation and to be encouraged to make informed decisions in collaboration with their medical team. Standardized recommendations for onco-fertility into guidelines should be taken into consideration in the future.
Collapse
Affiliation(s)
- Elena Chitoran
- School of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (E.C.); (S.-O.I.)
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology “Al. Trestioreanu”, 022328 Bucharest, Romania
| | - Vlad Rotaru
- School of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (E.C.); (S.-O.I.)
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology “Al. Trestioreanu”, 022328 Bucharest, Romania
| | | | - Cristiana-Elena Durdu
- Obstetrics and Gynecology Department, “Filantropia” Clinical Hospital, 011132 Bucharest, Romania
| | - Roxana-Elena Bohiltea
- School of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (E.C.); (S.-O.I.)
- Obstetrics and Gynecology Department, “Filantropia” Clinical Hospital, 011132 Bucharest, Romania
| | - Sinziana-Octavia Ionescu
- School of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (E.C.); (S.-O.I.)
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology “Al. Trestioreanu”, 022328 Bucharest, Romania
| | - Aisa Gelal
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology “Al. Trestioreanu”, 022328 Bucharest, Romania
| | - Ciprian Cirimbei
- School of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (E.C.); (S.-O.I.)
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology “Al. Trestioreanu”, 022328 Bucharest, Romania
| | - Mihnea Alecu
- School of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (E.C.); (S.-O.I.)
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology “Al. Trestioreanu”, 022328 Bucharest, Romania
| | - Laurentiu Simion
- School of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (E.C.); (S.-O.I.)
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology “Al. Trestioreanu”, 022328 Bucharest, Romania
| |
Collapse
|
3
|
Han R, Song Z, Li H, Wang C, Zhang L, Yang X. Analysis of the benefit of gonadotropin-releasing hormone agonist treatment in premenopausal women undergoing hematopoietic cell transplantation. Sci Rep 2023; 13:14497. [PMID: 37666835 PMCID: PMC10477207 DOI: 10.1038/s41598-023-40778-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 08/16/2023] [Indexed: 09/06/2023] Open
Abstract
Gonadotropin-releasing hormone agonist (GnRHa) appears to exhibit ovarian protection during chemotherapy for malignant tumors. The purpose of this study was to analyze the benefits of GnRHa in premenopausal women undergoing hematopoietic cell transplantation (HSCT). Candidates for myeloablative chemotherapy HSCT requiring fertility preservation in the Gynecological Endocrinology Clinic of Peking University People's Hospital from December 2011 to December 2021 were retrospectively analyzed. Patients who chose to receive GnRHa treatment were given at least 2 courses of a 3.75-mg dose of a GnRHa before myeloablative chemotherapy, and patients who chose not to receive GnRHa treatment were included in the control group. All patients were monitored for menstruation return and menopause-related symptoms, and ovarian function tests [follicle-stimulating hormone (FSH), luteinizing hormone, and estradiol] were performed 6-12 months after HSCT. In addition, we assessed the vaginal bleeding of patients in the laminar air-flow room (LAFR). A total of 234 cases were included in this study: 77 cases in the treatment group and 157 cases in the control group. The incidence of vaginal bleeding in the LAFR in the treatment group was significantly lower than that in the control group (24.68% vs. 79.62%, P < 0.001). The menopausal symptoms of the patients in the treatment group were reduced after transplantation (46.75% vs. 19.75%, P < 0.001). There was no difference in visible follicles by follow-up ultrasound in the two groups after HSCT (16.88% vs. 13.38%, P = 0.474). The level of FSH at 6-12 months after transplantation was lower (98.00 mIU/ml vs. 117.53 mIU/ml, P = 0.001). The proportion of patients with FSH < 40 mIU/ml did not differ between the two groups. One patient in the treatment group recovered spontaneous menstruation, while none recovered spontaneous menstruation in the control group (1.30% vs. 0%, P = 0.329). The use of GnRHa may relieve menopause-related symptoms and reduce vaginal bleeding in the LAFR and breakthrough bleeding after transplantation. GnRHa treatment can reduce the level of FSH after myeloablative chemotherapy, but it cannot reduce the incidence of premature ovarian failure in women of reproductive age following myeloablative HSCT.
Collapse
Affiliation(s)
- Ruxue Han
- Department of Gynecology and Obstetrics, Peking University People's Hospital, Beijing, China
| | - Ziyi Song
- Department of Gynecology and Obstetrics, Peking University People's Hospital, Beijing, China
| | - Huiling Li
- Department of Gynecology and Obstetrics, Peking University People's Hospital, Beijing, China
| | - Chaohua Wang
- Department of Gynecology and Obstetrics, Peking University People's Hospital, Beijing, China.
| | - Leping Zhang
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
| | - Xin Yang
- Department of Gynecology and Obstetrics, Peking University People's Hospital, Beijing, China.
| |
Collapse
|
4
|
Tanase A, Ciocoiu M, Tănase G. Fertility options after chemotherapy in young premenopausal patients. ACTA ENDOCRINOLOGICA (BUCHAREST, ROMANIA : 2005) 2023; 19:398-402. [PMID: 38356982 PMCID: PMC10863951 DOI: 10.4183/aeb.2023.398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2024]
Abstract
Context Infertility affects more and more younger patients nowadays. Life-saving cancer treatments - chemotherapy and radiation therapy unfortunately damage oocytes and reduce the window of fertility in women. There were an estimated 14.1 million cases of cancer worldwide in 2012, of which 6.7 million were in women. Aim We are reviewing fertility options and treatments for women fighting oncological diseases. It is important to promote that there is a possibility for these patients to know all they can do in order to preserve their fertility and our government should help all these young patients.The trend of delayed childbearing has increased worldwide duet o increased life costs, difficulty of finding a suitable partner and carier options, while the number of women facing a cancer diagnosis is rising. Material and methods We describe a review regarding the impact of these treatments for the fertility of these patients. Results Show that informed patients can better cope with their diagnostic and have a better prognosis. Conclusions Improving survival rates and current data have created a new medical field oncofertility. The term was first introduced in 2015 and is being used all over clinics that treat young patients nowadays.
Collapse
Affiliation(s)
- A. Tanase
- Obstetrics and Gynecology, “Grigore T Popa” University of Medicine and Pharmacy Iasi
| | - M. Ciocoiu
- Pathophysiology, “Grigore T Popa” University of Medicine and Pharmacy Iasi
| | - G.V. Tănase
- Anesthesiology and Intensive Care Unit, Iasi, Romania
| |
Collapse
|
5
|
Jespersen F, Petersen SL, Andersen P, Sellebjerg F, Magyari M, Sørensen PS, Blinkenberg M. Autologous hematopoietic stem cell transplantation of patients with aggressive relapsing-remitting multiple sclerosis: Danish nation-wide experience. Mult Scler Relat Disord 2023; 76:104829. [PMID: 37364374 DOI: 10.1016/j.msard.2023.104829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/28/2023] [Accepted: 06/09/2023] [Indexed: 06/28/2023]
Abstract
BACKGROUND Autologous hematopoietic stem cell treatment (AHSCT) is considered an effective treatment option for patients with aggressive relapsing-remitting multiple sclerosis (RRMS). Still there are few randomized and controlled studies of AHSCT to shed light on the safety and efficacy of the treatment, and therefore experiences from single centers are important. AIM To describe the Danish experience with AHSCT regarding patient characteristics, safety, and efficacy. METHOD Nationwide retrospective single center study of patients with multiple sclerosis (MS) treated with AHSCT. RESULTS A total of 32 patients were treated with AHSCT from May 2011 to May 2021. Seven were treated with carmustine, etoposide, cytarabine arabinoside, and melphalan (BEAM) as well as antithymocyte globulin (ATG). Twenty-five patients were treated with cyclophosphamide (CY) and ATG. In the whole cohort, relapse-free survival (RFS) was 77% (95% CI: 64-94%), worsening-free survival (WFS) was 79% (95% CI: 66-96%), MRI event-free survival (MFS) was 93% (95% CI: 85-100%), and no evidence of disease (NEDA-3) was 69% (95% CI: 54-89%) at the end of year two post-AHSCT. We had no treatment related mortality and only few severe adverse events (AEs). CONCLUSION AHSCT of patients with aggressive RRMS was an effective and relatively safe treatment with few serious AEs and no mortality in Danish patients.
Collapse
Affiliation(s)
- Freja Jespersen
- Danish Multiple Sclerosis Center, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark.
| | - Søren Lykke Petersen
- Department of Hematology Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Pernille Andersen
- Blood bank, Department of Clinical Immunology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Finn Sellebjerg
- Danish Multiple Sclerosis Center, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Melinda Magyari
- Danish Multiple Sclerosis Center, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Per Soelberg Sørensen
- Danish Multiple Sclerosis Center, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Morten Blinkenberg
- Danish Multiple Sclerosis Center, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
6
|
Wang Z, An J, Wang C. Gonadotropin-releasing hormone agonist for the preservation of ovarian function in survivors of haematopoietic stem cell transplantation for haematological diseases. BMC Womens Health 2022; 22:436. [DOI: 10.1186/s12905-022-02039-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 10/31/2022] [Indexed: 11/09/2022] Open
Abstract
Abstract
Objective
Administration of GnRH agonist (GnRHa) prior to chemotherapy may decreases the risk of gonadal dysfunction in patients with tumors. However, relevant data in haematopoietic stem cell transplantation (HSCT) recipients has not yet been established. Hence, the present study was designed to evaluate the clinical efficacy of GnRHa cotreatment prior to myeloablative regimens on ovarian protection in female survivors of HSCT for haematological diseases.
Patients and methods
Eligible patients were divided into a GnRHa group and a control group. Medical records regarding age at HSCT; diagnosis/indication for HSCT; pre- and posttransplantation serum sex hormone levels; menstruation and perimenopausal symptoms after HSCT were collected and compared. The primary and secondary outcome was the incidence of premature ovarian insufficiency (POI) symptoms associated with hypoestrogenism.
Results
A total of 330 patients were enrolled in the study: 19 patients were lost to follow-up, and clinical information was obtained in 311 patients. There was no significant difference in the primary outcome of follow-up between the two groups (78.50% [84 of 107] for the GnRHa group versus 83.33% [170 of 204] for the control group). The adjusted relative risks (RR) and 95% confidence interval (CI) were 1.19 and 0.73–1.93 (P = 0.487). Among patients who received cotreatment with GnRHa, 62.62% (67 of 107) complained of perimenopausal symptoms, which was significantly lower than the 74.51% (152 of 204) in the control group (adjusted RR: 1.46, 95% CI: 1.04–2.06, P = 0.031).
Conclusion
GnRHa cotreatment may not decrease the POI rate in HSCT survivors. However, it may reduce perimenopausal symptoms in this population, suggesting a potential benefit of GnRHa in clinical practice and warrant further researches.
Collapse
|
7
|
Arthur DE, Soliman ME, Adeniji SE, Adedirin O, Peter F. QSAR AND MOLECULAR DOCKING STUDY OF GONADOTROPIN-RELEASING HORMONE RECEPTOR INHIBITORS. SCIENTIFIC AFRICAN 2022. [DOI: 10.1016/j.sciaf.2022.e01291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022] Open
|
8
|
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: 3.0] [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
|
9
|
Odeh OM, Awwad J, Khalife D, Ghunaim S. The use of GnRH analogs in preserving ovarian function during chemotherapy. MIDDLE EAST FERTILITY SOCIETY JOURNAL 2021. [DOI: 10.1186/s43043-021-00088-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
The literature has always been controversial on the use of gonadotropin-releasing hormone agonists in preserving fertility in women of childbearing age after chemotherapy; thereby, in this article, we will be discussing its use in preserving fertility.
Main body of abstract
When it comes to preserving fertility, it is crucial to consider all available options in this topic due to its very sensitive nature, thereby we have found that while a lot of trials favor the use of gonadotropin-releasing hormone agonists, the lack of proper follow-up and long-term trials renders its use highly debatable, and since the longest follow-up trial showed non-significant results, it also opens the floor for debate on whether this short-term benefit is worth adding another drug to the regimen or not.
Short conclusion
As described in this review, while the use of gonadotropin-releasing hormone agonists is beneficial in a lot of studies, the lack of long-term reports still makes its use debatable, thereby more trials should be done.
Collapse
|
10
|
Del Castillo LM, Buigues A, Rossi V, Soriano MJ, Martinez J, De Felici M, Lamsira HK, Di Rella F, Klinger FG, Pellicer A, Herraiz S. The cyto-protective effects of LH on ovarian reserve and female fertility during exposure to gonadotoxic alkylating agents in an adult mouse model. Hum Reprod 2021; 36:2514-2528. [PMID: 34333622 PMCID: PMC8373474 DOI: 10.1093/humrep/deab165] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 05/07/2021] [Indexed: 12/15/2022] Open
Abstract
STUDY QUESTION Does LH protect mouse oocytes and female fertility from alkylating chemotherapy? SUMMARY ANSWER LH treatment before and during chemotherapy prevents detrimental effects on follicles and reproductive lifespan. WHAT IS KNOWN ALREADY Chemotherapies can damage the ovary, resulting in premature ovarian failure and reduced fertility in cancer survivors. LH was recently suggested to protect prepubertal mouse follicles from chemotoxic effects of cisplatin treatment. STUDY DESIGN, SIZE, DURATION This experimental study investigated LH effects on primordial follicles exposed to chemotherapy. Seven-week-old CD-1 female mice were randomly allocated to four experimental groups: Control (n = 13), chemotherapy (ChT, n = 15), ChT+LH-1x (n = 15), and ChT+LH-5x (n = 8). To induce primary ovarian insufficiency (POI), animals in the ChT and ChT+LH groups were intraperitoneally injected with 120 mg/kg of cyclophosphamide and 12 mg/kg of busulfan, while control mice received vehicle. For LH treatment, the ChT+LH-1x and ChT+LH-5x animals received a 1 or 5 IU LH dose, respectively, before chemotherapy, then a second LH injection administered with chemotherapy 24 h later. Then, two animals/group were euthanized at 12 and 24 h to investigate the early ovarian response to LH, while remaining mice were housed for 30 days to evaluate short- and long-term reproductive outcomes. The effects of LH and chemotherapy on growing-stage follicles were analyzed in a parallel experiment. Seven-week-old NOD-SCID female mice were allocated to control (n = 5), ChT (n = 5), and ChT+LH-1x (n = 6) groups. Animals were treated as described above, but maintained for 7 days before reproductive assessment. PARTICIPANTS/MATERIALS, SETTING, METHODS In the first experiment, follicular damage (phosphorylated H2AX histone (γH2AX) staining and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) assay), apoptotic biomarkers (western blot), and DNA repair pathways (western blot and RT-qPCR) were assessed in ovaries collected at 12 and 24 h to determine early ovarian responses to LH. Thirty days after treatments, remaining mice were stimulated (10 IU of pregnant mare serum gonadotropin (PMSG) and 10 IU of hCG) and mated to collect ovaries, oocytes, and embryos. Histological analysis was performed on ovarian samples to investigate follicular populations and stromal status, and meiotic spindle and chromosome alignment was measured in oocytes by confocal microscopy. Long-term effects were monitored by assessing pregnancy rate and litter size during six consecutive breeding attempts. In the second experiment, mice were stimulated and mated 7 days after treatments and ovaries, oocytes, and embryos were collected. Follicular numbers, follicular protection (DNA damage and apoptosis by H2AX staining and TUNEL assay, respectively), and ovarian stroma were assessed. Oocyte quality was determined by confocal analysis. MAIN RESULTS AND THE ROLE OF CHANCE LH treatment was sufficient to preserve ovarian reserve and follicular development, avoid atresia, and restore ovulation and meiotic spindle configuration in mature oocytes exposed at the primordial stage. LH improved the cumulative pregnancy rate and litter size in six consecutive breeding rounds, confirming the potential of LH treatment to preserve fertility. This protective effect appeared to be mediated by an enhanced early DNA repair response, via homologous recombination, and generation of anti-apoptotic signals in the ovary a few hours after injury with chemotherapy. This response ameliorated the chemotherapy-induced increase in DNA-damaged oocytes and apoptotic granulosa cells. LH treatment also protected growing follicles from chemotherapy. LH reversed the chemotherapy-induced depletion of primordial and primary follicular subpopulations, reduced oocyte DNA damage and granulosa cell apoptosis, restored mature oocyte cohort size, and improved meiotic spindle properties. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION This was a preliminary study performed with mouse ovarian samples. Therefore, preclinical research with human samples is required for validation. WIDER IMPLICATIONS OF THE FINDINGS The current study tested if LH could protect the adult mouse ovarian reserve and reproductive lifespan from alkylating chemotherapy. These findings highlight the therapeutic potential of LH as a complementary non-surgical strategy for preserving fertility in female cancer patients. STUDY FUNDING/COMPETING INTEREST(S) This study was supported by grants from the Regional Valencian Ministry of Education (PROMETEO/2018/137), the Spanish Ministry of Science and Innovation (CP19/00141), and the Spanish Ministry of Education, Culture and Sports (FPU16/05264). The authors declare no conflict of interest.
Collapse
Affiliation(s)
- L M Del Castillo
- IVI Foundation—IIS La Fe, Reproductive Medicine Research Group, Valencia, Spain
- Department of Pediatrics, Obstetrics and Gynecology, School of Medicine, University of Valencia, Valencia, Spain
| | - A Buigues
- IVI Foundation—IIS La Fe, Reproductive Medicine Research Group, Valencia, Spain
| | - V Rossi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - M J Soriano
- IVI Foundation—IIS La Fe, Reproductive Medicine Research Group, Valencia, Spain
| | - J Martinez
- IVI Foundation—IIS La Fe, Reproductive Medicine Research Group, Valencia, Spain
- Department of Pediatrics, Obstetrics and Gynecology, School of Medicine, University of Valencia, Valencia, Spain
| | - M De Felici
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - H K Lamsira
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - F Di Rella
- Clinical and Experimental Senology, Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, Naples, Italy
| | - F G Klinger
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - A Pellicer
- IVI Foundation—IIS La Fe, Reproductive Medicine Research Group, Valencia, Spain
- IVI-RMA Rome, Rome, Italy
| | - S Herraiz
- Correspondence address. IVI Foundation—IIS La Fe, Reproductive Medicine Research Group, Av. Fernando Abril Martorell, 106-Torre A-Planta1, 46026 Valencia, Spain. Tel: +34-96-390-33-05; E-mail: https://orcid.org/0000-0003-0703-6922
| |
Collapse
|
11
|
Male and Female Fertility: Prevention and Monitoring Hodgkin' Lymphoma and Diffuse Large B-Cell Lymphoma Adult Survivors. A Systematic Review by the Fondazione Italiana Linfomi. Cancers (Basel) 2021; 13:cancers13122881. [PMID: 34207634 PMCID: PMC8228520 DOI: 10.3390/cancers13122881] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 06/02/2021] [Accepted: 06/05/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Adult patients with Hodgkin lymphoma (HL) and diffuse large B-cell lymphoma (DLBCL) have prolonged survival but face the risk of treatment-induced impaired fertility. This systematic review, conducted by Fondazione Italiana Linfomi (FIL) researchers, aims to evaluate the incidence of treatment-related infertility, fertility preservation options, fertility assessment measures, and the optimal interval between the end of treatment and conception. METHODS MEDLINE, the Cochrane Library, and EMBASE were systematically searched up to September 2020 for published cohort, case-control, and cross-sectional studies on fertility issues. RESULTS Forty-five eligible studies were identified. Gonadotoxicity was related to sex, type and dosage of treatment, and, in females, to age. After receiving alkylating-agent-containing regimens, less than 30% of males recovered spermatogenesis, and 45% of females ≥30 years in age retained regular menstrual cycles. Sperm cryopreservation was offered to the majority of patients; sperm utilization resulted in a 33-61% pregnancy rate. After ovarian tissue transplantation, the spontaneous pregnancy and live birth rates were 38% and 23%; after IVF, the live birth rate was 38.4%. No data could be extracted on the utilization rate of cryopreserved mature oocytes. The results of studies on GnRH analogs are controversial; therefore, their use should not be considered an alternative to established cryopreservation techniques. Sperm count, FSH, and inhibin-B levels were appropriate measures to investigate male fertility; serum AMH levels and antral follicle count were the most appropriate markers for ovarian reserve. No data could be found regarding the optimal interval between the end of treatment and conception. CONCLUSIONS The risk of infertility should be discussed with adult lymphoma patients at the time of diagnosis, and fertility preservation options should be proposed before first-line treatment with alkylating-agent-containing regimens.
Collapse
|
12
|
Dolmans MM, Taylor HS, Rodriguez-Wallberg KA, Blumenfeld Z, Lambertini M, von Wolff M, Donnez J. Utility of gonadotropin-releasing hormone agonists for fertility preservation in women receiving chemotherapy: pros and cons. Fertil Steril 2021; 114:725-738. [PMID: 33040981 DOI: 10.1016/j.fertnstert.2020.08.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 08/06/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Marie-Madeleine Dolmans
- Pôle de Gynécologie, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium; Gynecology Department, Cliniques Universitaires Saint Luc, Brussels, Belgium
| | - Hugh S Taylor
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut
| | - Kenny A Rodriguez-Wallberg
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Reproductive Medicine, Division of Gynecology and Reproduction, Karolinska University Hospital, Stockholm, Sweden
| | - Zeev Blumenfeld
- Department of Reproductive Endocrinology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Matteo Lambertini
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
| | | | - Jacques Donnez
- Société de Recherche pour l'Infertilité, Brussels, Belgium; Catholic University of Louvain, Brussels, Belgium.
| |
Collapse
|
13
|
Brancati S, Gozzo L, Longo L, Vitale DC, Russo G, Drago F. Fertility Preservation in Female Pediatric Patients With Cancer: A Clinical and Regulatory Issue. Front Oncol 2021; 11:641450. [PMID: 33796467 PMCID: PMC8008167 DOI: 10.3389/fonc.2021.641450] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 01/29/2021] [Indexed: 12/22/2022] Open
Abstract
Fertility preservation represents one important goal of cancer patients’ management due to the high impact on health and quality of life of survivors. The available preventive measures cannot be performed in all patients and are not feasible in all health-care facilities. Therefore, the pharmacological treatment with GnRHa has become a valuable non-invasive and well-tolerated alternative, especially in those who cannot access to cryopreservation options due to clinical and/or logistic issues. Supporting data demonstrate a significant advantage for the survivors who received GnRHa in the long-term maintenance of ovarian function and preservation of fertility. The prevention of the risk of ovarian failure with GnRHa is a typical off-label use, defined as the administration of a medicinal product not in accordance with the authorized product information. Italy has officially recognized the off-label use of GnRHa in adult women at risk of premature and permanent menopause following chemotherapy. However, fertility preservation still represents an unmet medical need in adolescents who cannot access to other treatment options.
Collapse
Affiliation(s)
- Serena Brancati
- Clinical Pharmacology Unit/Regional Pharmacovigilance Centre, University Hospital of Catania, Catania, Italy
| | - Lucia Gozzo
- Clinical Pharmacology Unit/Regional Pharmacovigilance Centre, University Hospital of Catania, Catania, Italy.,Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Laura Longo
- Clinical Pharmacology Unit/Regional Pharmacovigilance Centre, University Hospital of Catania, Catania, Italy
| | - Daniela Cristina Vitale
- Clinical Pharmacology Unit/Regional Pharmacovigilance Centre, University Hospital of Catania, Catania, Italy
| | - Giovanna Russo
- Pediatric Onco-Hematology, University Hospital of Catania, Catania, Italy
| | - Filippo Drago
- Clinical Pharmacology Unit/Regional Pharmacovigilance Centre, University Hospital of Catania, Catania, Italy.,Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.,Centre for Research and Consultancy in HTA and drug Regulatory Affairs (CERD), University of Catania, Catania, Italy
| |
Collapse
|
14
|
Silvestris E, Cormio G, Skrypets T, Dellino M, Paradiso AV, Guarini A, Minoia C. Novel aspects on gonadotoxicity and fertility preservation in lymphoproliferative neoplasms. Crit Rev Oncol Hematol 2020; 151:102981. [PMID: 32485429 DOI: 10.1016/j.critrevonc.2020.102981] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 04/21/2020] [Accepted: 05/07/2020] [Indexed: 12/22/2022] Open
Abstract
The topic of fertility preservation in patients with a lymphoproliferative disease offers new aspects of debate, due to the introduction of novel chemotherapeutic regimens and small molecules in the clinical landscape. Cancer related infertility is mostly dependent on gonadotoxic treatments and fertile female patients are today addressed to the oocyte cryopreservation or to ovarian cortex fragment cryopreservation. These methods present advantages and disadvantages, which will be discussed in the present review, together with the options for male patients. The recent discovery of functional ovarian stem cells (OCSs) in woman ovarian cortex, opens new avenues offering a innovative procedure for fertility preservation through as model of regenerative medicine. Here, we review the gonadotoxic potential of "classical" chemotherapeutic treatments as well as of "novel" targeted therapies actually employed for lymphoproliferative neoplasms in young patients and revisit both the today available and future chances to preserve and restore fertility after the cancer healing.
Collapse
Affiliation(s)
- Erica Silvestris
- Gynecologic Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II" 70124 Bari, Italy.
| | - Gennaro Cormio
- Gynecologic Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II" 70124 Bari, Italy; Department of Biomedical Sciences and Human Oncology, Unit of Obstetrics and Gynecology, University of Bari Aldo Moro, Bari, Italy.
| | - Tetiana Skrypets
- Haematology Unit, National Cancer Center, IRCCS Istituto Tumori "Giovanni Paolo II", viale O. Flacco 65, Bari, Italy; Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Italy.
| | - Miriam Dellino
- Gynecologic Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II" 70124 Bari, Italy.
| | - Angelo Virgilio Paradiso
- Institutional BioBank, Experimental Oncology and Biobank Management Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy.
| | - Attilio Guarini
- Haematology Unit, National Cancer Center, IRCCS Istituto Tumori "Giovanni Paolo II", viale O. Flacco 65, Bari, Italy.
| | - Carla Minoia
- Haematology Unit, National Cancer Center, IRCCS Istituto Tumori "Giovanni Paolo II", viale O. Flacco 65, Bari, Italy.
| |
Collapse
|
15
|
Salama M, Anazodo A, Woodruff TK. Preserving fertility in female patients with hematological malignancies: a multidisciplinary oncofertility approach. Ann Oncol 2019; 30:1760-1775. [PMID: 31418765 DOI: 10.1093/annonc/mdz284] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023] Open
Abstract
Oncofertility is a new interdisciplinary field at the intersection of oncology and reproductive medicine that expands fertility options for young cancer patients. The most common forms of hematological malignancies that occur in girls and young women and therefore necessitate oncofertility care are acute lymphocytic leukemia, acute myeloid leukemia, non-Hodgkin's lymphoma, and Hodgkin's lymphoma. Aggressive gonadotoxic anticancer regimens including alkylating chemotherapy and total body irradiation are used often in treating girls and young women with hematological malignancies. The risks of gonadotoxicity and subsequent iatrogenic premature ovarian insufficiency and fertility loss depend mainly on the type and stage of the disease, dose of anticancer therapy as well as the age of the patient at the beginning of treatment. To avoid or at least mitigate the devastating complications of anticancer therapy-induced gonadotoxicity, effective and comprehensive strategies that integrate different options for preserving and restoring fertility ranging from established to experimental strategies should be offered before, during, and after chemotherapy or radiotherapy. A multidisciplinary approach that involves strong coordination and collaboration between hemato-oncologists, gynecologists, reproductive biologists, research scientists, and patient navigators is essential to guarantee high standard of care.
Collapse
Affiliation(s)
- M Salama
- Department of Obstetrics and Gynecology, Feinberg School of Medicine-Northwestern University, Chicago, USA
| | - A Anazodo
- Kids Cancer Centre, Sydney Children's Hospital, Sydney, Australia; Nelune Cancer Centre, Prince of Wales Hospital, Sydney, Australia; School of Women's and Children's Health, University of New South Wales, Sydney, Australia
| | - T K Woodruff
- Department of Obstetrics and Gynecology, Feinberg School of Medicine-Northwestern University, Chicago, USA.
| |
Collapse
|
16
|
Ciccarone M, Hohaus S, Pulsoni A, Cavaceppi P, Franzò S, Fabbri R, Cudillo L, Battistini R, Di Rocco A, Annibali O, Cox MC, Provenzano I, Abruzzese E, Renzi D, Tesei C, Anticoli Borza P, Cuccaro A, Andriani A, D'Elia GM, Facchiano A, Marchetti P, Cantonetti M. Preliminary results of a counselling programme for fertility preservation in female cancer patients: The experience of the GEMME DORMIENTI network. Eur J Cancer Care (Engl) 2019; 29:e13174. [PMID: 31571303 DOI: 10.1111/ecc.13174] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 08/09/2019] [Accepted: 09/04/2019] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To describe a population of patients referred for fertility preservation (FP), how to efficiently provide FP care, and how FP care changed over time. METHODS This longitudinal observational study enrolled 281 female cancer patients referred between 2013 and 2016 to the non-profit organisation Gemme Dormienti ONLUS (GD) for FP care. All patients underwent the same battery of instrumental and laboratory diagnostic tests. GnRHa therapy was started at least seven days before CTh treatment. RESULTS From 2013 to 2016, we observed a progressive increase in the number of patients referred for FP care. Out of 251 eligible patients, 135 patients were treated with GnRHa only, and 72 patients underwent GnRHa therapy and cryopreservation. The median time from GD referral to oocyte and ovarian tissue cryopreservation was 11 and 5 days respectively. Tissue cryopreservation requests increased during our study period (from four cases in 2013 to 17 cases in 2016). During follow-up, 17β-estradiol and FSH levels were significantly increased (p < .0001), and AMH levels were significantly decreased (p < .0001). CONCLUSION The rapid increase in the number of patients who requested FP care and in the complexity of FP procedures overtime reflects the need to improve quality of life for cancer patients.
Collapse
Affiliation(s)
- Mariavita Ciccarone
- Department of Gynaecology, San Carlo di Nancy Hospital, Rome, Italy.,Gemme Dormienti Association ONLUS, Rome, Italy
| | - Stefan Hohaus
- Policlinico Gemelli Foundation, Catholic University of the Sacred Heart, Rome, Italy
| | - Alessandro Pulsoni
- Department of Cellular Biotechnology and Haematology, Sapienza University, Rome, Italy
| | | | | | - Raffaella Fabbri
- Gynaecology and Physiopathology of Human Reproduction Unit, Department of Medical and Surgical Sciences, S.Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Laura Cudillo
- Haematology and Stem Cell Transplant Unit, Tor Vergata University, Roma, Italy
| | - Roberta Battistini
- A.O. San Camillo Forlanini U.O.C. Ematologia e Trapianti CSE, Rome, Italy
| | - Alice Di Rocco
- Department of Cellular Biotechnology and Haematology, Sapienza University, Rome, Italy
| | - Ombretta Annibali
- UOC Haematology Stem Cell Transplantation, University Campus Bio Medico, Rome, Italy
| | - Maria Christina Cox
- Haematology Unit, Azienda Ospedaliera-Universitaria Sant'Andrea, Rome, Italy
| | - Ida Provenzano
- UOC Oncoematologia Fondazione Policlinico Tor Vergata, Rome, Italy
| | | | - Daniela Renzi
- Haematology and Stem Cell Transplant Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | | | | | - Annarosa Cuccaro
- Policlinico Gemelli Foundation, Catholic University of the Sacred Heart, Rome, Italy
| | - Alessandro Andriani
- UOSD di Ematologia, Rome, Italy.,UOC di Ematologia, Osp. F. Spaziani, ASL Frosinone, Rome, Italy
| | - Gianna Maria D'Elia
- Department of Cellular Biotechnology and Haematology, Sapienza University, Rome, Italy
| | | | - Paolo Marchetti
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University, Rome, Italy
| | - Maria Cantonetti
- UOC Oncoematologia Fondazione Policlinico Tor Vergata, Rome, Italy
| |
Collapse
|
17
|
Blumenfeld Z. Fertility Preservation Using GnRH Agonists: Rationale, Possible Mechanisms, and Explanation of Controversy. CLINICAL MEDICINE INSIGHTS. REPRODUCTIVE HEALTH 2019; 13:1179558119870163. [PMID: 31488958 PMCID: PMC6710670 DOI: 10.1177/1179558119870163] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 07/18/2019] [Indexed: 12/15/2022]
Abstract
The only clinically accepted method of fertility preservation in young women facing gonadotoxic chemo- and/or radiotherapy for malignant or autoimmune diseases is cryopreservation of embryos or unfertilized ova, whereas cryopreservation of ovarian tissue for future reimplantation, or in vitro maturation of follicles, and the use of gonadotropin-releasing hormone agonists (GnRHa) are still considered investigational, by several authorities. Whereas previous publications have raised the fear of GnRHa's possible detrimental effects in patients with hormone receptor-positive breast cancers, recent randomized controlled trials (RCTs) have shown that it either improves or does not affect disease-free survival (DFS) in such patients. This review summarizes the pros and cons of GnRHa co-treatment for fertility preservation, suggesting 5 theoretical mechanisms for GnRHa action: (1) simulating the prepubertal hypogonadotropic milieu, (2) direct effect on GnRH receptors, (3) decreased ovarian perfusion, (4) upregulation of an ovarian-protecting molecule such as sphingosine-1-phosphate, and (5) protecting a possible germinative stem cell. We try to explain the reasons for the discrepancy between most publications that support the use of GnRHa for fertility preservation and the minority of publications that did not support its efficiency.
Collapse
Affiliation(s)
- Zeev Blumenfeld
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
18
|
Salama M, Anazodo A, Woodruff TK. Preserving fertility in female patients with hematological malignancies: the key points. Expert Rev Hematol 2019; 12:375-377. [PMID: 31058548 PMCID: PMC6666388 DOI: 10.1080/17474086.2019.1613150] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 04/26/2019] [Indexed: 02/07/2023]
Affiliation(s)
- Mahmoud Salama
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Antoinette Anazodo
- Kids Cancer Centre, Sydney Children’s Hospital, Sydney, Australia
- Nelune Cancer Centre, Prince of Wales Hospital, Sydney, Australia
- School of Women’s and Children’s Health, University of New South Wales, Sydney, Australia
| | - Teresa K. Woodruff
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
19
|
Abdel-Razeq H. Gonadotropin-releasing hormone agonists during chemotherapy for ovarian function and fertility preservation for patients with early-stage breast cancer. Cancer Manag Res 2019; 11:4273-4282. [PMID: 31190993 PMCID: PMC6514123 DOI: 10.2147/cmar.s204069] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 03/27/2019] [Indexed: 01/09/2023] Open
Abstract
Breast cancer remains the most common cancer among women worldwide. Many patients, especially in our region, are affected while young and during their child-bearing years. Chemotherapy, more commonly used in this age group, may result in premature ovarian failure and thus negatively impact their fertility. Several fertility-preservation methods are currently in use in this age group. Unfertilized ova cryopreservation and in vitro fertilization plus embryo cryopreservation are widely used in clinical practice. More recently, ovarian tissue cryopreservation is gaining in popularity. Several clinical trials and meta-analyses have shown that premenopausal women who received ovarian function suppression with gonadotropin-releasing hormone agonists while on chemotherapy were less likely to experience ovarian failure and had higher rates of menses resumption compared to those who did not. Some studies have also shown higher rates of successful pregnancies among treated patients. Given the conflicting results of the reported clinical trials and even the many published meta-analyses, this approach continues to be controversial and should only be used when other established fertility preservation methods cannot be utilized. The current review seeks to provide an updated summary on this controversial topic by reviewing all recently published clinical trials and meta-analyses.
Collapse
Affiliation(s)
- Hikmat Abdel-Razeq
- Department of Internal Medicine, Section of Hematology and Medical Oncology, King Hussein Cancer Center and School of Medicine, University of Jordan, Amman, Jordan
| |
Collapse
|
20
|
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: 3.8] [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
|
21
|
Pascuali N, Scotti L, Di Pietro M, Oubiña G, Bas D, May M, Gómez Muñoz A, Cuasnicú PS, Cohen DJ, Tesone M, Abramovich D, Parborell F. Ceramide-1-phosphate has protective properties against cyclophosphamide-induced ovarian damage in a mice model of premature ovarian failure. Hum Reprod 2019. [PMID: 29534229 DOI: 10.1093/humrep/dey045] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
STUDY QUESTION Is ceramide-1-phosphate (C1P) an ovarian protective agent during alkylating chemotherapy? SUMMARY ANSWER Local administration of C1P drastically reduces ovarian damage induced by cyclophosphamide (Cy) via protection of follicular reserve, restoration of hormone levels, inhibition of apoptosis and improvement of stromal vasculature, while protecting fertility, oocyte quality and uterine morphology. WHAT IS KNOWN ALREADY Cancer-directed therapies cause accelerated loss of ovarian reserve and lead to premature ovarian failure (POF). Previous studies have demonstrated that C1P regulates different cellular processes including cell proliferation, cell migration, angiogenesis and apoptosis. This sphingolipid may be capable of modulating vascular development and apoptosis in ovaries affected by chemotherapy. STUDY DESIGN, SIZE, DURATION The 6-8-week-old mice were weighed and administered either a single intraperitoneal injection of Cy (75 mg/kg) or an equal volume of saline solution only for control mice. Control and Cy mice underwent sham surgery and received an intrabursal injection of saline solution, while Cy + C1P animal groups received 5 μl C1P, either 0.5 or 1 mM, under the bursa of both ovaries 1 h prior to Cy administration. PARTICIPANTS/MATERIALS, SETTING, METHODS Animals were euthanized by cervical dislocation or cardiac puncture 2 weeks after surgery for collection of blood orovary and uterus samples, which were cleaned of adhering tissue in culture medium and used for subsequent assays. Ovaries were used for Western blotting or immunohistochemical and/or histological analyses or steroid extraction, as required (n = 5-8 per group). A set of mice (n = 3/group) was destined for oocyte recovery and IVF. Finally, another set (n = 5-6/group) was separated to study fertility parameters. MAIN RESULTS AND THE ROLE OF CHANCE The number of primordial (P < 0.01), primary (P < 0.05) and preantral follicles (P < 0.05) were decreased in Cy-treated mice compared to control animals, while atretic follicles were increased (P < 0.001). In Cy + C1P mice, the ovaries recovered control numbers of these follicular structures, in both C1P doses studied. Cy affected AMH expression, while it was at least partially recovered when C1P is administered as well. Cy caused an increase in serum FSH concentration (P < 0.01), which was prevented by C1P coadministration (P < 0.01). E2 levels in Cy-treated ovaries decreased significantly compared to control ovaries (P < 0.01), whilst C1P restored E2 levels to those of control ovaries (P < 0.01). Cy increased the expression of BAX (P < 0.01) and decreased the expression of BCLX-L compared to control ovaries (P < 0.01). The ovarian BCLX-L:BAX ratio was also lower in Cy-treated mice (P < 0.05). In the Cy + C1P group, the expression levels of BAX, BCLX-L and BCLX-L:BAX ratio were no different than those in control ovaries. In addition, acid sphingomyelinase (A-SMase) expression was higher in Cy-treated ovaries, whilst remaining similar to the control in the Cy + C1P group. Cy increased the apoptotic index (TUNEL-positive follicles/total follicles) in preantral and early antral stages, compared to control ovaries (P < 0.001 and P < 0.01, respectively). C1P protected follicles from this increase. No primordial or primary follicular cells stained for either cleaved caspase-3 or TUNEL when exposed to Cy, therefore, we have found no evidence for follicular reserve depletion in response to Cy being due to apoptosis. Cy caused evident vascular injury, especially in large cortical stromal vessels, and some neovascularization. In the Cy + C1P group, the disruptions in vascular wall continuity were less evident and the number of healthy stromal blood vessels seemed to be restored. In Cy-treated ovaries α-SMA-positive cells showed a less uniform distribution around blood vessels. C1P coadministration partially prevented this Cy-induced effect, with a higher presence of α-SMA-positive cells surrounding vessels. By H&E staining, Cy-treated mice showed endometrial alterations compared to controls, affecting both epithelial and stromal compartments. However, C1P allowed that the stromal tissue to maintain its loose quality and its glandular branches. Cy-treated animals had significantly lower pregnancy rates and smaller litter sizes compared with control mice (P = 0.013 and P < 0.05, respectively), whereas cotreatment with C1P preserved normal fertility. Furthermore, a higher (P < 0.05) proportion of abnormal oocytes was recovered from Cy-treated mice compared to the control, which was prevented by C1P administration. LARGE SCALE DATA N/A. LIMITATIONS REASONS FOR CAUTION The results of this study were generated from an in-vivo animal experimental model, already used by several authors. Further studies on C1P functions in female reproduction in pathological conditions such as chemotherapy-induced ovarian failure and on the safety of use of this sphingolipid are required. WIDER IMPLICATIONS OF THE FINDINGS The present findings showed that C1P administration prior to Cy might be a promising fertility preservation strategy in female patients who undergo chemotherapy. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by grants from ANPCyT (PICT 2015-1117), CONICET (PIP 380), Cancer National Institute (INC) and Roemmers Foundation, Argentina. The authors declare no conflicts of interest.
Collapse
Affiliation(s)
- Natalia Pascuali
- IInstituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Leopoldina Scotti
- IInstituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Mariana Di Pietro
- IInstituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Gonzalo Oubiña
- IInstituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Diana Bas
- IInstituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - María May
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Antonio Gómez Muñoz
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - Patricia S Cuasnicú
- IInstituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Débora J Cohen
- IInstituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Marta Tesone
- IInstituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Dalhia Abramovich
- IInstituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Fernanda Parborell
- IInstituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina
| |
Collapse
|
22
|
Smith KL, Gracia C, Sokalska A, Moore H. Advances in Fertility Preservation for Young Women With Cancer. Am Soc Clin Oncol Educ Book 2018; 38:27-37. [PMID: 30231357 DOI: 10.1200/edbk_208301] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Female patients of reproductive age with cancer often require treatment that can compromise their future fertility. Treatment-related infertility is an important cancer survivorship issue and is associated with depression and diminished quality of life. Recent advances in reproductive health care provide the opportunity to preserve fertility prior to the initiation of cancer therapy. Clinical guidelines recommend that oncology providers counsel patients about the risk of treatment-related infertility and fertility preservation options, and that they refer those who are interested in fertility preservation to fertility specialists. Guidelines endorse the use of assisted reproductive techniques (ART) provided by reproductive endocrinologists to preserve fertility in young female patients with cancer. In addition, ovarian suppression with gonadotropin-releasing hormone (GnRH) agonists may be considered for ovarian protection during chemotherapy. This article reviews currently available and emerging ART for fertility preservation in female patients of reproductive age with cancer and current data supporting the use of ovarian suppression for ovarian protection during chemotherapy in this population. We also review the uptake of fertility services and discuss barriers to fertility preservation in female patients of reproductive age with cancer.
Collapse
Affiliation(s)
- Karen Lisa Smith
- From The Johns Hopkins University School of Medicine, Baltimore, MD; Hospital of the University of Pennsylvania, Philadelphia, PA; Cleveland Clinic, Cleveland, OH
| | - Clarisa Gracia
- From The Johns Hopkins University School of Medicine, Baltimore, MD; Hospital of the University of Pennsylvania, Philadelphia, PA; Cleveland Clinic, Cleveland, OH
| | - Anna Sokalska
- From The Johns Hopkins University School of Medicine, Baltimore, MD; Hospital of the University of Pennsylvania, Philadelphia, PA; Cleveland Clinic, Cleveland, OH
| | - Halle Moore
- From The Johns Hopkins University School of Medicine, Baltimore, MD; Hospital of the University of Pennsylvania, Philadelphia, PA; Cleveland Clinic, Cleveland, OH
| |
Collapse
|
23
|
Sofiyeva N, Siepmann T, Barlinn K, Seli E, Ata B. Gonadotropin-Releasing Hormone Analogs for Gonadal Protection During Gonadotoxic Chemotherapy: A Systematic Review and Meta-Analysis. Reprod Sci 2018; 26:939-953. [PMID: 30270741 DOI: 10.1177/1933719118799203] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE A systematic review and meta-analysis was conducted to investigate whether gonadotropin-releasing hormone analogs (GnRHa) have a protective role in women treated with alkylating agents. DATA SOURCES Major databases (PubMED, EMBASE, Cochrane Central Register of Controlled Trials), systematic snowballing, and trial registries were screened from the inception dates until September 2017. METHODS AND STUDY SELECTION Comparative studies involving reproductive-aged women undergoing chemotherapy with or without coadministration of GnRHa were included. Spontaneous menstrual resumption was assessed as a main outcome. Statistical analyses were performed with STATA 14.2 statistical software. Effect estimates were presented as risk ratios (RR) with 95% confidence intervals (CIs). RESULTS The literature search yielded 25 436 citations and 84 papers were assessed in full text. Eighteen studies (11 randomized controlled trials [RCTs] and 7 cohort studies) published between 1987 and 2015 were included in the analysis, revealing a significant protective effect of GnRHa (n = 1043; RR:1.38; 95% CI: 1.18-1.63) although with high heterogeneity (I2 = 83.3%). Subgroup analyses revealed a significant benefit of GnRHa cotreatment both in RCTs and in cohort studies. Statistical significance was found in all subgroups by the underlying disease, that is, hematological malignancies, autoimmune diseases, and breast cancer. Sensitivity analyses in GnRH agonist-treated patients, in patients younger than 40 years old, and in patients without supradiaphragmatic radiotherapy also revealed a significant benefit of GnRHa cotreatment. CONCLUSION Our results indicate that concurrent GnRHa administration is an effective method to decrease gonadotoxicity of alkylating agents. The presence of low-quality evidence favoring gonadoprotective effect requires a strong recommendation for offering GnRHa coadministration to young women who are to undergo gonadotoxic chemotherapy. CAPSULE The present systematic review and meta-analysis shows a significant gonadoprotective effect of gonadotropin-releasing hormone analogs in women treated with alkylating agents.
Collapse
Affiliation(s)
- Nigar Sofiyeva
- 1 Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University, School of Medicine, New Haven, CT, USA.,2 Division of Health Care Sciences, Center for Clinical Research and Management Education, Dresden International University, Dresden, Germany
| | - Timo Siepmann
- 2 Division of Health Care Sciences, Center for Clinical Research and Management Education, Dresden International University, Dresden, Germany.,3 Department of Neurology, University Hospital Carl Gustav Carus, Technische Universitaet Dresden, Dresden, Germany
| | - Kristian Barlinn
- 3 Department of Neurology, University Hospital Carl Gustav Carus, Technische Universitaet Dresden, Dresden, Germany
| | - Emre Seli
- 1 Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University, School of Medicine, New Haven, CT, USA
| | - Baris Ata
- 1 Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University, School of Medicine, New Haven, CT, USA.,4 Department of Obstetrics and Gynecology, Koc University, School of Medicine, Istanbul, Turkey
| |
Collapse
|
24
|
ACOG Committee Opinion No. 747: Gynecologic Issues in Children and Adolescent Cancer Patients and Survivors. Obstet Gynecol 2018; 132:e67-e77. [DOI: 10.1097/aog.0000000000002763] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
25
|
Lee DY, Park YH, Lee JE, Choi D. Prediction of ovarian function recovery in young breast cancer patients after protection with gonadotropin-releasing hormone agonist during chemotherapy. Breast Cancer Res Treat 2018; 171:649-656. [PMID: 29943121 DOI: 10.1007/s10549-018-4863-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 06/22/2018] [Indexed: 11/24/2022]
Abstract
PURPOSE This study evaluated predictive factors for recovery of ovarian function after ovarian protection by GnRH agonist during chemotherapy in young breast cancer patients. METHODS This prospective cohort study analyzed 105 young breast cancer patients who were studied longitudinally after receiving GnRH agonist during cyclophosphamide-based chemotherapy for ovarian protection. Associations between pretreatment hormones, clinical factors, and recovery of ovarian function (resumption of menstruation or anti-Müllerian hormone (AMH) ≥ 1 ng/ml) were evaluated at 12 months and long-term follow-up after completion of chemotherapy. RESULTS Mean age was 32 years (range 23-42 years). In multivariate analyses, tamoxifen use (P = 0.035) and pretreatment follicle-stimulating hormone (FSH) (P = 0.032) were predictive of resumption of menstruation, and age (P = 0.019), tamoxifen use (P = 0.022), pretreatment FSH (P < 0.001), and AMH (P = 0.040) were predictors for AMH ≥ 1 ng/ml at 12 months. In addition, pretreatment AMH was a predictor for AMH ≥ 1 ng/ml after long-term follow-up. Receiver operating characteristic curve analyses gave area under the curve of 0.805 for resumption of menstruation and 0.903 for serum AMH concentration ≥ 1 ng/ml at 12 months, when age, tamoxifen use, pretreatment FSH, and AMH were combined. CONCLUSION Pretreatment AMH (3.26 ng/ml), age (33.9 years), pretreatment FSH (5.5 IU/l), and tamoxifen use are useful predictors for AMH ≥ 1 ng/ml at 12 months after GnRH agonist. This finding will support patient and clinician decision-making regarding fertility preservation.
Collapse
Affiliation(s)
- Dong-Yun Lee
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Yeon Hee Park
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jeong Eon Lee
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - DooSeok Choi
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea.
| |
Collapse
|
26
|
Abstract
BACKGROUND Triptorelin, a gonadotropin releasing hormone analogue, can be administered to postpubertal female individuals with cancer who receive chemotherapy to obtain menstrual suppression and decrease the risk of hemorrhage caused by thrombocytopenia. Our goal was to assess whether triptorelin also has a protective role against the gonadotoxicity of chemotherapy. PATIENTS AND METHODS This retrospective observational study includes all postmenarchal female patients who presented to our Unit from 2000 to 2015 and received chemotherapy for cancer. They were administered depot triptorelin. We evaluated long-term ovarian function in order to detect clinical signs of ovarian damage, miscarriages, and pregnancies. Laboratory follow-up consisted in dosing serum follicle stimulating hormone, luteinizing hormone, prolactin, estradiol, and progesterone. Ultrasound of the ovaries was performed as well. RESULTS Of 36 evaluable patients, 9 received hematopoietic stem cell transplantation (HSCT). The remaining 27 patients maintained normal ovarian function at clinical, laboratory, and ultrasound assessment. Five of them achieved spontaneous physiological pregnancy. Four of the 9 patients who underwent HSCT developed premature ovarian failure. CONCLUSION Our study suggests that gonadotropin releasing hormone-a administered during chemotherapy can prevent premature ovarian failure in patients treated without HSCT and that it is not enough to preserve the ovarian function during HSCT. Hence, a prospective randomized trial with a larger population would be recommended.
Collapse
|
27
|
A Case of Primary Uterina Lymphoma Presenting with Bleeding, Pelvic Pain, and Dysmenorrhea. Case Rep Obstet Gynecol 2018; 2018:5065738. [PMID: 29607236 PMCID: PMC5828537 DOI: 10.1155/2018/5065738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 10/26/2017] [Indexed: 11/19/2022] Open
Abstract
Primary non-Hodgkin's lymphoma (NHL) can arise from lymphatic cells located in solid organs (extranodal) and it accounts for 25 to 35% of all NHL. Primary lymphoma on the female genital tract (PLFGT) is a rare disease, comprising 0.2 to 1.1% of all extranodal lymphomas in the female population. In this paper, the authors report an extremely rare case of a 48-year-old woman who exhibited an abnormal uterine bleeding, pelvic pain, and dysmenorrhea history. The transvaginal ultrasound showed an anteverted uterus measuring 153 cm3 in volume, with intramural leiomyomas. She underwent a total laparoscopic hysterectomy with bilateral salpingectomy. The histologic evaluation of the specimen showed a follicular lymphoma with diffuse pattern in the endometrium. This report illustrates the difficulty in the diagnosis of primary lymphomas of the female genital tract.
Collapse
|
28
|
Abstract
Gonadotropin-releasing hormone (GnRH) is recognized as the central regulator of the functions of the pituitary-gonadal axis. The increasing knowledge on the mechanisms controlling the development and the function of GnRH-producing neurons is leading to a better diagnostic and therapeutic approach for hypogonadotropic hypogonadisms and for alterations of the puberty onset. During female life span, the function of the GnRH pulse generator may be affected by a number of inputs from other neuronal systems, offering alternative strategies for diagnostic and therapeutic interventions. Moreover, the identification of a GnRH/GnRH receptor system in both human ovary and endometrium has widened the spectrum of action of the peptide outside its hypothalamic functions. The pharmacological use of GnRH itself or its synthetic analogs (agonists and antagonists) provides a valid tool to either stimulate or block gonadotropin secretion and to modulate the female fertility in several reproductive disorders and in assisted reproduction technology. The use of GnRH agonists in young female patients undergoing chemotherapy is also considered a promising therapeutic approach to counteract iatrogenic ovarian failure.
Collapse
|
29
|
Affiliation(s)
- Zeev Blumenfeld
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
30
|
Sibson KR, Biss TT, Furness CL, Grainger JD, Hough RE, Macartney C, Payne JH, Chalmers EA. BSH Guideline: management of thrombotic and haemostatic issues in paediatric malignancy. Br J Haematol 2018; 180:511-525. [DOI: 10.1111/bjh.15112] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 10/28/2017] [Accepted: 11/07/2017] [Indexed: 01/19/2023]
Affiliation(s)
- Keith R. Sibson
- Department of Haematology; Great Ormond Street Hospital; London UK
| | - Tina T. Biss
- The Newcastle Upon Tyne Hospitals NHS Foundation Trust; Newcastle Upon Tyne UK
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW Fertility preservation has recently gained a worldwide interest among fertility specialists, oncologists, and all healthcare providers. Thus, the protection against iatrogenic infertility caused by chemotherapy assumes a high priority. Specifically, the issue of endocrine prevention of gonadotoxicity is still a controversial subject. RECENT FINDINGS During the last year, many publications on this subject appeared, swinging the pendulum toward the beneficial role of gonadotropin-releasing hormone agonist (GnRHa) cotreatment for fertility preservation despite gonadotoxic chemotherapy. Here, we summarize the recent publications on fertility preservation and minimizing chemotherapy-induced gonadotoxicity. SUMMARY More than 10 recent meta-analyses have concluded that GnRHa is beneficial and may decrease the risk of premature ovarian failure and increase the pregnancy rate in survivors. Furthermore, two recent international meetings of experts have concluded that GnRHa is effective and should be offered to every young woman facing gonadotoxic chemotherapy.
Collapse
|
32
|
Kim H, Kim SK, Lee JR, Hwang KJ, Suh CS, Kim SH. Fertility preservation for patients with breast cancer: The Korean Society for Fertility Preservation clinical guidelines. Clin Exp Reprod Med 2017; 44:181-186. [PMID: 29376014 PMCID: PMC5783914 DOI: 10.5653/cerm.2017.44.4.181] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 03/20/2017] [Accepted: 07/05/2017] [Indexed: 01/25/2023] Open
Abstract
With advances in the methods of cancer treatment used in modern medicine, the number of breast cancer survivors has been consistently rising. As the number of women who wish to become pregnant after being diagnosed with breast cancer increases, it is necessary to consider fertility preservation in these patients. However, medical doctors may be unaware of the importance of fertility preservation among cancer patients because most patients do not share their concerns about fertility with their doctors. Considering the time spent choosing and undergoing treatment, an early referral to a reproductive specialist is the best way to prevent a delay in cancer treatment. Since it is not easy to make decisions on matters related to cancer diagnosis and fertility, patients should be provided with enough time for decision-making, and to allow for this, an early referral will provide patients with sufficient time to choose an appropriate method of fertility preservation. The currently available options of fertility preservation for patients with breast cancer include cryopreservation of embryos, oocytes, and ovarian tissue and gonadotropin-releasing hormone agonist treatment before and during chemotherapy. An appropriate method of fertility preservation must be selected through consultations between individual patients and health professionals and analyses of the pros and cons of different options.
Collapse
Affiliation(s)
- Hoon Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea.,Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, Korea
| | - Seul Ki Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea.,Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jung Ryeol Lee
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea.,Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Kyung Joo Hwang
- Department of Obstetrics and Gynecology, Ajou University School of Medicine, Suwon, Korea
| | - Chang Suk Suh
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea.,Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, Korea
| | - Seok Hyun Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea.,Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
33
|
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: 0.7] [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
|
34
|
Pankiewicz K, Szewczyk G, Maciejewski TM, Szukiewicz D. Strategies for overcoming oncological treatment-related ovarian dysfunction - literature review. Gynecol Endocrinol 2017; 33:830-835. [PMID: 28604126 DOI: 10.1080/09513590.2017.1337095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
In the majority of developed countries, it is observed that the time for maternity is being postponed to over the 30th and even 40th year of life. A significant number of cancers are diagnosed during reproductive age, often before the first pregnancy. A fertility preservation is an important issue in oncological treatment, where there is a need for balancing between radicality and the preservation of function of reproductive organs. The authors discuss the problem of ovarian dysfunction after oncological treatment and present the possible strategies for saving the reproductive function of ovaries, including both invasive and pharmacological approaches.
Collapse
Affiliation(s)
- Katarzyna Pankiewicz
- a Department of Obstetrics and Gynecology , Institute of Mother and Child , Warsaw , Poland
| | - Grzegorz Szewczyk
- a Department of Obstetrics and Gynecology , Institute of Mother and Child , Warsaw , Poland
- b Department of General and Experimental Pathology , Warsaw Medical University , Warsaw , Poland
| | | | - Dariusz Szukiewicz
- b Department of General and Experimental Pathology , Warsaw Medical University , Warsaw , Poland
| |
Collapse
|
35
|
Salama M, Isachenko V, Isachenko E, Rahimi G, Mallmann P. Advances in fertility preservation of female patients with hematological malignancies. Expert Rev Hematol 2017; 10:951-960. [PMID: 28828900 DOI: 10.1080/17474086.2017.1371009] [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] [Indexed: 10/19/2022]
Abstract
INTRODUCTION The most common forms of hematological malignancies that occur in female reproductive years are lymphoma and leukemia. Areas covered: Several aggressive gonadotoxic regimens such as alkylating chemotherapy and total body irradiation are used frequently in treatment of lymphoma and leukemia leading to subsequent iatrogenic premature ovarian failure and fertility loss. In such cases, female fertility preservation options should be offered in advance. Expert commentary: In order to preserve fertility of young women and girls with lymphoma and leukemia, several established, experimental, and debatable options can be offered before starting chemotherapy and radiotherapy. However, each of those female fertility preservation options has both advantages and disadvantages and may not be suitable for all patients. That is why a fertility preservation strategy should be individualized and tailored distinctively for each patient in order to be effective. Artificial human ovary is a novel experimental in vitro technology to produce mature oocytes that could be the safest option to preserve and restore fertility of young women and girls with hematological malignancies especially when other fertility preservation options are not feasible or contraindicated. Further research and studies are needed to improve the results of artificial human ovary and establish it in clinical practice.
Collapse
Affiliation(s)
- Mahmoud Salama
- a Department of Gynecology and Obstetrics, Medical Faculty , University of Cologne , Cologne , Germany
| | - Vladimir Isachenko
- a Department of Gynecology and Obstetrics, Medical Faculty , University of Cologne , Cologne , Germany
| | - Evgenia Isachenko
- a Department of Gynecology and Obstetrics, Medical Faculty , University of Cologne , Cologne , Germany
| | - Gohar Rahimi
- a Department of Gynecology and Obstetrics, Medical Faculty , University of Cologne , Cologne , Germany
| | - Peter Mallmann
- a Department of Gynecology and Obstetrics, Medical Faculty , University of Cologne , Cologne , Germany
| |
Collapse
|
36
|
Fertility Preservation for Pediatric Patients: Current State and Future Possibilities. J Urol 2017; 198:186-194. [DOI: 10.1016/j.juro.2016.09.159] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2017] [Indexed: 11/19/2022]
|
37
|
Blumenfeld Z. Investigational and experimental GnRH analogs and associated neurotransmitters. Expert Opin Investig Drugs 2017; 26:661-667. [DOI: 10.1080/13543784.2017.1323869] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Zeev Blumenfeld
- Reproductive Endocrinology, Ob/Gyn, Rappaport Faculty of Medicine, Technion – Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
38
|
Nasioudis D, Kampaktsis PN, Frey M, Witkin SS, Holcomb K. Primary lymphoma of the female genital tract: An analysis of 697 cases. Gynecol Oncol 2017; 145:305-309. [PMID: 28284518 DOI: 10.1016/j.ygyno.2017.02.043] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 02/27/2017] [Accepted: 02/28/2017] [Indexed: 01/28/2023]
Abstract
OBJECTIVE Primary lymphoma of the female genital tract (PLFGT) is a rare entity. The aim of this population-based study was to elucidate the clinico-pathological, demographic characteristics and survival of women with PLFGT. MATERIALS AND METHODS The Surveillance, Epidemiology, and End Results database was accessed and cases of PLFGT diagnosed between 1988 and 2012 were identified. Five-year overall (OS) and cancer-specific (CSS) survival rates were calculated with the Kaplan-Meier method. The influence of demographic and clinical parameters on survival was examined with the log-rank test. Factors independently associated with cancer-specific mortality were evaluated with a Cox proportional hazard model. RESULTS A total of 697 women with PLFGT were identified with a median age of 54years. The most prevalent histological subtypes were diffuse large B-cell (59.8%) and follicular (11.9%) lymphoma. Tumors were most commonly located in the ovary (37%), cervix (21.4%), and uterus (16.5%). According to the Ann Arbor staging system, 42.6% and 17.9% of cases had stage I and stage II disease, respectively. Cancer-directed surgery (CDS) was performed in the majority of cases (62.8%). Five-year OS and CSS were 70.2% and 75.2% respectively. Localized disease, premenopausal age and follicular histology were associated with superior cancer-specific mortality while CDS did not confer any mortality benefit. CONCLUSIONS This is the largest cohort of PLFGT presented in literature. While in our study surgical treatment was not associated with improved outcomes, larger multi-institutional studies should address the optimal management of women with PLFGT.
Collapse
Affiliation(s)
- Dimitrios Nasioudis
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY, USA; Surgery Working Group, Society of Junior Doctors, Athens, Greece.
| | - Polydoros N Kampaktsis
- Surgery Working Group, Society of Junior Doctors, Athens, Greece; Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Melissa Frey
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY, USA
| | - Steven S Witkin
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY, USA
| | - Kevin Holcomb
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
39
|
Yoon JY, Park ES. Gonadal Function in Female Childhood Cancer Survivors. CLINICAL PEDIATRIC HEMATOLOGY-ONCOLOGY 2017. [DOI: 10.15264/cpho.2017.24.1.37] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Ju Young Yoon
- Department of Pediatrics, Gyeongsang National University Changwon Hospital, Changwon, Korea
| | - Eun Sil Park
- Department of Pediatrics, College of Medicine, Gyeongsang National University, Jinju, Korea
- Gyeongsang Institute of Health Science, Gyeongsang National University, Jinju, Korea
| |
Collapse
|
40
|
Batchvarov IS, Taylor RW, Bustamante-Marín X, Czerwinski M, Johnson ES, Kornbluth S, Capel B. A grafted ovarian fragment rescues host fertility after chemotherapy. Mol Hum Reprod 2016; 22:842-851. [PMID: 27698028 DOI: 10.1093/molehr/gaw064] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 07/15/2016] [Accepted: 09/30/2016] [Indexed: 12/21/2022] Open
Abstract
STUDY QUESTION Can host fertility be rescued by grafting of a fragment of a healthy ovary soon after chemotherapy? SUMMARY ANSWER We found that grafting a green fluorescent protein (GFP)-positive fragment from a healthy isogenic ovary to the left ovary of a chemo-treated host rescued function and fertility of the grafted host ovary, and resulted in the production of host-derived offspring as late as the sixth litter after chemotherapy (CTx) treatment, whereas none of the ungrafted controls produced a second litter. WHAT IS KNOWN ALREADY In women and girls undergoing chemotherapy, infertility and premature ovarian failure are frequent outcomes. There are accumulating reports of improved endocrine function after autotransplantation of an ovarian fragment, raising the possibility that the transplant is beneficial to the endogenous ovary. STUDY DESIGN, SIZE, DURATION We first established a CTx treatment regimen that resulted in the permanent loss of fertility in 100% of female mice of the FVB inbred strain. We grafted an isogenic ovary fragment from a healthy female homozygous for a GFP transgene to the left ovary of 100 CTx-treated hosts, and compared fertility to 39 ungrafted controls in 6 months of continuous matings, using GFP to distinguish offspring derived from the graft, and those derived from the host. PARTICIPANTS/MATERIALS, SETTING, METHODS Immunofluoresece and western blot analysis of 39 treated ovaries during and 15 days after CTx treatment revealed elevated apoptosis, rapid loss of granulosa cells and an increased recruitment of growing follicles. Using immunofluorescence and confocal imaging, we tracked the outcome of the grafted tissue over 4 months and its effect on the adjacent and contralateral ovary of the host. MAIN RESULTS AND THE ROLE OF CHANCE Fifty-three percent of grafted females produced a second litter whereas none of the ungrafted females produced a second litter. The likelihood that this could occur by chance is very low (P < 0.0001). LIMITATIONS, REASONS FOR CAUTION These results are shown only in mice, and whether or how they might apply to chemotherapy patients subjected to different CTx regimens is not yet clear. WIDER IMPLICATIONS OF THE FINDINGS Our experiments prove that rescue of a chemo-treated ovary is possible, and establish a system to investigate the mechanism of rescue and to identify the factors responsible with the long-term goal of developing therapies for preservation of ovarian endocrine function and fertility in women undergoing chemotherapy. LARGE SCALE DATA No large datasets were produced. STUDY FUNDING/COMPETING INTERESTS Duke University Medical Center Chancellor's Discovery Grant to BC; ESJ was supported by an NRSA 5F31CA165545; SK was supported by NIH RO1 GM08033; RWT was supported by the Duke University School of Medicine Ovarian Cancer Research Fellowship; XBM was supported by CONICYT. The authors have no conflicts of interest to declare.
Collapse
Affiliation(s)
| | - Rachel Williamson Taylor
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA.,School of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Ximena Bustamante-Marín
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA.,Facultad de Ciencias de la Salud, Departamento Biomédico Universidad de Antofagasta, Antofagasta, Chile
| | - Michael Czerwinski
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Erika Segear Johnson
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Sally Kornbluth
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Blanche Capel
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
41
|
Hickman LC, Valentine LN, Falcone T. Preservation of gonadal function in women undergoing chemotherapy: a review of the potential role for gonadotropin-releasing hormone agonists. Am J Obstet Gynecol 2016; 215:415-22. [PMID: 27422055 DOI: 10.1016/j.ajog.2016.06.053] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 06/24/2016] [Accepted: 06/27/2016] [Indexed: 01/08/2023]
Abstract
A cancer diagnosis in women of reproductive age has unique medical and psychosocial ramifications, especially with treatments that are known to cause gonadal toxicity. For patients who undergo chemotherapy, a multidisciplinary team approach is essential to ensure that the patients' reproductive wishes are addressed. Currently, embryo and oocyte cryopreservation are the standard of care for those who wish to preserve their fertility. The use of gonadotropin-releasing hormone agonists has been a source of debate with numerous studies that have investigated the efficacy on both fertility and ovarian function preservation. This review evaluates the current literature on the use of gonadotropin-releasing hormone agonists for preservation of gonadal function. Assisted reproductive technology is excellent for preservation of fertility but will not protect gonadal function. Protection of gonadal function is critical for the broader issues of health and quality of life as a result of a hypogonadal state. At this moment, gonadotropin-releasing hormone agonists are the only drug class available to protect gonadal function.
Collapse
|
42
|
Fertility preservation in pediatric and adolescent cancer patients in Switzerland: A qualitative cross-sectional survey. Cancer Epidemiol 2016; 44:141-146. [DOI: 10.1016/j.canep.2016.08.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 08/17/2016] [Accepted: 08/22/2016] [Indexed: 12/21/2022]
|
43
|
Andreoli L, Bertsias GK, Agmon-Levin N, Brown S, Cervera R, Costedoat-Chalumeau N, Doria A, Fischer-Betz R, Forger F, Moraes-Fontes MF, Khamashta M, King J, Lojacono A, Marchiori F, Meroni PL, Mosca M, Motta M, Ostensen M, Pamfil C, Raio L, Schneider M, Svenungsson E, Tektonidou M, Yavuz S, Boumpas D, Tincani A. EULAR recommendations for women's health and the management of family planning, assisted reproduction, pregnancy and menopause in patients with systemic lupus erythematosus and/or antiphospholipid syndrome. Ann Rheum Dis 2016; 76:476-485. [PMID: 27457513 PMCID: PMC5446003 DOI: 10.1136/annrheumdis-2016-209770] [Citation(s) in RCA: 484] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 06/13/2016] [Accepted: 06/25/2016] [Indexed: 12/26/2022]
Abstract
Objectives Develop recommendations for women's health issues and family planning in systemic lupus erythematosus (SLE) and/or antiphospholipid syndrome (APS). Methods Systematic review of evidence followed by modified Delphi method to compile questions, elicit expert opinions and reach consensus. Results Family planning should be discussed as early as possible after diagnosis. Most women can have successful pregnancies and measures can be taken to reduce the risks of adverse maternal or fetal outcomes. Risk stratification includes disease activity, autoantibody profile, previous vascular and pregnancy morbidity, hypertension and the use of drugs (emphasis on benefits from hydroxychloroquine and antiplatelets/anticoagulants). Hormonal contraception and menopause replacement therapy can be used in patients with stable/inactive disease and low risk of thrombosis. Fertility preservation with gonadotropin-releasing hormone analogues should be considered prior to the use of alkylating agents. Assisted reproduction techniques can be safely used in patients with stable/inactive disease; patients with positive antiphospholipid antibodies/APS should receive anticoagulation and/or low-dose aspirin. Assessment of disease activity, renal function and serological markers is important for diagnosing disease flares and monitoring for obstetrical adverse outcomes. Fetal monitoring includes Doppler ultrasonography and fetal biometry, particularly in the third trimester, to screen for placental insufficiency and small for gestational age fetuses. Screening for gynaecological malignancies is similar to the general population, with increased vigilance for cervical premalignant lesions if exposed to immunosuppressive drugs. Human papillomavirus immunisation can be used in women with stable/inactive disease. Conclusions Recommendations for women's health issues in SLE and/or APS were developed using an evidence-based approach followed by expert consensus.
Collapse
Affiliation(s)
- L Andreoli
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,Unit of Rheumatology and Clinical Immunology, Spedali Civili, Brescia, Italy
| | - G K Bertsias
- Department of Rheumatology, Clinical Immunology and Allergy, University of Crete Medical School, Heraklion, Greece
| | - N Agmon-Levin
- The Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel Hashomer, Israel.,The Faculty of Medicine, Tel Aviv University, Israel
| | - S Brown
- Royal National Hospital For Rheumatic Diseases, Bath, UK
| | - R Cervera
- Department of Autoimmune Diseases, Hospital Clínic, Barcelona, Catalonia, Spain
| | - N Costedoat-Chalumeau
- AP-HP, Hôpital Cochin, Centre de référence maladies auto-immunes et systémiques rares, Paris, France.,Université Paris Descartes-Sorbonne Paris Cité, Paris, France
| | - A Doria
- Rheumatology Unit, Department of Medicine, University of Padua, Italy
| | - R Fischer-Betz
- Policlinic of Rheumatology, Hiller Research Unit, University Clinic Duesseldorf, Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany
| | - F Forger
- Department of Rheumatology, Immunology and Allergology, University Hospital of Bern, Bern, Switzerland
| | - M F Moraes-Fontes
- Unidade de Doenças Auto-imunes-Serviço Medicina Interna 7.2, Hospital Curry Cabral/Centro Hospitalar Lisboa Central, NEDAI/SPMI, Lisboa, Portugal
| | - M Khamashta
- Lupus Research Unit, The Rayne Institute, St. Thomas Hospital, London, UK.,Department of Rheumatology, Dubai Hospital, Dubai, United Arab Emirates
| | - J King
- EULAR PARE Patient Research Partner, London, UK
| | - A Lojacono
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,Unit of Obstetrics and Gynaecology, Spedali Civili, Brescia, Italy
| | - F Marchiori
- EULAR PARE Patient Research Partner, Rome, Italy
| | - P L Meroni
- Department of Clinical Sciences and Community Health, University of Milan, Istituto Auxologico Italiano, Milan, Italy
| | - M Mosca
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - M Motta
- Neonatology and Neonatal Intensive Care Unit, Spedali Civili, Brescia, Italy
| | - M Ostensen
- Norwegian National Advisory Unit on Pregnancy and Rheumatic Diseases, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - C Pamfil
- Department of Rheumatology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - L Raio
- Department of Obstetrics and Gynaecology, University Hospital of Bern, Inselspital, Switzerland
| | - M Schneider
- Policlinic of Rheumatology, Hiller Research Unit, University Clinic Duesseldorf, Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany
| | - E Svenungsson
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - M Tektonidou
- Rheumatology Unit, Joint Academic Rheumatology Programme, 1st Department of Propaedeutic Internal Medicine Athens, National and Kapodistrian University of Athens, Athens, Greece
| | - S Yavuz
- Department of Rheumatology, Istanbul Bilim University, Istanbul Florence Nightingale Hospital, Esentepe-Istanbul, Turkey
| | - D Boumpas
- 4th Department of Internal Medicine, 'Attikon' University Hospital, Medical School, University of Athens, Athens, Greece.,Joint Academic Rheumatology Program, National and Kapodestrian University of Athens, Athens, Greece
| | - A Tincani
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,Unit of Rheumatology and Clinical Immunology, Spedali Civili, Brescia, Italy
| |
Collapse
|
44
|
Blumenfeld Z. Endocrine prevention of chemotherapy-induced ovarian failure. Future Oncol 2016; 12:1671-4. [DOI: 10.2217/fon-2016-0182] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Zeev Blumenfeld
- Reproductive Endocrinology, RAMBAM Health Care Campus, The Rappaport Faculty of Medicine, Technion – Israel Institute of Technology, Haifa, 31096 Israel
| |
Collapse
|
45
|
Harada M, Osuga Y. Where are oncofertility and fertility preservation treatments heading in 2016? Future Oncol 2016; 12:2313-21. [PMID: 27328888 DOI: 10.2217/fon-2016-0161] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
An improvement in the survival rates of cancer patients and recent advancements in assisted reproductive technologies have led to remarkable progress in oncofertility and fertility preservation treatments. Although there are several available or emerging approaches for fertility preservation, the limited evidence for each strategy is the greatest concern. In this review, we discuss the concerns on currently available options, and propose new approaches for fertility preservation that may be available in the future.
Collapse
Affiliation(s)
- Miyuki Harada
- Department of Obstetrics & Gynecology, Faculty of Medicine, University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, 113-8655, Japan
| | - Yutaka Osuga
- Department of Obstetrics & Gynecology, Faculty of Medicine, University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, 113-8655, Japan
| |
Collapse
|
46
|
Salama M, Isachenko V, Isachenko E, Rahimi G, Mallmann P. Updates in preserving reproductive potential of prepubertal girls with cancer: Systematic review. Crit Rev Oncol Hematol 2016; 103:10-21. [PMID: 27184425 DOI: 10.1016/j.critrevonc.2016.04.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 03/10/2016] [Accepted: 04/07/2016] [Indexed: 11/27/2022] Open
Abstract
INTRODUCTION With increasing numbers of adult female survivors of childhood cancers due to advances in early diagnosis and treatment, the issue of preserving the reproductive potential of prepubertal girls undergoing gonadotoxic treatments has gained greater attention. METHODS According to PRISMA guidelines, a systematic review of the literature was performed for all relevant full-text articles published in PubMed in English throughout the past 15 years to explore the significant updates in preserving the reproductive potential of prepubertal girls with cancer. RESULTS The two established fertility preservation options, embryo freezing and egg freezing, cannot be offered routinely to prepubertal girls as these options necessitate prior ovarian stimulation and subsequent mature oocytes retrieval that are contraindicated or infeasible before puberty. Therefore, the most suitable fertility preservation options to prepubertal girls are (1) ovarian tissue freezing and autotransplantation, (2) in vitro maturation, and (3) ovarian protection techniques. In this review, we discuss in detail those options as well as their success rates, advantages, disadvantages and future directions. We also suggest a new integrated strategy to preserve the reproductive potential of prepubertal girls with cancer. CONCLUSION Although experimental, ovarian tissue slow freezing and orthotopic autotransplantation may be the most feasible option to preserve the reproductive potential of prepubertal girls with cancer. However, this technique has two major and serious disadvantages: (1) the risk of reintroducing malignant cells, and (2) the relatively short lifespan of ovarian tissue transplants. Several medical and ethical considerations should be taken into account before applying this technique to prepubertal girls with cancer.
Collapse
Affiliation(s)
- Mahmoud Salama
- Department of Gynecology and Obstetrics, Medical Faculty, University of Cologne, Germany; Department of Reproductive Medicine, Medical Division, National Research Center of Egypt, Egypt.
| | - Vladimir Isachenko
- Department of Gynecology and Obstetrics, Medical Faculty, University of Cologne, Germany.
| | - Evgenia Isachenko
- Department of Gynecology and Obstetrics, Medical Faculty, University of Cologne, Germany.
| | - Gohar Rahimi
- Department of Gynecology and Obstetrics, Medical Faculty, University of Cologne, Germany.
| | - Peter Mallmann
- Department of Gynecology and Obstetrics, Medical Faculty, University of Cologne, Germany.
| |
Collapse
|
47
|
Bénard J, Calvo J, Comtet M, Benoit A, Sifer C, Grynberg M. [Fertility preservation in women of the childbearing age: Indications and strategies]. ACTA ACUST UNITED AC 2016; 45:424-44. [PMID: 27021926 DOI: 10.1016/j.jgyn.2016.02.005] [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: 07/08/2015] [Revised: 02/15/2016] [Accepted: 02/24/2016] [Indexed: 12/21/2022]
Abstract
Advances on cryopreservation techniques now allow considering oocyte, embryo or ovarian tissue freezing for female fertility preservation. Originally developed for patients suffering from cancer, fertility preservation has rapidly invaded others medical fields, and represents now the standard of care for all young patient diagnosed with a disease that could impair fertility or having to receive possibly gonadotoxic treatment. As a result, autoimmune diseases, some genetic pathologies or iterative pelvic surgeries, at risk of premature ovarian failure, have become common indications of fertility preservation. In addition, the social egg freezing aiming at preventing the age-related fertility decline is still debated in France, although authorized in numerous countries. This review will discuss the different strategies of fertility preservation in young girls and women of reproductive age, regarding different medical or non-medical indications.
Collapse
Affiliation(s)
- J Bénard
- Service de médecine de la reproduction, hôpital Jean-Verdier, avenue du 14-Juillet, 93140 Bondy, France; Université Paris XIII, 93000 Bobigny, France
| | - J Calvo
- Service de médecine de la reproduction, hôpital Jean-Verdier, avenue du 14-Juillet, 93140 Bondy, France
| | - M Comtet
- Service de médecine de la reproduction, hôpital Jean-Verdier, avenue du 14-Juillet, 93140 Bondy, France
| | - A Benoit
- Service de médecine de la reproduction, hôpital Jean-Verdier, avenue du 14-Juillet, 93140 Bondy, France
| | - C Sifer
- Service de cytogénétique et biologie de la reproduction, hôpital Jean-Verdier, avenue du 14-Juillet, 93140 Bondy, France
| | - M Grynberg
- Service de médecine de la reproduction, hôpital Jean-Verdier, avenue du 14-Juillet, 93140 Bondy, France; Université Paris XIII, 93000 Bobigny, France; Unité Inserm U1133, université Paris-Diderot, 75013 Paris, France.
| |
Collapse
|
48
|
Blumenfeld Z. Ovarian suppression during chemotherapy increases pregnancy rate in survivors. Ann Oncol 2016; 27:953. [PMID: 26811348 DOI: 10.1093/annonc/mdw038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Z Blumenfeld
- Reproductive Endocrinology, RAMBAM Health Care Campus, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
49
|
Affiliation(s)
- Christopher L R Barratt
- Editor In Chief, MHR Reproductive and Developmental Biology, Medical School, Ninewells Hospital, University of Dundee, Dundee DD1 9SY, UK Assisted Conception Unit, NHS Tayside, Ninewells Hospital, Dundee DD1 9SY, UK
| |
Collapse
|
50
|
Del Mastro L, Lambertini M. Temporary Ovarian Suppression With Gonadotropin-Releasing Hormone Agonist During Chemotherapy for Fertility Preservation: Toward the End of the Debate? Oncologist 2015; 20:1233-5. [PMID: 26463868 DOI: 10.1634/theoncologist.2015-0373] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 09/21/2015] [Indexed: 12/11/2022] Open
Affiliation(s)
- Lucia Del Mastro
- Department Of Medical Oncology, U.O. Sviluppo Terapie Innovative, IRCCS AOU San Martino-IST, Genova, Italy
| | - Matteo Lambertini
- Department of Medical Oncology, U.O. Oncologia Medica 2, IRCCS AOU San Martino-IST, Genova, Italy
| |
Collapse
|