1
|
Erden M, Gayete-Lafuente S, Vural NA, Oktay KH. Utility and Outcomes of Ovarian Tissue Cryopreservation and Transplantation for Gynecologic Cancers: A Systematic Review and Meta-analysis. Obstet Gynecol 2024; 144:481-492. [PMID: 39173181 PMCID: PMC11499045 DOI: 10.1097/aog.0000000000005708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 07/11/2024] [Indexed: 08/24/2024]
Abstract
OBJECTIVE To evaluate the utility, success, and safety of ovarian tissue cryopreservation and autologous cryopreserved ovarian tissue transplantation for fertility preservation in patients with gynecologic cancers. DATA SOURCES A comprehensive search was performed of the MEDLINE, EMBASE, ClinicalTrials.gov , and Cochrane Library databases to identify relevant studies on the utility and outcomes of ovarian tissue cryopreservation and autologous cryopreserved ovarian tissue transplantation for gynecologic cancers from inception until January 23, 2024. METHODS OF STUDY SELECTION Two reviewers independently performed the study selection, data extraction, and risk-of-bias assessment, and the results were then reviewed together. Twenty-three studies were included in the current systematic review. TABULATION, INTEGRATION, AND RESULTS The resultant data were meta-analyzed to produce a pooled-effect estimate of the utility of ovarian tissue cryopreservation and autologous transplantation in gynecologic cancers as a proportion of all indications. We found that 7.5% and 9.6% of women undergoing ovarian tissue cryopreservation and autologous transplantation, respectively, had gynecologic cancers. In comparison, hematologic malignancies and breast cancer accounted for approximately 66.0% of all indications for these procedures. The return rate for autologous cryopreserved ovarian tissue transplantation in gynecologic cancers (6.0%) was not statistically different from those for other indications. Among women with gynecologic cancer who underwent ovarian stimulation, 27.3% had at least one child, and the ovarian endocrine function was restored in 78.1% of the women after autologous transplantation. The median graft longevity was 32 months, and no graft-site recurrence was reported after autologous transplantation in women with gynecologic cancer. CONCLUSION Our results suggest that ovarian tissue cryopreservation and autologous transplantation are feasible options for preserving ovarian function in women with gynecologic cancers, although ovarian tissue cryopreservation is underutilized, and further studies are needed to determine the longer-term outcomes of autologous transplantation. SYSTEMATIC REVIEW REGISTRATION PROSPERO, CRD42024498522.
Collapse
Affiliation(s)
- Murat Erden
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT
| | - Sonia Gayete-Lafuente
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT
| | - Nazli Aylin Vural
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT
- Department of Gynecological Oncology, Yozgat State Hospital, Yozgat, Turkey
| | - Kutluk H. Oktay
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT
- Innovation Institute for Fertility Preservation New York, NY and New Haven, CT
| |
Collapse
|
2
|
Fouks Y, Bortoletto P, Chang J, Penzias A, Vaughan D, Sakkas D. Looking into the future: a machine learning powered prediction model for oocyte return rates after cryopreservation. Reprod Biomed Online 2024; 50:104432. [PMID: 39642702 DOI: 10.1016/j.rbmo.2024.104432] [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: 01/23/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 12/09/2024]
Abstract
RESEARCH QUESTION Could a predictive model, using data from all US fertility clinics reporting to the Society for Assisted Reproductive Technology, estimate the likelihood of patients using their stored oocytes? DESIGN Multiple learner algorithms, including penalized regressions, random forests, gradient boosting machine, linear discriminant analysis and bootstrap aggregating decision trees were used. Data were split into training and test datasets. Patient demographics, medical and fertility diagnoses, partner information and geographic locations were analysed. RESULTS A total of 77,631 oocyte-cryopreservation cycles (2014-2020) were analysed. Patient age averaged 34.5 years. Treatment indications varied: planned (35.6%), gender-related (0.1%), medically indicated (15.5%), oncologic (5.7%) and unknown (42.3%). Infertility diagnoses were less common: unexplained infertility (1.8%), age-related infertility (3.2%), diminished ovarian reserve (9.9%) and endometriosis (1.6%). An ensemble model combining bootstrap aggregation classification and regression trees, stochastic gradient boosting and linear discriminant analysis yielded the highest predictive accuracy on test set (balanced accuracy: 0.83, sensitivity: 0.76, specificity: 0.91), with a receiver operating characteristic curve of 0.90 and precision-recall curve and area under the curve of 0.57. Key factors influencing the likelihood of returning for oocyte use included patient age, presence of a partner, race or ethnicity, the clinic's geographic region and oocyte cryopreservation indication. CONCLUSIONS This model demonstrated significant predictive accuracy, and is a valuable tool for patient counselling on oocyte cryopreservation. It helps to identify patients more likely to use stored oocytes, enhancing healthcare decision-making and the efficiency of gamete storage programmes. The model can be applied to self-financed and insurance-funded cycles.
Collapse
Affiliation(s)
- Yuval Fouks
- Boston IVF - IVIRMA Global Research Alliance, Waltham, MA, USA; Harvard T.H. Chan School of Public Health, Boston MA, USA; Reproductive Services Royal Women's Hospital, Melbourne Parkville, VIC; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Pietro Bortoletto
- Boston IVF - IVIRMA Global Research Alliance, Waltham, MA, USA; Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Boston MA, USA; Department of Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Boston MA, USA
| | - Jeffrey Chang
- Harvard T.H. Chan School of Public Health, Boston MA, USA
| | - Alan Penzias
- Boston IVF - IVIRMA Global Research Alliance, Waltham, MA, USA; Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Boston MA, USA; Department of Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Boston MA, USA
| | - Denis Vaughan
- Boston IVF - IVIRMA Global Research Alliance, Waltham, MA, USA; Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Boston MA, USA; Department of Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Boston MA, USA
| | - Denny Sakkas
- Boston IVF - IVIRMA Global Research Alliance, Waltham, MA, USA
| |
Collapse
|
3
|
Gazzo I, Moffa F, Ferrero S. Fertility preservation in women with endometriosis: Oocyte cryopreservation and other techniques. Best Pract Res Clin Obstet Gynaecol 2024; 95:102503. [PMID: 38777734 DOI: 10.1016/j.bpobgyn.2024.102503] [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: 10/23/2023] [Accepted: 05/02/2024] [Indexed: 05/25/2024]
Abstract
In recent years, advancements in cryopreservation techniques for oocytes, embryos, and ovarian tissue have enabled offering fertility preservation (FP) options to women with endometriosis. It is recommended to always conduct specialized counselling on FP, especially before considering surgical interventions for endometriosis. The decision regarding the methods of FP, the timing, and to which women affected by endometriosis these techniques should be offered are still subjects of discussion. However, several studies suggest that it can be proposed before surgical interventions for endometriosis, particularly if the patient is undergoing mono or bilateral endometrioma surgery. The most recommended technique is ovarian stimulation, followed by oocyte cryopreservation. Nevertheless, the literature contains various studies describing FP through embryo cryopreservation or the retrieval and cryopreservation of ovarian tissue.
Collapse
Affiliation(s)
- Irene Gazzo
- DINOGMI, University of Genova, Genova, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | | | - Simone Ferrero
- DINOGMI, University of Genova, Genova, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| |
Collapse
|
4
|
Wang YL, Zhai QJ, Wang ZH, Yang X, Wang JL, Zhu HL. A retrospective study of ovarian tissue cryopreservation in female patients with hematological diseases for fertility preservation. Arch Gynecol Obstet 2024; 309:2863-2880. [PMID: 38575798 DOI: 10.1007/s00404-024-07484-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 03/16/2024] [Indexed: 04/06/2024]
Abstract
PURPOSES To investigate the effect and safety of ovarian tissue cryopreservation (OTC) for fertility preservation in female patients with hematological diseases. METHODS We designed a retrospective study. The clinical data of patients with hematological diseases undergoing OTC admitted to Peking University People's Hospital from April 2017 to January 2023 were analyzed and summarized. RESULTS A total of 24 patients were included in the study, including 19 patients with malignant hematological diseases and 5 patients with non-malignant hematological diseases. The former included 14 patients with acute leukemia, 1 patient with chronic leukemia, and 4 patients with myelodysplastic syndrome, while the latter 5 patients were aplastic anemia (AA). 16 patients had received chemotherapy before OTC. The average age of 24 patients was 22.80 ± 6.81 years. The average anti-Mullerian hormone (AMH) was 1.97 ± 2.12 ng/mL, and the average follicle-stimulating hormone (FSH) was 7.01 ± 4.24 IU/L in examination before OTC. FSH was greater than 10.0 IU/L in 4 cases. The pre-OTC laboratory tests showed that the average white blood cell (WBC) count was (3.33 ± 1.35) × 109/L, the average hemoglobin was 91.42 ± 22.84 g/L, and the average platelet was (147.38 ± 114.46) × 109/L. After injection of recombinant human granulocyte colony-stimulating factor (rhG-CSF), blood transfusion, and iron supplementation in pre-OTC treatment, the average WBC count was (4.91 ± 3.07) × 109/L, the average hemoglobin was 98.67 ± 15.43 g/L, and the average platelet was (156.38 ± 103.22) × 109/L. Of the 24 patients, 22 underwent laparoscopic bilateral partial oophorectomy and oophoroplasty, and 2 underwent laparoscopic unilateral oophorectomy. The average duration of OTC was 59.54 ± 17.58 min, and the average blood loss was 32.1 ± 41.6 mL. The maximum blood loss was 200 mL. There was no significant difference in WBC count and hemoglobin concentration after OTC compared to pre-OTC period. Only the platelet count after OTC surgery was significantly different from that before surgery ([134.54 ± 80.84 vs. 156.38 ± 103.22] × 109/L, p < 0.05). None of the 24 patients had serious complications after OTC. 2 patients had mild infection symptoms, but both recovered well. 23 patients underwent hematopoietic stem cell transplantation (HSCT) after OTC. The median and interquartile range from OTC to the pretreatment of HSCT was 33 (57) days, and the median and interquartile range from OTC to HSCT was 41 (57) days. Seven of them began pretreatment of HSCT within 20 days and began HSCT within 30 days after OTC. All patients were followed up. Of the 23 patients who underwent HSCT after surgery, 22 presented with amenorrhea and 1 with scanty menstrual episodes. Seven patients underwent hormone replacement therapy (HRT) after HSCT. A patient with AA underwent ovarian tissue transplantation (OTT) 3 years after HSCT and resumed regular menstruation 6 months after OTT. CONCLUSIONS Ovarian tissue cryopreservation has a promising future in fertility protection in patients with hematological diseases. However, patients with hematological malignancies often have received gonadotoxic therapy before OTC, which may be accompanied by myelosuppression while patients with non-malignant hematological diseases often present with severe hemocytopenia. So perioperative complete blood count of patients should be paid attention to. There was no significant difference in the WBC count and hemoglobin concentration in patients with hematological diseases before and after OTC surgery, and the platelet count decreased slightly within the normal range. Infection is the most common post-OTC complication, and HSCT pretreatment can be accepted as early as the 10th day after OTC. OTC has no adverse effects on patients with hematological diseases and does not delay HSCT treatment. For young patients with hematological diseases, OTC is an effective method of fertility preservation.
Collapse
Affiliation(s)
- Yi-Ling Wang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Peking University, Beijing, 100044, China
| | - Qing-Jie Zhai
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Peking University, Beijing, 100044, China
| | - Zhao-Hua Wang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Peking University, Beijing, 100044, China
| | - Xin Yang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Peking University, Beijing, 100044, China
| | - Jian-Liu Wang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Peking University, Beijing, 100044, China
| | - Hong-Lan Zhu
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Peking University, Beijing, 100044, China.
| |
Collapse
|
5
|
Ruan X, Xu C, Huang H, Xu B, Du J, Cheng J, Jin F, Gu M, Kong W, Yin C, Wu Y, Tian Q, Cao Y, Wu R, Xu L, Jin J, Li Y, Dai Y, Ju R, Ma F, Wang G, Wei W, Huang X, Qin M, Lin Y, Sun Y, Liu R, Zhang W, Li X, Zou L, Hao M, Ye X, Wang F, Wang Y, Hu Z, Huang Y, Zhu T, Yang C, Wang J, Yang X, Ni R, Wang L, Luo G, Min A, Zhang S, Li P, Cheng L, Li L, Jin Q, Shi D, Li Y, Ren F, Cheng Y, Niu J, Tian Y, Mueck AO. Practice guideline on ovarian tissue cryopreservation and transplantation in the prevention and treatment of iatrogenic premature ovarian insufficiency. Maturitas 2024; 182:107922. [PMID: 38325136 DOI: 10.1016/j.maturitas.2024.107922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 02/09/2024]
Abstract
Premature ovarian insufficiency (POI) refers to the decline of ovarian function before the age of 40. POI causes a reduction in or loss of female fertility, accompanied by different degrees of menopausal symptoms, which increases the risk of chronic diseases related to early menopause and seriously affects patients' quality of life and health. It is conservatively estimated that at least one million prepubertal girls and women of reproductive age in China are at risk of iatrogenic POI caused by radiotherapy and chemotherapy every year. With the development of medical technology and the breakthrough of scientific and technological advances, preventing and treating iatrogenic POI have become possible. International and national guidelines consider cryopreserved ovarian tissue transplantation to be the most promising method of preserving the ovarian function and fertility of prepubertal girls and women of reproductive age who cannot delay radiotherapy and chemotherapy. In order to guide the clinical application of ovarian tissue cryopreservation and transplantation technology in China, the Guideline Working Group finally included 14 scientific questions and 18 recommendations through a questionnaire survey, field investigation, and consultation of a large number of Chinese and English literature databases in order to provide a reference for colleagues in clinical practice.
Collapse
Affiliation(s)
- Xiangyan Ruan
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China; Department for Women's Health, University Women's Hospital and Research Center for Women's Health, University of Tuebingen, Tuebingen, Germany.
| | - Che Xu
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China; Fuxing Hospital, Capital Medical University, Beijing, China
| | - Hefeng Huang
- Ministry of Education Key Laboratory of Reproductive Genetics, Shool of Medicine, Zhejiang University, Hangzhou, China
| | - Binghe Xu
- National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Juan Du
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Jiaojiao Cheng
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Fengyu Jin
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Muqing Gu
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Weimin Kong
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Chenghong Yin
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yurui Wu
- Children's Hospital, Capital Institute of Pediatrics, Beijing, China
| | - Qinjie Tian
- Peking Union Medical College Hospital, Peking Union Medical College/Chinese Academy of Medical Sciences, Beijing, China
| | - Yunxia Cao
- The First Affiliated Hospital of Anhui Medical University, Anhui, China
| | - Ruifang Wu
- Peking University Shenzhen Hospital, Shenzhen, China
| | - Liangzhi Xu
- West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jing Jin
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yanglu Li
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yinmei Dai
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Rui Ju
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Fei Ma
- National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Gang Wang
- Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, China
| | - Wei Wei
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | | | - Maoquan Qin
- National Center for Children's Health, Hematology Center, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Yuan Lin
- Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fujian, China
| | - Yuan Sun
- Beijing Jingdu Children's Hospital, Beijing, China
| | - Rong Liu
- Children's Hospital, Capital Institute of Pediatrics, Beijing, China
| | - Wei Zhang
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Xiaodong Li
- The First Hospital of Hebei Medical University, Hebei, China
| | - Lin Zou
- Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Min Hao
- Second Hospital of Shanxi Medical University, Shanxi, China
| | - Xiyang Ye
- Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Fuling Wang
- The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yue Wang
- Henan Provincial People's Hospital, Zhengzhou, China
| | - Zhuoying Hu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yanhong Huang
- Xi'an International Medical Center Hospital, Xi'an, China
| | - Tianyuan Zhu
- Gansu Provincial Maternal and Child-care Hospital/Gansu Province Central Hospital, Lanzhou, China
| | - Caihong Yang
- The General Hospital of Ningxia Medical University, Ningxia, China
| | - Jinping Wang
- Zibo Maternal And Child Health Hospital, Zibo, China
| | - Xiaomin Yang
- Liuzhou Maternity and Child Healthcare Hospital, Liuzhou, China
| | - Rong Ni
- The Central Hospital of Enshi Tu Jia and Miao Autonomous Prefecture, Enshi, China
| | - Liqun Wang
- Jiangxi Maternal and Child Health Hospital, Jiangxi, China
| | - Guangxia Luo
- The First People's Hospital of Huaihua (Hunan University of Medicine General Hospital), Huaihua, China
| | - Aiping Min
- People's Hospital of Leshan City, Leshan, China
| | - Siyou Zhang
- The First People's Hospital of Foshan, Foshan, China
| | - Peiling Li
- The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Linghui Cheng
- The First Affiliated Hospital of Anhui Medical University, Anhui, China
| | - Lianfang Li
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Huairou Maternal and Child Health Care Hospital, Huairou, China
| | - Quanfang Jin
- Shanghai Baoshan Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, China
| | - Dongmei Shi
- Maternal and Child Health Hospital of Yinchuan, Yinchuan, China
| | - Yan Li
- Luoyang Anhe Hospital, Luoyang, China
| | | | | | - Jumin Niu
- Shenyang Women's and Children's Hospital, Shenyang, China
| | - Ying Tian
- XiangXi Ninger Obstetrics and Gynecology Hospital, Xiangxi, China
| | - Alfred O Mueck
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China; Department for Women's Health, University Women's Hospital and Research Center for Women's Health, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
6
|
Un B, Cetinkaya-Un B, Akpolat M, Andic F, Yazir Y. The effects of human amnion membrane-derived mesenchymal stem cells conditioned medium on ionizing radiation-induced premature ovarian failure and endoplasmic reticulum stress-related apoptosis mechanism. Eur J Obstet Gynecol Reprod Biol 2023; 288:191-197. [PMID: 37566962 DOI: 10.1016/j.ejogrb.2023.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 05/07/2023] [Accepted: 08/02/2023] [Indexed: 08/13/2023]
Abstract
OBJECTIVES Premature ovarian failure (POF) is defined as the cessation of menstrual periods for at least 4-6 months before the age of 40 years, accompanied by FSH values measuring over 40 IU/L for a month. Radiation therapy, one of the cancer treatment methods, is known to accelerate ovarian aging by reducing and eliminating the number of primordial follicles in the ovarian follicle pool. Ionizing radiation has been reported to cause POF. The objective of this study is to investigate the impact of mesenchymal stem cell conditioned medium (hAMSCs-CM), which is isolated from the amniotic membrane of human placenta, on premature ovarian failure (POF) caused by whole-body irradiation. The study will focus on the ER stress and apoptosis mechanisms in the process. STUDY DISAYN A POF model was created by exposing rats to 7 Gy of whole-body irradiation. Serum-free hAMSCs-CM were then administered via the tail vein. Follicle count was performed on the ovaries, and immunohistochemistry was used to determine the expressions of GRP78, CHOP, IRE-1, caspase-12, caspase-9, caspase-3. TUNEL was also carried out, and levels of serum FSH, LH, E2, AMH, and oxidative stress marker 8-OHdG were measured. RESULTS AND CONCLUSION The application of hAMSCs-CM has been found to have a positive impact on follicles affected by radiation. After treatment, the number of primordial, primary, secondary, and graafian follicles, which had previously decreased due to radiation, showed an increase. Furthermore, the number of atretic follicles, which had been increasing due to radiation, showed a decrease. ER is one of the targets affected by ionizing radiation. After ionizing radiation, the expressions of ER stress-related markers and apoptosis markers increased in the ovary. After hAMSCs-CM administration, the expressions of these markers and number of TUNEL-positive cells decreased. Following irradiation, anti-mullerian hormone (AMH) and estradiol (E2) levels decreased, while follicle stimulating hormone (FSH) and luteinizing hormone (LH) levels increased. After administration of hAMSCs-CM, AMH and E2 levels increased, while FSH and LH levels decreased. Amnion membrane-derived mesenchymal stem cell conditioned medium can play a therapeutic role in ionizing radiation-induced premature ovarian failure by reducing endoplasmic reticulum stress and apoptosis.
Collapse
Affiliation(s)
- Burak Un
- Department of Obstetrics and Gynecology, Adana City Training and Research Hospital, 01370 Adana, Turkey; Department of Histology and Embryology, Medicine Faculty, Zonguldak Bulent Ecevit University, 67600 Zonguldak, Turkey.
| | - Busra Cetinkaya-Un
- IVF Unit, Adana City Training and Research Hospital, 01370 Adana, Turkey; Department of Histology and Embryology, Medicine Faculty, Zonguldak Bulent Ecevit University, 67600 Zonguldak, Turkey.
| | - Meryem Akpolat
- Department of Histology and Embryology, Medicine Faculty, Zonguldak Bulent Ecevit University, 67600 Zonguldak, Turkey.
| | - Fundagul Andic
- Department of Radiation Oncology, Medicine Faculty, Cukurova University, 01380 Adana, Turkey.
| | - Yusufhan Yazir
- Department of Histology and Embryology, Medicine Faculty, Kocaeli University, 41001 Kocaeli, Turkey.
| |
Collapse
|
7
|
Schallmoser A, Einenkel R, Färber C, Hüren V, Pougin A, Emrich N, John J, Sänger N. Cryostorage of human ovarian tissue: evaluating the storage and disposal pattern over a 22-year period in 2475 patients. Reprod Biomed Online 2023; 47:103239. [PMID: 37400319 DOI: 10.1016/j.rbmo.2023.05.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 07/05/2023]
Abstract
RESEARCH QUESTION What are the parameters of age, indications for ovarian tissue cryopreservation, storage characteristics and reasons for tissue disposal in a large cohort of individuals undertaking cryopreservation? DESIGN The relevant parameters in a single university centre were revised and digitalized in the period from 2019 to 2021. To assess patients' motivation at the end of storage, patients were contacted by letter, e-mails and telephone calls. RESULTS A group of 2475 patients with stored ovarian tissue were analysed in the time period between 2000 and 2021; the response rate for contact calls and letters was 28.8% (224/777). Where storage had ended (n = 1155), patients had on average stored for 3.8 years and begun storing at age 30 years; the main indications were breast cancer (53%) and lymphoma (17.5%). Of these participants, 2.5% had a transplantation on site, 10.3% transferred their tissue to another cryobank and 11.5% were deceased. The majority of the group (75.7%) ended their storage due to pregnancy (49.1%), a lack of desire to have children (25.9%), storage fees that were too expensive (8.9%), death (8.5%), recurrence of cancer (8.5%), lack of a partner (4%) and fear of surgery in the future (3.1%); 6.7% retrospectively regretted ending storage. CONCLUSIONS The pregnancy rate of 49.1%, resulting from ovarian tissue that was not removed during surgery for scheduled ovarian tissue cryopreservation supports the clinical approach of removing and cryopreserving only 25-50% of one ovary. It is proposed that interdisciplinary counselling should be implemented not only prior to fertility preservation, but also when intending to end storage.
Collapse
Affiliation(s)
- Andreas Schallmoser
- Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital of Bonn, Germany.
| | - Rebekka Einenkel
- Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital of Bonn, Germany
| | - Cara Färber
- Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital of Bonn, Germany
| | - Vanessa Hüren
- Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital of Bonn, Germany
| | - Anna Pougin
- Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital of Bonn, Germany
| | - Norah Emrich
- Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital of Bonn, Germany
| | - Julia John
- Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital of Bonn, Germany
| | - Nicole Sänger
- Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital of Bonn, Germany.
| |
Collapse
|
8
|
Rangi S, Hur C, Richards E, Falcone T. Fertility Preservation in Women with Endometriosis. J Clin Med 2023; 12:4331. [PMID: 37445365 DOI: 10.3390/jcm12134331] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 06/20/2023] [Accepted: 06/24/2023] [Indexed: 07/15/2023] Open
Abstract
Several mechanisms have been implicated in the pathogenesis of endometriosis-related infertility. For patients considering surgery, the risk of iatrogenic injury is among the most important factors in the context of fertility preservation, along with age and individual reproductive goals. In the case of endometrioma excision, evidence overwhelmingly demonstrates the negative impact of surgery on ovarian reserve, with significant reductions in antimullerian hormone (up to 30% in unilateral versus up to 44% in bilateral endometriomas). The surgical endometriosis patient should be thoroughly counseled regarding fertility preservation and discussion should include tissue, embryo, and oocyte cryopreservation options. For the latter, data support cryopreservation of 10-15 oocytes in women ≤35 years and over 20 for those >35 years for a realistic chance to achieve one or more live births. When performing surgical interventions for endometriosis, reproductive surgeons should employ fertility-conserving surgical methods to reduce the likelihood of postoperative iatrogenic diminished ovarian reserve.
Collapse
Affiliation(s)
- Sabrina Rangi
- Cleveland Clinic Department of Obstetrics and Gynecology, Women's Health Institute, 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Christine Hur
- Cleveland Clinic Department of Obstetrics and Gynecology, Women's Health Institute, 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Elliott Richards
- Cleveland Clinic Department of Obstetrics and Gynecology, Women's Health Institute, 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Tommaso Falcone
- Cleveland Clinic Department of Obstetrics and Gynecology, Women's Health Institute, 9500 Euclid Ave, Cleveland, OH 44195, USA
| |
Collapse
|
9
|
Hartman N, James C, Barrera EP, Grimstad FW, El-Arabi AM. Effects on Fertility and the Preservation of Fertility in the Transgender Patient. CURRENT SEXUAL HEALTH REPORTS 2023. [DOI: 10.1007/s11930-023-00355-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
|
10
|
Fraison E, Huberlant S, Labrune E, Cavalieri M, Montagut M, Brugnon F, Courbiere B. Live birth rate after female fertility preservation for cancer or haematopoietic stem cell transplantation: a systematic review and meta-analysis of the three main techniques; embryo, oocyte and ovarian tissue cryopreservation. Hum Reprod 2023; 38:489-502. [PMID: 36421038 PMCID: PMC9977128 DOI: 10.1093/humrep/deac249] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 10/21/2022] [Indexed: 11/25/2022] Open
Abstract
STUDY QUESTION What are the chances of achieving a live birth after embryo, oocyte and ovarian tissue cryopreservation (OTC) in female cancer survivors? SUMMARY ANSWER The live birth rates (LBRs) following embryo and oocyte cryopreservation are 41% and 32%, respectively, while for IVF and spontaneous LBR after tissue cryopreservation and transplantation, these rates are 21% and 33%, respectively. WHAT IS KNOWN ALREADY Currently, fertility preservation (FP) has become a major public health issue as diagnostic and therapeutic progress has made it possible to achieve an 80% survival rate in children, adolescents and young adults with cancer. In the latest ESHRE guidelines, only oocyte and embryo cryopreservation are considered as established options for FP. OTC is still considered to be an innovative method, while it is an acceptable FP technique in the American Society for Reproductive Medicine guidelines. However, given the lack of studies on long-term outcomes after FP, it is still unclear which technique offers the best chance to achieve a live birth. STUDY DESIGN, SIZE, DURATION We performed a systematic review and meta-analysis of published controlled studies. Searches were conducted from January 2004 to May 2021 in Medline, Embase and the Cochrane Library using the following search terms: cancer, stem cell transplantation, FP, embryo cryopreservation, oocyte vitrification, OTC and reproductive outcome. PARTICIPANTS/MATERIALS, SETTING, METHODS A total of 126 full-text articles were preselected from 1436 references based on the title and abstract and assessed via the Newcastle-Ottawa Quality Assessment Scale. The studies were selected, and their data were extracted by two independent reviewers according to the Cochrane methods. A fixed-effect meta-analysis was performed for outcomes with high heterogeneity. MAIN RESULTS AND THE ROLE OF CHANCE Data from 34 studies were used for this meta-analysis. Regarding cryopreserved embryos, the LBR after IVF was 41% (95% CI: 34-48, I2: 0%, fixed effect). Concerning vitrified oocytes, the LBR was 32% (95% CI: 26-39, I2: 0%, fixed effect). Finally, the LBR after IVF and the spontaneous LBR after ovarian tissue transplantation were 21% (95% CI: 15-26, I2: 0%, fixed-effect) and 33% (95% CI: 25-42, I2: 46.1%, random-effect), respectively. For all outcomes, in the sensitivity analyses, the maximum variation in the estimated percentage was 1%. LIMITATIONS, REASONS FOR CAUTION The heterogeneity of the literature prevents us from comparing these three techniques. This meta-analysis provides limited data which may help clinicians when counselling patients. WIDER IMPLICATIONS OF THE FINDINGS This study highlights the need for long-term follow-up registries to assess return rates, as well as spontaneous pregnancy rates and birth rates after FP. STUDY FUNDING/COMPETING INTEREST(S) This work was sponsored by an unrestricted grant from GEDEON RICHTER France. The authors have no competing interests to declare. REGISTRATION NUMBER CRD42021264042.
Collapse
Affiliation(s)
- E Fraison
- Service de Médecine de la Reproduction, Hospices Civils de Lyon, Hôpital Mère Enfant, Bron, France.,Université Claude Bernard, Faculté de Médecine Laennec, Lyon, France.,INSERM Unité 1208, Bron, France
| | - S Huberlant
- Service de Gynécologie Obstétrique et Médecine de la Reproduction, CHU Carémeau, Nîmes, France.,Université de Montpellier-Nîmes, Nîmes Cedex 2, France
| | - E Labrune
- Service de Médecine de la Reproduction, Hospices Civils de Lyon, Hôpital Mère Enfant, Bron, France.,Université Claude Bernard, Faculté de Médecine Laennec, Lyon, France.,INSERM Unité 1208, Bron, France
| | - M Cavalieri
- Service de Gynécologie-Obstétrique et Médecine de la Reproduction, CHU François Mitterrand, Dijon, France
| | - M Montagut
- Service de Médecine de la Reproduction, Clinique Croix du Sud, Quint-Fonsegrives, France
| | - F Brugnon
- Assistance Médicale à la Procréation, CECOS, CHU Clermont Ferrand, CHU Estaing, Clermont-Ferrand, France.,Université Clermont Auvergne, IMoST, INSERM 1240, Faculté de Médecine, Clermont-Ferrand, France
| | - B Courbiere
- Service d'Assistance Médicale à la Procréation, Plateforme Cancer & Fertilité OncoPACA-Corse, AP-HM, Hôpital La Conception, Marseille, France.,Aix-Marseille Université, IMBE, CNRS, IRD, Avignon Université, Marseille, France
| |
Collapse
|
11
|
Yildiz S, Bildik G, Benlioglu C, Turan V, Dilege E, Ozel M, Kim S, Oktem O. Breast cancer treatment and ovarian function. Reprod Biomed Online 2023; 46:313-331. [PMID: 36400663 DOI: 10.1016/j.rbmo.2022.09.014] [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: 06/13/2022] [Revised: 09/14/2022] [Accepted: 09/14/2022] [Indexed: 02/07/2023]
Abstract
The aim of this study was to provide an update on ovarian function and the mechanisms of gonadal damage after exposure to chemotherapy in breast cancer survivors. The alkylating agents are toxic to both primordial and growing follicles. However, anti-metabolite drugs are more likely to destroy preantral and antral follicles. Younger patients are more likely to have a higher ovarian reserve, and therefore, more likely to retain some residual ovarian function after exposure to gonadotoxic regimens. However, there can be significant variability in ovarian reserve among patients of the same age. Furthermore, patients with critically diminished ovarian reserve may continue to menstruate regularly. Therefore age and menstrual status are not reliable indicators of good ovarian reserve and might give a false sense of security and result in an adverse outcome if the patient is consulted without considering more reliable quantitative markers of ovarian reserve (antral follicle count and anti-Müllerian hormone) and fertility preservation is not pursued. In contrast to well-documented ovarian toxicity of older chemotherapy regimens, data for newer taxane-containing protocols have only accumulated in the last decade and data are still very limited regarding the impact of targeted therapies on ovarian function.
Collapse
Affiliation(s)
- Sule Yildiz
- The Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Koç University Hospital, Koç University School of Medicine, Istanbul, Turkey
| | - Gamze Bildik
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston TX 77054, USA
| | - Can Benlioglu
- Department of Obstetrics and Gynecology, Koç University Hospital, Istanbul, Turkey
| | - Volkan Turan
- Istanbul Tema Hospital, Assisted Reproduction Unit, Istanbul
| | - Ece Dilege
- Department of General Surgery, Koç University Hospital, Koç University School of Medicine, Istanbul, Turkey
| | - Melis Ozel
- Department of Gynecology and Obstetrics Klinikum Ingolstadt, Bavaria, Germany
| | - Samuel Kim
- Eden Centers for Advanced Fertility, Fullerton CA 92835, USA
| | - Ozgur Oktem
- The Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Koç University Hospital, Koç University School of Medicine, Istanbul, Turkey.
| |
Collapse
|
12
|
Bonavina G, Taylor HS. Endometriosis-associated infertility: From pathophysiology to tailored treatment. Front Endocrinol (Lausanne) 2022; 13:1020827. [PMID: 36387918 PMCID: PMC9643365 DOI: 10.3389/fendo.2022.1020827] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/06/2022] [Indexed: 11/13/2022] Open
Abstract
Despite the clinically recognized association between endometriosis and infertility, the mechanisms implicated in endometriosis-associated infertility are not fully understood. Endometriosis is a multifactorial and systemic disease that has pleiotropic direct and indirect effects on reproduction. A complex interaction between endometriosis subtype, pain, inflammation, altered pelvic anatomy, adhesions, disrupted ovarian reserve/function, and compromised endometrial receptivity as well as systemic effects of the disease define endometriosis-associated infertility. The population of infertile women with endometriosis is heterogeneous, and diverse patients' phenotypes can be observed in the clinical setting, thus making difficult to establish a precise diagnosis and a single mechanism of endometriosis related infertility. Moreover, clinical management of infertility associated with endometriosis can be challenging due to this heterogeneity. Innovative non-invasive diagnostic tools are on the horizon that may allow us to target the specific dysfunctional alteration in the reproduction process. Currently the treatment should be individualized according to the clinical situation and to the suspected level of impairment. Here we review the etiology of endometriosis related infertility as well as current treatment options, including the roles of surgery and assisted reproductive technologies.
Collapse
Affiliation(s)
- Giulia Bonavina
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, United States
| | - Hugh S Taylor
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
13
|
Dietl AK, Dittrich R, Hoffmann I, Denschlag D, Hanjalic-Beck A, Müller A, Beckmann MW, Lotz L. Does it make sense to refreeze ovarian tissue after unexpected occurrence of endometriosis when transplanting the tissue? J Ovarian Res 2022; 15:53. [PMID: 35513873 PMCID: PMC9074247 DOI: 10.1186/s13048-022-00972-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 03/26/2022] [Indexed: 11/10/2022] Open
Abstract
Background Ovarian insufficiency is a major concern for long-term cancer survivors. Ovarian tissue cryopreservation for fertility preservation is an emerging technique that has proven successful over the past decade through transplantation of frozen-thawed ovarian tissue. Compared to other established techniques, such as oocyte freezing, ovarian tissue cryopreservation preserves actual organ function and thus the production of sex hormones. Endometriosis in perimenopausal women is rare, however it can be surprising diagnosis in the planned transplantation of cryopreserved ovarian tissue and the already thawed tissue may not be transplanted, so that it has to be refrozen. Results Ovarian function returned in the patient two months after transplantation, as shown by estrogen production. Ten months after the ovarian tissue transplantation mild stimulation with FSH was initiated in accordance with a low-dose protocol. When ultrasonography revealed a follicle 17 mm in size in the ovarian graft, hCG was added and after follicular puncture one oocyte was obtained. The oocyte could be fertilized by IVF and transferred to the uterus. On day 14 after embryo-transfer, a positive hCG-Level was detected and after an uncomplicated pregnancy a healthy child was delivered. Conclusions We report the first pregnancy and live birth achieved using transplantation of thawed and refrozen ovarian tissue in a woman treated by chemotherapy and subsequent endometriosis surgery. Refreezing of cryopreserved ovarian tissue is not a hindrance to successful transplantation of ovarian tissue. Against the background of increasing numbers of candidates for transplantation of ovarian tissue is expected that the combination chemotherapy followed by endometriosis will increase.
Collapse
Affiliation(s)
- Anna K Dietl
- Department of Obstetrics and Gynecology, Erlangen University Hospital, Friedrich-Alexander University of Erlangen-Nürnberg, Universitätsstrasse 21-23, 91054, Erlangen, Germany
| | - Ralf Dittrich
- Department of Obstetrics and Gynecology, Erlangen University Hospital, Friedrich-Alexander University of Erlangen-Nürnberg, Universitätsstrasse 21-23, 91054, Erlangen, Germany.
| | - Inge Hoffmann
- Department of Obstetrics and Gynecology, Erlangen University Hospital, Friedrich-Alexander University of Erlangen-Nürnberg, Universitätsstrasse 21-23, 91054, Erlangen, Germany
| | | | - Aida Hanjalic-Beck
- Center for Gynecologic Endocrinology and Reproductive Medicine, Freiburg, Germany
| | - Andreas Müller
- Department of Obstetrics and Gynecology, Municipal-Hospital, Karlsruhe, Germany
| | - Matthias W Beckmann
- Department of Obstetrics and Gynecology, Erlangen University Hospital, Friedrich-Alexander University of Erlangen-Nürnberg, Universitätsstrasse 21-23, 91054, Erlangen, Germany
| | - Laura Lotz
- Department of Obstetrics and Gynecology, Erlangen University Hospital, Friedrich-Alexander University of Erlangen-Nürnberg, Universitätsstrasse 21-23, 91054, Erlangen, Germany
| |
Collapse
|
14
|
Khattak H, Malhas R, Craciunas L, Afifi Y, Amorim CA, Fishel S, Silber S, Gook D, Demeestere I, Bystrova O, Lisyanskaya A, Manikhas G, Lotz L, Dittrich R, Colmorn LB, Macklon KT, Hjorth IMD, Kristensen SG, Gallos I, Coomarasamy A. Fresh and cryopreserved ovarian tissue transplantation for preserving reproductive and endocrine function: a systematic review and individual patient data meta-analysis. Hum Reprod Update 2022; 28:400-416. [PMID: 35199164 PMCID: PMC9733829 DOI: 10.1093/humupd/dmac003] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 12/29/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Ovarian tissue cryopreservation involves freezing and storing of surgically retrieved ovarian tissue in liquid or vapour nitrogen below -190°C. The tissue can be thawed and transplanted back with the aim of restoring fertility or ovarian endocrine function. The techniques for human ovarian tissue freezing and transplantation have evolved over the last 20 years, particularly in the context of fertility preservation in pre-pubertal cancer patients. Fresh ovarian tissue transplantation, using an autograft or donor tissue, is a more recent development; it has the potential to preserve fertility and hormonal function in women who have their ovaries removed for benign gynaecological conditions. The techniques of ovarian tissue cryopreservation and transplantation have progressed rapidly since inception; however, the evidence on the success of this intervention is largely based on case reports and case series. OBJECTIVE AND RATIONALE The aim of this study was to systematically review the current evidence by incorporating study-level and individual patient-level meta-analyses of women who received ovarian transplants, including frozen-thawed transplant, fresh or donor graft. SEARCH METHODS The review protocol was registered with PROSPERO (CRD42018115233). A comprehensive literature search was performed using MEDLINE, EMBASE, CINAHL and Cochrane Central Register of Controlled Trials from database inception to October 2020. Authors were also contacted for individual patient data if relevant outcomes were not reported in the published manuscripts. Meta-analysis was performed using inverse-variance weighting to calculate summary estimates using a fixed-effects model. OUTCOMES The review included 87 studies (735 women). Twenty studies reported on ≥5 cases of ovarian transplants and were included in the meta-analysis (568 women). Fertility outcomes included pregnancy, live birth and miscarriage rates, and endocrine outcomes included oestrogen, FSH and LH levels. The pooled rates were 37% (95% CI: 32-43%) for pregnancy, 28% (95% CI: 24-34%) for live birth and 37% (95% CI: 30-46%) for miscarriage following frozen ovarian tissue transplantation. Pooled mean for pre-transplant oestrogen was 101.6 pmol/l (95% CI: 47.9-155.3), which increased post-transplant to 522.4 pmol/l (95% CI: 315.4-729; mean difference: 228.24; 95% CI: 180.5-276). Pooled mean of pre-transplant FSH was 66.4 IU/l (95% CI: 52.8-84), which decreased post-transplant to 14.1 IU/l (95% CI: 10.9-17.3; mean difference 61.8; 95% CI: 57-66.6). The median time to return of FSH to a value <25 IU/l was 19 weeks (interquartile range: 15-26 weeks; range: 0.4-208 weeks). The median duration of graft function was 2.5 years (interquartile range: 1.4-3.4 years; range: 0.7-5 years). The analysis demonstrated that ovarian tissue cryopreservation and transplantation could restore reproductive and hormonal functions in women. Further studies with larger samples of well-characterized populations are required to define the optimal retrieval, cryopreservation and transplantation processes. WIDER IMPLICATIONS Ovarian tissue cryopreservation and transplantation may not only be effective in restoring fertility but also the return of reproductive endocrine function. Although this technology was developed as a fertility preservation option, it may have the scope to be considered for endocrine function preservation.
Collapse
Affiliation(s)
- Hajra Khattak
- Tommy’s National Centre for Miscarriage Research, Institute of Metabolism and
Systems Research, University of Birmingham, Birmingham, UK
| | - Rosamund Malhas
- Birmingham Women’s and Children’s NHS Foundation Trust,
Birmingham, UK
| | - Laurentiu Craciunas
- Population Health Sciences Institute, Newcastle University, Newcastle upon
Tyne, UK
| | - Yousri Afifi
- Birmingham Women’s and Children’s NHS Foundation Trust,
Birmingham, UK
| | - Christiani A Amorim
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et
Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Simon Fishel
- CARE Fertility Group, Nottingham, UK
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores
University, Liverpool, UK
| | | | - Debra Gook
- Reproductive Services/Melbourne IVF, The Royal Women’s Hospital,
Parkville, VIC, Australia
| | - Isabelle Demeestere
- Research Laboratory on Human Reproduction, Faculty of Medicine, Université
Libre de Bruxelles (ULB), Brussels, Belgium
| | - Olga Bystrova
- AVA-PETER Fertility Clinic, Saint-Petersburg, Russia
| | - Alla Lisyanskaya
- Division of Gynecologic Oncology, Saint-Petersburg City Oncology
Clinic, Saint-Petersburg, Russia
| | - Georgy Manikhas
- Department of Oncology of the First Pavlov State Medical University of
Saint-Petersburg, Saint-Petersburg, Russia
| | - Laura Lotz
- Department of Obstetrics and Gynecology, Erlangen University Hospital,
Friedrich Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Ralf Dittrich
- Department of Obstetrics and Gynecology, Erlangen University Hospital,
Friedrich Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Lotte Berdiin Colmorn
- The Fertility Clinic, University Hospital of Copenhagen,
Rigshospitalet, Copenhagen, Denmark
| | - Kirsten Tryde Macklon
- The Fertility Clinic, University Hospital of Copenhagen,
Rigshospitalet, Copenhagen, Denmark
| | | | - Stine Gry Kristensen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women,
Children and Reproduction, University Hospital of Copenhagen,
Rigshospitalet, Copenhagen, Denmark
| | - Ioannis Gallos
- Tommy’s National Centre for Miscarriage Research, Institute of Metabolism and
Systems Research, University of Birmingham, Birmingham, UK
| | - Arri Coomarasamy
- Tommy’s National Centre for Miscarriage Research, Institute of Metabolism and
Systems Research, University of Birmingham, Birmingham, UK
| |
Collapse
|
15
|
Maher JY, Islam MS, Yin O, Brennan J, Gough E, Driggers P, Segars J. The role of Hippo pathway signaling and A-kinase anchoring protein 13 in primordial follicle activation and inhibition. F&S SCIENCE 2022; 3:118-129. [PMID: 35560009 PMCID: PMC11096729 DOI: 10.1016/j.xfss.2022.03.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/18/2022] [Accepted: 03/25/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVE To determine whether the mechanotransduction and pharmacomanipulation of A-kinase anchoring protein 13 (AKAP13) altered Hippo signaling pathway transcription and growth factors in granulosa cells. Primary ovarian insufficiency is the depletion or dysfunction of primordial ovarian follicles. In vitro activation of ovarian tissue in patients with primary ovarian insufficiency alters the Hippo and phosphatase and tensin homolog/phosphatidylinositol 3-kinase/protein kinase B/forkhead box O3 pathways. A-kinase anchoring protein 13 is found in granulosa cells and may regulate the Hippo pathway via F-actin polymerization resulting in altered nuclear yes-associated protein (YAP)/transcriptional coactivator with PDZ-binding motif coactivators and Tea domain family (TEAD) transcription factors. DESIGN Laboratory studies. SETTING Translational science laboratory. PATIENT(S) None. INTERVENTION(S) COV434 cells, derived from a primary human granulosa tumor cell line, were studied under different cell density and well stiffness conditions. Cells were transfected with a TEAD-luciferase (TEAD-luc) reporter as well as expression constructs for AKAP13 or AKAP13 mutants and then treated with AKAP13 activators, inhibitors, and follicle-stimulating hormone. MAIN OUTCOME MEASURE(S) TEAD gene activation or inhibition was measured by TEAD-luciferase assays. The messenger ribonucleic acid levels of Hippo pathway signaling molecules, including connective tissue growth factor (CTGF), baculoviral inhibitors of apoptosis repeat-containing 5, Ankyrin repeat domain-containing protein 1, YAP1, and TEAD1, were measured by quantitative real-time polymerase chain reaction. Protein expressions for AKAP13, CTGF, YAP1, and TEAD1 were measured using Western blot. RESULT(S) Increased TEAD-luciferase activity and expression of markers for cellular growth were associated with decreased cell density, increased well stiffness, and AKAP13 activator (A02) treatment. Additionally, decreased TEAD-luc activity and expression of markers for cellular growth were associated with AKAP13 inhibitor (A13) treatment, including a reduced expression of the BIRC5 and ANKRD1 (YAP-responsive genes) transcript levels and CTGF protein levels. There were no changes in TEAD-luc with follicle-stimulating hormone treatment, supporting Hippo pathway involvement in the gonadotropin-independent portion of folliculogenesis. CONCLUSION(S) These findings suggest that AKAP13 mediates Hippo-regulated changes in granulosa cell growth via mechanotransduction and pharmacomanipulation. The AKAP13 regulation of the Hippo pathway may represent a potential target for regulation of follicle activation.
Collapse
Affiliation(s)
- Jacqueline Yano Maher
- Johns Hopkins School of Medicine, Baltimore, Maryland; Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Children's National Medical Center, Washington, D.C..
| | | | - Ophelia Yin
- David Geffen School of Medicine, University of California, Los Angeles, California
| | | | - Ethan Gough
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Paul Driggers
- Johns Hopkins School of Medicine, Baltimore, Maryland
| | - James Segars
- Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
16
|
Ní Dhonnabháin B, Elfaki N, Fraser K, Petrie A, Jones BP, Saso S, Hardiman PJ, Getreu N. A comparison of fertility preservation outcomes in patients who froze oocytes, embryos, or ovarian tissue for medically indicated circumstances: a systematic review and meta-analysis. Fertil Steril 2022; 117:1266-1276. [PMID: 35459522 DOI: 10.1016/j.fertnstert.2022.03.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 03/04/2022] [Accepted: 03/04/2022] [Indexed: 02/08/2023]
Abstract
OBJECTIVE To compare obstetric outcomes in patients cryopreserving reproductive cells or tissues before gonadotoxic therapy. DESIGN A literature search was conducted following PRISMA guidelines on Embase, Medline, and Web of Science. Studies reporting obstetric outcomes in cancer patients who completed cryopreservation of oocyte, embryo, or ovarian tissue were included. SETTING Not applicable. PATIENT(S) Cancer patients attempting pregnancy using cryopreserved cells or tissues frozen before cancer therapy. INTERVENTION(S) Oocyte, embryo, or ovarian tissue cryopreservation for fertility preservation in cancer. MAIN OUTCOME MEASURE(S) The total numbers of clinical pregnancies, live births, and miscarriages in women attempting pregnancy using cryopreserved reproductive cells or tissues were calculated. A meta-analysis determined the effect size of each intervention. RESULT(S) The search returned 4,038 unique entries. Thirty-eight eligible studies were analyzed. The clinical pregnancy rates were 34.9%, 49.0%, and 43.8% for oocyte, embryo, and ovarian tissue cryopreservation, respectively. No significant differences were found among groups. The live birth rates were 25.8%, 35.3%, and 32.3% for oocyte, embryo, and ovarian tissue cryopreservation, respectively, with no significant differences among groups. The miscarriage rates were 9.2%, 16.9%, and 7.5% for oocyte, embryo, and ovarian tissue cryopreservation, respectively. Significantly fewer miscarriages occurred with ovarian tissue cryopreservation than with embryo cryopreservation. CONCLUSION(S) This enquiry is required to counsel cancer patients wishing to preserve fertility. Although the limitations of this study include heterogeneity, lack of quality studies, and low utilization rates, it serves as a starting point for comparison of reproductive and obstetric outcomes in patients returning for family-planning after gonadotoxic therapy.
Collapse
Affiliation(s)
- Bríd Ní Dhonnabháin
- Institute for Women's Health, University College London, London, United Kingdom
| | - Nagla Elfaki
- Department of Obstetrics and Gynaecology, University College London Hospital, London, United Kingdom
| | - Kyra Fraser
- Department of Surgery, The Royal Free Hospital, London, United Kingdom
| | - Aviva Petrie
- Biostatistics Unit, Eastman Dental Institute, University College London, London, United Kingdom
| | - Benjamin P Jones
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Srdjan Saso
- Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
| | - Paul J Hardiman
- Department of Gynaecology, The Royal Free Hospital, London, United Kingdom
| | - Natalie Getreu
- Institute for Women's Health, University College London, London, United Kingdom.
| |
Collapse
|
17
|
Khattak H, Gallos I, Coomarasamy A, Topping AE. Why are women considering ovarian tissue cryopreservation to preserve reproductive and hormonal ovarian function? A qualitative study protocol. BMJ Open 2022; 12:e051288. [PMID: 35418423 PMCID: PMC9013984 DOI: 10.1136/bmjopen-2021-051288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 03/28/2022] [Indexed: 11/26/2022] Open
Abstract
INTRODUCTION Current fertility preservation options available to women are oocyte cryopreservation (egg freezing) or embryo cryopreservation. A newer procedure, ovarian tissue cryopreservation (OTC), has become available in some centres, which offers another option for women and girls considering fertility preservation. These procedures are commonly offered to women about to undergo treatments for cancer. OTC involves removing sections of ovarian tissue and cryopreserving it for future reimplantation, often several years later. OTC offers girls and women who may become infertile with optionality and the possibility of pregnancy. OTC has potential for other applications, including restoring ovarian endocrine function beyond biological menopause. This is not without controversy but has led to some women considering undergoing the procedure for purposes of ovarian hormonal preservation (conservation of ovarian endocrine function). OTC is invasive, involves two surgical procedures with concomitant risks and can be costly. Understanding why women may consider and ultimately undergo OTC is timely, so that evidence-based and women-centred care can be provided. METHODS A pragmatic narrative qualitative design will be used. A purposive sample of women aged 18-45 who are considering, or have sought, OTC will be recruited over 1-year period. Potential participants will be approached via a clinic that offers OTC on a private basis or via social media. ANALYSIS Participant interviews will be audio and, if consented, video recorded. These will be conducted face-to-face or virtually. The recordings will be transcribed verbatim and analysed using a thematic analysis approach supported by NVivo software. ETHICS AND DISSEMINATION Ethical approval has been granted by the Institutional Ethical Review ERN_19-1578A. We expect to disseminate the findings of this study through journal articles, conference presentations and multimedia to public.
Collapse
Affiliation(s)
- Hajra Khattak
- Tommy's National Centre for Miscarriage Research, Institute of Metabolism and Systems Research, University of Birmingham College of Medical and Dental Sciences, Birmingham, UK
| | - Ioannis Gallos
- Tommy's National Centre for Miscarriage Research, Institute of Metabolism and Systems Research, University of Birmingham College of Medical and Dental Sciences, Birmingham, UK
| | - Arri Coomarasamy
- Tommy's National Centre for Miscarriage Research, Institute of Metabolism and Systems Research, University of Birmingham College of Medical and Dental Sciences, Birmingham, UK
| | - A E Topping
- Institute of Clinical Sciences, School of Nursing, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, UK
| |
Collapse
|
18
|
Ekpo MD, Xie J, Hu Y, Liu X, Liu F, Xiang J, Zhao R, Wang B, Tan S. Antifreeze Proteins: Novel Applications and Navigation towards Their Clinical Application in Cryobanking. Int J Mol Sci 2022; 23:2639. [PMID: 35269780 PMCID: PMC8910022 DOI: 10.3390/ijms23052639] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 02/16/2022] [Accepted: 02/25/2022] [Indexed: 12/04/2022] Open
Abstract
Antifreeze proteins (AFPs) or thermal hysteresis (TH) proteins are biomolecular gifts of nature to sustain life in extremely cold environments. This family of peptides, glycopeptides and proteins produced by diverse organisms including bacteria, yeast, insects and fish act by non-colligatively depressing the freezing temperature of the water below its melting point in a process termed thermal hysteresis which is then responsible for ice crystal equilibrium and inhibition of ice recrystallisation; the major cause of cell dehydration, membrane rupture and subsequent cryodamage. Scientists on the other hand have been exploring various substances as cryoprotectants. Some of the cryoprotectants in use include trehalose, dimethyl sulfoxide (DMSO), ethylene glycol (EG), sucrose, propylene glycol (PG) and glycerol but their extensive application is limited mostly by toxicity, thus fueling the quest for better cryoprotectants. Hence, extracting or synthesizing antifreeze protein and testing their cryoprotective activity has become a popular topic among researchers. Research concerning AFPs encompasses lots of effort ranging from understanding their sources and mechanism of action, extraction and purification/synthesis to structural elucidation with the aim of achieving better outcomes in cryopreservation. This review explores the potential clinical application of AFPs in the cryopreservation of different cells, tissues and organs. Here, we discuss novel approaches, identify research gaps and propose future research directions in the application of AFPs based on recent studies with the aim of achieving successful clinical and commercial use of AFPs in the future.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Songwen Tan
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China; (M.D.E.); (J.X.); (Y.H.); (X.L.); (F.L.); (J.X.); (R.Z.); (B.W.)
| |
Collapse
|
19
|
Diaz AA, Kubo H, Handa N, Hanna M, Laronda MM. A Systematic Review of Ovarian Tissue Transplantation Outcomes by Ovarian Tissue Processing Size for Cryopreservation. Front Endocrinol (Lausanne) 2022; 13:918899. [PMID: 35774145 PMCID: PMC9239173 DOI: 10.3389/fendo.2022.918899] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/10/2022] [Indexed: 01/18/2023] Open
Abstract
UNLABELLED Ovarian tissue cryopreservation (OTC) is the only pre-treatment option currently available to preserve fertility for prepubescent girls and patients who cannot undergo ovarian stimulation. Currently, there is no standardized method of processing ovarian tissue for cryopreservation, despite evidence that fragmentation of ovaries may trigger primordial follicle activation. Because fragmentation may influence ovarian transplant function, the purpose of this systematic review was (1) to identify the processing sizes and dimensions of ovarian tissue within sites around the world, and (2) to examine the reported outcomes of ovarian tissue transplantation including, reported duration of hormone restoration, pregnancy, and live birth. A total of 2,252 abstracts were screened against the inclusion criteria. In this systematic review, 103 studies were included for analysis of tissue processing size and 21 studies were included for analysis of ovarian transplantation outcomes. Only studies where ovarian tissue was cryopreserved (via slow freezing or vitrification) and transplanted orthotopically were included in the review. The size of cryopreserved ovarian tissue was categorized based on dimensions into strips, squares, and fragments. Of the 103 studies, 58 fertility preservation sites were identified that processed ovarian tissue into strips (62%), squares (25.8%), or fragments (31%). Ovarian tissue transplantation was performed in 92 participants that had ovarian tissue cryopreserved into strips (n = 51), squares (n = 37), and fragments (n = 4). All participants had ovarian tissue cryopreserved by slow freezing. The pregnancy rate was 81.3%, 45.5%, 66.7% in the strips, squares, fragment groups, respectively. The live birth rate was 56.3%, 18.2%, 66.7% in the strips, squares, fragment groups, respectively. The mean time from ovarian tissue transplantation to ovarian hormone restoration was 3.88 months, 3.56 months, and 3 months in the strips, squares, and fragments groups, respectively. There was no significant difference between the time of ovarian function' restoration and the size of ovarian tissue. Transplantation of ovarian tissue, regardless of its processing dimensions, restores ovarian hormone activity in the participants that were reported in the literature. More detailed information about the tissue processing size and outcomes post-transplant are required to identify a preferred or more successful processing method. SYSTEMATIC REVIEW REGISTRATION [https://www.crd.york.ac.uk], identifier [CRD42020189120].
Collapse
Affiliation(s)
- Ashley A. Diaz
- Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, United States
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Hana Kubo
- Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, United States
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Nicole Handa
- Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, United States
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Maria Hanna
- Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, United States
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Monica M. Laronda
- Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, United States
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- *Correspondence: Monica M. Laronda,
| |
Collapse
|
20
|
Khaleghi S, Fathi R, Eivazkhani F, Moini A, Novin MG, Ebrahimi B, Nazarian H. Two-Decade Experience of Royan Institute in Obtaining Mature Oocyte from Cryopreserved Ovarian Tissue: In Vitro and In Vivo Approaches. Reprod Sci 2021; 29:1685-1696. [PMID: 34533785 DOI: 10.1007/s43032-021-00728-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 08/26/2021] [Indexed: 12/14/2022]
Abstract
Ovarian tissue cryopreservation (OTC) holds promise for preservation of fertility among women who have lost their fertility due to diseases such as cancer. OTC has significantly assisted such cases by helping them maintain normal hormonal levels and menstrual cycles. Appropriate strategies to develop and extract mature oocytes from OTC could overcome a range of complications that are associated with ovarian dysfunction, caused by aging, and primary or secondary ovarian insufficiency. Scientists from different departments at The Royan Institute (Tehran, Iran) have been conducting studies to find the best way to extract mature oocytes from animal and human cryopreserved ovarian tissues. The various studies conducted in this area in the past 20 years, by researchers of the Royan Institute, are collated and provided in this review, in order to provide an idea of where we are now in the area of fertility preservation.
Collapse
Affiliation(s)
- Sara Khaleghi
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rouhollah Fathi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.
| | - Farideh Eivazkhani
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Ashraf Moini
- Department of Endocrinology and Female Infertility, Royan Institute of Reproductive Biomedicine, ACECR, Tehran, Iran.,Breast Disease Research Center (BDRC), Tehran University of Medical Science, Tehran, Iran
| | - Marefat Ghaffari Novin
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bita Ebrahimi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Hamid Nazarian
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
21
|
Lee S, Ozkavukcu S, Ku SY. Current and Future Perspectives for Improving Ovarian Tissue Cryopreservation and Transplantation Outcomes for Cancer Patients. Reprod Sci 2021; 28:1746-1758. [PMID: 33791995 PMCID: PMC8144135 DOI: 10.1007/s43032-021-00517-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 02/21/2021] [Indexed: 02/06/2023]
Abstract
Although advances in cancer treatment and early diagnosis have significantly improved cancer survival rates, cancer therapies can cause serious side effects, including ovarian failure and infertility, in women of reproductive age. Infertility following cancer treatment can have significant adverse effects on the quality of life. However, established methods for fertility preservation, including embryo or oocyte cryopreservation, are not always suitable for female cancer patients because of complicated individual conditions and treatment methods. Ovarian tissue cryopreservation and transplantation is a promising option for fertility preservation in pre-pubertal girls and adult patients with cancer who require immediate treatment, or who are not eligible to undergo ovarian stimulation. This review introduces various methods and strategies to improve ovarian tissue cryopreservation and transplantation outcomes, to help patients and clinicians choose the best option when considering the potential complexity of a patient's situation. Effective multidisciplinary oncofertility strategies, involving the inclusion of a highly skilled and experienced oncofertility team that considers cryopreservation methods, thawing processes and devices, surgical procedures for transplantation, and advances in technologies, are necessary to provide high-quality care to a cancer patient.
Collapse
Affiliation(s)
- Sanghoon Lee
- Moores Cancer Center, University of California San Diego, San Diego, CA, USA.
- Department of Obstetrics and Gynecology, Korea University College of Medicine, 73, Inchon-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea.
| | - Sinan Ozkavukcu
- Center for Assisted Reproduction, Department of Obstetrics and Gynecology, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Seung-Yup Ku
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
22
|
Decellularization Methods of Ovary in Tissue Engineering. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1345:129-139. [PMID: 34582019 DOI: 10.1007/978-3-030-82735-9_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The ovaries or female gonads are situated in the ovarian fossa of the abdominal cavity. These are paired, almond-shaped organs measuring about 3.5 cm long and 1.5 cm thick and exist out of a central medullary zone and a peripheral cortex that are enclosed in a fibrous capsule called the tunica albuginea. The ovaries serve 2 main functions, the first one being the production of female gametes called oocytes (oogenesis). Interestingly, the number of primary oocytes that reside in the ovary is determined at birth. About 400 oocyte-containing follicles successfully go through all the developmental stages from this limited pool during folliculogenesis throughout the female reproductive life. In this process, primordial follicles grow and advance until forming a mature or Graafian follicle; during ovulation, secondary oocytes are released and the remaining follicular wall collapses and forms the highly vascularized corpus luteum or luteal gland. This ovarian cycle is regulated by several hormones secreted from the adenohypophysis and lasts about 28 days. During this cycle, the ovaries also serve as endocrine glands and produce female sex hormones such as estrogens and progesterone (steroidogenesis), influencing the growth and development of tissues sensitive to these hormones such as the endometrium. Hence, the endometrial cycle goes synchronized with the ovarian cycle.
Collapse
|
23
|
Is fertility preservation a necessity before endometriosis surgical treatment? GINECOLOGIA.RO 2021. [DOI: 10.26416/gine.31.1.2021.4329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
24
|
Calagna G, Della Corte L, Giampaolino P, Maranto M, Perino A. Endometriosis and strategies of fertility preservation: a systematic review of the literature. Eur J Obstet Gynecol Reprod Biol 2020; 254:218-225. [PMID: 33011504 DOI: 10.1016/j.ejogrb.2020.09.045] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 12/16/2022]
Abstract
Endometriosis is a common chronic inflammatory disorder, often causing both pain and infertility. It is estimated that 25-50 % of patients undergoing fertility treatments have had endometriosis as it involves an impairment of the ovarian reserve. For these reasons, endometriosis has been highlighted as a condition that may require a fertility preservation procedure, while being benign in nature. The aim of this review is to summarize the current evidence on fertility preservation techniques for patients affected by endometriosis, focusing on the main characteristics of the different approaches. A systematic review of literature was performed by searching in the main electronic databases (MEDLINE, EMBASE, Web of Science, Scopus, ClinicalTrial.gov, OVID and Cochrane Library), from their inception to February 2020 for studies testing fertility preservation (FP) techniques. Only scientific publications in English were included. Risk of Bias Assessment was performed. Eight articles were included in the study: 3 case reports (one paper reporting 2 cases), 1 prospective longitudinal analysis with only 1 reported case with criteria for inclusion in the review, 2 retrospective observational studies and 2 retrospective cohort studies. The results of our analysis showed embryo (1 article), oocyte (4 articles) and ovarian tissue cryopreservation (3 articles) as preservation methods proposed in international literature for patients with endometriosis. Only few authors reported details on successive outcomes. After ovarian tissue transplantation, 1 pregnancy with IVF and 1 case of endocrine function/ovulation recovery were described; six of 16 endometriotic patients, who underwent embryo cryopreservation, experienced livebirths after successive embryo-transfer; in a large series of 485 patients performing oocyte cryopreservation, an overall cumulative live-birth rate/patient of 46.4 % (225 babies) was reported. Based on the risk of premature ovarian failure related to endometriosis, the offer of FP techniques to these patients has significantly increased, as well as the reported experiences in recent medical literature. However, further studies concerning risks, benefits, effectiveness and cost-effectiveness are needed.
Collapse
Affiliation(s)
- Gloria Calagna
- Department of Obstetrics and Gynecology, "Villa Sofia Cervello" University of Palermo, Palermo, Italy
| | - Luigi Della Corte
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, University of Naples Federico II, Naples, Italy.
| | | | - Marianna Maranto
- Department of Obstetrics and Gynecology, "Villa Sofia Cervello" University of Palermo, Palermo, Italy
| | - Antonino Perino
- Department of Obstetrics and Gynecology, "Villa Sofia Cervello" University of Palermo, Palermo, Italy
| |
Collapse
|
25
|
Barberet J, Barry F, Choux C, Guilleman M, Karoui S, Simonot R, Bruno C, Fauque P. What impact does oocyte vitrification have on epigenetics and gene expression? Clin Epigenetics 2020; 12:121. [PMID: 32778156 PMCID: PMC7418205 DOI: 10.1186/s13148-020-00911-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 07/21/2020] [Indexed: 02/07/2023] Open
Abstract
Children conceived by assisted reproductive technologies (ART) have a moderate risk for a number of adverse events and conditions. The question whether this additional risk is associated with specific procedures used in ART or whether it is related to the intrinsic biological factors associated with infertility remains unresolved. One of the main hypotheses is that laboratory procedures could have an effect on the epigenome of gametes and embryos. This suspicion is linked to the fact that ART procedures occur precisely during the period when there are major changes in the organization of the epigenome. Oocyte freezing protocols are generally considered safe; however, some evidence suggests that vitrification may be associated with modifications of the epigenetic marks. In this manuscript, after describing the main changes that occur during epigenetic reprogramming, we will provide current information regarding the impact of oocyte vitrification on epigenetic regulation and the consequences on gene expression, both in animals and humans. Overall, the literature suggests that epigenetic and transcriptomic profiles are sensitive to the stress induced by oocyte vitrification, and it also underlines the need to improve our knowledge in this field.
Collapse
Affiliation(s)
- Julie Barberet
- CHU Dijon Bourgogne, Laboratoire de Biologie de la Reproduction, CECOS, 14 rue Gaffarel, 21079 Dijon Cedex, France
| | - Fatima Barry
- CHU Dijon Bourgogne, Laboratoire de Biologie de la Reproduction, CECOS, 14 rue Gaffarel, 21079 Dijon Cedex, France
| | - Cécile Choux
- Gynécologie-Obstétrique, CHU Dijon Bourgogne, 14 rue Gaffarel, 21079 Dijon Cedex, France
| | - Magali Guilleman
- CHU Dijon Bourgogne, Laboratoire de Biologie de la Reproduction, CECOS, 14 rue Gaffarel, 21079 Dijon Cedex, France
| | - Sara Karoui
- CHU Dijon Bourgogne, Laboratoire de Biologie de la Reproduction, CECOS, 14 rue Gaffarel, 21079 Dijon Cedex, France
| | - Raymond Simonot
- CHU Dijon Bourgogne, Laboratoire de Biologie de la Reproduction, CECOS, 14 rue Gaffarel, 21079 Dijon Cedex, France
| | - Céline Bruno
- CHU Dijon Bourgogne, Laboratoire de Biologie de la Reproduction, CECOS, 14 rue Gaffarel, 21079 Dijon Cedex, France
| | - Patricia Fauque
- CHU Dijon Bourgogne, Laboratoire de Biologie de la Reproduction, CECOS, 14 rue Gaffarel, 21079 Dijon Cedex, France
| |
Collapse
|
26
|
Fouks Y, Hamilton E, Cohen Y, Hasson J, Kalma Y, Azem F. In-vitro maturation of oocytes recovered during cryopreservation of pre-pubertal girls undergoing fertility preservation. Reprod Biomed Online 2020; 41:869-873. [PMID: 32843309 DOI: 10.1016/j.rbmo.2020.07.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 07/14/2020] [Accepted: 07/16/2020] [Indexed: 11/28/2022]
Abstract
RESEARCH QUESTION In-vitro maturation (IVM) of oocytes recovered during ovarian tissue cryopreservation (OTC) is often practised, although it is still considered experimental. To date, only a few studies have examined the success of this maturation process in pre-menarche girls. The aim of this study was to examine the outcomes of IVM of oocytes recovered during OTC in pre-menarche patients scheduled for onco-therapy. DESIGN A retrospective cohort study in a tertiary university-affiliated hospital. A total of 93 patients aged 0-25 years who underwent OTC as part of onco-fertility preservation between 2007 and 2019 were included in the study. Oocytes were recovered from the medium after OTC and matured over 48 h. Oocyte development and maturation rate were recorded and compared between different age groups. RESULTS Patient's age was not correlated linearly with the total number of mature oocytes R = 0.2. The absolute maturation rate in post-menarche and pre-menarche patients differed significantly (38.0% versus 25.3%, respectively; P > 0.001), whereas the degeneration rate of the oocytes did not (39.8% versus 33.5%; P = 0.167). The pre-menarche group had significantly lower mean number of metaphase II oocytes compared with the post-menarche group (2.8 [±2.3] versus 5.6 [±4.6]; P = 0.01; 95% CI -4.62 to -0.46). Oocytes recovered from patients aged 1-5 years demonstrated low maturation rate. CONCLUSIONS Oocytes recovered from pre-menarche girls, and especially those younger than the age of 5 years who undergo fertility preservation, have a lower chance of reaching maturity in IVM compared with older women. This may indicate a need for alternative methods for preserving fertility in these young patients.
Collapse
Affiliation(s)
- Yuval Fouks
- Lis Maternity Hospital, Department of Obstetrics and Gynecology, Fertility Division, Tel-Aviv Medical Center affiliated to the Sackler Faculty of Medicine, 6 Weizmann Street, Tel-Aviv 6423906, Israel.
| | - Emily Hamilton
- Lis Maternity Hospital, Department of Obstetrics and Gynecology, Fertility Division, Tel-Aviv Medical Center affiliated to the Sackler Faculty of Medicine, 6 Weizmann Street, Tel-Aviv 6423906, Israel
| | - Yoni Cohen
- Lis Maternity Hospital, Department of Obstetrics and Gynecology, Fertility Division, Tel-Aviv Medical Center affiliated to the Sackler Faculty of Medicine, 6 Weizmann Street, Tel-Aviv 6423906, Israel
| | - Joseph Hasson
- Lis Maternity Hospital, Department of Obstetrics and Gynecology, Fertility Division, Tel-Aviv Medical Center affiliated to the Sackler Faculty of Medicine, 6 Weizmann Street, Tel-Aviv 6423906, Israel
| | - Yael Kalma
- Lis Maternity Hospital, Department of Obstetrics and Gynecology, Fertility Division, Tel-Aviv Medical Center affiliated to the Sackler Faculty of Medicine, 6 Weizmann Street, Tel-Aviv 6423906, Israel
| | - Foad Azem
- Lis Maternity Hospital, Department of Obstetrics and Gynecology, Fertility Division, Tel-Aviv Medical Center affiliated to the Sackler Faculty of Medicine, 6 Weizmann Street, Tel-Aviv 6423906, Israel
| |
Collapse
|
27
|
Alhadad LJ, Harisa GI, Alanazi FK. Design and encapsulation of anticancer dual HSP27 and HER2 inhibitor into low density lipoprotein to target ovarian cancer cells. Saudi Pharm J 2020; 28:387-396. [PMID: 32273796 PMCID: PMC7132616 DOI: 10.1016/j.jsps.2020.01.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 01/29/2020] [Indexed: 01/13/2023] Open
Abstract
Tumor cells overexpress low-density lipoprotein (LDL) receptors (LDL-r). Hence, LDL is proposed as a targeting shuttle of anticancer drugs. Therefore, the objective of this study was to synthesize a dual inhibitor of heat shock protein 27 (HSP27) and human epidermal growth factor receptor 2 (HER2) conjugated with cholesterol and encapsulated into LDL for selective targeting of ovarian cancer cells. In the present study, the anticancer agent and its cholesterol conjugate were successfully prepared and characterized physically for color, shape, and melting point. Moreover, the compounds were chemically characterized for 1H NMR and 13C NMR spectra using FTIR and LCMS/MS. Our results revealed that the prepared anticancer agent and its cholesterol conjugate elicited dual HSP27 and HER2 inhibition, as confirmed using western blotting. The anticancer agent (compound D) entered cells and targeted the HSP27 function, thereby reducing HER2 expression. However, a cholesterol-conjugated anticancer agent (compound F) had high cellular uptake and inhibited the growth of SKOV3 cells after encapsulation into LDL. The obtained results concluded that the design of an LDL-encapsulated cholesterol-conjugated HSP27-HER2 dual inhibitor may be a promising approach to realize specific targeted achieve killing of ovarian cancer.
Collapse
Affiliation(s)
- Laila J Alhadad
- Department of Chemistry, College of Science, Kuwait University, Safat, Kuwait
| | - Gamaleldin I Harisa
- Kayyali Chair for Pharmaceutical Industry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.,Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.,Department of Biochemistry, College of Pharmacy, Al-Azhar University, Nasr City, Cairo, Egypt
| | - Fars K Alanazi
- Kayyali Chair for Pharmaceutical Industry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.,Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
28
|
Corkum KS, Rhee DS, Wafford QE, Demeestere I, Dasgupta R, Baertschiger R, Malek MM, Aldrink JH, Heaton TE, Weil BR, Madonna MB, Lautz TB. Fertility and hormone preservation and restoration for female children and adolescents receiving gonadotoxic cancer treatments: A systematic review. J Pediatr Surg 2019; 54:2200-2209. [PMID: 30773394 DOI: 10.1016/j.jpedsurg.2018.12.021] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 12/01/2018] [Accepted: 12/07/2018] [Indexed: 10/27/2022]
Abstract
BACKGROUND/PURPOSE The purpose of this systematic review by the American Pediatric Surgical Cancer Committee was to summarize evidence from the current medical literature regarding fertility restoration and hormone replacement for female children and adolescents treated with gonadotoxic treatments. METHODS Using PRISMA guidelines, questions were addressed by searching Medline, Cochrane, Embase Central and National clearing house databases using relevant search terms. Eligible studies included those that addressed ovarian tissue cryopreservation (OTC), oocyte harvest, ovarian transposition, and ovarian tissue auto-transplantation for females under the age of 20. Four reviewers independently screened studies for eligibility, extracted data and assessed the risk of bias. Study outcomes were summarized in a narrative synthesis. RESULTS Two thousand two hundred seventy-six studies were identified by database search and manual review and 2185 were eliminated based on defined exclusion criteria. Ninety-one studies served as the basis for the systematic review. There were 1019 patients who underwent OTC with ages ranging from 0.4 to 20.4 years old, with 298 under the age of 13. Twenty patients aged 13-20 years old underwent successful oocyte harvest. Thirty-seven children underwent ovarian transposition as a means of fertility preservation. Eighteen patients underwent auto-transplantation of thawed ovarian cortical tissue that was harvested before the age of 21 years resulting in 10 live births. CONCLUSIONS Clinically accepted and experimental fertility preservation options such as OTC, oocyte cryopreservation, and ovarian transposition are available to females aged 20 years and younger who are at risk for premature ovarian insufficiency and infertility due to gonadotoxic treatments. There is a large cohort of pediatric-aged patients, with a wide variety of diagnoses and treatments, who have undergone fertility preservation. Currently, fertility and hormone restoration experience for patients who were 20- years of age or younger at the time of fertility preservation remains limited. LEVEL OF EVIDENCE IV.
Collapse
Affiliation(s)
- Kristine S Corkum
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Division of Pediatric Surgery, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Daniel S Rhee
- Department of Surgery, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Q Eileen Wafford
- Galter Health Sciences Library, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Isabelle Demeestere
- Research Laboratory in Human Reproduction, Université Libre de Bruxelles, (ULB), Brussels, Belgium
| | | | - Reto Baertschiger
- Division of Pediatric Surgery, Department of Surgery, Dartmouth Hitchcock Medical Center, Lebanon, NH, USA
| | - Marcus M Malek
- Division of Pediatric General and Thoracic Surgery, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Jennifer H Aldrink
- Division of Pediatric Surgery, Nationwide Children's Hospital, Department of Surgery, The Ohio State University College of Medicine, Columbus, OH
| | - Todd E Heaton
- Division of Pediatric Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Brent R Weil
- Department of Surgery, Boston Children's Hospital, Boston, MA
| | | | - Timothy B Lautz
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Division of Pediatric Surgery, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA.
| |
Collapse
|
29
|
Chapron C, Marcellin L, Borghese B, Santulli P. Rethinking mechanisms, diagnosis and management of endometriosis. Nat Rev Endocrinol 2019; 15:666-682. [PMID: 31488888 DOI: 10.1038/s41574-019-0245-z] [Citation(s) in RCA: 495] [Impact Index Per Article: 99.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/18/2019] [Indexed: 02/08/2023]
Abstract
Endometriosis is a chronic inflammatory disease defined as the presence of endometrial tissue outside the uterus, which causes pelvic pain and infertility. This disease should be viewed as a public health problem with a major effect on the quality of life of women as well as being a substantial economic burden. In light of the considerable progress with diagnostic imaging (for example, transvaginal ultrasound and MRI), exploratory laparoscopy should no longer be used to diagnose endometriotic lesions. Instead, diagnosis of endometriosis should be based on a structured process involving the combination of patient interviews, clinical examination and imaging. Notably, a diagnosis of endometriosis often leads to immediate surgery. Therefore, rethinking the diagnosis and management of endometriosis is warranted. Instead of assessing endometriosis on the day of the diagnosis, gynaecologists should consider the patient's 'endometriosis life'. Medical treatment is the first-line therapeutic option for patients with pelvic pain and no desire for immediate pregnancy. In women with infertility, careful consideration should be made regarding whether to provide assisted reproductive technologies prior to performing endometriosis surgery. Modern endometriosis management should be individualized with a patient-centred, multi-modal and interdisciplinary integrated approach.
Collapse
Affiliation(s)
- Charles Chapron
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France.
- Assistance Publique - Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, Department of Gynecology Obstetrics II and Reproductive Medicine, Paris, France.
- Department 'Development, Reproduction and Cancer', Institut Cochin, INSERM U1016, Université Paris Descartes, Paris, France.
| | - Louis Marcellin
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
- Assistance Publique - Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, Department of Gynecology Obstetrics II and Reproductive Medicine, Paris, France
- Department 'Development, Reproduction and Cancer', Institut Cochin, INSERM U1016, Université Paris Descartes, Paris, France
| | - Bruno Borghese
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
- Assistance Publique - Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, Department of Gynecology Obstetrics II and Reproductive Medicine, Paris, France
- Department 'Development, Reproduction and Cancer', Institut Cochin, INSERM U1016, Université Paris Descartes, Paris, France
| | - Pietro Santulli
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
- Assistance Publique - Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, Department of Gynecology Obstetrics II and Reproductive Medicine, Paris, France
- Department 'Development, Reproduction and Cancer', Institut Cochin, INSERM U1016, Université Paris Descartes, Paris, France
| |
Collapse
|
30
|
Lantsberg D, Fernando S, Cohen Y, Rombauts L. The Role of Fertility Preservation in Women with Endometriosis: A Systematic Review. J Minim Invasive Gynecol 2019; 27:362-372. [PMID: 31546067 DOI: 10.1016/j.jmig.2019.09.780] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 09/01/2019] [Accepted: 09/16/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To summarize the available evidence concerning fertility preservation techniques in the context of women with endometriosis. DATA SOURCES We searched for studies published between 1984 and 2019 on endometriosis and Assisted Reproductive Technology outcomes. We searched MEDLINE and PubMed and performed a manual search of reference lists within identified studies. METHODS OF STUDY SELECTION A total of 426 articles were identified, and 7 studies were eligible to be included for the systematic review. We included all published studies, excluding reviews, case reports, and animal studies. TABULATION, INTEGRATION, AND RESULTS Despite a significant increase in the number of studies addressing fertility preservation over the study period, we found a relative lack of evidence addressing the use of fertility preservation techniques in women with endometriosis. The studies identified included 2 case reports, 1 histological science study, and 4 retrospective cohort studies. CONCLUSION Women with endometriosis may benefit from fertility preservation techniques. However, there currently is a paucity of data in this population, especially when compared with other indications for fertility preservation. Although much knowledge can be translated from the oncofertility discipline, we have identified and discussed endometriosis-related changes to ovarian reserve and oocyte health that justify further well-designed research to confirm that fertility preservation outcomes are similar for women with endometriosis.
Collapse
Affiliation(s)
- Daniel Lantsberg
- Department of Obstetrics, Gynecology and Fertility, Lis Maternity Hospital (Drs. Lantsberg and Cohen); Sourasky Tel-Aviv Medical Center (Drs. Lantsberg and Cohen), and Sackler Faculty of Medicine (Drs. Lantsberg and Cohen), Tel-Aviv University, Israel.
| | - Shavi Fernando
- Department of Obstetrics and Gynecology, Monash University, Clayton (Prof. Rombauts and Dr. Fernando), Australia; Women's Health, Monash Health, Clayton (Prof. Rombauts and Dr. Fernando), Australia
| | - Yoni Cohen
- Sourasky Tel-Aviv Medical Center (Drs. Lantsberg and Cohen), and Sackler Faculty of Medicine (Drs. Lantsberg and Cohen), Tel-Aviv University, Israel
| | - Luk Rombauts
- Department of Obstetrics and Gynecology, Monash University, Clayton (Prof. Rombauts and Dr. Fernando), Australia; Women's Health, Monash Health, Clayton (Prof. Rombauts and Dr. Fernando), Australia; Monash IVF Group, Richmond (Prof Rombauts), Australia
| |
Collapse
|
31
|
Advances in fertility-preservation surgery: navigating new frontiers. Fertil Steril 2019; 112:438-445. [DOI: 10.1016/j.fertnstert.2019.06.029] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 06/11/2019] [Accepted: 06/19/2019] [Indexed: 11/21/2022]
|
32
|
Rivas Leonel EC, Lucci CM, Amorim CA. Cryopreservation of Human Ovarian Tissue: A Review. Transfus Med Hemother 2019; 46:173-181. [PMID: 31244585 PMCID: PMC6558345 DOI: 10.1159/000499054] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 02/01/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Cryopreservation of human ovarian tissue has been increasingly applied worldwide to safeguard fertility in cancer patients, notably in young girls and women who cannot delay the onset of their treatment. Moreover, it has been proposed to patients with benign pathologies with a risk of premature ovarian insufficiency. So far, more than 130 live births have been reported after transplantation of cryopreserved ovarian tissue, and almost all patients recovered their ovarian function after tissue reimplantation. SUMMARY This review aims to summarize the recent results described in the literature regarding human ovarian tissue cryopreservation in terms of methods and main results obtained so far. To cryopreserve human ovarian tissue, most studies describe a slow freezing/rapid thawing protocol, which is usually an adaptation of a protocol developed for sheep ovarian tissue. Since freezing has been shown to have a deleterious effect on ovarian stroma and granulosa cells, various research groups have been vitrifying ovarian tissue. Despite promising results, only 2 babies have been born after transplantation of vitrified/warmed ovarian tissue. Optimization of both cryopreservation strategies as well as thawing/warming protocols is therefore necessary to improve the survival of follicles in cryopreserved ovarian tissue. KEY MESSAGES Human ovarian tissue cryopreservation has been successfully applied worldwide to preserve fertility in patients with malignant or nonmalignant pathologies that have a detrimental effect on fertility. Human ovarian tissue cryopreservation could also be applied as an alternative to postpone pregnancy or menopause in healthy women. Slow freezing and vitrification procedures have been applied to cryopreserve human ovarian tissue, but both alternatives require optimization.
Collapse
Affiliation(s)
- Ellen Cristina Rivas Leonel
- Institut de Recherche Expérimentale et Clinique, Pôle de Recherche en Gynécologie, Université Catholique de Louvain, Brussels, Belgium
- Institute of Biosciences, Department of Biology, Humanities and Exact Sciences, São Paulo State University, São José do Rio Preto, Brazil
| | - Carolina M. Lucci
- Institute of Biological Sciences, Department of Physiology, University of Brasília, Brasília, Brazil
| | - Christiani A. Amorim
- Institut de Recherche Expérimentale et Clinique, Pôle de Recherche en Gynécologie, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
33
|
Arfi A, Owen C, Zilberman S, Ferrier C, Boudy AS, Bendifallah S, Darai E. [How do I…an intra-ovarian ovarian cortex transplant by laparoscopy]. ACTA ACUST UNITED AC 2019; 47:603-605. [PMID: 31003019 DOI: 10.1016/j.gofs.2019.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Indexed: 10/27/2022]
Affiliation(s)
- A Arfi
- Département de gynécologie et obstétrique, hôpital Tenon, université Pierre-et-Marie-Curie, Assistance publique des Hôpitaux de Paris (AP-HP), 4, rue de la chine, 75020 Paris 6, France.
| | - C Owen
- Département de gynécologie et obstétrique, hôpital Tenon, université Pierre-et-Marie-Curie, Assistance publique des Hôpitaux de Paris (AP-HP), 4, rue de la chine, 75020 Paris 6, France
| | - S Zilberman
- Département de gynécologie et obstétrique, hôpital Tenon, université Pierre-et-Marie-Curie, Assistance publique des Hôpitaux de Paris (AP-HP), 4, rue de la chine, 75020 Paris 6, France
| | - C Ferrier
- Département de gynécologie et obstétrique, hôpital Tenon, université Pierre-et-Marie-Curie, Assistance publique des Hôpitaux de Paris (AP-HP), 4, rue de la chine, 75020 Paris 6, France
| | - A-S Boudy
- Département de gynécologie et obstétrique, hôpital Tenon, université Pierre-et-Marie-Curie, Assistance publique des Hôpitaux de Paris (AP-HP), 4, rue de la chine, 75020 Paris 6, France
| | - S Bendifallah
- Département de gynécologie et obstétrique, hôpital Tenon, université Pierre-et-Marie-Curie, Assistance publique des Hôpitaux de Paris (AP-HP), 4, rue de la chine, 75020 Paris 6, France; Inserm UMR_S_707, « Epidémiologie, Information des Systèmes, Modèles », université Pierre-et-Marie-Curie, 75012 Paris 6, France; Groupe de Recherche Clinique 6 (GRC6-UPMC) : Centre Expert En Endométriose (C3E), 75020 Paris, France; UMR_S938, université Pierre-et-Marie-Curie, Paris 6, France
| | - E Darai
- Département de gynécologie et obstétrique, hôpital Tenon, université Pierre-et-Marie-Curie, Assistance publique des Hôpitaux de Paris (AP-HP), 4, rue de la chine, 75020 Paris 6, France; Inserm UMR_S_707, « Epidémiologie, Information des Systèmes, Modèles », université Pierre-et-Marie-Curie, 75012 Paris 6, France; Groupe de Recherche Clinique 6 (GRC6-UPMC) : Centre Expert En Endométriose (C3E), 75020 Paris, France; UMR_S938, université Pierre-et-Marie-Curie, Paris 6, France
| |
Collapse
|
34
|
Ubaldi FM, Cimadomo D, Vaiarelli A, Fabozzi G, Venturella R, Maggiulli R, Mazzilli R, Ferrero S, Palagiano A, Rienzi L. Advanced Maternal Age in IVF: Still a Challenge? The Present and the Future of Its Treatment. Front Endocrinol (Lausanne) 2019; 10:94. [PMID: 30842755 PMCID: PMC6391863 DOI: 10.3389/fendo.2019.00094] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 02/01/2019] [Indexed: 12/13/2022] Open
Abstract
Advanced maternal age (AMA; >35 year) is associated with a decline in both ovarian reserve and oocyte competence. At present, no remedies are available to counteract the aging-related fertility decay, however different therapeutic approaches can be offered to women older than 35 year undergoing IVF. This review summarizes the main current strategies proposed for the treatment of AMA: (i) oocyte cryopreservation to conduct fertility preservation for medical reasons or "social freezing" for non-medical reasons, (ii) personalized controlled ovarian stimulation to maximize the exploitation of the ovarian reserve in each patient, (iii) enhancement of embryo selection via blastocyst-stage preimplantation genetic testing for aneuploidies and frozen single embryo transfer, or (iv) oocyte donation in case of minimal/null residual chance of pregnancy. Future strategies and tools are in the pipeline that might minimize the risks of AMA through non-invasive approaches for embryo selection (e.g., molecular analyses of leftover products of IVF, such as spent culture media). These are yet challenging but potentially ground-breaking perspectives promising a lower clinical workload with a higher cost-effectiveness. We also reviewed emerging experimental therapeutic approaches to attempt at restoring maternal reproductive potential, e.g., spindle-chromosomal complex, pronuclear or mitochondrial transfer, and chromosome therapy. In vitro generation of gametes is also an intriguing challenge for the future. Lastly, since infertility is a social issue, social campaigns, and education among future generations are desirable to promote the awareness of the impact of age and lifestyle habits upon fertility. This should be a duty of the clinical operators in this field.
Collapse
Affiliation(s)
- Filippo Maria Ubaldi
- Clinica Valle Giulia, G.en.e.r.a. Centers for Reproductive Medicine, Rome, Italy
| | - Danilo Cimadomo
- Clinica Valle Giulia, G.en.e.r.a. Centers for Reproductive Medicine, Rome, Italy
- *Correspondence: Danilo Cimadomo
| | - Alberto Vaiarelli
- Clinica Valle Giulia, G.en.e.r.a. Centers for Reproductive Medicine, Rome, Italy
| | - Gemma Fabozzi
- Clinica Valle Giulia, G.en.e.r.a. Centers for Reproductive Medicine, Rome, Italy
| | - Roberta Venturella
- Department of Experimental and Clinical Medicine, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Roberta Maggiulli
- Clinica Valle Giulia, G.en.e.r.a. Centers for Reproductive Medicine, Rome, Italy
| | - Rossella Mazzilli
- Clinica Valle Giulia, G.en.e.r.a. Centers for Reproductive Medicine, Rome, Italy
- Andrology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Susanna Ferrero
- Clinica Valle Giulia, G.en.e.r.a. Centers for Reproductive Medicine, Rome, Italy
| | - Antonio Palagiano
- Department of Gynecological, Obstetrical and Reproductive Sciences, University of Campania Luigi Vanvitelli, Caserta, Italy
| | - Laura Rienzi
- Clinica Valle Giulia, G.en.e.r.a. Centers for Reproductive Medicine, Rome, Italy
| |
Collapse
|
35
|
Guzel Y, Bildik G, Dilege E, Oktem O. Sphingosine-1-phosphate reduces atresia of primordial follicles occurring during slow-freezing and thawing of human ovarian cortical strips. Mol Reprod Dev 2018; 85:858-864. [PMID: 29995320 DOI: 10.1002/mrd.23043] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 07/07/2018] [Indexed: 11/07/2022]
Abstract
We aimed in this study to explore if sphingosine-1-phosphate (S1P) reduces apoptosis of primordial follicles during cryopreservation of human ovarian cortical samples. Ovarian cortical tissue fragments obtained from young patients who underwent laparoscopic excision of benign ovarian cysts were used for the experiments. The samples were slow-frozen and thawed with and without S1P at 200 and 400 μM, cultured for 1 day, and then were fixed and processed for both histomorphological assessment and detection of apoptosis with immunohistochemistry using apoptosis marker cleaved caspase-3. Follicle counts were expressed as the mean number of follicles per mm2 . The mean number of primordial follicles and in vitro estradiol (E2) and anti-mullerian hormone (AMH) production of the slow-frozen and thawed samples were significantly reduced compared with fresh unfrozen samples. S1P treatment at 400 μM but not 200 μM concentration resulted in a significant increase in the number of surviving primordial follicles and in vitro E2 and AMH productions of the samples compared with their counterparts slow-frozen without S1P. We found that that there was a significant decrease in the number of primordial follicles with their oocytes stained positive for cleaved caspase-3 in the slow-frozen samples S1P 400 μM in comparison with the samples slow-frozen without S1P. These results suggest that S1P may ameliorate follicle atresia occurring in human ovarian cortical samples during cryopreservation.
Collapse
Affiliation(s)
- Yilmaz Guzel
- Department of Obstetrics and Gynecology, Istanbul Aydin University School of Medicine, Istanbul, Turkey
| | - Gamze Bildik
- Cellular and Molecular Medicine, Graduate School of Health Sciences, Koc University, Istanbul, Turkey
| | - Ece Dilege
- Department of Surgery, Koc University School of Medicine, Istanbul, Turkey
| | - Ozgur Oktem
- Cellular and Molecular Medicine, Graduate School of Health Sciences, Koc University, Istanbul, Turkey.,Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, School of Medicine, Koc University, Istanbul, Turkey
| |
Collapse
|
36
|
Nikiforov D, Russo V, Nardinocchi D, Bernabò N, Mattioli M, Barboni B. Innovative multi-protectoral approach increases survival rate after vitrification of ovarian tissue and isolated follicles with improved results in comparison with conventional method. J Ovarian Res 2018; 11:65. [PMID: 30086787 PMCID: PMC6081856 DOI: 10.1186/s13048-018-0437-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/26/2018] [Indexed: 12/22/2022] Open
Abstract
Background In recent years, autotransplantation of cryopreserved ovarian tissue became a promising approach to preserve female fertility. The slow freezing is the most effective technique which resulted in greater live birth incidence so far. Despite that, interest to vitrification of the ovarian tissue is swiftly growing, thereby undermining the necessity for further improvements in the technique. In present study, we evaluated possibilities to increase follicle survival rates adopting innovative multi-protectoral vitrification protocols, applied to the slivers of ovarian cortex or isolated early-antral follicles, frozen individually. These experimental protocols have been compared with with validated vitrification and slow freezing ones, clinically used for female fertility preservation. Results The results showed that third tested variation of experimental vitrification protocol, with four cryoprotectants in relatively low concentrations and applied to pieces of ovarian tissue at 0 °C during equilibration, increased survival rate of ovine ovarian tissue and improved results in comparison with conventional vitrification method. This variation of experimental protocol showed significant increase in percentage of follicles with good morphology (69,3%) in comparison with only commercially available vitrification protocol for ovarian tissue (62,1%). Morphology results were confirmed by TUNEL assay. Analysis of estradiol and progesterone production by cultured individual follicles after freezing/thawing revealed that steroids secretion remained significantly higher after multi-protectoral vitrification and slow freezing protocol, when follicles after standard vitrification protocol demonstrated decline in steroidogenic activity. Conclusions The multi-protectoral approach represents a workable solution to improve vitrification outcome on ovarian tissue and isolated follicles. The reduction of individual cryoprotectants concentrations, while maintaining their sufficient cumulative level in the final freezing solution, helps to increase efficiency of the procedure. Moreover, equilibration with lower temperatures helped to decrease even further the toxic effects of cryoprotectants and preserve original quality of ovarian tissue. Therefore, multi-protectoral vitrification can be suggested as an improved method for the clinical cryopreservation of ovarian tissue.
Collapse
Affiliation(s)
- Dmitry Nikiforov
- Faculty of Bioscience, Unit of Basic and Applied Biosciences, University of Teramo, 64100, via R. Balzarini 1, Teramo, Italy.
| | - Valentina Russo
- Faculty of Bioscience, Unit of Basic and Applied Biosciences, University of Teramo, 64100, via R. Balzarini 1, Teramo, Italy
| | - Delia Nardinocchi
- Faculty of Bioscience, Unit of Basic and Applied Biosciences, University of Teramo, 64100, via R. Balzarini 1, Teramo, Italy
| | - Nicola Bernabò
- Faculty of Bioscience, Unit of Basic and Applied Biosciences, University of Teramo, 64100, via R. Balzarini 1, Teramo, Italy
| | - Mauro Mattioli
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise "Giuseppe Caporale" (IZSAM), 64100, Teramo, Italy
| | - Barbara Barboni
- Faculty of Bioscience, Unit of Basic and Applied Biosciences, University of Teramo, 64100, via R. Balzarini 1, Teramo, Italy
| |
Collapse
|
37
|
Ovarian tissue cryopreservation and transplantation in patients with cancer. Obstet Gynecol Sci 2018; 61:431-442. [PMID: 30018897 PMCID: PMC6046360 DOI: 10.5468/ogs.2018.61.4.431] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 10/11/2017] [Accepted: 10/20/2017] [Indexed: 12/31/2022] Open
Abstract
Chemotherapy and radiotherapy improved survival rates of patients with cancer. However, they can cause ovarian failure and infertility in women of reproductive age. Infertility following cancer treatment is considered a major quality of life issue. Ovarian tissue cryopreservation and transplantation is an important option for fertility preservation in adult patients with cancer who need immediate chemotherapy or do not want to undergo ovarian stimulation. Ovarian tissue freezing is the only option for preserving the fertility of prepubertal patients with cancer. In a recent review, it was reported that frozen-thawed ovarian transplantation has lead to about 90 live births and the conception rate was about 30%. Endocrine function recovery was observed in 92.9% between 3.5 and 6.5 months after transplantation. Based on our review, ovarian tissue cryopreservation and transplantation may be carefully considered before cancer treatment in order to preserve fertility and endocrine function in young cancer survivors.
Collapse
|
38
|
Vinolas C, Raad J, Sonigo C, Sifer C, Sermondade N, Grynberg M. Medical techniques of fertility preservation in the male and female. J Visc Surg 2018; 155 Suppl 1:S3-S9. [PMID: 29784583 DOI: 10.1016/j.jviscsurg.2018.03.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Therapeutic advances in many medical fields have led to the need to consider patient quality of life after curative medico-surgical treatments for malignancy. Thus, it has become a major issue for young patients to preserve the ability to become "genetic" parents, with their own gametes. While the preservation of male fertility has been an established technique for more than 30 years, it is only in the last decade that progress in cryopreservation techniques has allowed surgeons to offer successful oocyte and ovarian tissue cryobanking. However, in addition to the still experimental nature of some fertility preservation techniques, this practice also raises many ethical and moral questions.
Collapse
Affiliation(s)
- C Vinolas
- Service de Médecine de la Reproduction & Préservation de la Fertilité, Hôpital Jean Verdier, Avenue du 14 Juillet, 93140 Bondy, France
| | - J Raad
- Service de Médecine de la Reproduction & Préservation de la Fertilité, Hôpital Jean Verdier, Avenue du 14 Juillet, 93140 Bondy, France
| | - C Sonigo
- Service de Médecine de la Reproduction & Préservation de la Fertilité, 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
| | - N Sermondade
- 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 & Préservation de la Fertilité, 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
|
39
|
Decanter C, d'Argent EM, Boujenah J, Poncelet C, Chauffour C, Collinet P, Santulli P. [Endometriosis and fertility preservation: CNGOF-HAS Endometriosis Guidelines]. ACTA ACUST UNITED AC 2018. [PMID: 29530556 DOI: 10.1016/j.gofs.2018.02.010] [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] [Indexed: 12/01/2022]
Abstract
Fertility preservation (FP) techniques are progressing rapidly these past few years thanks to the oocyte vitrification. Indication of FP techniques is now extended to non-oncological situation that may induce risk of premature ovarian failure. Ovarian endometriosis can lead to premature ovarian failure and further infertility due to the high risk of ovarian cysts recurrence and surgery. To date, there is no cohort study regarding FP and endometriosis as well as no recommendation. Our purpose is to review the arguments in favor of FP in this specific area and to elaborate strategies according to each clinical form.
Collapse
Affiliation(s)
- C Decanter
- Service d'assistance médicale à la procréation et de préservation de la fertilité, hôpital Jeanne-de-Flandre, CHRU de Lille, 1, rue Eugène Avinée, 59037 Lille cedex, France; EA 4308 gamétogenèse et qualité du gamète, CHRU de Lille, 59037 Lille cedex, France.
| | - E M d'Argent
- Service de gynécologie-obstétrique et médecine de la reproduction, CHU Tenon, AP-HP, 4, rue de la Chine, 75020 Paris, France; Université Pierre-et-Marie-Curie, 75005 Paris 6, France; Hôpital Tenon, GRC6-UPMC: centre expert en endométriose (C3E), 75020 Paris, France
| | - J Boujenah
- Service de gynécologie-obstétrique, CHU Bondy, avenue du 14-Juillet, 93140 Bondy, France; Centre médical du Château, 22, rue Louis-Besquel, 94300 Vincennes, France
| | - C Poncelet
- Service de gynécologie-obstétrique, CH Renée-Dubos, 6, avenue de l'Ile-de-France, 95300 Pontoise, France; Université Paris-13, Sorbonne-Paris-Cité, UFR-SMBH, 93022 Bobigny, France
| | - C Chauffour
- Service de gynécologie-obstétrique et reproduction humaine, CHU d'Estaing, 1, place Lucie-Aubrac, 63003 Clermont-Ferrand, France
| | - P Collinet
- Clinique de gynécologie, hôpital Jeanne-de-Flandre, CHRU Lille, 59000 Lille, France; Université Lille-Nord-de-France, 59000 Lille, France
| | - P Santulli
- Service de chirurgie gynécologie obstétrique 2 et médecine de la reproduction, CHU Cochin, AP-HP, 27, rue du Faubourg-Saint-Jacques, 75014 Paris, France; Équipe génomique, épigénétiques et physiopathologie de la reproduction, département développement, reproduction, cancer, Inserm U1016, université Paris-Descartes, Sorbonne-Paris-Cité, 12, rue de l'École-de-Médecine, 75270 Paris cedex 06, France
| |
Collapse
|
40
|
Transplantation of frozen-thawed ovarian tissue: an update on worldwide activity published in peer-reviewed papers and on the Danish cohort. J Assist Reprod Genet 2018; 35:561-570. [PMID: 29497953 DOI: 10.1007/s10815-018-1144-2] [Citation(s) in RCA: 168] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 02/19/2018] [Indexed: 02/07/2023] Open
Abstract
PURPOSE The purpose of the study is to review all peer-reviewed published reports of women receiving ovarian tissue transplantation (OTT) with frozen/thawed tissue (OTC) with respect to age, diagnosis, transplantation site, fertility outcome, and potential side effects, including data from all women in the Danish program. METHODS A systematic review of the literature was performed in PubMed combined with results from all patients who had received OTT in Denmark up to December 2017. RESULTS OTT has been reported from 21 different countries comprising a total of 360 OTT procedures in 318 women. In nine women, malignancy was diagnosed after OTT; none were considered to be directly caused by the OTT. Despite a potential under reporting of cancer recurrence, there is currently no evidence to suggest that OTT causes reseeding of the original cancer. Renewed ovarian endocrine function was reported in 95% of the women. Half of all children born following OTT resulted from natural conception, and newborns were reported to be healthy except for one neonate with a chromosome anomaly with a family disposition. Women who conceived after OTT were significantly younger than those who failed. CONCLUSION This study found no indications of sufficient numbers of malignant cells present in the ovarian tissue to cause recurrence of cancer after OTT. Further, it is unlikely that OTC affects the well-being of children born. OTC is now an established method of fertility preservation in Denmark with public reimbursement. The current data encourage that women who require gonadotoxic treatment should be offered an individual evaluation considering fertility preservation.
Collapse
|
41
|
Chaput L, Grémeau AS, Vorilhon S, Pons H, Chabrot C, Grèze V, Pouly JL, Brugnon F. Préservation de la fertilité en cancérologie. Bull Cancer 2018; 105:99-110. [DOI: 10.1016/j.bulcan.2017.11.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 11/06/2017] [Indexed: 11/17/2022]
|
42
|
Solomonov E, Marcus Braun N, Siman-Tov Y, Ben-Shachar I. Team preparation for human uterus transplantation: Autologous transplantation in sheep model. Clin Transplant 2017; 31. [DOI: 10.1111/ctr.13137] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2017] [Indexed: 12/31/2022]
Affiliation(s)
- Evgeny Solomonov
- General and Hepato-Biliary Surgery Department; Ziv Medical Center; Safed Israel
- Faculty of Medicine in the Galilee; Bar-Ilan University; Safed Israel
| | - Naama Marcus Braun
- Faculty of Medicine in the Galilee; Bar-Ilan University; Safed Israel
- Obstetrics & Gynecology Department; Ziv Medical Center; Safed Israel
| | - Yariv Siman-Tov
- Pre-Clinical Research Unit; Assaf Harofeh Medical Center; Tzrifin Israel
| | - Inbar Ben-Shachar
- Faculty of Medicine in the Galilee; Bar-Ilan University; Safed Israel
- Obstetrics & Gynecology Department; Ziv Medical Center; Safed Israel
| |
Collapse
|
43
|
Oktem O, Kim SS, Selek U, Schatmann G, Urman B. Ovarian and Uterine Functions in Female Survivors of Childhood Cancers. Oncologist 2017; 23:214-224. [PMID: 29158370 DOI: 10.1634/theoncologist.2017-0201] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 09/21/2017] [Indexed: 01/27/2023] Open
Abstract
Adult survivors of childhood cancers are more prone to developing poor reproductive and obstetrical outcomes than their siblings and the general population as a result of previous exposure to chemotherapy and radiation during childhood. Chemotherapy drugs exert cytotoxic effects systemically and therefore can damage the ovaries, leading to infertility, premature ovarian failure, and, to a lesser extent, spontaneous abortions. They have very limited or no deleterious effects on the uterus that can be recognized clinically. By contrast, radiation is detrimental to both the ovaries and the uterus, thereby causing a greater magnitude of adverse effects on the female reproductive function. These include infertility, premature ovarian failure, miscarriage, fetal growth restrictions, perinatal deaths, preterm births, delivery of small-for-gestational-age infants, preeclampsia, and abnormal placentation. Regrettably, the majority of these adverse outcomes arise from radiation-induced uterine injury and are reported at higher incidence in the adult survivors of childhood cancers who were exposed to uterine radiation during childhood in the form of pelvic, spinal, or total-body irradiation. Recent findings of long-term follow-up studies evaluating reproductive performance of female survivors provided some reassurance to female cancer survivors by documenting that pregnancy and live birth rates were not significantly compromised in survivors, including those who had been treated with alkylating agents and had not received pelvic, cranial, and total-body irradiation. We aimed in this narrative review article to provide an update on the impact of chemotherapy and radiation on the ovarian and uterine function in female survivors of childhood cancer. IMPLICATIONS FOR PRACTICE Adult survivors of childhood cancers are more prone to developing a number of poor reproductive and obstetrical outcomes than their siblings and the general population as a result of previous exposure to chemotherapy and radiation during childhood. The impact of radiation therapy on the female genital system is greater than chemotherapy regimens because radiation is detrimental to both the uterus and the ovaries, whereas toxic effects of chemotherapy drugs are confined to the ovaries. Therefore, radiation-induced uterine damage accounts for most poor obstetrical outcomes in the survivors. These include infertility, miscarriages, stillbirths, fetal growth restrictions, preeclampsia, and preterm deliveries.
Collapse
Affiliation(s)
- Ozgur Oktem
- Department of Obstetrics and Gynecology, School of Medicine, Koc University, Istanbul, Turkey
- Assisted Reproduction Unit, Fertility Preservation Program, American Hospital Women's Health Center, Istanbul, Turkey
| | - Samuel S Kim
- Department of Obstetrics and Gynecology, School of Medicine, Kansas University, Kansas City, Kansas, USA
| | - Ugur Selek
- Department of Radiation Oncology, Koc University School of Medicine and MD Anderson Cancer Center, Houston, Texas, USA
- Department of Radiation Oncology, Comprehensive Cancer Care Program, American Hospital, Istanbul, Turkey
| | - Glenn Schatmann
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Medical College of Cornell University, New York, New York, USA
| | - Bulent Urman
- Department of Obstetrics and Gynecology, School of Medicine, Koc University, Istanbul, Turkey
- Assisted Reproduction Unit, Fertility Preservation Program, American Hospital Women's Health Center, Istanbul, Turkey
| |
Collapse
|
44
|
d'Argent EM, Antoine JM. [IVF and endometriosis, oocyte donation and fertility preservation]. Presse Med 2017; 46:1192-1198. [PMID: 29129415 DOI: 10.1016/j.lpm.2017.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 09/14/2017] [Indexed: 10/18/2022] Open
Abstract
Endometriosis is a common condition, causing pain and infertility. In infertile woman with superficial peritoneal endometriosis and patent tubes, laparoscopy is recommended, followed by ovarian stimulation alone or in combination with intrauterine inseminations. In case of ovarian or deep endometriosis, the indications of surgery and assisted reproductive technologies remain to be defined precisely. In vitro fertilization is generally proposed after the failure of up to three inseminations, directly for ovarian or deep endometriosis, or in case of an associated factor of infertility, mainly male. Before ovarian stimulation in view to in vitro fertilization, a pretreatment by GnRH agonist for 2 to 6 months or combined contraceptive for 6 to 8 weeks would improve the pregnancy rate. Egg donation is effective in patients with advanced ovarian failure or lack of ovarian response to stimulation. Fertility preservation, especially by oocytes vitrified, must be proposed preventively to women with endometriosis at risk of ovarian failure, without close wish to be pregnant.
Collapse
Affiliation(s)
- Emmanuelle Mathieu d'Argent
- Assistance publique-Hôpitaux de Paris, hôpital Tenon, université Pierre-et-Marie-Curie-Paris 6, GRC6-UPMC, centre expert en endométriose (C3E), service de gynécologie obstétrique et médecine de la reproduction, Paris, France.
| | - Jean-Marie Antoine
- Assistance publique-Hôpitaux de Paris, hôpital Tenon, université Pierre-et-Marie-Curie-Paris 6, GRC6-UPMC, centre expert en endométriose (C3E), service de gynécologie obstétrique et médecine de la reproduction, Paris, France
| |
Collapse
|
45
|
Gavish Z, Spector I, Peer G, Schlatt S, Wistuba J, Roness H, Meirow D. Follicle activation is a significant and immediate cause of follicle loss after ovarian tissue transplantation. J Assist Reprod Genet 2017; 35:61-69. [PMID: 29098533 DOI: 10.1007/s10815-017-1079-z] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Accepted: 10/24/2017] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Extensive follicle loss has been demonstrated in ovarian grafts post transplantation, reducing their productivity and lifespan. Several mechanisms for this loss have been proposed, and this study aims to clarify when and how the massive follicle loss associated with transplantation of ovarian tissue graft occurs. An understanding of the mechanisms of follicle loss will pinpoint potential new targets for optimization and improvement of this important fertility preservation technique. METHODS Frozen-thawed marmoset (n = 15), bovine (n = 37), and human (n = 46) ovarian cortical tissue strips were transplanted subcutaneously into immunodeficient castrated male mice for 3 or 7 days. Histological (H&E, Masson's trichrome) analysis and immunostaining (Ki-67, GDF9, cleaved caspase-3) were conducted to assess transplantation-associated follicle dynamics, with untransplanted frozen-thawed tissue serving as a negative control. RESULTS Evidence of extensive primordial follicle (PMF) activation and loss was observed already 3 days post transplantation in marmoset, bovine, and human tissue grafts, compared to frozen-thawed untransplanted controls (p < 0.001). No significant additional PMF loss was observed 7 days post transplantation. Recovered grafts of all species showed markedly higher rates of proliferative activity and progression from dormant to growing follicles (Ki-67 and GDF9 staining) as well as higher growing/primordial (GF/PMF) ratio (p < 0.02) and higher collagen levels compared with untransplanted controls. CONCLUSIONS This multi-species study demonstrates that follicle activation plays an important role in transplantation-induced follicle loss, and that it occurs within a very short time frame after grafting. These results underline the need to prevent this activation at the time of transplantation in order to retain the maximal possible follicle reserve and extend graft lifespan.
Collapse
Affiliation(s)
- Zohar Gavish
- Fertility Preservation Laboratory, Sheba Medical Center, Tel Hashomer, Israel
| | - Itay Spector
- Fertility Preservation Laboratory, Sheba Medical Center, Tel Hashomer, Israel
| | - Gil Peer
- IVF Division, Carmel Medical Center, the Ruth & Bruce Rappaport Faculty of Medicine, Technion Institute of Technology, Haifa, Israel
| | - Stefan Schlatt
- Centre of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Muenster, Germany
| | - Joachim Wistuba
- Centre of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, Muenster, Germany
| | - Hadassa Roness
- Fertility Preservation Laboratory, Sheba Medical Center, Tel Hashomer, Israel
| | - Dror Meirow
- Fertility Preservation Laboratory, Sheba Medical Center, Tel Hashomer, Israel. .,Sackler School of Medicine, Tel Aviv University, P.O. Box 39040, 6997801, Tel Aviv, Israel.
| |
Collapse
|
46
|
Pacheco F, Oktay K. Current Success and Efficiency of Autologous Ovarian Transplantation: A Meta-Analysis. Reprod Sci 2017; 24:1111-1120. [PMID: 28701069 DOI: 10.1177/1933719117702251] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
CONTEXT Ovarian cryopreservation followed by autotransplantation is still considered an experimental strategy for fertility preservation (FP) mainly because the success rates are unknown. OBJECTIVE To determine cohort epidemiologic characteristics and success rates of autologous ovarian tissue transplantation (OTT) with previously cryopreserved tissue. MATERIALS AND METHODS Literature review from 1999 to October 1, 2016. Additional cases were retrieved from meeting abstracts and own database. We selected studies that reported autologous OTT with previously banked tissue in humans. We did not include any cases involving fresh ovarian tissue transplantation or those performed to treat idiopathic premature ovarian failure/insufficiency. Both authors reviewed and selected studies for eligibility, which resulted in 59 full-text studies assessed for eligibility. Cases were extracted from original reports and reviews by the junior author, and the senior author reviewed and verified the extracted data. RESULTS Nineteen reports were included for qualitative synthesis. In 10 studies, detailed data were available to determine clinical and live birth + ongoing (LB + OG) pregnancy as well as endocrine restoration rates. Three hundred nine OTTs were performed with cryopreserved tissue, resulting in the birth of 84 children and 8 OG pregnancies. The cumulative clinical and LB + OG rates were 57.5% and 37.7%, respectively, and the endocrine restoration rate was 63.9%. CONCLUSION Success rates with cryopreserved OTT have reached promising levels. Given these recent data, ovarian tissue cryopreservation should be considered as a viable option for FP.
Collapse
Affiliation(s)
- Fernanda Pacheco
- 1 Innovation Fertility Preservation & IVF, New York, NY, USA.,2 Division of Reproductive Medicine, Department of Obstetrics and Gynecology, New York Medical College, Valhalla, NY, USA.,3 Classiclínica, Porto Alegre, Rio Grande do Sul, Brazil
| | - Kutluk Oktay
- 1 Innovation Fertility Preservation & IVF, New York, NY, USA.,2 Division of Reproductive Medicine, Department of Obstetrics and Gynecology, New York Medical College, Valhalla, NY, USA
| |
Collapse
|
47
|
Hammarberg K, Kirkman M, Stern C, McLachlan RI, Gook D, Rombauts L, Vollenhoven B, Fisher JRW. Cryopreservation of reproductive material before cancer treatment: a qualitative study of health care professionals' views about ways to enhance clinical care. BMC Health Serv Res 2017; 17:343. [PMID: 28490359 PMCID: PMC5424377 DOI: 10.1186/s12913-017-2292-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 05/04/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cancer treatment can diminish fertility in women and men. The need for fertility preservation is growing as increasing numbers of people survive cancer. Cryostorage of reproductive material to preserve potential for conception for cancer survivors has moved from being experimental to being a part of clinical management of women and men who are diagnosed with cancer in their reproductive years. There is little existing evidence about how fertility preservation services can be enhanced to meet the complex needs of patients who are diagnosed with cancer in their reproductive years. The aim of this research was to inform clinical practice development by drawing on the collective experience and knowledge of staff at well-established clinics that offer fertility preservation before cancer treatment. METHODS A qualitative research model was adopted using semi-structured interviews with professionals involved in the care of people who freeze reproductive material before cancer treatment. In the state of Victoria, Australia, two large assisted reproductive technology (ART) centres have been providing fertility preservation services for more than two decades. An invitation to participate in a semi-structured interview about clinical care in the context of fertility preservation was emailed to past and current staff members. To capture diverse perspectives, informants were sought from all relevant professions: fertility specialists, andrologists, nurses, embryologists/scientists, counsellors, and administrative staff. Transcripts were analysed thematically. RESULTS Thirteen key informants were interviewed from August 2013 to February 2014. The identified themes relating to enhancing clinical care in a fertility preservation service were communication between oncology and ART specialists; managing urgency; managing patients' expectations; establishing and implementing protocols, systems, and data bases; and maintaining contact with patients. CONCLUSION The collective knowledge of this study's informants, who represent multidisciplinary teams with more than two decades' experience in fertility preservation, yields important insights into strategies that fertility preservation services can employ to promote the integration of oncology and fertility care, the psychosocial care of patients, data recording and monitoring, and reporting of outcomes.
Collapse
Affiliation(s)
- Karin Hammarberg
- Jean Hailes Research Unit, School of Public Health and Preventive Medicine, Monash University, 549 St Kilda Rd, Melbourne, Victoria 3004 Australia
| | - Maggie Kirkman
- Jean Hailes Research Unit, School of Public Health and Preventive Medicine, Monash University, 549 St Kilda Rd, Melbourne, Victoria 3004 Australia
| | - Catharyn Stern
- Melbourne IVF, Melbourne, Victoria 3002 Australia
- Reproductive Services, Royal Women’s Hospital, Parkville, Victoria 3052 Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, Victoria 3010 Australia
| | - Robert I. McLachlan
- Monash IVF, Clayton, Victoria 3163 Australia
- Andrology Australia, Melbourne, Victoria 3163 Australia
- Hudson Institute of Medical Research, Clayton, Victoria 3163 Australia
| | - Debra Gook
- Melbourne IVF, Melbourne, Victoria 3002 Australia
- Reproductive Services, Royal Women’s Hospital, Parkville, Victoria 3052 Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, Victoria 3010 Australia
| | - Luk Rombauts
- Monash IVF, Clayton, Victoria 3163 Australia
- Hudson Institute of Medical Research, Clayton, Victoria 3163 Australia
- Monash Health, Clayton, Victoria 3163 Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria 3163 Australia
| | - Beverley Vollenhoven
- Monash IVF, Clayton, Victoria 3163 Australia
- Monash Health, Clayton, Victoria 3163 Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria 3163 Australia
| | - Jane R. W. Fisher
- Jean Hailes Research Unit, School of Public Health and Preventive Medicine, Monash University, 549 St Kilda Rd, Melbourne, Victoria 3004 Australia
| |
Collapse
|
48
|
Peters ITA, van der Steen MA, Huisman BW, Hilders CGJM, Smit VTHBM, Vahrmeijer AL, Sier CFM, Trimbos JB, Kuppen PJK. Morphological and phenotypical features of ovarian metastases in breast cancer patients. BMC Cancer 2017; 17:206. [PMID: 28327103 PMCID: PMC5361796 DOI: 10.1186/s12885-017-3191-y] [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] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 03/11/2017] [Indexed: 12/23/2022] Open
Abstract
Background Autotransplantation of frozen-thawed ovarian tissue is a method to preserve ovarian function and fertility in patients undergoing gonadotoxic therapy. In oncology patients, the safety cannot yet be guaranteed, since current tumor detection methods can only exclude the presence of malignant cells in ovarian fragments that are not transplanted. We determined the need for a novel detection method by studying the distribution of tumor cells in ovaries from patients with breast cancer. Furthermore, we examined which cell-surface proteins are suitable as a target for non-invasive tumor-specific imaging of ovarian metastases from invasive breast cancer. Methods Using the nationwide database of the Dutch Pathology Registry (PALGA), we identified a cohort of 46 women with primary invasive breast cancer and ovarian metastases. The localization and morphology of ovarian metastases were determined on hematoxylin-and-eosin-stained sections. The following cell-surface markers were immunohistochemically analyzed: E-cadherin, epithelial membrane antigen (EMA), human epidermal growth receptor type 2 (Her2/neu), carcinoembryonic antigen (CEA), αvβ6 integrin and epithelial cell adhesion molecule (EpCAM). Results The majority of ovarian metastases (71%) consisted of a solitary metastasis or multiple distinct nodules separated by uninvolved ovarian tissue, suggesting that ovarian metastases might be overlooked by the current detection approach. Combining the targets E-cadherin, EMA and Her2/neu resulted in nearly 100% detection of ductal ovarian metastases, whereas the combination of EMA, Her2/neu and EpCAM was most suitable to detect lobular ovarian metastases. Conclusions Examination of the actual ovarian transplants is recommended. A combination of targets is most appropriate to detect ovarian metastases by tumor-specific imaging. Electronic supplementary material The online version of this article (doi:10.1186/s12885-017-3191-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Inge T A Peters
- Department of Gynecology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Bertine W Huisman
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Vincent T H B M Smit
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Cornelis F M Sier
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - J Baptist Trimbos
- Department of Gynecology, Leiden University Medical Center, Leiden, the Netherlands
| | - Peter J K Kuppen
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
49
|
Robotic-assisted laparoscopy in reproductive surgery: a contemporary review. J Robot Surg 2017; 11:97-109. [PMID: 28194637 DOI: 10.1007/s11701-017-0682-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 02/03/2017] [Indexed: 10/20/2022]
Abstract
Robotic surgery is a conceptual fusion of the conventional open surgery and the minimally invasive laparoscopic surgery. We reviewed the current role of robotic-assisted laparoscopy in the field of reproductive surgery by a literature search in PubMed database. We analyzed the reported advantages and limitations of the use of robotics in reproductive surgeries like myomectomy, tubal reanastomosis, endometriosis, ovarian tissue cryopreservation, and ovarian transposition. Overall, robotic assistance in reproductive surgery resulted in decreased blood loss, less post-operative pain, shorter hospital stay, and faster convalescence, whereas reproductive outcomes were similar to open/laparoscopic approaches. The main drawbacks of robotic surgery were higher cost and longer operating times. It is as safe and effective as the conventional laparoscopy and represents a reasonable alternate to abdominal approach. Procedures that are technically challenging with the conventional laparoscopy can be performed with robotic assistance. It has advantages of improved visualization and Endowrist™ movements allowing precise suturing. This helps to overcome the limitations of laparoscopy, especially in complicated procedures, and may shorten the steep learning curve in minimal invasive surgery. Randomized controlled trials looking at both short- and long-term outcomes are warranted to strengthen the role of robotic surgery in the field of reproductive surgery.
Collapse
|
50
|
Bastings L, Westphal JR, Beerendonk CCM, Bekkers RLM, Zusterzeel PLM, Hendriks JCM, Braat DDM, Peek R. Clinically applied procedures for human ovarian tissue cryopreservation result in different levels of efficacy and efficiency. J Assist Reprod Genet 2016; 33:1605-1614. [PMID: 27714478 PMCID: PMC5171896 DOI: 10.1007/s10815-016-0816-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 09/15/2016] [Indexed: 02/04/2023] Open
Abstract
PURPOSE Different protocols are being used worldwide for the cryopreservation of human ovarian tissue for fertility preservation purposes. The efficiency and efficacy of the majority of these protocols has not been extensively evaluated, possibly resulting in sub-optimally cryopreserved ovarian tissue. To address the impact of this issue, we assessed the effects of two clinically successful human ovarian tissue slow-freezing cryopreservation procedures on the quality of the cryopreserved tissue. METHODS To differentiate between cryopreservation (C) versus thawing (T) related effects, four combinations of these two (A and B) very different cryopreservation/thawing protocols (ACAT, ACBT, BCAT, BCBT) were studied. Before and after cryopreservation and thawing, the percentage of living and morphologically normal follicles, as well as the overall tissue viability, was assessed. RESULTS Our experiments revealed that the choice of the cryopreservation protocol noticeably affected the overall tissue viability and percentage of living follicles, with a higher viability after protocol BC when compared to AC. No statistically significant differences in tissue viability were observed between the two thawing protocols, but thawing protocol BT required considerably more human effort and materials than thawing protocol AT. Tissue morphology was best retained using the BCAT combination. CONCLUSION Our results indicate that extensive and systematical evaluation of clinically used protocols is warranted.
Collapse
Affiliation(s)
- Lobke Bastings
- Department of Obstetrics and Gynecology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Johan R Westphal
- Department of Obstetrics and Gynecology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Catharina C M Beerendonk
- Department of Obstetrics and Gynecology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Ruud L M Bekkers
- Department of Obstetrics and Gynecology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Petra L M Zusterzeel
- Department of Obstetrics and Gynecology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Jan C M Hendriks
- Department for Health Evidence, Section Biostatistics, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Didi D M Braat
- Department of Obstetrics and Gynecology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Ronald Peek
- Department of Obstetrics and Gynecology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|