IMR Press / CEOG / Volume 48 / Issue 4 / DOI: 10.31083/j.ceog4804126
Open Access Review
Fertility-sparing in cancer patients
Show Less
1 Department of Gynecological Oncology, University of Health Sciences, Bakirkoy Dr. Sadi Konuk Training and Research Hospital, 34147 Istanbul, Turkey
2 Department of Obstetrics and Gynecology, Acibadem Mehmet Ali Aydinlar University, Acibadem Bakirkoy Hospital, 34147 Istanbul, Turkey
3 Department of Obstetrics and Gynecology, University of Münster, 48149 Münster, Germany
4 Department of Obstetrics and Gynecology, University Hospital Center Zagreb, 10000 Zagreb, Croatia
5 Department of Medicine, Nazarbayev University School of Medicine, 020000 Nur-Sultan, Kazakhstan
6 Clinical Academic Department of Women’s Health, National Research Center of Mother and Child Health, University Medical Center, 020000 Nur-Sultan, Kazakhstan
7 Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
8 Department of International Relations and Scientific Development, Akfa University, 100190 Tashkent, Uzbekistan
9 Department of Women’s and Children’s Health, University of Padua, 35122 Padua, Italy
*Correspondence: semaakyildiz@hotmail.com (Sema Karakaş)
Clin. Exp. Obstet. Gynecol. 2021, 48(4), 787–794; https://doi.org/10.31083/j.ceog4804126
Submitted: 8 February 2021 | Revised: 30 March 2021 | Accepted: 31 March 2021 | Published: 15 August 2021
(This article belongs to the Special Issue Updates in Obstetrics and Gynecology)
Copyright: © 2021 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license (https://creativecommons.org/licenses/by/4.0/).
Abstract

Objective: This review aimed to investigate and summarize the current evidence on fertility-sparing options in cancer patients. Mechanism: Fertility preservation methods are becoming popular through the improved prognosis of cancer patients at a younger age and early diagnostic tools. However, currently, more and more women are suffering from iatrogenic ovarian failure and fertility loss because of cancer treatment. Most treatments have been used for hematological malignancies, but different gynecological cancers can be eligible. Findings in brief: Fertility preserving strategies such as oocyte and embryo cryopreservation, ovarian tissue preservation, ovarian transposition, and aspiration of ovarian follicles are the methods that could be suggested to the patients. The current knowledge supports oocyte and embryo cryopreservation as feasible, safe, and effective treatment approaches for cancer patients seeking fertility preservation. Conclusions: Robust evidence is still needed to prove the effectiveness of cryopreservation of the ovarian tissue and ovarian follicle aspiration approaches since these techniques are still in early their steps.

Keywords
Cryopreservation
Infertility
Organ transplants
Ovarian neoplasms
1. Introduction

Patients with malignancy have an increased life span due to success rates in early diagnosis and satisfactory standards of treatments [1]. It is suggested that 6% of reproductive-age women continue to live with different types of malignancies [2]. Moreover, in some cases, cancers are diagnosed in pregnancy [3, 4, 5]. However, the effects of cytotoxic chemotherapy and radiotherapy on fertility have raised significant concerns, such as the burden of ovarian follicles, the fibrosis of the ovarian cortex, the deterioration of ovarian vascularization, and the induction of apoptotic processes of oocytes and granulosa cells [6, 7, 8].

In patients who require chemotherapy, the extent of ovarian damage depends on the type, length, and dosing schedule of the anticancer treatment combined with patients’ age [9, 10]. Instead, the iatrogenic effects of radiation depend upon the closeness of ovaries to the radiation field [9, 10]. Regarding the radiotherapy effect, 10% of patients who underwent vaginal brachytherapy and 40% of the patients who required external radiation therapy with or without vaginal brachytherapy lost their ovarian function [11, 12, 13]. The risk of damage determined by radiation increases with the total dose. A dose lower than 2 Gy is expected to decrease approximately 50% of primordial follicles; a quantity of 5–10 Gy is nearly toxic for all follicles [14]. Anticancer therapies may also affect genetic material and embryonic development [15]. An experimental study was conducted to investigate the cytotoxic effect of cyclophosphamide in an in vitro fertilization model. The study reported that the fertilization rate and the embryonic development were significantly reduced, and the percentage of aneuploid embryos was considerably increased compared to controls [15].

Considering the detrimental effects of anticancer therapies on fertility, fertility-preserving strategies have gained importance in cancer patients. Different methods ranging from pharmacological protection to surgical interventions, such as ovarian transposition, oocyte cryopreservation, and embryo and ovarian tissue freezing, have been implemented, combining assisted reproductive techniques and surgical methods [12, 16, 17].

Fertility-sparing strategies have to be personalized to the patient and cancer type [12, 17]. Most treatments have been used for hematological malignancies [11, 18], but different cancers can be eligible. In this regard, considering the peculiar characteristics of gynecologic pathologies, which even in the case of the benign disease require fertility-sparing approaches [19, 20], fertility-sparing methods are used with specific consideration for the patients with gynecological malignancies, such as cervical, vaginal, ovarian, and uterine cancers [21, 22]. To emphasize the importance of fertility-preserving strategies, an up-to-date guideline published by the American Society of Clinical Oncology suggested that in all women who need gonadotoxic treatments, fertility-preserving options should be considered regardless of age, parity, or prognosis [23]. This review aimed to investigate and summarize the current evidence on fertility-sparing options in cancer patients.

2. Pharmacological fertility-sparing strategies

Chemotherapy has detrimental effects on both primordial follicles and growing follicles, resulting in infertility and premature ovarian failure [6]. The “ovarian protection” therapies preserve ovarian reserve, especially in prepubertal females, compared to other surgical alternatives [23]. Medical therapies have been investigated employing GnRH agonists, Sphingosine-1-phosphate, imatinib, thalidomide, tamoxifen, G-CSF, and AS101 to overcome this unintended burden of anticancer treatments. Nevertheless, these drugs were studied only in rodent models [24]. Only GnRH agonists and Sphingosine-1-phosphate have been used in humans.

2.1 GnRH agonists

GnRH agonists prompt the release of luteinizing hormone (LH) and follicle-stimulating hormone (FSH) from the anterior pituitary gland, resulting in the long-term downregulation of gonadotrophin secretion [25, 26]. Different studies assessed the follicle preserving effect of GnRH agonists, particularly in lymphoma and estrogen-receptor-positive breast cancer patients [27]. Various mechanisms have been proposed to explain the follicle preserving effect, such as reducing primordial follicles entering the differentiation stage, the lower recruitment of primordial follicles, and the decrease of apoptosis [25]. Besides, GnRH agonists may lower the vascularity of gonads and cause reduced levels of gonadotoxic agents in the targeted organs. Nevertheless, data on the protective effects of this treatment are questioned due to the heterogeneity of study populations and procedures and the lack of a proven mechanism of action for ovarian protection with GnRH agonist [26, 27, 28].

In a study with 257 premenopausal women with operable hormone-receptor-negative breast cancer, patients received standard chemotherapy with the GnRH agonist goserelin [29]. The study reported an ovarian failure rate of 8% in the goserelin plus chemotherapy group and 22% in the chemotherapy-alone group after a 2-year follow-up. The pregnancy rates were also higher in women in the goserelin plus chemotherapy group than in the chemotherapy alone group (21% vs. 11%) [29]. Conversely, a two-center, four-arm, open-label randomized controlled trial (RCT) of GnRH analog co-treatment (GnRH antagonist and agonist combination) in breast cancer patients undergoing cyclophosphamide chemotherapy alone reported no significant protective effect on ovarian function [30]. In another RCT, no protective effect of GnRH agonists was observed in preserving ovarian reserves after cyclophosphamide, cisplatin, or paclitaxel in co-treatment [24].

2.2 Sphingosine-1-phosphate

The other ovarian-preserving agent, studied in both human and experimental investigations, is Sphingosine-1-phosphate, which inhibits apoptosis via the sphingomyelin pathway. Sphingosine-1-phosphate pretreatment was shown to reduce the radiation-related burden of primordial follicles in rats, primates, and xenografted human ovarian tissue [31]. A translational research study evaluating the protective effect of Sphingosine-1-phosphate on human ovarian tissue concluded that Sphingosine-1-phosphate might promote follicle survival in human ovarian cortical samples in in vitro environment. However, there are still concerns regarding the administration route of Sphingosine-1-phosphate, and further human studies are needed [32].

3. Surgical fertility-sparing strategies

Ovarian transposition can be considered for young patients who require pelvic irradiation [27, 33]. The procedure consists of mobilizing ovaries out of the pelvis, keeping gonads out of the radiation field. The adnexal pedicle should be fixed to the peritoneal surface of the lateral abdominal wall, 3–4 cm above the umbilical line [34]. The operation’s essential purpose is to ensure adequate ovarian blood, and metallic clips can be utilized to identify ovaries during radiotherapy.

Apart from surgery-related complications, another primary concern with ovarian transposition is ovarian metastases. Moreover, complicate future oocyte retrieval due to moving ovaries mostly out of the pelvic cavity is an additional concern, although a few studies reported spontaneous conception. Finally, adjuvant chemotherapy and the scattering effect of radiation may cause a decrease in ovarian blood supply, which can be particularly detrimental for patients with advanced age [33].

4. Assisted reproductive techniques as fertility-sparing strategies
4.1 Mature oocyte cryopreservation

Mature oocyte cryopreservation is regularly involved in assisted reproduction. However, in cancer patients, the method is not practicable for prepubertal girls [35]. Moreover, the required fourteen days of ovarian stimulation are the major limitation in reproductive age women, considering that anticancer treatments may require immediate action [36]. For this reason, alternative protocols have been proposed for an “emergent ovarian stimulation”, such as the random [37] and the dual stimulation protocols [35]. The dual stimulation protocol is a protocol in which ovaries are stimulated in both the follicular and luteal phases of the same cycle, yielding a higher oocyte number [35]. These strategies are practical in young patients who do not require an immediate life-saving chemo or radiation therapy, allowing patients at higher risk of iatrogenic premature ovarian insufficiency (POI) to have a possibility of pregnancy.

Even in cancer patients, age is the primary criterion predicting pregnancy rate and success of this treatment. Cobo et al. [38] reported retrospective data on the expected live birth rate per oocyte in women aged 35 years and 36 years. There was a 60.5% probability of live birth in patients 35 years old compared to a rate of 29.7% in patients 36 years old [38]. Goldman et al. [39] evaluated live birth rates of thawed oocytes and found that physicians should educate their patients about the real and probable pregnancy rates considering their age and ovarian reserves. Also, in some studies, the live birth rate per vitrified oocyte was reported around 5.7%, with approximately the need of 10 oocytes for a patient to have a fair chance of pregnancy [2, 38, 39].

Concerning patients with malignancy, further considerations are required. Women with BRCA mutations could have a decreased ovarian reserve [40]. Stimulation of oocytes in patients with hormone-sensitive cancers is questionable [41]; conventional ovarian stimulation causes supra-physiological estradiol levels, and women with estrogen receptor-positive (ER+) tumors may be unsuitable [41, 42]. In this regard, reliable data suggest that combination protocols with aromatase inhibitors and standard GnRH antagonist stimulation decrease the supra-physiological levels of estrogens [43, 44]. A recent systematic review found that letrozole had no detrimental effect on the disease-free survival period in breast cancer patients. However, there was a lack of good-quality evidence [45].

4.2 Stimulation protocols in fertility preservation

Patients who undergo controlled ovarian stimulation for fertility-preservation are usually responsive to moderate stimulation schedules for gonadotropins [46, 47, 48]. In conventional assisted reproductive technology, controlled ovarian stimulation begins at the early or mid-luteal follicular phase and takes 2–4 weeks. Therefore, traditional controlled ovarian stimulation can require up to 2–6 weeks to be completed from the moment of a cancer diagnosis.

Nevertheless, controlled ovarian stimulation therapy can start at any point of the menstrual cycle [49]. This possibility is adopted in the random-start ovarian stimulation protocol, which induces controlled ovarian stimulation immediately and regardless of the menstrual cycle phase. This method is becoming an established method in fertility preservation strategies for cancer patients, enabling oocyte retrieval in most cases with no more than two weeks.

A systematic review (251 patients) comparing ovarian stimulation cycles initiated in the luteal phase and the follicular phase reported that the luteal phase group required longer stimulation days and higher gonadotropin doses [50]. However, the number of retrieved oocytes did not differ, and oocytes obtained in the luteal phase were fertilized more efficiently [50]. Two studies involving 347 cancer patients undergoing ovarian stimulation for fertility preservation compared conventional vs. random-start ovarian stimulation [51, 52]. They reported no significant differences in the number of retrieved oocytes and gonadotropin doses [51, 52].

In a study by Cavagna et al. [53], they reported outcomes of random-start protocols in different cycle phases such as early follicular phase (n = 41), late follicular phase (n = 21), and luteal phase (n = 47). They found that the number of retrieved oocytes and the maturity rates were similar, although significantly higher gonadotropin doses were required in cycles initiated in the luteal phase.

An alternative controlled ovarian stimulation protocol is the double stimulation or so-called “dual stimulation” proposed for poor responder patients. It involves two stimulation protocols within the same menstrual cycle [54]. The first protocol is initiated in the follicular phase, then the second protocol begins immediately after the oocyte pick-up, in the luteal phase of the same cycle. Double stimulation provides two oocyte pick-ups that are performed approximately two weeks apart. It allows gathering more oocytes in a shorter period, which is ideal for women with a cancer diagnosis [54]. The current evidence shows that the double stimulation protocol is safe and provides sufficient oocyte quality. A study comparing 100 patients in the dual stimulation group and 197 in the conventional single-cycle stimulation group concluded that the cumulative live birth rate (LBR) (15% vs. 7%) and the rate of euploid blastocysts (31% vs. 14%) are higher in the dual stimulation group [54].

4.3 In vitro maturation of oocytes and cryopreservation

In vitro maturation (IVM) of human germinal vesicle (GV)-stage oocytes were defined by Edwards et al. [55] in 1969. In 1994, immature oocytes were surgically obtained from patients with polycystic ovary syndrome (PCOS), and IVM was consequently applied. A live birth took place after this treatment [56]. Live births were achieved with frozen-thawed embryos acquired from IVM oocytes of patients who overcame cancer in Singapore in 2014 [57]. This procedure has the advantage to removes the risk of reintroducing malignant cells related to the use of ovarian tissue [58]. Furthermore, IVM of GV-stage oocytes prevents any delay in anticancer treatments, given that no hormonal stimulation is required [59]. IVM is currently used in many centers, with a 20 to 35% live birth rate from cryopreserved IVM oocytes, improved cryopreservation protocols, and culture media [60, 61]. However, there is still not clear evidence comparing the success rate between IVM oocytes and vitrified-warmed mature oocytes [61, 62].

4.4 Embryo cryopreservation

Since the first pregnancy in 1985 [63], embryos’ cryopreservation is widely used in assisted reproduction [64]. The technical changeover from slow freezing to vitrification has led to successful results [65]. However, as compared to other approaches, this technique requires sperm donors and mature oocytes. Accordingly, the method may not be applicable for single women or prepubertal girls. Moreover, similar to other procedures, this technique is limited by the required administration of ovarian stimulation agents for 10–15 days [36].

4.5 Ovarian tissue cryopreservation and reimplantation

Small pieces of frozen-thawed ovarian tissue allowed the ovarian function restoration in oophorectomized animal models, reporting follicular survival and progression to the antral stage [66]. With the progressive developments of tissue cryopreservation techniques and the application in humans, more than 130 live births have been reported worldwide [67] after the first live birth reported by Donnez et al. in 2004 [68].

The recommended ovarian tissue retrieval consists of gathering 4–5 slices of the ovarian cortex of about 1 cm per 4–5 mm per 1.0–1.5 cm in size with the laparoscopic approach. The use of extensive electrocoagulation should be refrained since primordial follicles might be damaged [69]. Unilateral oophorectomy can be used to acquire sufficient tissue samples in prepubertal patients due to the lower ovarian tissue volume [2, 70]. Also, an ovary should be left in situ to enable future orthotopic transplantation. One of the fragments can be used for histopathologic analysis to exclude the risk of malignancy.

Initially, sliced ovarian grafts were transplanted heterotopically [66]. However, in the subsequent trials, ovarian tissue was more frequently transplanted in the pelvis [71]. Orthotopic transplantation technique includes the placement of thawed slices into a peritoneal pocket acquired through a peritoneal incision in the ovarian fossa. Another transplantation technique might be performed by placing tissue slices into the peri-medullar area through a limited ovarian cortex incision [72]. Later, sliced ovarian tissues can be fixed by suturing [2].

Heterotopic transplantation is defined as grafting thawed ovarian tissue outside the pelvic cavity in regions such as abdominal subcutaneous tissue or forearm. Heterotopic transplantation is less used but may be considered for patients with severe pelvic adhesions caused by earlier operations or patients who underwent previous pelvic radiation. Oocytes cultivated in controlled ovarian stimulation cycles can easily be accessed through this technique. Live birth rates varied between 23% and 57.5% in studies reporting fertility rates after ovarian tissue transplantation [2, 70, 72, 73, 74, 75].

One of the primary concerns regarding ovarian tissue reimplantation is that the ovarian graft functions are expected to last five to ten years based on the patient’s age, and type and duration of gonadotoxic treatments [73]. Another drawback of the procedure is the possible immediate follicular failure due to ischemia/reperfusion (I/R) injury [67, 76], which causes the loss of almost 50% of follicles. For this reason, different studies are investigating vascular growth factors and antioxidants as a treatment to reduce ischemia-reperfusion injury. Moreover, Oktay et al. [77] reported that the transplantation of cryopreserved ovarian tissue with a human decellularized extracellular tissue matrix scaffold could reduce the oxidative stress damage. However, regardless of the growing body of evidence, the technique continues to be an experimental method [41]. Noteworthy, there is still an ongoing debate on the efficiency of the vitrification method for freezing ovarian tissue [78].

One of several significant issues in ovarian tissue preservation is the risk of reseeding malignant cells that may cause re-occurrence of primary cancer [41]. A high risk of malignant cell reimplantation has been primarily observed in hematological malignancies [10, 79, 80], such as leukemia, Hodgkin lymphoma, and non-Hodgkin lymphoma [11]. Ovarian metastases have been additionally observed in gastric cancer (55.8%), colon cancer (26.6%), and lung cancer (23.4%) [81]. Moreover, frequent are the ovarian metastasis of gynecological malignancies, such as endometrial cancers, cervical adenocarcinomas, and tubal cancers [82]. In adenocarcinomas of the cervix, the rate of ovarian metastasis is relatively high, with an incidence of 6.8%, while in squamous cell cervical cancer, it is about 0.7–2.5%. However, no ovarian metastasis has been shown in cervical cancer patients with grafted ovarian tissue until now [83]. The risk of ovarian metastasis in the initial phase of endometrial cancer is relatively low, with an incidence of 1.9% [81]. Nevertheless, breast cancer is the most frequent malignancy in reproductive age women, with approximately 55% of the cases occurring in women under 40 years. In these patients, only a few studies assessed the effects of cryopreserved ovarian cortex transplantation techniques [2], and for late-stage breast cancer, the risk of ovarian metastasis is about 13.2–37.8%; therefore, caution should be exercised in these cases.

To overcome this issue, researchers should focus on specific molecular markers or other methods to identify minimal residual disease in grafted tissue [10, 84]. In this regard, some researchers have proposed transplanting the ovarian tissue into immune-deficient mice before implantation to identify the presence of cancer [85]. Moreover, some researchers suggested using the in vitro maturation of the oocytes derived from the collected ovarian tissue. The technique consists of the aspiration of immature oocytes from the ovarian tissue samples with subsequent in vitro maturation and vitrification [86]. This procedure is considered ideal for cancer patients with transplanted ovarian tissue at high risk of tumor seeding [27, 33]. Hourvitz et al. [87] reported data from more than 100 patients who have cancer. According to the report, the success rate of this approach was more than 50%. Published data from more than 40 cancer patients aged between 2 and 18 years reported an oocyte maturation rate of 10–30% [9, 88, 89]. Two live births were reported using in vitro-matured oocytes from ovarian tissue [57, 90].

5. Borderline ovarian tumors

Borderline ovarian tumors (BOT) are ovarian malignancies in which maintaining fertility is an option. These tumors have low malignant potential with an overall good prognosis and account for 10–15% of all ovarian epithelial cancers [91, 92, 93]. Although most patients diagnosed with BOT need follow-up with little or no chemotherapy, the surgical procedure of reference induces fertility loss or reduce ovarian reserve. Moreover, the delay of pregnancies for 2–5 years is recommended [91]. Therefore, fertility-sparing considerations are required. Regarding controlled ovarian hyperstimulation in patients with BOT, who are candidates for cryopreservation of oocytes, one concern is the possible tumor spillage during the oocyte pick-up. In this regard, preoperative confirmation of BOT is recommended, and some researchers have described that preoperative diagnosis with imaging studies is reliable [92, 93, 94, 95]. Controlled ovarian hyperstimulation may also increase BOT progression. Moreover, surgery following a controlled ovarian hyperstimulation treatment may increase ovarian cortical bleeding due to the substantial electrocoagulation for increased vascularity. In these cases, surgery should be postponed until 2–6 weeks after controlled ovarian hyperstimulation for corpora luteal resolution [91, 94, 95, 96, 97].

Regarding cryopreservation of ovarian tissue, it can be collected during the surgical treatment of BOT. Fain-Kahn et al. [97] stated that cryopreservation of the ovarian cortex should be performed in women with BOT recurrence in one ovary and women with bilateral BOT. However, a case series showed that the macroscopic inspection of affected ovaries was not always useful in identifying the best unaffected ovarian cortex site [97]. Moreover, concerns of tumor seeding are still present [98].

Regarding the effect of fertility-sparing surgery on oncological outcomes in women with BOTs, a study involving 2946 patients reported that fertility-preserving surgery was significantly associated with worse disease-specific survival in patients aged 50 years, but not in younger aged patients [96]. However, there is still a lack of sufficient evidence regarding the safety of fertility-sparing surgery for BOT patients.

6. Conclusions

Fertility preservation methods are becoming more common thanks to the improved effectiveness of oncologic treatments and early diagnosis implementation. More and more females are being affected by iatrogenic ovarian failure and loss of fertility due to these procedures. In this scenario, oocyte and embryo cryopreservation appear feasible, safe, and effective treatment able to preserve fertility in cancer patients of reproductive age. Conversely, cryopreservation of the ovarian tissue may be used for prepubertal girls and patients needing urgent cancer care. The procedure by which aspirated ovarian follicles can be matured in vitro and then used is still experimental and still needs more evidence on its effectiveness. Indeed, different steps are yet required to improve the effectiveness of available fertility preservation options and develop new fertility-sparing strategies for young cancer patients, such as developing substances for ovarian protection, improving ovarian grafting and post-implantation follicular survival, reducing the risk of reseeding malignancies with ovarian biopsies, developing more effective in vitro maturation system for primordial follicles, implementing the aspiration of immature oocytes from preantral and small antral ovarian follicles independently from cycle phase, and investigating new medications able to limit the ischemia/reperfusion injury.

Author contributions

SK, CK, SDS, MM and MT designed the research study. SK, CK and SDS performed the research. MT, BA and AA provided help and advice on the writing. SK and CK analyzed the data. SK and CK wrote the manuscript. All authors contributed to editorial changes in the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Acknowledgment

Thanks to all the peer reviewers for their opinions and suggestions.

Funding

This research received no external funding.

Conflict of interest

The authors declare no conflict of interest.

References
[1]
Bedoschi G, Oktay K. Current approach to fertility preservation by embryo cryopreservation. Fertility and Sterility. 2013; 99: 1496–1502.
[2]
Silber S. Ovarian tissue cryopreservation and transplantation: scientific implications. Journal of Assisted Reproduction and Genetics. 2016; 33: 1595–1603.
[3]
Sartori E, Franchi M, Capelli G, Cicinelli E, Colacurci N, De Vincenzo R, et al. Cancer in pregnancy: proposal of an italian multicenter study. Gynecologic oncology group of the Italian Society of Gynecology and Obstetrics (SIGO). Italian Journal of Gynaecology & Obstetrics. 2018; 30: 37–44.
[4]
Simonetto C, Garzon S, Laganà AS, Casarin J, Raffaelli R, Cromi A, et al. Non-obstetric surgery during pregnancy: current perspectives and future directions. Italian Journal of Gynaecology & Obstetrics. 2019; 31: 57–68.
[5]
Bapayeva G, Terzic M, Togyzbayeva K, Bekenova A, Terzic S, Garzon S, et al. Late diagnosis of pheochromocytoma in pregnancy with poor fetal outcome. Archive of Oncology. 2021; 27: 9–11.
[6]
Oktem O, Oktay K. A novel ovarian xenografting model to characterize the impact of chemotherapy agents on human primordial follicle reserve. Cancer Research. 2007; 67: 10159–10162.
[7]
Morgan S, Anderson RA, Gourley C, Wallace WH, Spears N. How do chemotherapeutic agents damage the ovary? Human Reproduction Update. 2012; 18: 525–535.
[8]
Parissone F, Di Paola R, Balter R, Garzon S, Zaffagnini S, Neri M, et al. Female adolescents and young women previously treated for pediatric malignancies: assessment of ovarian reserve and gonadotoxicity risk stratification for early identification of patients at increased infertility risk. Journal of Pediatric Endocrinology and Metabolism. 2020; 34: 25–33.
[9]
Abir R, Ben-Aharon I, Garor R, Yaniv I, Ash S, Stemmer SM, et al. Cryopreservation of in vitro matured oocytes in addition to ovarian tissue freezing for fertility preservation in paediatric female cancer patients before and after cancer therapy. Human Reproduction. 2016; 31: 750–762.
[10]
Chung K, Donnez J, Ginsburg E, Meirow D. Emergency IVF versus ovarian tissue cryopreservation: decision making in fertility preservation for female cancer patients. Fertility and Sterility. 2013; 99: 1534–1542.
[11]
Donnez J, Dolmans M. Fertility preservation in women. Nature Reviews Endocrinology. 2013; 9: 735-749.
[12]
Moawad NS, Santamaria E, Rhoton-Vlasak A, Lightsey JL. Laparoscopic ovarian transposition before pelvic cancer treatment: ovarian function and fertility preservation. Journal of Minimally Invasive Gynecology. 2017; 24: 28–35.
[13]
Laganà AS, Garzon S, Raffaelli R, Ban Frangež H, Lukanovič D, Franchi M. Vaginal stenosis after cervical cancer treatments: challenges for reconstructive surgery. Journal of Investigative Surgery. 2019; 1–2.
[14]
Wallace WHB, Thomson AB, Kelsey TW. The radiosensitivity of the human oocyte. Human Reproduction. 2003; 18: 117–121.
[15]
Koike M, Kanda A, Kido K, Goto K, Kumasako Y, Nagaki M, et al. Effects of cyclophosphamide administration on the in vitro fertilization of mice. Reproductive Medicine and Biology. 2018; 17: 262–267.
[16]
Fisch B, Abir R. Female fertility preservation: past, present and future. Reproduction. 2018; 156: F11–F27.
[17]
Arian SE, Goodman L, Flyckt RL, Falcone T. Ovarian transposition: a surgical option for fertility preservation. Fertility and Sterility. 2017; 107: e15.
[18]
Donnez J, Dolmans M, Pellicer A, Diaz-Garcia C, Sanchez Serrano M, Schmidt KT, et al. Restoration of ovarian activity and pregnancy after transplantation of cryopreserved ovarian tissue: a review of 60 cases of reimplantation. Fertility and Sterility. 2013; 99: 1503–1513.
[19]
Sleiman Z, Karaman E, Terzic M, Terzic S, Falzone G, Garzon S. Fertility preservation in benign gynecological diseases: current approaches and future perspectives. Journal of Reproduction & Infertility. 2019; 20: 201–208.
[20]
Terzic M, Aimagambetova G, Garzon S, Bapayeva G, Ukybassova T, Terzic S, et al. Ovulation induction in infertile women with endometriotic ovarian cysts: current evidence and potential pitfalls. Minerva Medica. 2020; 111: 50–61.
[21]
La Rosa VL, Garzon S, Gullo G, Fichera M, Sisti G, Gallo P, et al. Fertility preservation in women affected by gynaecological cancer: the importance of an integrated gynaecological and psychological approach. Ecancermedicalscience. 2020; 14: 1035.
[22]
Casarin J, Laganà AS, Uccella S, Cromi A, Pinelli C, Gisone B, et al. Surgical treatment of large adnexal masses: a retrospective analysis of 330 consecutive cases. Minimally Invasive Therapy & Allied Technologies. 2020; 29: 366–374.
[23]
Loren AW, Mangu PB, Beck LN, Brennan L, Magdalinski AJ, Partridge AH, et al. Fertility preservation for patients with cancer: American society of clinical oncology clinical practice guideline update. Journal of Clinical Oncology. 2013; 31: 2500–2510.
[24]
Bildik G, Akin N, Senbabaoglu F, Sahin GN, Karahuseyinoglu S, Ince U, et al. GnRH agonist leuprolide acetate does not confer any protection against ovarian damage induced by chemotherapy and radiation in vitro. Human Reproduction. 2015; 30: 2912–2925.
[25]
Blumenfeld Z, Evron A. Preserving fertility when choosing chemotherapy regimens—the role of gonadotropin-releasing hormone agonists. Expert Opinion on Pharmacotherapy. 2015; 16: 1009–1020.
[26]
Hickman LC, Valentine LN, Falcone T. Preservation of gonadal function in women undergoing chemotherapy: a review of the potential role for gonadotropin-releasing hormone agonists. American Journal of Obstetrics and Gynecology. 2016; 215: 415–422.
[27]
Rodriguez-Wallberg KA, Oktay K. Options on fertility preservation in female cancer patients. Cancer Treatment Reviews. 2012; 38: 354–361.
[28]
Salama M, Isachenko V, Isachenko E, Rahimi G, Mallmann P. Updates in preserving reproductive potential of prepubertal girls with cancer: systematic review. Critical Reviews in Oncology/Hematology. 2016; 103: 10–21.
[29]
Moore HCF, Unger JM, Phillips K, Boyle F, Hitre E, Porter D, et al. Goserelin for ovarian protection during breast-cancer adjuvant chemotherapy. New England Journal of Medicine. 2015; 372: 923–932.
[30]
Elgindy EA, El-Haieg DO, Khorshid OM, Ismail EI, Abdelgawad M, Sallam HN, et al. Gonadatrophin suppression to prevent chemotherapy-induced ovarian damage: a randomized controlled trial. Obstetrics and Gynecology. 2013; 121: 78–86.
[31]
Zelinski MB, Murphy MK, Lawson MS, Jurisicova A, Pau KYF, Toscano NP, et al. In vivo delivery of FTY720 prevents radiation-induced ovarian failure and infertility in adult female nonhuman primates. Fertility and Sterility. 2011; 95: 1440–1445.e1–7.
[32]
Guzel Y, Bildik G, Oktem O. Sphingosine-1-phosphate protects human ovarian follicles from apoptosis in vitro. European Journal of Obstetrics, Gynecology, and Reproductive Biology. 2018; 222: 19–24.
[33]
Moorland MT. Cancer in Female Adolescents. New York: Nova Publishers. 2008.
[34]
Huang K, Lee C, Tsai C, Han C, Hwang L. A new approach for laparoscopic ovarian transposition before pelvic irradiation. Gynecologic Oncology. 2007; 105: 234–237.
[35]
Ubaldi F, Vaiarelli A, D’Anna R, Rienzi L. Management of poor responders in IVF: is there anything new? BioMed Research International. 2014; 2014: 352098.
[36]
Massarotti C, Scaruffi P, Lambertini M, Remorgida V, Del Mastro L, Anserini P. State of the art on oocyte cryopreservation in female cancer patients: a critical review of the literature. Cancer Treatment Reviews. 2017; 57: 50–57.
[37]
Simi G, Obino MER, Casarosa E, Litta P, Artini PG, Cela V. Different stimulation protocols for oocyte cryropreservation in oncological patients: a retrospective analysis of single university centre. Gynecological Endocrinology. 2015; 31: 966–970.
[38]
Cobo A, García-Velasco JA, Coello A, Domingo J, Pellicer A, Remohí J. Oocyte vitrification as an efficient option for elective fertility preservation. Fertility and Sterility. 2016; 105: 755–764.e8.
[39]
Goldman RH, Racowsky C, Farland LV, Munné S, Ribustello L, Fox JH. Predicting the likelihood of live birth for elective oocyte cryopreservation: a counseling tool for physicians and patients. Human Reproduction. 2017; 32: 853–859.
[40]
Friedler S, Koc O, Gidoni Y, Raziel A, Ron-El R. Ovarian response to stimulation for fertility preservation in women with malignant disease: a systematic review and meta-analysis. Fertility and Sterility. 2012; 97: 125–133.
[41]
Martinez F. Update on fertility preservation from the barcelona international society for fertility preservation-ESHRE-ASRM 2015 expert meeting: indications, results and future perspectives. Fertility and Sterility. 2017; 108: 407–415.e11.
[42]
Key T, Appleby P, Barnes I, Reeves G. Endogenous sex hormones and breast cancer in postmenopausal women: reanalysis of nine prospective studies. Journal of the National Cancer Institute. 2002; 94: 606–616.
[43]
Oktay K, Turan V, Bedoschi G, Pacheco FS, Moy F. Fertility preservation success subsequent to concurrent aromatase inhibitor treatment and ovarian stimulation in women with breast cancer. Journal of Clinical Oncology. 2015; 33: 2424–2429.
[44]
Kim J, Turan V, Oktay K. Long-term safety of letrozole and gonadotropin stimulation for fertility preservation in women with breast cancer. Journal of Clinical Endocrinology and Metabolism. 2016; 101: 1364–1371.
[45]
Rodgers RJ, Reid GD, Koch J, Deans R, Ledger WL, Friedlander M, et al. The safety and efficacy of controlled ovarian hyperstimulation for fertility preservation in women with early breast cancer: a systematic review. Human Reproduction. 2017; 32: 1033–1045.
[46]
Papaleo E, Zaffagnini S, Munaretto M, Vanni VS, Rebonato G, Grisendi V, et al. Clinical application of a nomogram based on age, serum FSH and AMH to select the FSH starting dose in IVF/ICSI cycles: a retrospective two-centres study. European Journal of Obstetrics & Gynecology and Reproductive Biology. 2016; 207: 94–99.
[47]
La Marca A, Papaleo E, Grisendi V, Argento C, Giulini S, Volpe A. Development of a nomogram based on markers of ovarian reserve for the individualisation of the follicle-stimulating hormone starting dose in in vitro fertilisation cycles. BJOG: An International Journal of Obstetrics and Gynaecology. 2012; 119: 1171–1179.
[48]
Di Paola R, Garzon S, Giuliani S, Laganà AS, Noventa M, Parissone F, et al. Are we choosing the correct FSH starting dose during controlled ovarian stimulation for intrauterine insemination cycles? Potential application of a nomogram based on woman’s age and markers of ovarian reserve. Archives of Gynecology and Obstetrics. 2018; 298: 1029–1035.
[49]
Cakmak H, Katz A, Cedars MI, Rosen MP. Effective method for emergency fertility preservation: random-start controlled ovarian stimulation. Fertility and Sterility. 2013; 100: 1673–1680.
[50]
Boots CE, Meister M, Cooper AR, Hardi A, Jungheim ES. Ovarian stimulation in the luteal phase: systematic review and meta-analysis. Journal of Assisted Reproduction and Genetics. 2016; 33: 971–980.
[51]
Muteshi C, Child T, Ohuma E, Fatum M. Ovarian response and follow-up outcomes in women diagnosed with cancer having fertility preservation: comparison of random start and early follicular phase stimulation—ohort study. European Journal of Obstetrics & Gynecology and Reproductive Biology. 2018; 230: 10–14.
[52]
Pereira N, Hancock K, Cordeiro CN, Lekovich JP, Schattman GL, Rosenwaks Z. Comparison of ovarian stimulation response in patients with breast cancer undergoing ovarian stimulation with letrozole and gonadotropins to patients undergoing ovarian stimulation with gonadotropins alone for elective cryopreservation of oocytes. Gynecological Endocrinology. 2016; 32: 823–826.
[53]
Cavagna F, Pontes A, Cavagna M, Dzik A, Donadio NF, Portela R, et al. Specific protocols of controlled ovarian stimulation for oocyte cryopreservation in breast cancer patients. Current Oncology. 2018; 25: e527–e532.
[54]
Vaiarelli A, Cimadomo D, Trabucco E, Vallefuoco R, Buffo L, Dusi L, et al. Double stimulation in the same ovarian cycle (duostim) to maximize the number of oocytes retrieved from poor prognosis patients: a multicenter experience and SWOT analysis. Frontiers in Endocrinology. 2018; 9: 317.
[55]
Edwards RG, Bavister BD, Steptoe PC. Early stages of fertilization in vitro of human oocytes matured in vitro. Nature. 1969; 221: 632–635.
[56]
Trounson A, Wood C, Kausche A. In vitro maturation and the fertilization and developmental competence of oocytes recovered from untreated polycystic ovarian patients. Fertility and Sterility. 1994; 62: 353–362.
[57]
Prasath EB, Chan MLH, Wong WHW, Lim CJW, Tharmalingam MD, Hendricks M, et al. First pregnancy and live birth resulting from cryopreserved embryos obtained from in vitro matured oocytes after oophorectomy in an ovarian cancer patient. Human Reproduction. 2014; 29: 276–278.
[58]
Chian R, Xu C, Huang JYJ, Ata B. Obstetric outcomes and congenital abnormalities in infants conceived with oocytes matured in vitro. Facts, Views and Vision in Obstetrics and Gynaecology. 2014; 6: 15–18.
[59]
Huang JYJ, Chian R, Gilbert L, Fleiszer D, Holzer H, Dermitas E, et al. Retrieval of immature oocytes from unstimulated ovaries followed by in vitro maturation and vitrification: a novel strategy of fertility preservation for breast cancer patients. American Journal of Surgery. 2010; 200: 177–183.
[60]
Park CW, Lee SH, Yang KM, Lee IH, Lim KT, Lee KH, et al. Cryopreservation of in vitro matured oocytes after ex vivo oocyte retrieval from gynecologic cancer patients undergoing radical surgery. Clinical and Experimental Reproductive Medicine. 2016; 43: 119–125.
[61]
Ellenbogen A, Shavit T, Shalom-Paz E. IVM results are comparable and may have advantages over standard IVF. Facts, Views and Vision in Obstetrics and Gynaecology. 2014; 6: 77–80.
[62]
Chian R, Huang JYJ, Gilbert L, Son W, Holzer H, Cui SJ, et al. Obstetric outcomes following vitrification of in vitro and in vivo matured oocytes. Fertility and Sterility. 2009; 91: 2391–2398.
[63]
Cohen J, Simons RF, Fehilly CB, Fishel SB, Edwards RG, Hewitt J, et al. Birth after replacement of hatching blastocyst cryopreserved at expanded blastocyst stage. Lancet. 1985; 1: 647.
[64]
Herrero L, Martínez M, Garcia-Velasco JA. Current status of human oocyte and embryo cryopreservation. Current Opinion in Obstetrics & Gynecology. 2011; 23: 245–250.
[65]
Turner NH, Partridge A, Sanna G, Di Leo A, Biganzoli L. Utility of gonadotropin-releasing hormone agonists for fertility preservation in young breast cancer patients: the benefit remains uncertain. Annals of Oncology. 2013; 24: 2224–2235.
[66]
Oktay K, Newton H, Mullan J, Gosden RG. Development of human primordial follicles to antral stages in SCID/hpg mice stimulated with follicle stimulating hormone. Human Reproduction. 1998; 13: 1133–1138.
[67]
Donnez J, Dolmans M. Fertility Preservation in Women. New England Journal of Medicine. 2017; 377: 1657–1665.
[68]
Donnez J, Dolmans M, Demylle D, Jadoul P, Pirard C, Squifflet J, et al. Livebirth after orthotopic transplantation of cryopreserved ovarian tissue. Lancet. 2004; 364: 1405–1410.
[69]
Donnez J, Martinez-Madrid B, Jadoul P, Van Langendonckt A, Demylle D, Dolmans M. Ovarian tissue cryopreservation and transplantation: a review. Human Reproduction Update. 2006; 12: 519–535.
[70]
Andersen CY, Rosendahl M, Byskov AG, Loft A, Ottosen C, Dueholm M, et al. Two successful pregnancies following autotransplantation of frozen/thawed ovarian tissue. Human Reproduction. 2008; 23: 2266–2272.
[71]
Meirow D, Levron J, Eldar-Geva T, Hardan I, Fridman E, Zalel Y, et al. Pregnancy after transplantation of cryopreserved ovarian tissue in a patient with ovarian failure after chemotherapy. New England Journal of Medicine. 2005; 353: 318–321.
[72]
Ladanyi C, Mor A, Christianson MS, Dhillon N, Segars JH. Recent advances in the field of ovarian tissue cryopreservation and opportunities for research. Journal of Assisted Reproduction and Genetics. 2017; 34: 709–722.
[73]
Demeestere I, Simon P, Emiliani S, Delbaere A, Englert Y. Orthotopic and heterotopic ovarian tissue transplantation. Human Reproduction Update. 2009; 15: 649–665.
[74]
Jensen AK, Kristensen SG, Macklon KT, Jeppesen JV, Fedder J, Ernst E, et al. Outcomes of transplantations of cryopreserved ovarian tissue to 41 women in Denmark. Human Reproduction. 2015; 30: 2838–2845.
[75]
Van der Ven H, Liebenthron J, Beckmann M, Toth B, Korell M, Krüssel J, et al. Ninety-five orthotopic transplantations in 74 women of ovarian tissue after cytotoxic treatment in a fertility preservation network: tissue activity, pregnancy and delivery rates. Human Reproduction. 2016; 31: 2031–2041.
[76]
Friedman O, Orvieto R, Fisch B, Felz C, Freud E, Ben-Haroush A, et al. Possible improvements in human ovarian grafting by various host and graft treatments. Human Reproduction. 2012; 27: 474–482.
[77]
Oktay K, Bedoschi G, Pacheco F, Turan V, Emirdar V. First pregnancies, live birth, and in vitro fertilization outcomes after transplantation of frozen-banked ovarian tissue with a human extracellular matrix scaffold using robot-assisted minimally invasive surgery. American Journal of Obstetrics and Gynecology. 2016; 214: 94.e1–94.e9.
[78]
Abir R, Fisch B, Fisher N, Samara N, Lerer-Serfaty G, Magen R, et al. Attempts to improve human ovarian transplantation outcomes of needle-immersed vitrification and slow-freezing by host and graft treatments. Journal of Assisted Reproduction and Genetics. 2017; 34: 633–644.
[79]
Abir R, Aviram A, Feinmesser M, Stein J, Yaniv I, Parnes D, et al. Ovarian minimal residual disease in chronic myeloid leukaemia. Reproductive BioMedicine Online. 2014; 28: 255–260.
[80]
Grèze V, Brugnon F, Chambon F, Halle P, Canis M, Amiot C, et al. Highly sensitive assessment of neuroblastoma minimal residual disease in ovarian tissue using RT-qPCR-a strategy for improving the safety of fertility restoration. Pediatric Blood & Cancer. 2017; 64.
[81]
Rosendahl M, Greve T, Andersen CY. The safety of transplanting cryopreserved ovarian tissue in cancer patients: a review of the literature. Journal of Assisted Reproduction and Genetics. 2013; 30: 11–24.
[82]
Casey L, Singh N. Metastases to the ovary arising from endometrial, cervical and fallopian tube cancer: recent advances. Histopathology. 2020; 76: 37–51.
[83]
Donnez J, Dolmans M. Ovarian tissue freezing: current status. Current Opinion in Obstetrics & Gynecology. 2015; 27: 222–230.
[84]
Abir R, Feinmesser M, Yaniv I, Fisch B, Cohen IJ, Ben-Haroush A, et al. Occasional involvement of the ovary in Ewing sarcoma. Human Reproduction. 2010; 25: 1708–1712.
[85]
Dolmans M, Luyckx V, Donnez J, Andersen CY, Greve T. Risk of transferring malignant cells with transplanted frozen-thawed ovarian tissue. Fertility and Sterility. 2013; 99: 1514–1522.
[86]
Huang JYJ, Tulandi T, Holzer H, Tan SL, Chian R. Combining ovarian tissue cryobanking with retrieval of immature oocytes followed by in vitro maturation and vitrification: an additional strategy of fertility preservation. Fertility and Sterility. 2008; 89: 567–572.
[87]
Hourvitz A, Yerushalmi GM, Maman E, Raanani H, Elizur S, Brengauz M, et al. Combination of ovarian tissue harvesting and immature oocyte collection for fertility preservation increases preservation yield. Reproductive Biomedicine Online. 2015; 31: 497–505.
[88]
Revel A, Revel-Vilk S, Aizenman E, Porat-Katz A, Safran A, Ben-Meir A, et al. At what age can human oocytes be obtained? Fertility and Sterility. 2009; 92: 458–463.
[89]
Fasano G, Dechène J, Antonacci R, Biramane J, Vannin A, Van Langendonckt A, et al. Outcomes of immature oocytes collected from ovarian tissue for cryopreservation in adult and prepubertal patients. Reproductive Biomedicine Online. 2017; 34: 575–582.
[90]
Uzelac PS, Delaney AA, Christensen GL, Bohler HCL, Nakajima ST. Live birth following in vitro maturation of oocytes retrieved from extracorporeal ovarian tissue aspiration and embryo cryopreservation for 5 years. Fertility and Sterility. 2015; 104: 1258–1260.
[91]
Mangili G, Somigliana E, Giorgione V, Martinelli F, Filippi F, Petrella MC, et al. Fertility preservation in women with borderline ovarian tumours. Cancer Treatment Reviews. 2016; 49: 13–24.
[92]
Terzic M, Rapisarda AMC, Della Corte L, Manchanda R, Aimagambetova G, Norton M, et al. Diagnostic work-up in paediatric and adolescent patients with adnexal masses: an evidence-based approach. Journal of Obstetrics and Gynaecology. 2020; 1–13.
[93]
Terzic M, Aimagambetova G, Norton M, Della Corte L, Marín-Buck A, Lisón JF, et al. Scoring systems for the evaluation of adnexal masses nature: current knowledge and clinical applications. Journal of Obstetrics and Gynaecology. 2020; 1–8.
[94]
Van Calster B, Timmerman D, Valentin L, McIndoe A, Ghaem-Maghami S, Testa AC, et al. Triaging women with ovarian masses for surgery: observational diagnostic study to compare RCOG guidelines with an International Ovarian Tumour Analysis (IOTA) group protocol. BJOG: An International Journal of Obstetrics and Gynaecology. 2012; 119: 662–671.
[95]
Sayasneh A, Wynants L, Preisler J, Kaijser J, Johnson S, Stalder C, et al. Multicentre external validation of IOTA prediction models and RMI by operators with varied training. British Journal of Cancer. 2013; 108: 2448–2454.
[96]
Sun H, Chen X, Zhu T, Liu N, Yu A, Wang S. Age-dependent difference in impact of fertility preserving surgery on disease-specific survival in women with stage I borderline ovarian tumors. Journal of Ovarian Research. 2018; 11: 54.
[97]
Fain-Kahn V, Poirot C, Uzan C, Prades M, Gouy S, Genestie C, et al. Feasibility of ovarian cryopreservation in borderline ovarian tumours. Human Reproduction. 2009; 24: 850–855.
[98]
De Vos M, Smitz J, Woodruff TK. Fertility preservation in women with cancer. Lancet. 2014; 384: 1302–1310.
Share
Back to top