Next Article in Journal
Clinical Management of Neuroendocrine Neoplasms in Clinical Practice: A Formal Consensus Exercise
Previous Article in Journal
Characterization of the Immune Response to PD-1 Blockade during Chemoradiotherapy for Head and Neck Squamous Cell Carcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Fertility after Cancer: Risks and Successes

Department of Obstetrics and Gynecology, “Sapienza” University, 00185 Rome, Italy
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(10), 2500; https://doi.org/10.3390/cancers14102500
Submission received: 29 March 2022 / Revised: 8 May 2022 / Accepted: 16 May 2022 / Published: 19 May 2022

Abstract

:

Simple Summary

Approximately one million new cases of cancer are diagnosed in women of reproductive age every year. In the last few decades, advances in early diagnosis and treatment have improved the survival rate. However, the adverse effects of anticancer therapy on the ovaries and uterus have a significant impact on future fertility and may affect the quality of life of cancer survivors. Unfortunately, evidence about the trend of ovarian reserve loss over time is insufficient for predicting the duration of the fertile period. Currently, impaired fertility in cancer survivors is a growing issue that is complicated by an increasing number of women delaying childbearing. This review focuses on the detrimental effects of chemotherapy, radiotherapy, and surgery on reproductive functions and describes the mechanisms causing reduced fertility in cancer survivors. Moreover, in this review, the available fertility preservation strategies to guarantee the chance of motherhood in cancer survivors are illustrated.

Abstract

The incidence of cancer in reproductive-aged women is 7%, but, despite the increased number of cancer cases, advances in early diagnosis and treatment have raised the survival rate. Furthermore, in the last four decades, there has been a rising trend of delaying childbearing. There has been an increasing number of couples referred to Reproductive Medicine Centers for infertility problems after one partner has been treated for cancer. In these cases, the main cause of reduced fertility derives from treatments. In this review, we describe the effects and the risks of chemotherapy, radiotherapy, and surgery in women with cancer, and we will focus on available fertility preservation techniques and their efficacy in terms of success in pregnancy and live birth rates.

Graphical Abstract

1. Introduction

The incidence of any type of cancer in 15–39 year old women is 52.3 rate per 100,000 [1].
In Italy, 3% of cancer cases are diagnosed in women under the age of 40. The most common types of cancer in women under 40 are breast cancer, thyroid cancer, melanoma, cervical cancer, endometrial cancer, ovarian cancer, leukemia/lymphomas, and colorectal cancer [2].
Breast cancer (BC) is the most common cancer in women under 49 (40% of all cancer cases). Despite the increased incidence of BC with age, approximately 7–10% of women diagnosed with BC are under 40 [3]. The incidence trend in Italy is slightly increasing (0.3% per year), whereas mortality continues to decline (−0.8% per year). The 5-year survival rate in young women (15–44 years) is 91% [4].
Thyroid cancer is common between the ages of 0–49 years (15% of all cancer cases). The most important prognostic factor is represented by the following histotype: the 20-year survival rate is 98–99% for papillary carcinomas and 80–90% for follicular ones (together they constitute 90% of thyroid cancer with papillary/follicular ratio 10:1), whereas it drops to 50–75% at 10 years for medullary carcinomas [5].
Melanoma is the third most frequent cancer in women aged between 18 and 39 years, with an increasing incidence trend (3.1% per year) [6].
Cervical cancer is the second for incidence in women. Its incidence increases with age up to 45 years with a peak between 45–55 years. The incidence rate is rising in developing countries but is decreasing in high resource countries [7].
Endometrial cancer has a very low incidence in reproductive age. Only 20% of endometrial cancers affect premenopausal women, and of these, no more than 5–8% affect women under 40 [8].
As for ovarian cancer, 80–90% occurs amongst women between the ages of 20 and 65 years old, and 90% of malignant tumors are diagnosed in women over the age of 40. A total of 5–10% of ovarian cancers are of intermediate malignancy (borderline) and approximately 30% of them affect women under 40. However, the diagnosis of epithelial ovarian cancer in women of reproductive age has become more frequent with increasing gynecological physical checkups [2].
Colorectal cancer’s (CRC) incidence is increasing in women of reproductive age. Fortunately, the 5-year survival rate from CRC is improving, with a survival rate of 65% [9].
Considering pediatric cancer patients, a recent study of over 3500 young women who survived childhood cancer such as leukemia, central nervous system cancer, Hodgkin lymphoma, non-Hodgkin lymphoma, Wilms tumor, neuroblastoma, soft tissue sarcoma, or bone tumor, shows a significantly higher risk of infertility than in the control group (RR: 1.48) [10,11]. There are still limited modalities available to preserve prepubertal fertility. Ovarian tissue reimplantation is the only fertility preservation technique that can be used to preserve prepubertal fertility, whereas mature oocyte cryopreservation is the main technique used for fertility preservation in post-menarche adolescents. However, a growing awareness and competence on the subject is beginning to emerge, especially amongst oncology pediatricians in Northern Europe [12,13].
In the United States, the 5-year overall survival rate for all invasive cancers between ages 15–39 is about 82.5% [14]. Despite the increased incidence of cancer cases, advances in early diagnosis and treatment have increased the survival rate [15].
Furthermore, in the last four decades, there has been a rising trend of delaying childbearing [16]. Hence, there is an increasing number of couples referred to Reproductive Medicine Centers for infertility problems after one partner has been treated for cancer. In these cases, the main cause of reduced fertility derives from the gonadotoxic effects of chemo/radiotherapy treatment [17]. As far as young women are concerned, there are two main concerns: the possible harmful effects of previous anticancer treatments on a future pregnancy, and the consequences pregnancy may have on the patient, particularly in the case of endocrine-sensitive neoplasms, even if to this day, there is no evidence of such possible adverse effects [18,19,20,21,22,23]. All reproductive-aged cancer patients must therefore be adequately informed of the risk of fertility loss/reduction as a consequence of anticancer treatments, and at the same time the strategies available to reduce this risk.

Hematological Malignancies

Hematological malignancies are 7–9% of cancers in women. Acute lymphocytic leukemia, acute myeloid leukemia, non-Hodgkin’s, and Hodgkin’s lymphoma are the most common hematological cancers in reproductive age. Therapies used to treat these cancers are often gonadotoxic action. Gonadotoxicity and risk of premature ovarian failure (POI) depend on the type and stage of disease, the type and dose of anticancer therapy, and the patient’s age at the beginning of treatment. Cytogenetic and molecular abnormalities can influence the response to treatment, making additional treatments necessary in refractory or relapsing cases. With increasing overall survival rates in hematological cancers, preservation of fertility in these patients has gained increasing attention. [24,25,26,27,28]
Validated options are freezing embryos and oocytes. In many cases, however, immediate cancer therapy is required, making it impossible to apply these options. Furthermore, ovarian stimulation is not possible in prepubertal age due to an inactive hypothalamus-pituitary-ovarian axis. Questionable options are: use of gonadotropin-releasing hormone analogues during chemotherapy, which, however, don’t protect the ovaries when TBI is used; surgical transposition of the ovary (oophoropexy) and gonadal shielding, which, however, does not protect ovaries when systemic chemotherapy is used.
Experimental options are: freezing ovarian tissues and following autotransplantation with potential contamination (especially in case of leukemia) of ovarian tissue with malignant haematological cells; in vitro maturation of oocytes in prepubertal age; an artificial ovary; stamina cells; neoadjuvant cytoprotective pharmacotherapy, and others [29,30,31,32,33,34,35,36]. Each of these options has advantages and disadvantages, so not all of them may be possible for all patients.
Preservation of fertility must be adapted to the patient [37,38,39,40,41]. A multidisciplinary approach between oncologists, gynecologists, reproductive biologists, and researchers is essential to ensure a high standard of care. Regarding fertility outcomes, ovarian stimulation followed by in vitro fertilization and embryo transfer is the most established strategy in postpubertal age. Pregnancy rates after frozen embryo transfer range from 36% to 61%, based on the patient’s age. Alternatively, oocyte freezing can be performed but randomized controlled trials show a pregnancy rate from 4.5 to 12% [42].

2. Ovarian Reserve in Female Cancer Survivors

Clinically, cancer treatments may compromise ovarian function, triggering delayed or arrested puberty, subfertility, infertility, and premature ovarian insufficiency (POI: menopause before the age of 40) [43,44].
Velez et al. first demonstrated, thanks to a population-based cohort study, that when comparing two groups of infertile women of the same age (34.8 years and 34.9 years, respectively), the infertility diagnosis was higher in cancer survivors (RR 1.30; 95% CI 1.23–1.37) compared to healthy women [45].
The reduction of ovarian reserve is a common cause of female infertility, especially among cancer survivors.
Cancer treatments may alter ovarian reserve for different reasons [46]. The ovary is particularly sensitive to the side effects of chemotherapy and radiotherapy. The risk of ovarian failure depends on the agent used and the dose administered [47], and ovarian failure can happen during or after treatment [48].
Traditionally, the ovarian reserve was studied with Follicle Stimulating Hormone (FSH). However, FSH has several limitations, such as variability between cycles and during the same menstrual cycle. Recently, Anti-Mullerian Hormone (AMH) and ovarian antral follicle count (AFC) have been used as the main indicators of ovarian reserve. In particular, AMH is produced by the granulosa cells of preantral and antral follicles and inhibits recruitment of primordial follicles into the follicular pool [49].
Typically, AMH levels decrease during chemotherapy with slight recovery 3–6 months later. AMH, before and after treatment, can be helpful in the management of young women with cancer [50]. In women with breast cancer, AMH levels before the treatment have recently been shown to be a useful predictor of the post-chemotherapy loss of ovarian function, in addition to age, which is the only other established individual predictor [51,52].
Other studies have evaluated the impact of cancer therapy on the ovarian reserve of cancer survivors. In 2012, Gracia et al. compared ovarian reserve markers in a population of reproductive-aged cancer survivors and similar-aged controls. This study revealed a significant reduction of AMH and AFC in cancer survivors and a dose-dependent relationship between cancer therapies and markers of ovarian reserve. Furthermore, AMH and AFC levels were lower among cancer survivors than in the same age control group, although FSH values and menstrual cycles were regular, hence reflecting subclinical follicular depletion. Therefore, according to these authors, AMH and AFC seem to be the most sensitive markers to evaluate ovarian reserve depletion. It seems likely, but uncertain, that decline of those markers is a good reflection of fertility and/or predictor of time of menopause in cancer survivors) [53].
According to the study by Nielsen et al., female childhood cancer survivors may be at risk of infertility. The study compared 10-year cancer survivors to controls and found that patients treated with gonadotoxic drugs had statistically significantly lower AFC values than the control group (median 15 vs. 18, p = 0.047) On the other hand, the anti-Mullerian hormone values were not significantly lower (AMH) (median 13.0 vs. 17.8 pmol/L) [54].
In 2017, Wenners et al. studied breast cancer patients undergoing chemotherapy and confirmed the importance of AMH, AFC, and age as predictors of ovarian function. Additionally, they studied the effect of smoking, demonstrating its negative impact on ovarian activity and reserve [55].
In 2018 Van den Berg et al. evaluated, on a large scale, the long-term effects of childhood cancer treatment on both ultrasonographic and hormonal markers of ovarian reserve. Their findings showed that the ovarian reserve markers were remarkably low only in a minority of childhood cancer survivors (CCSs), even after treatment with alkylating CT (6.5–13.0%). However, the proportions of CCSs with abnormal ovarian reserve markers increased faster after age 35, compared to the controls. Hence, these women should be advised to try to conceive earlier in their reproductive lifespan, as it might prove shorter than anticipated [56].
Moreover, they found several discrepancies between combinations of ovarian reserve markers. In particular, low AMH levels were not associated with abnormal AFC, FSH, or inhibin B values, especially in young women; therefore, AMH may decline earlier than other markers in the sequence of events leading to menopause.
In conclusion, further studies are needed to confirm recent evidence regarding the reproductive potential of these patients.
Future studies should focus on the clinical value of each marker of ovarian reserve, in order to predict the chance of future pregnancy or to make a firm diagnosis of premature menopause in cancer survivors.

3. Influence of Cancer on Ovarian Function

If the cancer “per se” may induce alterations of ovarian functions, these should be investigated.
Several studies have examined the fertility of women with cancer before chemo/radiotherapy and show mixed results. Different studies found a negative interference of cancer on ovarian function even before starting cancer treatment [57,58]. Pal et al. were the first that found an adverse effect of the tumor on ovarian function [59]. An observational study described how women with hormone-dependent cancer have a poorer response to controlled ovarian stimulation, with fewer oocytes retrieved than those with non-hormone-dependent cancer [60].
Alvarez and Ramanathan found significantly fewer oocytes in breast cancer patients than in patients with hematological cancer [61]. In a retrospective cohort study that included 155 women, Volodarsky-Perel et al. showed that women with grade G3 and stage III-IV breast cancer had significantly fewer numbers of mature oocytes than patients with grade G1-2 and at stage I-II of the disease. Also, the number of cryopreserved embryos was lower in women with grade G3. The same authors demonstrated that patients with high-grade cancer have fewer oocytes and embryos retrieved than those with low-grade cancer [62].
Similarly, Decanter et al. found fewer oocytes in women with cancer than in the age-matched control group [63]. Moria et al. showed that breast cancer patients had fewer retrieved oocytes than the control group [64].
However, most of the studies have not demonstrated relevant differences in ovarian reserve and oocytes retrieved between women with cancer and the healthy group. Almog et al., in their study with 81 women, stated that the ovarian function was not affected by the type of cancer [65,66,67,68,69].
These observations are thought to be linked to the systemic effect of cancer, causing higher catabolic status, increased stress hormones levels, and impaired function of the granulosa cells. Furthermore, invasive cancer infiltrates and destroys the surrounding tissue causing immune system reactions involving distant organs [70,71]. This systemic response conditions folliculogenesis, follicular cell proliferation, and oocyte maturation through the release of metalloproteases and growth factors [72,73].
Due to the close relationship between cancer and BRCA1 and 2 mutations, it is important to evaluate whether the cancer itself or the BRCA mutations underlie the reduced ovarian reserve in these women [74,75]. Porcu et al. demonstrated that BRCA 1 patients have a higher risk of premature ovarian failure compared to non-BRCA-mutated women with breast cancer and the healthy controls. BRCA1 groups have lower AMH levels and a significantly lower rate of mature oocytes. This effect seems to be independent of the probable interference of cancer. Therefore, the ovarian response after ovarian stimulation may be influenced by the presence of cancer [76].
More studies are needed to understand if there is a negative effect of cancer on ovarian function even before cancer treatment is started and if the type of cancer influences the ovarian response.

4. Effects of Cancer Treatment on Female Reproductive Function

The adverse effects of cancer treatment on female reproductive function are an increasing problem that affects the quality of life in survivors of childhood, adolescent, and young adult cancer.
The three principal anti-cancer treatments are surgery, radiotherapy, and chemotherapy.
In most cases, primary ovarian or uterine cancers are surgically removed together with these organs, leading to sterility. For these women, using donated eggs or surrogacy are the only chance to have a child [77].
Chemotherapy and radiotherapy may damage the reproductive system by destroying the hypothalamic–pituitary axis, the uterus, or the primordial and growing follicles within the ovaries.
The effects of chemotherapy and radiotherapy on the ovaries and uterus have a significant impact on the future fertility of childhood cancer patients as well as women up to the age of menopause.

4.1. Ovarian Damage

Cancer therapy can involve the administration of a wide variety of therapeutic protocols [78].
The American Society of Clinical Oncology published a useful classification of cancer treatments based on their level of gonadotoxicity and their consequent risk of permanent amenorrhea [79].
The level of gonadotoxicity depends on chemotherapeutic classes, dose, method of administration (oral versus intravenous), and combination of drugs; moreover, the toxicity changes with the type of disease, the woman’s age at the time of treatment, and the woman’s pre-treatment fertility.
“Highly gonadotoxic treatments” cause permanent amenorrhea in at least an 80% of cases; the most common of them are, for example, chemotherapy used to treat breast cancer in women over 40 (combinations of cyclophosphamide, methotrexate, fluorouracil, doxorubicin, epirubicin), external beam radiation to a field that includes the ovaries, or myeloablative conditioning for hematopoietic stem cell transplantation with high-dose alkylating agents (cyclophosphamide/busulfan) in combination with total body irradiation [79,80]. Different studies have reported that the risk of gonadal insufficiency after stem cell transplantation is related to a woman’s age; the risk of infertility is 65–95% in adult women, higher than in prepubertal girls (around 50%) because of higher ovarian reserve [81,82].
Treatments classified as “intermediate gonadotoxicity” involve a 40–60% risk of amenorrhoea. These include adjuvant chemotherapy for breast cancer in women aged 30–39 (combinations of cyclophosphamide, methotrexate, fluorouracil, doxorubicin, epirubicin) and escalated (second-line) chemotherapy used for Hodgkin’s lymphoma [60].
“Low” gonadotoxic cancer treatments, with a risk of amenorrhea < 20%, include first-line treatment for Hodgkin’s lymphoma (ABVD therapy) and treatment for acute lymphoblastic and myeloid leukemia [77].
Treatments considered “very low risk” or “no risk” are antimetabolites (such as methotrexate, cytarabine) and vinca alkaloids (vincristine, vinblastine), which do not cause damage to human follicles [77,80].

4.1.1. Impact of Chemotherapy

Many studies have investigated the type of damage of each chemotherapeutic agent on different cell types of the ovary.
These findings have reported that ovarian damage can occur via several mechanisms [83].
Apoptotic death of primordial follicle and growing follicles are caused by direct DNA damage (via DNA double-strand breaks and inter-strand crosslinking) [80,84].
Direct damage to the ovarian stroma causes fibrosis and hyalinisation of small blood vessels, resulting in ischemia and necrosis due to a reduction in ovarian blood volume and consequently indirect damage to follicles growth [80,85].
Indirect damage to primordial follicles is due to increased follicle activation. Meirow’s group explained this mechanism of enhanced follicular demise owing to accelerated folliculogenesis by proposing the “burnout theory” [86,87]. In particular, the destruction of growing follicles and thus the local reduction in AMH concentrations causes an upregulation in the PI3K/PTEN/Akt signaling pathway. The direct consequence is that waves of activated primordial follicles were excessively recruited into growing follicles, which are more susceptible to the direct damage of chemotherapy, resulting in follicle burnout [80]. Table 1.

4.1.2. Impact of Radiotherapy

Radiation therapy is one treatment modality for various types of malignancies, but unfortunately, exposure to ionizing radiation can lead to acute and long-term damage.
The effects of radiation to the abdominopelvic region depends on the dose intensity, fractionation, field of irradiation, and the age of the patient [77,88,89]. Women in the prepubertal period have ovaries relatively more resistant to gonadotoxicity [90].
The radiosensitivity of oocytes is high and differs according to their growth phase. In particular, dividing granular cells appear to be the main target of radiation-related gonadotoxicity.
Radiotherapy-induced ovarian injury also involves the stroma with vascular damage, leading to tissue atrophy and fibrosis [91].
The underlying mechanism induced by radiotherapy is both direct and indirect. The radiation induces a direct ionization of the cellular macromolecules, such as gonadal DNA causing multiple lesions within the helical turns of the DNA, which is referred to as “direct” damage. Radiotherapy also leads to the generation of reactive oxygen species (ROS) in cells, increasing oxidative stress and diminishing antioxidant defense mechanisms.
This imbalance may play a role in the etiology of radiotherapy-induced gonadotoxicity, which is defined as “indirect” damage [92].
Through these pathways, radiotherapy can affect healthy normal tissues in the ovaries and can influence the length of a women’s fertile lifespan and the timing of menopause.

4.2. Uterine Damage

Recent trials have reported that anti-cancer treatments can cause permanent injury to the uterus and compromise its ability to allow and sustain a healthy pregnancy [93].

4.2.1. Impact of Chemotherapy

The percentage of pregnancies obtained through egg donation or using one’s own eggs before anticancer treatment is lower than in the control group. Even though patients underwent assisted reproductive technologies (ART), the implantation rate and clinical pregnancy rate (4.9% and 9.5%, respectively) were statistically significantly lower, underlining that cancer therapy-induced damage to the uterus may contribute to infertility [93,94].
A small number of studies have demonstrated that chemotherapy exposure during childhood (especially to alkylating agents) is associated with a smaller uterine size and volume [95,96].
Despite this indirect evidence of uterine damage induced by chemotherapy, there is a paucity of data regarding the specific pathological mechanism through which these drugs can act. Currently, the damage to endometrial epithelium, myometrium, uterine vasculature, and the endometrial stem cell niche can only be extrapolated from animal models or laboratory and clinical findings on the analogous cell line of other human tissue (for example intestinal stem cells, cardiomyocites, skeletal muscle) [93].

4.2.2. Impact of Radiotherapy

Radiation to the uterus can impair reproductive function. Evidence has reported that radiotherapy can cause microvascular injury with endothelial damage and myometrial fibrosis compromising uterine growth and distensibility. Radiation may also damage muscle fibers and decrease pelvic floor muscle function [80]; when the woman’s exposure happens before puberty, stunted uterine growth and fibrosis can only be partially rescued by hormone replacement therapy [70]. Critchley et al. reported a shorter uterine length and a rare uterine blood flow in patients who received radiation during childhood compared to women with a history of POF (Primary Ovarian Failure) without radiation exposure.
The radiation consequences on the uterus are greater in the case of exposure at a young age, leading to greater risks for future pregnancy. The radiation dose that poses the greatest risk of reproductive failure is >45 Gy in adults or >25 Gy in childhood [80].

5. Oncofertility

Approximately one million new cases of cancer are diagnosed in reproductive-aged women every year [93,97].
In the last few decades, life expectancy of these patients has increased thanks to anticancer treatments. The increased survival rate, combined with increased age for childbearing, has led to the occurrence of side effects such as fertility problems [98].
Diagnosis and treatment of tumors can often cause fertility problems in women. It has been estimated that 70–75% of cancer survivors are interested in parenthood and that 80% of them are affected by reduced fertility [99].
Despite patients’ interest in parenthood, the percentage of patients who receive correct information varies from 51% to 95%, and the percentage of patients who access fertility preservation techniques is low [100,101].
Fertility loss can cause devastating emotional reactions in women impacting their plans for the future. Various studies demonstrate that discussing fertility preservation options can improve quality of life and can contribute to psychological health [102,103].
The creation of an oncofertility team around the patient would allow this conversation to happen at the appropriate time and would reduce fertility loss in cancer patients [36].
Oncologists should inform patients about impaired fertility risk and should provide information on strategies available to preserve it.
Fertility preservation strategies in females depend on age, type of treatment planned, diagnosis, presence of a partner, time available before starting treatment, and potential for cancer to metastasize to the ovaries [79].
After consultation with a hematologist, oncologist, and specialist in reproductive medicine, an adequate consult can be carried out to evaluate ovarian reserve and gonadotoxicity of the therapies and to propose an appropriate fertility preservation technique [97].

5.1. Fertility Preservation Options

Different fertility preservation strategies can be proposed (Figure 1):
  • Hormone Protection by Suppressing Ovaries
  • Oophoropexy
  • Embryo Storage, Oocyte Storage
  • Ovarian Tissue Storage
  • Fertility Sparing Surgery

5.1.1. GnRH-Analogues

In cancer patients who are candidates for chemotherapy, the use of GnRH analogues should be proposed but not considered as an alternative to cryopreservation [104].
GnRH analogues are used as chemoprotectors; used during chemotherapy they induce menopause, suppressing the ovarian cycle.
In 2018 and 2020, ASCO Guidelines and the European Society for Medical Oncology, respectively, recommended that GnRHa use should be offered to all cancer patients who desire to preserve fertility. [105]
In 2018, the British Fertility Society affirmed that GnRHa should be started immediately before chemotherapy and continued for the duration of therapy. [105]
GnRH analogues arrest ovarian cells in the G0 phase inducing cellular quiescence and making these cells less responsive to chemotherapy [106,107]; this treatment has shown effects in reducing primary ovarian insufficiency (POI) risk, increasing pregnancy rates, and having no negative effects on the cancer’s outcome.
The use of GnRH analogues can be also proposed in patients with hormone receptor-positive disease with safety [108].
Several studies have shown that the use of Goserelin has preserved fertility in a high percentage of patients affected by breast cancer [109,110].
The use of Goserelin reduces premature ovarian failure risk as well as prevalence of amenorrhoea and also improves disease-free survival and overall survival [111,112].
AMH can be used to evaluate the GnRHa protective effect on fertility [105].

5.1.2. Oophoropexy

Ovarian transposition (oophoropexy) consists of surgical removal of the ovaries from the irradiation site.
This strategy can be proposed to patients who are candidates for pelvic radiotherapy (children and pre-menopausal women who desire to preserve fertility and prevent early menopause), in cases of gynecological or hematological cancers, such as cervical cancer and Hodgkin’s lymphomas, and in cases of medulloblastoma, urogenital rhabdomyosarcoma, pelvic sarcomas, Wilm’s tumor, and rectal cancer [107,113,114].
Before the procedure, a pelvic MRI should be performed to ensure that the tumor does not involve the ovarian region.
Ovarian transposition can be performed by laparoscopy or laparotomy (in case of concomitant resection of the tumor). One or both ovaries are relocated, either medially (behind the uterus in the case of Hodgkin’s Lymphoma), laterally, near the inguinal ring, in the paracolic gutters, or near the lower kidney pole (in the case of urogenital tumors, medulloblastoma, and rhabdomyosarcoma), or to any distant site.
At the end of the procedure, two metal clips should be applied to the transposed ovaries to make them visible on imaging.
Possible complications of oophoropexy are: torsion of the ovarian blood vessels, development of benign ovarian cysts, and subsequent chronic pain and ovarian and abdominal wall metastases at the trocar site [113,114,115].
Success rate of this treatment, in terms of preserved ovarian function, varies between 60% and 83% and several spontaneous pregnancies after this procedure are described. Nevertheless, some authors claim that these patients could require assisted reproductive technologies because of increased distance between the ovary and the fallopian tube.
This increased distance compromises oocyte migration through the Fallopian tube and impairs fertility [114].

5.1.3. Embryo and Oocyte Cryopreservation

Embryo cryopreservation is one of the options to preserve fertility.
An oocyte and a sperm cell (obtained from a male partner or sperm donor) are needed to create an embryo [107].
Embryo cryopreservation is a good choice for patients with a stable relationship because of better pregnancy outcomes [116]. However, not all women have a stable relationship at the time of diagnosis; in this case, oocyte cryopreservation is a valid alternative, giving women an opportunity to procreate with a chosen partner in the future, without the need to fertilize the oocyte after retrieval [117,118].
Embryo and oocyte cryopreservation cause a two-week delay in chemotherapy initiation [35,36].
Ovaries are stimulated with daily injections of follicle-stimulating hormone and stimulation can be started at any point in the menstrual cycle.
Follicle growth is monitored by ultrasounds and blood tests (serum estradiol and progesterone levels). Ovulation is induced with an HCG injection when appropriate and oocytes are collected by transvaginal aspiration (with ultrasound guidance). The oocytes retrieved can be cryopreserved or fertilized in vitro to obtain embryos [79,119].
Oocyte and embryo cryopreservation, performed prior to anti-cancer therapies, are defined by the American Society of Clinical Oncology (ASCO) and the European Society for Medical Oncology (ESMO) as the most appropriate procedures to ensure motherhood in cancer survivors [120].
Among reproductive-aged women, breast cancer is one of the most frequent. In some cases, this is a hormone-sensitive cancer because of the expression of estrogen receptors.
In patients with these tumors, exposure to high estrogen levels can be risky. In order to avoid this exposure, oocytes can be recovered from a natural cycle or from ovarian stimulation protocol with aromatase inhibitor or tamoxifen (chemoprotective agents that have ovulation-inducing properties) [106,121].

5.1.4. New Strategies

Among new strategies, immature oocytes retrieval is an experimental technique that involves cryopreservation of immature oocytes or matured in vitro.
These oocytes can be used for vitrification or to obtain embryos by ICSI with partner sperm. This strategy allows for a reduction in the time needed for preservation and avoids exposure to hyperestrogenism caused by stimulation [122].
Mature oocytes can be obtained by in vitro maturation of immature oocytes (IVM) or in vitro activation of dormant follicles (IVA) [120].
Achieving pregnancy is possible using oocytes maturated in vitro [123] but pregnancy rates are lower in patients who have used embryos obtained from immature oocytes or oocytes matured in vitro than those who have used embryos obtained from mature oocytes [122].
After treatment, patients should be informed about their ovarian function using different parameters (AMH, FSH, estradiol) in order to decide whether to use cryopreserved oocytes or to start a new cycle [123].
The discovery of ovarian stem cells in the ovarian cortex, first found in mammals and then also in women, opened new chances to preserve fertility.
Despite these findings, in vitro maturation of ovarian stem cells (OSC)s to oocyte-like cells (OLCs) still needs to be investigated for future clinical use in female cancer survivors [120].

5.1.5. Ovarian Tissue Cryopreservation

Ovarian tissue cryopreservation (OTC) is the only fertility preservation strategy available in prepubertal patients and those who cannot postpone treatment [35,124].
The entire ovary or part of this can be collected laparoscopically at any period of menstrual cycle. Obtained tissue is sliced and cryopreserved.
When patients are declared free from cancer with a good prognosis, the ovarian tissue is thawed, tested to assess the absence of cancer cells, and reimplanted orthotopically or heterotopically [125].
Although the use of this technique has shown good results in adult patients, ex vivo maturation of ovarian tissue taken in childhood and the subsequent auto-transplantation is still considered experimental [126].
One of the problems of ovarian tissue transplantation is that revascularization occurs after a few days from the time of the procedure. This causes tissue ischemia and a loss of more than 60% of the primordial follicles [106].
Local administration of antiapoptotic and angiogenic factors can improve the revascularization of ovarian tissue [127].
Another problem with ovarian tissue transplantation is the possibility of transferring cancer cells in patients. Even though the tissue is controlled before freezing and before transplantation, the risk of tumor cell transmission remains.
This risk increases in certain tumors, leukemia being a case in point [128,129].
Therefore, this treatment is only proposed to patients with a low risk of ovarian metastasis [107].
Tumor cell transmission can be reduced by transferring primordial follicles onto an artificial tissue to replace native organs [130].
Depending on the number of follicles in cryopreserved tissue, ovarian function resumes after transplantation for 4–5 years on average and in some cases up to 7 years [118,131].
The first pregnancy after this fertility preservation technique was obtained in 2004; pregnancy and live birth rates are growing exponentially over the years. [35,132].
Patients who have undergone this fertility preservation strategy often have undetectable AMH levels, but spontaneous pregnancies after orthotopic transplantation have been reported [105].
AMH level is not associated with the duration of ovarian graft function or the possibility to achieve pregnancy in these women [133].

5.2. Success Rates

Cryopreservation of embryos, oocytes, ovarian tissue, or fertility preservation does not guarantee achieving a pregnancy in the future [36].
Among survivors, pregnancy rates are about 40% lower than the general population [16].
Several studies have described success rates after fertility preservation techniques (Table 2):
  • Live birth rate (LBR) ranges from 20% to 45% in patients undergoing embryos-cryopreservation [134,135].
  • Live birth rate varies from 20% to 50%, depending on age, in women undergoing oocytes cryopreservation [136,137,138].
  • Live birth rate (LBR) ranges from 18.2% to 41.6% in patients undergoing ovarian tissue cryopreservation and reimplantation [132,137,139,140,141,142,143].
  • Live birth rate (LBR) revolves around 8.9% in women undergoing in vitro maturation (IVM) [120].
  • Live birth rate (LBR) revolves around 7% in patients undergoing in vitro activation (IVA) [120].
  • Few studies have been carried out on pregnancy rates after oophoropexis. Despite this, pregnancies have been reported after this procedure by various authors [114,144].

6. Fertility Sparing Surgery for Gynecologic Cancer and Reproductive Outcomes

All standard therapies for gynecologic cancer strongly impact a woman’s childbearing potential, with severe social and psychological effects. Preservation of fertility in some cases is possible and should be discussed during treatment planning, with reproductive-aged women with early-stage gynecologic cancer and with those who desire to preserve fertility.

6.1. Cervical Cancer

The standard surgical treatment for cervical cancer is a destructive and disabling surgery consisting of a radical hysterectomy and pelvic lymphadenectomy. However, in selected young patients, fertility preservation may be possible. In particular, it could be offered to patients with early-stage disease (tumor diameter ≤ 2 cm), negative nodes, and non-aggressive histological subtypes [145]. The CONTESSA/NEOCON-F multi-center study is intended to demonstrate the possibility of a conservative surgery to preserve fertility in patients with tumors diameter of 2–4 cm (FIGO 2018 stage IB2) with no lymphovascular disease after neoadjuvant chemotherapy [146] as well. According to preliminary data, live birth rates after ART in stage IB2 is only 9% [147]. Moreover, a retrospective study by Tesfai et al. evaluated the safety and the oncological and obstetric outcomes in patients with cervical tumors > 2 cm (FIGO stage IB-IIA) pre-treated with neoadjuvant chemotherapy before abdominal radical trachelectomy. In 15 out of the 19 patients treated (74%), fertility was successfully preserved and three of them became pregnant [148]. There are several fertility preservation techniques available: Dargent’s procedure; simple trachelectomy or cone resection; neoadjuvant chemotherapy with conservative surgery; and laparotomic, laparoscopic, and robot-assisted abdominal radical trachelectomy. In a recent systematic review, Bentivegna et al. examined these six fertility-sparing options to establish the best approach in terms of oncological outcomes and live birth success. All of these procedures showed optimum fertility results and were also dependent on the experience of the surgical team, but the best outcomes have been seen in patients with excellent prognostic factors, such as small tumor size, negative or little lymphovascular space involvement, and absence of positive margins [149]. These patients should be treated with conventional surgery alone, as their cases carry a minimum risk of recurrence.

Reproductive Outcomes

In a systematic review by the same group, the fertility rates and obstetric outcomes in a total of 2777 patients submitted to FSS (Fertility-Sparing Surgery) and 944 ensuing pregnancies were evaluated. The pregnancy and live birth rates were promising: 55% and 70%, respectively. The best approach in terms of fertility was found to be vaginal or minimally invasive radical trachelectomy. However, the live birth rate was similar in both invasive and non-invasive FSS. On the other hand, the prematurity rate was significantly lower in patients who had undergone a simple trachelectomy/cone resection rather than other conservative procedures [150]. In fact, obstetric complications seem to be higher in patients submitted to FSS. In a recent study, complications such as cervical stenosis and Asherman syndrome emerged as probable negative factors on fertility rate [151].
Nezhat et al. evaluated the reproductive outcomes in 3044 patients who underwent fertility-sparing surgery for early-stage cervical cancer (stage IA1–IB1), with encouraging results: the mean clinical pregnancy rate was 55.4%. The best surgical approach with the highest clinical pregnancy rate was vaginal radical trachelectomy (67.5%). The risk of recurrence and cancer death were very low (3.2% and 0.6%, respectively) after a median period of follow-up of 39.7% [152].
Schuurman et al. reported similar results: a vaginal approach of FSS in cervical cancer turned out to be the best procedure in terms of pregnancy and live birth rates. A possible explanation may be a less invasive resection of the cervix and parametrium [153].
Nevertheless, in a recent study, Tamauchi et al. investigated the response to controlled ovarian stimulation in patients after radical trachelectomy.
They compared a group of 14 patients who underwent ovarian stimulation after radical trachelectomy with a control group who had male infertility or unexplained infertility. Estradiol levels and number of oocytes retrieved were lower in patients undergoing a trachelectomy than in the control group, despite the increased use of gonadotropin (3527.5 IU, SD 1313.4 vs. 2670.8 IU, SD 905.1, p = 0.001) [154].
In conclusion, fertility-sparing surgery must be considered a valid alternative to traditional radical hysterectomy for early-stage cervical cancer in women who desire fertility preservation. Adequate reproductive counseling before FSS is mandatory since many women are not conscious of the complications that could follow this kind of surgery and that could negatively impact the reproductive and pregnancy experience (Table 3).

6.2. Endometrial Cancer

Although rare, the incidence of endometrial cancer (EC) and atypical complex hyperplasia (ACH) is increasing in reproductive-aged women [155]. For both EC and ACH, the standard of treatment consists of a hysterectomy with bilateral salpingo-oophorectomy (ESGO/ESTRO/ESP guidelines for the management of patients with EC). However, the Society of Gynecologic Oncology’s Clinical Practice Committee describes as a perfect candidate for conservative treatment a patient who firmly desires fertility-sparing management with a well-differentiated (grade 1) tumor, stage FIGO IA without invasion of myometrium on MRI, absence of lymphovascular invasion, without intra-abdominal disease or adnexal mass, and no contraindications for medical management [156,157]. In these cases, according to the guidelines for the management of patients with EC, the options for conservative management are: progestogen (PG) therapy, LNG-IUD alone, or in combination with other hormonal agents [158]. The most successful fertility-sparing option is hormone therapy since ACH and EC, which express receptors for estrogen and progesterone, showed a higher chance of retaining fertility [159,160]. Medroxyprogesterone acetate (MPA) and Megestrol acetate (MA) are progestogen drugs commonly used in different doses (MPA 400–600 mg/day or MA 160–320 mg/day for a minimum of 6 months) for AEH and early-stage EC, but there is no consensus on the optimal treatment regimen. Nevertheless, in a recent systematic review, MA has been associated with higher rates of remission compared to MPA and other hormonal treatments [161]. Several studies have been conducted with the aim of estimating the oncological and reproductive outcomes of reproductive-aged women with EC or ACH.
Wei et al., demonstrated that in these patients, conservative treatments can achieve good complete response rates (71% with PG and 76% with IUD alone); however, the pregnancy outcomes are worse in patients treated with IUD alone, rather than with progestin (18% vs. 34%) [162]. The same group reported a response rate for patients with LNG-IUD of 76%, and the recurrence rate was 9%. The pregnancy rate was 18%, and 14% for delivering a live newborn. When a combined therapy of oral PG plus LNG-IUD was used, then the complete response rate was 87%. [162].
However, a significant portion of patients experience recurrence after achieving complete remission. Traditional surgery is recommended in this case, but some patients who have not obtained pregnancy at the time of recurrence want to preserve their fertility. When recurrent diseases involve well-differentiated tumors confined to the endometrium, the second round of fertility-sparing therapy could be performed. A recent work conducted by Chen described fertility-preserving re-treatment that can have a promising response, with 88.6% of CR and a pregnancy rate after CR of 26.5% [163].
In the case of hysterectomy, or after radiation that will preserve the uterus but cause non-functionality in terms of future implantation and pregnancy, uterine transplantation should be offered to give women a chance to regain motherhood [164].

Reproductive Outcomes

Fertility outcomes in patients affected by endometrial cancer are not well described. A study by Park at al reviewed the medical history of 141 women with stage IA, grade 1 endometrioid adenocarcinoma who received progestin therapy. Seventy women (49.6%) tried to conceive, with a pregnancy rate of 73% (51 patients) and 58 live neonates [165]. On the other hand, Maggiore examined the fertility outcomes of levonorgestrel-releasing intra-uterine system in patients with EC or ACH. Among the 41 women who obtained a total surgical response, only 14 (34.14%) achieved a pregnancy (6 spontaneously and 8 through IVF) [166]. In a recent metanalysis by Harrison et al., a cohort of women aged ≤ 45 with EC or ACH was investigated. They received a standard treatment or a conservative treatment by progestin therapy. The live birth rate in these women was lower than that described in the literature, accounting for 11.6% of patients who received fertility-sparing management. In addition, among patients who achieved a pregnancy, 54% used some form of assisted fertility treatment [167] (Table 3).

6.3. Ovarian Cancer

Approximately 3 to 13% of women with epithelial ovarian cancer are younger than 40 [168]. Premenopausal women with early-stage epithelial ovarian cancer may benefit from the feasibility and safety of FSS as they have a good cancer prognosis. [169]. In a systematic review, Bentivegna et al. described the conservative treatments that preserve reproductive function can be safe in women with unilateral epithelial cancers for stage IA grade 1 and grade 2, and IC grade 1, according to the FIGO staging system. Other patients with less favorable prognostic factors (bilateral ovarian involvement, any grade 3 disease, and stages IB, IC2, and IC3) must be informed that the safety of FSS is not confirmed. FSS is contraindicated for stage II/III (any histologic subtype) [170] anyway. The surgical approach consists of unilateral salpingo-oophorectomy with peritoneal washings and pelvic and para-aortic lymphadenectomy, as well as omentectomy. The risk of recurrence is about 8% and 10% in stages IA and IC. However, several retrospective studies confirmed that the overall survival in patients with early-stage epithelial ovarian cancer 10 years after diagnosis is up to 90% and found that survival was similar when comparing women who underwent fertility-sparing surgery with patients who underwent conventional surgery [171,172,173]. Borderline tumors represent a small but significant part of all primary ovarian tumors. It is possible to treat women with borderline tumors in clinical stage I with cystectomy or unilateral oophorectomy. The recurrence rates are 10–15% in a median 5-year follow-up, and the mortality rate is <1%. For women with bilateral borderline ovarian tumors, unilateral oophorectomy and contralateral cystectomy have been proposed as a reasonable technique in women with the desire to preserve fertility [174]. Ultraconservative management consisting of bilateral cystectomy has been proposed with excellent fertility results [175]. Non-epithelial cancers of the ovary represent approximately 5% of all ovarian malignancies and include ovarian germ cell tumors and ovarian sex cord-stromal tumors [176]. Malignant ovarian germ cell tumors (MOGCTs) are the most commonly found in women of reproductive age, and the FSS is generally the first choice of approach since these tumors are chemosensitive [177]. Experience to date indicates that the cure rates for women with early-stage MOGCTs approach 100% and at least 75% in advanced-stage disease [178].
Sex cord-stromal tumors generally have an indolent course and occur more likely in postmenopausal women. Frequently, if diagnosed in young women, these tumors are often seen as an early-stage disease and FSS is possible in a high percentage of young patients [179]. The oncological outcomes of tumors treated conservatively are reassuring, with similar results of conservative and radical surgery [180].
After fertility sparing treatment for non-epithelial cancer, there is a risk of POF. For this reason, it is necessary to accurately identify, through the Edinburgh selection criteria, the few girls and young women who will develop premature ovarian insufficiency, and who will be candidates for ovarian tissue cryopreservation (OTC). In the criteria, the conditions listed for OTC are as follows: a realistic chance of surviving for 5 years; a high risk of POI > 50%; and mild, non-gonadotoxic chemotherapy is acceptable if the patient is younger than 15 years of age [124].

Reproductive Outcomes

Any surgery that involves the ovaries may impact ovarian reserve and reduce the possibility of spontaneous conception. For these reasons, women who undergo fertility-sparing surgery may require the help of assisted reproductive technology, even if spontaneous conceptions have also been reported. Literature data regarding the pregnancy rate of women after FSS accounts for approximately 30% of all patients; however, if one includes only women of reproductive age, pregnancy rate increases and range from 66 to 100%, showing no relevant impairments after FSS [151]. A recent systematic review reported the reproductive outcomes of patients diagnosed with early-stage epithelial ovarian carcinoma (EOC), borderline ovarian tumors (BOT), or nonepithelial ovarian carcinoma (NEOC), are candidates for fertility-sparing surgery or conventional surgery. The fertility rates vary depending on the tumor type and were 76 to 96% for EOC, 65 to 95% for NEOC, and 23 to 100% BOT. The authors confirmed the safety of FSS procedures since the 5-year overall survival rates were almost overlapped between patients who underwent FSS (87.5%) and those who had conventional surgery (91.8%).
Data in the literature regarding reproductive outcomes after MOGCTs are reassuring, between 66.6% and 95%. Regarding the pregnancy rate in the case of juvenile granulosa cell tumors, the data is encouraging, compared to adult granulosa cells tumors (42.3% vs. 28.5%) [180,181]. In fact, although preserving reproductive function is a goal of FSS, it has been reported that one in four women want to conceive after surgery [182]. These data require careful advice for women who decide to undergo FSS procedures rather than traditional ones, because while exposing themselves to an increased risk of cancer, these treatments do not necessarily guarantee future pregnancy for women. (Table 3).

6.4. Vulvar Cancer

The risk of vulvar cancer increases with a women’s age and it is 10 times higher in patients over 75 years of age [183]. Less than 20% of cases are in women younger than age 50 [156]. FSS should include less destructive surgery, and oophoropexy could be proposed to patients who are candidates for pelvic radiotherapy and OTC before chemotherapy, if possible. Chemotherapy can be associated with a lower risk of local long-term complications due to reduced local tissue damage [183]. No literature data are available about fertility outcomes in young women affected by vulvar cancer.

7. Special Categories

7.1. Fertility Preservation Post Cancer Diagnosis in Young Women

After cancer diagnosis, there is no suitable period to get pregnant.
Completion of treatments, age of the patient, risk of relapse, ovarian function, and patient’s desire should be considered to determine the most appropriate time to conceive [16].
Discussion on patient fertility should be held both at the time of cancer diagnosis and at the end of treatments.
AMH should be used to evaluate chemotherapy-associated ovarian dysfunction after treatments, but it is well known that AMH does not predict the probability to achieve pregnancy [105,184].
In the event of a reduced ovarian reserve (DOR), women should be advised that a low ovarian reserve does not affect the spontaneous conception rate if they wish to seek a short-term pregnancy. The oncologist should only reassess the safety of conception, with the patient, if the appropriate conditions are met and discuss the possibility of undergoing assisted reproductive technology (ART) procedures in case of infertility [36].
Women who presented cancer at a young age and who survived hide their fears about their reproductive health. Counseling about post-cancer preservation should be proposed and should not be seen as an alternative to fertility preservation at the time of cancer diagnosis, but as an additional option.
Lehman et al. described fertility preservation in women after cancer with preserved ovarian reserve, diminished ovarian reserve (DOR), and POI [185]. Eleven subjects with DOR (52%) stored their eggs. Filippi et al. described oocyte cryopreservation in female cancer survivors, aged >18 years, who were diagnosed with DOR. Oocyte cryostorage was performed in 9 (69%) women [186].

7.2. Fertility Preservation in Women with Hereditary Cancer Syndromes

Hereditary cancer syndromes (HCS) are a group of genetic conditions associated with an increased risk of developing cancer during lifespan. Fertility preservation to prevent the effects of cancer treatments on ovarian function should be preceded by personalized and early fertility counseling. Carriers should be informed about oocytes cryopreservation, either before or after tumor diagnosis, but should also be aware of the risk of transmission [187]. There are no specific guidelines about fertility preservation in HCS, and only few papers, mostly focusing on BRCA-1/2 carriers, are reported. The ASRM guidelines give few recommendations for BRCA carriers. They recommend informing carriers of the possibility to undergo prenatal diagnosis in the case of natural conception or preimplantation genetic testing (PGT) and oocytes donation in the case of IVF [188].

8. Conclusions

Counseling of cancer diagnosis and treatment should be associated with adequate counseling about fertility preservation before treatment initiation, but also after cancer treatments. Women with cancer, regardless of their age, should be educated on the risk of endocrine dysfunction, cessation of ovarian activity, and depletion of the ovarian reserve. Furthermore, they should be assigned to fertility specialists to decide on fertility preservation options. In postpubertal women, where the beginning of chemotherapy can be postponed by at least 10–12 days, oocyte cryopreservation should be offered. OTC should be proposed instead when immediate chemotherapy is necessary both in prepubertal and postpubertal women. Evidence of the trend over time of ovarian reserve loss is insufficient to predict the duration of the fertile period. Therefore, it is important to inform women about the post-cancer depletion of the ovarian reserve in an attempt to induce them to modify their reproductive plans. If they have a partner, motherhood could be anticipated. In cases of single women not seeking pregnancy in the near future, one should consider the option of social freezing, as incidence of pregnancy decreases significantly over the years amongst cancer-treated patients.

Author Contributions

Conceptualization, C.D.T. and L.M.; investigation, L.M., C.D.T.; writing—original draft preparation, L.M., C.D.T., G.G., G.M., A.C. and A.F.; writing—review and editing C.D.T., G.G., A.C., A.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

All data involved in this study will be made available by the corresponding author upon request.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. You, L.; Lv, Z.; Li, C.; Ye, W.; Zhou, Y.; Jin, J.; Han, Q. Worldwide cancer statistics of adolescents and young adults in 2019: A systematic analysis of the Global Burden of Disease Study 2019. ESMO Open 2021, 6, 100255. [Google Scholar] [CrossRef] [PubMed]
  2. Mahajan, N. Fertility preservation in female cancer patients: An overview. J. Hum. Reprod. Sci. 2015, 8, 3–13. [Google Scholar] [CrossRef] [PubMed]
  3. Rossi, L.; Mazzara, C.; Pagani, O. Diagnosis and Treatment of Breast Cancer in Young Women. Curr. Treat. Options Oncol. 2019, 20, 86. [Google Scholar] [CrossRef] [PubMed]
  4. Francis, P.A.; Regan, M.M.; Fleming, G.F.; Láng, I.; Ciruelos, E.; Bellet, M.; Bonnefoi, H.R.; Climent, M.A.; Da Prada, G.A.; Burstein, H.J.; et al. Adjuvant ovarian suppression in premenopausal breast cancer. N. Engl. J. Med. 2015, 372, 436–446. [Google Scholar] [CrossRef] [Green Version]
  5. Smallridge, R.C.; Copland, J.A. Anaplastic thyroid carcinoma: Pathogenesis and emerging therapies. Clin. Oncol. 2010, 22, 486–497. [Google Scholar] [CrossRef] [Green Version]
  6. DiSano, J.A.; Schaefer, E.W.; Kjerulff, K.; Hollenbeak, C.S.; Pameijer, C.R. Pregnancy after a melanoma diagnosis in women in the United States. J. Surg. Res. 2018, 231, 133–139. [Google Scholar] [CrossRef]
  7. Arbyn, M.; Weiderpass, E.; Bruni, L.; de Sanjosé, S.; Saraiya, M.; Ferlay, J.; Bray, F. Estimates of incidence and mortality of cervical cancer in 2018: A worldwide analysis. Lancet Glob. Health 2020, 8, e191–e203. [Google Scholar] [CrossRef] [Green Version]
  8. Guillon, S.; Popescu, N.; Phelippeau, J.; Koskas, M. A systematic review and meta-analysis of prognostic factors for remission in fertility-sparing management of endometrial atypical hyperplasia and adenocarcinoma. Int. J. Gynaecol. Obstet. 2019, 146, 277–288. [Google Scholar] [CrossRef]
  9. Shandley, L.M.; McKenzie, L.J. Recent Advances in Fertility Preservation and Counseling for Reproductive-Aged Women with Colorectal Cancer: A Systematic Review. Dis. Colon Rectum 2019, 62, 762–771. [Google Scholar] [CrossRef]
  10. Salama, M.; Anazodo, A.; Woodruff, T.K. Preserving fertility in female patients with hematological malignancies: A multidisciplinary oncofertility approach. Ann. Oncol. 2019, 30, 1760–1775. [Google Scholar] [CrossRef] [Green Version]
  11. Barton, S.E.; Najita, J.S.; Ginsburg, E.S.; Leisenring, W.M.; Stovall, M.; Weathers, R.E.; Sklar, C.A.; Robison, L.L.; Diller, L. Infertility, infertility treatment, and achievement of pregnancy in female survivors of childhood cancer: A report from the Childhood Cancer Survivor Study cohort. Lancet Oncol. 2013, 14, 873–881. [Google Scholar] [CrossRef] [Green Version]
  12. Overbeek, A.; van den Berg, M.; Louwé, L.; Wendel, E.; Ter Kuile, M.; Kaspers, G.; Stiggelbout, A.; van Dulmen-Den Broeder, E.; Hilders, C. Practice, attitude and knowledge of Dutch paediatric oncologists regarding female fertility. Neth. J. Med. 2014, 72, 264–270. [Google Scholar] [PubMed]
  13. De Lambert, G.; Poirot, C.; Guérin, F.; Brugières, L.; Martelli, H. Preservation of fertility in children with cancer. Bull. Cancer 2015, 102, 436–442. [Google Scholar] [CrossRef] [PubMed]
  14. Keegan, T.H.; Ries, L.A.; Barr, R.D.; Geiger, A.M.; Dahlke, D.V.; Pollock, B.H.; Bleyer, W.A.; National Cancer Institute Next Steps for Adolescent and Young Adult Oncology Epidemiology Working Group. Comparison of cancer survival trends in the United States of adolescents and young adults with those in children and older adults. Cancer 2016, 122, 1009–1016. [Google Scholar] [CrossRef]
  15. De Melo, A.S.; de Paula, C.T.V.; Rufato, M.A.F.; Rufato, M.C.A.C.; Rodrigues, J.K.; Ferriani, R.A.; Barreto, J. Fertility optimization in women with cancer: From preservation to contraception. JBRA Assist. Reprod. 2019, 23, 418–429. [Google Scholar]
  16. Peccatori, F.A.; Azim, H.A., Jr.; Orecchia, R.; Hoekstra, H.J.; Pavlidis, N.; Kesic, V.; Pentheroudakis, G.; ESMO Guidelines Working Group. Cancer, pregnancy and fertility: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2013, 24, 160–170. [Google Scholar] [CrossRef]
  17. Barton, S.E.; Missmer, S.A.; Berry, K.F.; Ginsburg, E.S. Female cancer survivors are low responders and have reduced success compared with other patients undergoing assisted reproductive technologies. Fertil. Steril. 2012, 97, 381–386. [Google Scholar] [CrossRef] [PubMed]
  18. Boyle, K.E.; Vlahos, N.; Jarow, J.P. Assisted reproductive technology in the new millennium: Part II. Urology 2004, 63, 217–224. [Google Scholar] [CrossRef]
  19. Howell, S.; Shalet, S. Gonadal damage from chemotherapy and radiotherapy. Endocrinol. Metab. Clin. N. Am. 1998, 27, 927–943. [Google Scholar] [CrossRef]
  20. Gelber, S.; Coates, A.S.; Goldhirsch, A.; Castiglione-Gertsch, M.; Marini, G.; Lindtner, J.; Edelmann, D.Z.; Gudgeon, A.; Harvey, V.; Gelber, R.D. International Breast Cancer Study Group. Effect of pregnancy on overall survival after the diagnosis of early-stage breast cancer. J. Clin. Oncol. 2001, 19, 1671–1675. [Google Scholar] [CrossRef]
  21. Córdoba, O.; Bellet, M.; Vidal, X.; Cortés, J.; Llurba, E.; Rubio, I.T.; Xercavins, J. Pregnancy after treatment of breast cancer in young women does not adversely affect the prognosis. Breast 2012, 21, 272–275. [Google Scholar] [CrossRef] [PubMed]
  22. Azim Jr, H.A.; Santoro, L.; Pavlidis, N.; Gelber, S.; Kroman, N.; Azim, H.; Peccatori, F.A. Safety of pregnancy following breast cancer diagnosis: A meta-analysis of 14 studies. Eur. J. Cancer 2011, 47, 74–83. [Google Scholar] [CrossRef] [PubMed]
  23. Azim Jr, H.A.; Kroman, N.; Paesmans, M.; Gelber, S.; Rotmensz, N.; Ameye, L.; De Mattos-Arruda, L.; Pistilli, B.; Pinto, A.; Jensen, M.B. Prognostic impact of pregnancy after breast cancer according to estrogen receptor status: A multicenter retrospective study. J. Clin. Oncol. 2013, 31, 73–79. [Google Scholar] [CrossRef]
  24. Leader, A.; Lishner, M.; Michaeli, J.; Revel, A. Fertility considerations and preservation in haemato-oncology patients undergoing treatment. Br. J. Haematol. 2011, 153, 291–308. [Google Scholar] [CrossRef] [PubMed]
  25. Donnez, J.; Dolmans, M.M. Preservation of fertility in females with haematological malignancy. Br. J. Haematol. 2011, 154, 175–184. [Google Scholar] [CrossRef]
  26. Shapira, M.; Raanani, H.; Cohen, Y.; Meirow, D. Fertility preservation in young females with hematological malignancies. Acta Haematol. 2014, 132, 400–413. [Google Scholar] [CrossRef]
  27. Loren, A.W. Fertility issues in patients with hematologic malignancies. Hematol. Am. Soc. Hematol. Educ. Program 2015, 2015, 138–145. [Google Scholar] [CrossRef] [Green Version]
  28. Anderson, R.A.; Remedios, R.; Kirkwood, A.A.; Patrick, P.; Stevens, L.; Clifton-Hadley, L.; Roberts, T.; Hatton, C.; Kalakonda, N.; Milligan, D.W.; et al. Determinants of ovarian function after response-adapted therapy in patients with advanced Hodgkin’s lymphoma (RATHL): A secondary analysis of a randomized phase 3 trial. Lancet Oncol. 2018, 19, 1328–1337. [Google Scholar] [CrossRef] [Green Version]
  29. Salama, M.; Winkler, K.; Murach, K.F.; Seeber, B.; Ziehr, S.C.; Wildt, L. Female fertility loss and preservation: Threats and opportunities. Ann. Oncol. 2013, 24, 598–608. [Google Scholar] [CrossRef]
  30. De Vos, M.; Smitz, J.; Woodruff, T.K. Fertility preservation in women with cancer. Lancet 2014, 384, 1302–1310. [Google Scholar] [CrossRef] [Green Version]
  31. Metzger, M.L.; Meacham, L.R.; Patterson, B.; Casillas, J.S.; Constine, L.S.; Hijiya, N.; Kenney, L.B.; Leonard, M.; Lockart, B.A.; Likes, W.; et al. Female reproductive health after childhood, adolescent, and young adult cancers: Guidelines for the assessment and management of female reproductive complications. J. Clin. Oncol. 2013, 31, 1239–1247. [Google Scholar] [CrossRef] [PubMed]
  32. Hirshfeld-Cytron, J.; Gracia, C.; Woodruff, T.K. Nonmalignant diseases and treatments associated with primary ovarian failure: An expanded role for fertility preservation. J. Womens Health 2011, 20, 1467–1477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. West, E.R.; Zelinski, M.B.; Kondapalli, L.A.; Gracia, C.; Chang, J.; Coutifaris, C.; Critser, J.; Stouffer, R.L.; Shea, L.D.; Woodruff, T.K. Preserving female fertility following cancer treatment: Current options and future possibilities. Pediatr. Blood Cancer 2009, 53, 289–295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Roberts, J.; Ronn, R.; Tallon, N.; Holzer, H. Fertility preservation in reproductive-age women facing gonadotoxic treatments. Curr. Oncol. 2015, 22, e294–e304. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Donnez, J.; Dolmans, M.M. Fertility preservation in women. N. Engl. J. Med. 2017, 377, 1657–1665. [Google Scholar] [CrossRef]
  36. Dolmans, M.M.; Lambertini, M.; Macklon, K.T.; Almeida Santos, T.; Ruiz-Casado, A.; Borini, A.; Bordes, V.; Frith, L.; Van Moer, E.; Germeyer, A. EUropean REcommendations for female FERtility preservation (EU-REFER): A joint collaboration between oncologists and fertility specialists. Crit. Rev. Oncol. Hematol. 2019, 138, 233–240. [Google Scholar] [CrossRef]
  37. Gardino, S.L.; Emanuel, L.L. Choosing life when facing death: Understanding fertility preservation decision-making for cancer patients. Cancer Treat. Res. 2010, 156, 447–458. [Google Scholar]
  38. Li, N.; Jayasinghe, Y.; Kemertzis, M.A.; Moore, P.; Peate, M. Fertility preservation in pediatric and adolescent oncology patients: The decision-making process of parents. J. Adolesc. Young Adult Oncol. 2017, 6, 213–222. [Google Scholar] [CrossRef]
  39. Cohn, F. Oncofertility and informed consent: Addressing beliefs, values, and future decision making. Cancer Treat. Res. 2010, 156, 249–258. [Google Scholar]
  40. Harada, M.; Osuga, Y. Where are oncofertility and fertility preservation treatments heading in 2016? Future Oncol. 2016, 12, 2313–2321. [Google Scholar] [CrossRef] [Green Version]
  41. Salama, M.; Anazodo, A.; Woodruff, T.K. Preserving fertility in female patients with hematological malignancies: The key points. Expert Rev. Hematol. 2019, 12, 375–377. [Google Scholar] [CrossRef] [PubMed]
  42. Pfeifer, S.; Goldberg, I.; McClure, R.; Lobo, R.; Thomas, M.; Widra, E.; Licht, M.; Collins, I.; Cedars, M.; Racowsky, C.; et al. Mature oocyte cryopreservation: A guideline. Fertil. Steril. 2013, 99, 37–43. [Google Scholar]
  43. Thomas-Teinturier, C.; Allodji, R.S.; Svetlova, E.; Frey, M.A.; Oberlin, O.; Millischer, A.E.; Epelboin, S.; Decanter, C.; Pacquement, H.; Tabone, M.D.; et al. Ovarian reserve after treatment with alkylating agents during childhood. Hum. Reprod. 2015, 30, 1437–1446. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Chow, E.J.; Stratton, K.L.; Leisenring, W.M.; Oeffinger, K.C.; Sklar, C.A.; Donaldson, S.S.; Ginsberg, J.P.; Kenney, L.B.; Levine, J.M.; Robison, L.L.; et al. Pregnancy after chemotherapy in male and female survivors of childhood cancer treated between 1970 and 1999: A report from the Childhood Cancer Survivor Study cohort. Lancet Oncol. 2016, 17, 567–576. [Google Scholar] [CrossRef] [Green Version]
  45. Velez, M.P.; Richardson, H.; Baxter, N.N.; McClintock, C.; Greenblatt, E.; Barr, R.; Green, M. Risk of infertility in female adolescents and young adults with cancer: A population-based cohort study. Hum. Reprod. 2021, 36, 1981–1988. [Google Scholar] [CrossRef]
  46. Mackie, E.J.; Radford, M.; Shalet, S.M. Gonadal function following chemotherapy for childhood Hodgkin’s disease. Med. Pediatr. Oncol. 1996, 27, 74–78. [Google Scholar] [CrossRef]
  47. Bath, L.E.; Wallace, W.H.; Critchley, H.O. Late effects of the treatment of childhood cancer on the female reproductive system and the potential for fertility preservation. BJOG 2002, 109, 107–114. [Google Scholar] [CrossRef]
  48. Whitehead, E.; Shalet, S.M.; Blackledge, G.; Todd, I.; Crowther, D.; Beardwell, C.G. The effect of combination chemotherapy on ovarian function in women treated for Hodgkin’s disease. Cancer 1983, 52, 988–993. [Google Scholar] [CrossRef]
  49. Johnson, L.N.; Sammel, M.D.; Dillon, K.E.; Lechtenberg, L.; Schanne, A.; Gracia, C.R. Antimüllerian hormone and antral follicle count are lower in female cancer survivors and healthy women taking hormonal contraception. Fertil. Steril. 2014, 102, 774–781.e3. [Google Scholar] [CrossRef] [Green Version]
  50. Dillon, K.E.; Sammel, M.D.; Prewitt, M.; Ginsberg, J.P.; Walker, D.; Mersereau, J.E.; Gosiengfiao, Y.; Gracia, C.R. Pretreatment antimüllerian hormone levels determine rate of posttherapy ovarian reserve recovery: Acute changes in ovarian reserve during and after chemotherapy. Fertil. Steril. 2013, 99, 477–483.e1. [Google Scholar] [CrossRef] [Green Version]
  51. Anderson, R.A.; Wallace, W.H. Antimullerian hormone, the assessment of the ovarian reserve, and the reproductive outcome of the young patient with cancer. Fertil. Steril. 2013, 99, 1469–1475. [Google Scholar] [CrossRef] [PubMed]
  52. Broer, S.L.; Broekmans, F.J.; Laven, J.S.; Fauser, B.C. Anti-Müllerian hormone: Ovarian reserve testing and its potential clinical implications. Hum. Reprod. Update 2014, 20, 688–701. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Gracia, C.R.; Sammel, M.D.; Freeman, E.; Prewitt, M.; Carlson, C.; Ray, A.; Vance, A.; Ginsberg, J.P. Impact of cancer therapies on ovarian reserve. Fertil. Steril. 2012, 97, 134–140.e1. [Google Scholar] [CrossRef] [Green Version]
  54. Nielsen, S.N.; Andersen, A.N.; Schmidt, K.T.; Rechnitzer, C.; Schmiegelow, K.; Bentzen, J.G.; Larsen, E.C. A 10-year follow up of reproductive function in women treated for childhood cancer. Reprod. Biomed. Online 2013, 27, 192–200. [Google Scholar] [CrossRef] [Green Version]
  55. Wenners, A.; Grambach, J.; Koss, J.; Maass, N.; Jonat, W.; Schmutzler, A.; Mundhenke, C. Reduced ovarian reserve in young early breast cancer patients: Preliminary data from a prospective cohort trial. BMC Cancer 2017, 17, 632. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Van den Berg, M.H.; Overbeek, A.; Lambalk, C.B.; Kaspers, G.; Bresters, D.; van den Heuvel-Eibrink, M.M.; Kremer, L.C.; Loonen, J.J.; van der Pal, H.J.; Ronckers, C.M.; et al. Long-term effects of childhood cancer treatment on hormonal and ultrasound markers of ovarian reserve. Hum. Reprod. 2018, 33, 1474–1488. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. 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. Fertil. Steril. 2012, 97, 125–133. [Google Scholar] [CrossRef]
  58. Garcia-Velasco, J.A.; Domingo, J.; Cobo, A.; Martinez, M.; Carmona, L.; Pellicer, A. Five years’ experience using oocyte vitrification to pre- serve fertility for medical and nonmedical indications. Fertil. Steril. 2013, 99, 1994–1999. [Google Scholar] [CrossRef]
  59. Pal, L.; Leykin, L.; Schifren, J.L.; Isaacson, K.B.; Chang, Y.C.; Nikruil, N.; Chen, Z.; Toth, T.L. Malignancy may adversely influence the quality and behaviour of oocytes. Hum. Reprod. 1998, 13, 1837–1840. [Google Scholar] [CrossRef] [Green Version]
  60. Domingo, J.; Guillen, V.; Ayllon, Y.; Martınez, M.; Munoz, E.; Pellicer, A.; Garcia-Velasco, J.A. Ovarian response to controlled ovarian hyperstimulation in cancer patients is diminished even before oncological treatment. Fertil. Steril. 2012, 97, 930–934. [Google Scholar] [CrossRef]
  61. Alvarez, R.M.; Ramanathan, P. Fertility preservation in female oncology patients: The influence of the type of cancer on ovarian stimulation response. Hum. Reprod. 2018, 33, 2051–2059. [Google Scholar] [CrossRef] [PubMed]
  62. Volodarsky-Perel, A.; Cohen, Y.; Arab, S.; Son, W.Y.; Suarthana, E.; Dahan, M.H.; Tulandi, T.; Buckett, W. Effects of cancer stage and grade on fertility preservation outcome and ovarian stimulation response. Hum. Reprod. 2019, 34, 530–538. [Google Scholar] [CrossRef] [PubMed]
  63. Decanter, C.; Robin, G.; Mailliez, A.; Sigala, J.; Morschhauser, F.; Ramdane, N.; Devos, P.; Dewailly, D.; Leroy-Martin, B.; Keller, L. Prospective assessment of follicular growth and the oocyte cohort after ovarian stimulation for fertility preservation in 90 cancer patients versus 180 matched controls. Reprod. Biomed. Online 2018, 36, 543–551. [Google Scholar] [CrossRef] [PubMed]
  64. Moria, A.; Das, M.; Shehata, F.; Holzer, H.; Son, W.Y.; Tulandi, T. Ovarian reserve and oocyte maturity in women with malignancy undergoing in vitro maturation treatment. Fertil. Steril. 2011, 95, 1621–1623. [Google Scholar] [CrossRef]
  65. Johnson, L.N.; Dillon, K.E.; Sammel, M.D.; Efymow, B.L.; Mainigi, M.A.; Dokras, A.; Gracia, C.R. Response to ovarian stimulation in patients facing gonadotoxic therapy. Reprod. Biomed. Online 2013, 26, 337–344. [Google Scholar] [CrossRef] [Green Version]
  66. Levin, I.; Almog, B. Effect of cancer on ovarian function in patients undergoing in vitro fertilization for fertility preservation: A reappraisal. Curr. Oncol. 2013, 20, e1–e33. [Google Scholar] [CrossRef] [Green Version]
  67. Nurudeen, S.K.; Douglas, N.C.; Mahany, E.L.; Sauer, M.V.; Choi, J.M. Fertility preservation decisions among newly diagnosed oncology patients: A single-center experience. Am. J. Clin. Oncol. 2016, 39, 154–159. [Google Scholar] [CrossRef]
  68. Cardozo, E.R.; Thomson, A.P.; Karmon, A.E.; Dickinson, K.A.; Wright, D.L.; Sabatini, M.E. Ovarian stimulation and in-vitro fertilization outcomes of cancer patients undergoing fertility preservation compared to age matched controls: A 17-year experience. J. Assist. Reprod. Genet. 2015, 32, 587–596. [Google Scholar] [CrossRef] [Green Version]
  69. Almog, B.; Azem, F.; Gordon, D.; Pauzner, D.; Amit, A.; Barkan, G.; Levin, I. Effects of cancer on ovarian response in controlled ovarian stimulation for fertility preservation. Fertil. Steril. 2012, 98, 957–960. [Google Scholar] [CrossRef]
  70. Liu, D.; Yan, J.; Qiao, J. Effects of malignancies on fertility preservation outcomes and relevant cryobiological advances. Sci. China Life Sci. 2020, 63, 217–227. [Google Scholar] [CrossRef]
  71. Roxburgh, C.S.; McMillan, D.C. Cancer and systemic inflammation: Treat the tumour and treat the host. Br. J. Cancer 2014, 110, 1409–1412. [Google Scholar] [CrossRef] [PubMed]
  72. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Ingman, W.V.; Robertson, S.A. Defining the actions of transforming growth factor beta in reproduction. Bioessays 2002, 24, 904–914. [Google Scholar] [CrossRef]
  74. Derks-Smeets, I.A.P.; Van Tilborg, T.C.; Van Montfoort, A.; Smits, L.; Torrance, H.L.; Meijer-Hoogeveen, M.; Broekmans, F.; Dreesen, J.C.F.M.; Paulussen, A.D.C.; Tjan-Heijnen, V.C.G. BRCA1 mutation carriers have a lower number of mature oocytes after ovarian stimulation for IVF/PGD. J. Assist. Reprod. Genet. 2017, 34, 1475–1482. [Google Scholar] [CrossRef] [PubMed]
  75. Lekovich, J.; Lobel, A.L.S.; Stewart, J.D.; Pereira, N.; Kligman, I.; Rosenwaks, Z. Female patients with lymphoma demonstrate diminished ovarian reserve even before initiation of chemotherapy when compared with healthy controls and patients with other malignancies. J. Assist. Reprod. Genet. 2016, 33, 657–662. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Porcu, E.; Cillo, G.M.; Cipriani, L.; Sacilotto, F.; Notarangelo, L.; Damiano, G.; Dirodi, M.; Roncarati, I. Impact of BRCA1 and BRCA2 mutations on ovarian reserve and fertility preservation outcomes in young women with breast cancer. J. Assist. Reprod. Genet. 2020, 37, 709–715. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Balachandren, N.; Davies, M. Fertility, ovarian reserve and cancer. Maturitas 2017, 105, 64–68. [Google Scholar] [CrossRef]
  78. Spears, N.; Lopes, F.; Stefansdottir, A.; Rossi, V.; De Felici, M.; Anderson, R.A.; Klinger, F.G. Ovarian damage from chemotherapy and current approaches to its protection. Hum. Reprod. Update 2019, 25, 673–693. [Google Scholar] [CrossRef]
  79. Lee, S.J.; Schover, L.R.; Partridge, A.H.; Patrizio, P.; Wallace, W.H.; Hagerty, K.; Beck, L.N.; Brennan, L.V.; Oktay, K. American Society of Clinical Oncology recommendations on fertility preservation in cancer patients. J. Clin. Oncol. 2006, 24, 2917–2931. [Google Scholar] [CrossRef]
  80. Jayasinghe, J.L.; Wallace, W.H.B.; Anderson, R.A. Expert Review of Endocrinology & Metabolism Ovarian function, fertility and reproductive lifespan in cancer patients. Expert Rev. Endocrinol. Metab. 2018, 13, 125–136. [Google Scholar]
  81. Tauchmanova, L.; Selleri, C.; De Rosa, G.; Esposito, M.; Orio, J.F.; Palomba, S.; Bifulco, G.; Nappi, C.; Lombardi, G.; Rotoli, B.; et al. Gonadal status in reproductive age women after haematopoietic stem cell transplantation for haematological malignancies. Hum. Reprod. 2003, 18, 1410–1416. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Vatanen, A.; Wilhelmsson, M.; Borgström, B.; Gustafsson, B.; Taskinen, M.; Saarinen-Pihkala, U.M.; Winiarski, J.; Jahnukainen, K. Ovarian function after allogeneic hematopoietic stem cell transplantation in childhood and adolescence. Eur. J. Endocrinol. 2014, 170, 211–218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Szymanska, K.J.; Tan, X.; Oktay, K. Unraveling the mechanisms of chemotherapy-induced damage to human primordial follicle reserve: Road to developing therapeutics for fertility preservation and reversing ovarian aging. Mol. Hum. Reprod. 2020, 26, 553–566. [Google Scholar] [CrossRef] [PubMed]
  84. Bedoschi, G.; Navarro, P.A.; Oktay, K. Chemotherapy-induced damage to ovary: Mechanisms and clinical impact. Future Oncol. 2016, 12, 2333–2344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Cho, H.W.; Lee, S.; Min, K.J.; Hong, J.H.; Song, J.Y.; Lee, J.K.; Lee, N.W.; Kim, T. Advances in the Treatment and Prevention of Chemotherapy-Induced Ovarian Toxicity. Int. J. Mol. Sci. 2020, 21, 7792. [Google Scholar] [CrossRef]
  86. Kalich-Philosoph, L.; Roness, H.; Carmely, A.; Fishel-Bartal, M.; Ligumsky, H.; Paglin, S.; Wolf, I.; Kanety, H.; Sredni, B.; Meirow, D. Cyclophosphamide triggers follicle activation and “burnout”; AS101 prevents follicle loss and preserves fertility. Sci. Transl. Med. 2013, 5, 185ra162. [Google Scholar] [CrossRef]
  87. Gavish, Z.; Peer, G.; Roness, H.; Cohen, Y.; Meirow, D. Follicle activation and ‘burn-out’ contribute to post-transplantation follicle loss in ovarian tissue grafts: The effect of graft thickness. Hum. Reprod. 2014, 29, 989–996. [Google Scholar] [CrossRef] [Green Version]
  88. Wallace, W.H.; Shalet, S.M.; Hendry, J.H.; Morris-Jones, P.H.; Gattamaneni, H.R. Ovarian failure following abdominal irradiation in childhood: The radiosensitivity of the human oocyte. Br. Inst. Radiol. 2014, 62, 995–998. [Google Scholar] [CrossRef]
  89. Kim, S.; Kim, S.W.; Han, S.J.; Lee, S.; Park, H.T.; Song, J.Y.; Kim, T. Molecular Mechanism and Prevention Strategy of Chemotherapy-and Radiotherapy-Induced Ovarian Damage. Int. J. Mol. Sci. 2021, 22, 7484. [Google Scholar] [CrossRef]
  90. Beerendonk, C.C.M.; Braat, D.D.M. Present and future options for the preservation of fertility in female adolescents with cancer. Endocr. Dev. 2005, 8, 166–175. [Google Scholar]
  91. Stroud, J.S.; Mutch, D.; Rader, J.; Powell, M.; Thaker, P.H.; Grigsby, P.W. Effects of cancer treatment on ovarian function. Fertil. Steril. 2009, 92, 417–427. [Google Scholar] [CrossRef] [PubMed]
  92. Devine, P.J.; Perreault, S.D.; Luderer, U. Roles of reactive oxygen species and antioxidants in ovarian toxicity. Biol. Reprod. 2012, 86, 27. [Google Scholar] [CrossRef] [PubMed]
  93. Griffiths, M.J.; Winship, A.L.; Hutt, K.J. Do cancer therapies damage the uterus and compromise fertility? Hum. Reprod. Update 2020, 26, 161–173. [Google Scholar] [CrossRef] [PubMed]
  94. Fujimoto, A.; Ichinose, M.; Harada, M.; Hirata, T.; Osuga, Y.; Fujii, T. The outcome of infertility treatment in patients undergoing assisted reproductive technology after conservative therapy. J. Assist. Reprod. Genet. 2014, 31, 1189–1194. [Google Scholar] [CrossRef] [Green Version]
  95. Beneventi, F.; Locatelli, E.; Giorgiani, G.; Zecca, M.; Mina, T.; Simonetta, M.; Cavagnoli, C.; Albanese, M.; Spinillo, A. Adolescent and adult uterine volume and uterine artery Doppler blood flow among subjects treated with bone marrow transplantation or chemotherapy in pediatric age: A case-control study. Fertil. Steril. 2015, 103, 455–461. [Google Scholar] [CrossRef]
  96. Van de Loo, L.; Van den Berg, M.H.; Overbeek, A.; van Dijk, M.; Damen, L.; Lambalk, C.B.; Ronckers, C.M.; van den Heuvel-Eibrink, M.M.; Kremer, L.; van der Pal, H.J.; et al. Uterine function, pregnancy complications, and pregnancy outcomes among female childhood cancer survivors. Fertil. Steril. 2019, 111, 372–380. [Google Scholar] [CrossRef] [Green Version]
  97. Massarotti, C.; Scaruffi, P.; Lambertini, M.; Sozzi, F.; Remorgida, V.; Anserini, P. Beyond fertility preservation: Role of the oncofertility unit in the reproductive and gynecological follow-up of young cancer patients. Hum. Reprod. 2019, 34, 1462–1469. [Google Scholar] [CrossRef]
  98. Lambertini, M.; Del Mastro, L.; Pescio, M.C.; Andersen, C.Y.; Azim, H.A.; Peccatori, F.A.; Costa, M.; Revelli, A.; Salvagno, F.; Gennari, A.; et al. Cancer and fertility preservation: International recommendations from an expert meeting. BMC Med. 2016, 14, 1. [Google Scholar] [CrossRef]
  99. Linkeviciute, A.; Boniolo, G.; Chiavari, L.; Peccatori, F.A. Fertility preservation in cancer patients: The global framework. Cancer Treat. Rev. 2014, 40, 1019–1027. [Google Scholar] [CrossRef]
  100. Van den Berg, M.; Baysal, Ö.; Nelen, W.L.D.M.; Braat, D.D.M.; Beerendonk, C.C.M.; Hermens, R.P.M.G. Professionals’ barriers in female oncofertility care and strategies for improvement. Hum. Reprod. 2019, 34, 1074–1082. [Google Scholar] [CrossRef] [Green Version]
  101. Goodman, L.R.; Balthazar, U.; Kim, J.; Mersereau, J.E. Trends of socioeconomic disparities in referral patterns for fertility preservation consultation. Hum. Reprod. 2012, 27, 2076–2081. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Vindrola-Padros, C.; Dyer, K.E.; Cyrus, J.; Lubker, I.M. Healthcare professionals’ views on discussing fertility preservation with young cancer patients: A mixed method systematic review of the literature. Psychooncology 2017, 26, 4–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Lewin, J.; Ma, J.M.Z.; Mitchell, L.; Tam, S.; Puri, N.; Stephens, D.; Srikanthan, A.; Bedard, P.; Razak, A.; Crump, M.; et al. The positive effect of a dedicated adolescent and young adult fertility program on the rates of documentation of therapy-associated infertility risk and fertility preservation options. Support. Care Cancer 2017, 25, 1915–1922. [Google Scholar] [CrossRef] [PubMed]
  104. Lambertini, M.; Cinquini, M.; Moschetti, I.; Peccatori, F.A.; Anserini, P.; Menada, M.V.; Tomirotti, M.; Del Mastro, L. Temporary ovarian suppression during chemotherapy to preserve ovarian function and fertility in breast cancer patients: A GRADE approach for evidence evaluation and recommendations by the Italian Association of Medical Oncology. Eur. J. Cancer 2017, 71, 25–33. [Google Scholar] [CrossRef]
  105. Lee, J.H.; Choi, Y.S. The role of gonadotropin-releasing hormone agonists in female fertility preservation. Clin. Exp. Reprod. Med. 2021, 48, 11–16. [Google Scholar] [CrossRef]
  106. Roberts, J.E.; Oktay, K. Fertility preservation: A comprehensive approach to the young woman with cancer. JNCI Monogr. 2005, 2005, 57–59. [Google Scholar] [CrossRef]
  107. Ajala, T.; Rafi, J.; Larsen-Disney, P.; Howell, R. Fertility preservation for cancer patients: A review. Obstet. Gynecol. Int. 2010, 2010, 160386. [Google Scholar] [CrossRef]
  108. Lambertini, M.; Boni, L.; Michelotti, A.; Magnolfi, E.; Cogoni, A.A.; Mosconi, A.M.; Giordano, M.; Garrone, O.; Arpino, G.; Poggio, F.; et al. Long-term outcomes with pharmacological ovarian suppression during chemotherapy in premenopausal early breast cancer patients. J. Natl. Cancer. Inst. 2021, 114, 400–408. [Google Scholar] [CrossRef]
  109. Lambertini, M.; Moore, H.C.; Leonard, R.C.; Loibl, S.; Munster, P.; Bruzzone, M.; Boni, L.; Unger, J.M.; Anderson, R.A.; Mehta, K.; et al. Gonadotropin-releasing hormone agonists during chemotherapy for preservation of ovarian function and fertility in premenopausal patients with early breast cancer: A systematic review and meta-analysis of individual patient–level data. J. Clin. Oncol. 2018, 36, 1981–1990. [Google Scholar] [CrossRef] [Green Version]
  110. Urruticoechea, A.; Arnedos, M.; Walsh, G.; Dowsett, M.; Smith, I.E. Ovarian protection with goserelin during adjuvant chemotherapy for pre-menopausal women with early breast cancer (EBC). Breast Cancer Res. Treat. 2008, 110, 411–416. [Google Scholar] [CrossRef]
  111. Moore, H.C.; Unger, J.M.; Albain, K.S. Ovatian protection during adjuvant chemotherapy. N. Eng. J. Med. 2015, 372, 2268–2269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Leonard, R.C.F.; Adamson, D.J.A.; Bertelli, G.; Mansi, J.; Yellowlees, A.; Dunlop, J.; Thomas, A.; Coleman, R.E.; Anderson, R.A. GnRH agonist for protection against ovarian toxicity during chemotherapy for early breast cancer: The Anglo Celtic Group OPTION trial. Ann. Oncol. 2017, 28, 1811–1816. [Google Scholar] [CrossRef] [PubMed]
  113. Huang, K.G.; Lee, C.L.; Tsai, C.S.; Han, C.M.; Hwang, L.L. A new approach for laparoscopic ovarian transposition before pelvic irradiation. Gynecol. Oncol. 2007, 105, 234–237. [Google Scholar] [CrossRef] [PubMed]
  114. Irtan, S.; Orbach, D.; Helfre, S.; Sarnacki, S. Ovarian transposition in prepubescent and adolescent girls with cancer. Lancet Oncol. 2013, 14, e601–e608. [Google Scholar] [CrossRef]
  115. Laios, A.; Portela, S.D.; Papadopoulou, A.; Gallos, I.D.; Otify, M.; Ind, T. Ovarian transposition and cervical cancer. Best Pract. Res. Clin. Obstet. Gynaecol. 2021, 75, 37–53. [Google Scholar] [CrossRef]
  116. Corney, R.H.; Swinglehurst, A.J. Young childless women with breast cancer in the UK: A qualitative study of their fertility-related experiences, options, and the information given by health professionals. Psychooncology 2014, 23, 20–26. [Google Scholar] [CrossRef]
  117. Jones, G.; Hughes, J.; Mahmoodi, N.; Smith, E.; Skull, J.; Ledger, W. What factors hinder the decision-making process for women with cancer and contemplating fertility preservation treatment? Hum. Reprod. Update 2017, 23, 433–457. [Google Scholar] [CrossRef] [Green Version]
  118. Dolmans, M.M.; Donnez, J. Fertility preservation in women for medical and social reasons: Oocytes vs ovarian tissue. Best Pract. Res. Clin. Obstet. Gynaecol. 2021, 70, 63–80. [Google Scholar] [CrossRef]
  119. Vu, J.V.; Llarena, N.C.; Estevez, S.L.; Moravek, M.B.; Jeruss, J.S. Oncofertility program implementation increases access to fertility preservation options and assisted reproductive procedures for breast cancer patients. J. Surg. Oncol. 2017, 115, 116–121. [Google Scholar] [CrossRef] [Green Version]
  120. Silvestris, E.; De Palma, G.; Canosa, S.; Palini, S.; Dellino, M.; Revelli, A.; Paradiso, A.V. Human ovarian cortex biobanking: A fascinating resource for fertility preservation in cancer. Int. J. Mol. Sci. 2020, 21, 3245. [Google Scholar] [CrossRef]
  121. Kasum, M.; Beketić-Orešković, L.; Peddi, P.F.; Orešković, S.; Johnson, R.H. Fertility after breast cancer treatment. Eur. J. Obstet. Gynecol. Reprod. Biol. 2014, 173, 13–18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Creux, H.; Monnier, P.; Son, W.Y.; Buckett, W. Thirteen years’ experience in fertility preservation for cancer patients after in vitro fertilization and in vitro maturation treatments. J. Assist. Reprod. Genet. 2018, 35, 583–592. [Google Scholar] [CrossRef] [PubMed]
  123. Segers, I.; Bardhi, E.; Mateizel, I.; Van Moer, E.; Schots, R.; Verheyen, G.; Tournaye, H.; De Vos, M. Live births following fertility preservation using in-vitro maturation of ovarian tissue oocytes. Hum. Reprod. 2020, 35, 2026–2036. [Google Scholar] [CrossRef] [PubMed]
  124. Wallace, W.H.B.; Smith, A.G.; Kelsey, T.W.; Edgar, A.E.; Anderson, R.A. Fertility preservation for girls and young women with cancer: Population-based validation of criteria for ovarian tissue cryopreservation. Lancet Oncol. 2014, 15, 1129–1136. [Google Scholar] [CrossRef] [Green Version]
  125. Levine, J.; Stern, C.J. Fertility preservation in adolescents and young adults with cancer. J. Clin. Oncol. 2010, 28, 4831–4841. [Google Scholar] [CrossRef] [PubMed]
  126. Van der Perk, M.M.; Van der Kooi, A.L.L.; Van de Wetering, M.D.; IJgosse, I.M.; Van Dulmen-den Broeder, E.; Broer, S.L.; Klijn, A.J.; Versluys, A.B.; Arends, B.; Oude Ophuis, R.J.A. Oncofertility care for newly diagnosed girls with cancer in a national pediatric oncology setting, the first full year experience from the Princess Máxima Center, the PEARL study. PLoS ONE 2021, 16, e0246344. [Google Scholar] [CrossRef] [PubMed]
  127. Donnez, J.; Dolmans, M.M. Fertility preservation in women. Nat. Rev. Endocrinol. 2013, 9, 735–749. [Google Scholar] [CrossRef]
  128. Oktay, K. Ovarian tissue cryopreservation and transplantation: Preliminary findings and implications for cancer patients. Hum. Reprod. Update 2001, 7, 526–534. [Google Scholar] [CrossRef] [Green Version]
  129. Seshadri, T.; Gook, D.; Lade, S.; Spencer, A.; Grigg, A.; Tiedemann, K.; McKedrick, J.; Mitchell, J.; Stern, C.; Seymour, J.F. Lack of evidence of disease contamination in ovarian tissue harvested for cryopreservation from patients with Hodgkin lymphoma and analysis of factors predictive of oocyte yield. Br. J. Cancer 2006, 94, 1007–1010. [Google Scholar] [CrossRef] [Green Version]
  130. Dolmans, M.M.; Donnez, J.; Cacciottola, L. Fertility preservation: The challenge of freezing and transplanting ovarian tissue. Trends Mol. Med. 2021, 27, 777–791. [Google Scholar] [CrossRef]
  131. Donnez, J.; Dolmans, M.M. Ovarian cortex transplantation: 60 reported live births brings the success and worldwide expansion of the technique towards routine clinical practice. J. Assist. Reprod. Genet. 2015, 32, 1167–1170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Donnez, J.; Dolmans, M.M.; Demylle, D.; Jadoul, P.; Pirard, C.; Squifflet, J. Livebirth after orthotopic transplantation of cryopreserved ovarian tissue. Lancet 2004, 364, 1405–1410. [Google Scholar] [CrossRef]
  133. Janse, F.; Donnez, J.; Anckaert, E.; De Jong, F.H.; Fauser, B.C.; Dolmans, M.M. Limited value of ovarian function markers following orthotopic transplantation of ovarian tissue after gonadotoxic treatment. J. Clin. Endocrinol. Metab. 2011, 96, 1136–1144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Dolmans, M.M.; De Ouderaen, S.H.; Demylle, D.; Pirard, C. Utilization rates and results of long-term embryo cryopreservation before gonadotoxic treatment. J. Assist. Reprod. Genet. 2015, 32, 1233–1237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Oktay, K.; Turan, V.; Bedoschi, G.; Pacheco, F.S.; Moy, F. Fertility preservation success subsequent to concurrent aromatase inhibitor treatment and ovarian stimulation in women with breast cancer. J. Clin. Oncol. 2015, 33, 2424. [Google Scholar] [CrossRef] [Green Version]
  136. Cobo, A.; García-Velasco, J.A.; Coello, A.; Domingo, J.; Pellicer, A.; Remohí, J. Oocyte vitrification as an efficient option for elective fertility preservation. Fertil. Steril. 2016, 105, 755–764. [Google Scholar] [CrossRef] [Green Version]
  137. Diaz-Garcia, C.; Domingo, J.; Garcia-Velasco, J.A.; Herraiz, S.; Mirabet, V.; Iniesta, I.; Cobo, A.; Remohí, J.; Pellicer, A. Oocyte vitrification versus ovarian cortex transplantation in fertility preservation for adult women undergoing gonadotoxic treatments: A prospective cohort study. Fertil. Steril. 2018, 109, 478–485. [Google Scholar] [CrossRef] [Green Version]
  138. Specchia, C.; Baggiani, A.; Immediata, V.; Ronchetti, C.; Cesana, A.; Smeraldi, A.; Scaravelli, G.; Levi-Setti, P.E. Oocyte cryopreservation in oncological patients: Eighteen years experience of a tertiary care referral center. Front. Endocrinol. 2019, 10, 600. [Google Scholar] [CrossRef] [Green Version]
  139. Donnez, J.; Dolmans, M.M.; Diaz, C.; Pellicer, A. Ovarian cortex transplantation: Time to move on from experimental studies to open clinical application. Fertil. Steril. 2015, 104, 1097–1098. [Google Scholar] [CrossRef]
  140. Van der Ven, H.; Liebenthron, J.; Beckmann, M.; Toth, B.; Korell, M.; Krüssel, J.; Frambach, T.; Kupka, M.; Hohl, M.K.; Winkler-Crepaz, K.; 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. Hum. Reprod. 2016, 31, 2031–2041. [Google Scholar] [CrossRef]
  141. Meirow, D.; Ra’anani, H.; Shapira, M.; Brenghausen, M.; Chaim, S.D.; Aviel-Ronen, S. Transplantations of frozen-thawed ovarian tissue demonstrate high reproductive performance and the need to revise restrictive criteria. Fertil. Steril. 2016, 106, 467–474. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Jensen, A.K.; Macklon, K.T.; Fedder, J.; Ernst, E.; Humaidan, P.; Andersen, C.Y. 86 successful births and 9 ongoing pregnancies worldwide in women transplanted with frozen-thawed ovarian tissue: Focus on birth and perinatal outcome in 40 of these children. J. Assist. Reprod. Genet. 2017, 34, 325–336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Shapira, M.; Dolmans, M.M.; Silber, S.; Meirow, D. Evaluation of ovarian tissue transplantation: Results from three clinical centers. Fertil. Steril. 2020, 114, 388–397. [Google Scholar] [CrossRef]
  144. Terenziani, M.; Piva, L.; Meazza, C.; Gandola, L.; Cefalo, G.; Merola, M. Oophoropexy: A relevant role in preservation of ovarian function after pelvic irradiation. Fertil. Steril. 2009, 91, 935.e15–935.e16. [Google Scholar] [CrossRef] [PubMed]
  145. Abu-Rustum, N.R.; Yashar, C.M.; Bean, S.; Bradley, K.; Campos, S.M.; Chon, H.S.; Chu, C.; Cohn, D.; Crispens, M.A.; Damast, S.; et al. NCCN Guidelines Insights: Cervical Cancer, Version 1. 2020. J. Natl. Compr. Cancer Netw. 2020, 18, 660–666. [Google Scholar] [CrossRef] [PubMed]
  146. Plante, M.; Van Trommel, N.; Lheureux, S.; Oza, A.M.; Wang, L.; Sikorska, K.; Ferguson, S.E.; Han, K.; Amant, F. Cervical cancer treated with Neo-adjuvant chemotherapy followed by fertility Sparing Surgery (CONTESSA); Neo-Adjuvant Chemotherapy and Conservative Surgery in Cervical Cancer to Preserve Fertility (NEOCON-F). A PMHC, DGOG, GCIG/CCRN and multicenter study. Int. J. Gynecol. Cancer 2019, 29, 969–975. [Google Scholar] [CrossRef] [PubMed]
  147. Cao, D.Y.; Yang, J.X.; Wu, X.H.; Chen, Y.L.; Li, L.; Liu, K.J.; Cui, M.H.; Xie, X.; Wu, Y.M.; Kong, B.H.; et al. Comparisons of vaginal and abdominal radical trachelectomy for early-stage cervical cancer: Preliminary results of a multi-center research in China. Br. J. Cancer 2013, 109, 2778–2782. [Google Scholar] [CrossRef] [Green Version]
  148. Tesfai, F.M.; Kroep, J.R.; Gaarenstroom, K.; De Kroon, C.; Van Loenhout, R.; Smit, V.; Trimbos, B.; Nout, R.A.; Van Poelgeest, M.I.E.; Beltman, J.J. Fertility-sparing surgery of cervical cancer >2 cm (International Federation of Gynecology and Obstetrics 2009 stage IB1-IIA) after neoadjuvant chemotherapy. Int. J. Gynecol. Cancer. 2020, 30, 115–121. [Google Scholar] [CrossRef]
  149. Bentivegna, E.; Gouy, S.; Maulard, A.; Chargari, C.; Leary, A.; Morice, P. Oncological outcomes after fertility-sparing surgery for cervical cancer: A systematic review. Lancet Oncol. 2016, 17, e240–e253. [Google Scholar] [CrossRef]
  150. Bentivegna, E.; Maulard, A.; Pautier, P.; Chargari, C.; Gouy, S.; Morice, P. Fertility results and pregnancy outcomes after conservative treatment of cervical cancer: A systematic review of the literature. Fertil. Steril. 2016, 106, 1195–1211. [Google Scholar] [CrossRef] [Green Version]
  151. Floyd, J.L.; Campbell, S.; Rauh-Hain, J.A.; Woodard, T. Fertility preservation in women with early-stage gynecologic cancer: Optimizing oncologic and reproductive outcomes. Int. J. Gynecol. Cancer 2021, 31, 345–351. [Google Scholar] [CrossRef] [PubMed]
  152. Nezhat, C.; Roman, R.A.; Rambhatla, A.; Nezhat, F. Reproductive and oncologic outcomes after fertility-sparing surgery for early stage cervical cancer: A systematic review. Fertil. Steril. 2020, 113, 685–703. [Google Scholar] [CrossRef] [PubMed]
  153. Schuurman, T.; Zilver, S.; Samuels, S.; Schats, W.; Amant, F.; van Trommel, N.; Lok, C. Fertility-Sparing Surgery in Gynecologic Cancer: A Systematic Review. Cancers 2021, 28, 1008. [Google Scholar] [CrossRef] [PubMed]
  154. Tamauchi, S.; Kajiyama, H.; Osuka, S.; Moriyama, Y.; Yoshihara, M.; Kikkawa, F. Reduced response to controlled ovarian stimulation after radical trachelectomy: A pitfall of fertility-sparing surgery for cervical cancer. Int. J. Gynaecol. Obstet. 2021, 154, 162–168. [Google Scholar] [CrossRef]
  155. Li, J. Improving pregnancy outcomes in fertility preserved cervical cancer patients: Big challenge after radical trachelectomy. J. Gynecol. Oncol. 2019, 30, 1–3. [Google Scholar] [CrossRef]
  156. Obermair, A.; Baxter, E.; Brennan, D.J.; McAlpine, J.N.; Muellerer, J.J.; Amant, F.; van Gent, M.D.J.M.; Coleman, R.L.; Westin, S.N.; Yates, M.S.; et al. Fertility-sparing treatment in early endometrial cancer: Current state and future strategies. Obstet. Gynecol. Sci. 2020, 63, 417–431. [Google Scholar] [CrossRef]
  157. SGO Clinical Practice Endometrial Cancer Working Group; Burke, W.M.; Orr, J.; Leitao, M.; Salom, E.; Gehrig, P.; Olawaiye, A.B.; Brewer, M.; Boruta, D.; Villella, J.; et al. Endometrial cancer: A review and current management strategies: Part I. Gynecol. Oncol. 2014, 134, 385–392. [Google Scholar] [CrossRef]
  158. Concin, N.; Matias-Guiu, X.; Vergote, I.; Cibula, D.; Mirza, M.R.; Marnitz, S.; Ledermann, J.; Bosse, T.; Chargari, C.; Fagotti, A.; et al. ESGO/ESTRO/ESP guidelines for the management of patients with endometrial carcinoma. Int. J. Gynecol. Cancer. 2021, 31, 12–39. [Google Scholar] [CrossRef]
  159. SGO Clinical Practice Endometrial Cancer Working Group; Burke, W.M.; Orr, J.; Leitao, M.; Salom, E.; Gehrig, P.; Olawaiye, A.B.; Brewer, M.; Boruta, D.; Villella, J.; et al. Endometrial cancer: A review and current management strategies: Part II. Gynecol. Oncol. 2014, 134, 393–402. [Google Scholar] [CrossRef]
  160. Zhang, Q.; Qi, G.; Kanis, M.J.; Dong, R.; Cui, B.; Yang, X.; Kong, B. Comparison among fertility-sparing therapies for well differentiated early-stage endometrial carcinoma and complex atypical hyperplasia. Oncotarget 2017, 8, 57642–57653. [Google Scholar] [CrossRef] [Green Version]
  161. Lucchini, S.M.; Esteban, A.; Nigra, M.A.; Palacios, A.T.; Alzate-Granados, J.P.; Borla, H.F. Updates on conservative management of endometrial cancer in patients younger than 45 years. Gynecol. Oncol. 2021, 161, 802–809. [Google Scholar] [CrossRef] [PubMed]
  162. Wei, J.; Zhang, W.; Feng, L.; Gao, W. Comparison of fertility-sparing treatments in patients with early endometrial cancer and atypical complex hyperplasia: A meta-analysis and systematic review. Medicine 2017, 96, e8034. [Google Scholar] [CrossRef] [PubMed]
  163. Chen, J.; Cao, D.; Yang, J.; Yu, M.; Zhou, H.; Cheng, N.; Wang, J.; Zhang, Y.; Peng, P.; Shen, K. Management of Recurrent Endometrial Cancer or Atypical Endometrial Hyperplasia Patients After Primary Fertility-Sparing Therapy. Front. Oncol. 2021, 11, 3470. [Google Scholar] [CrossRef] [PubMed]
  164. Brannstrom, M.; Kahler, P.D. Uterus transplantation and fertility preservation. Best. Pract. Res. Clin. Obstet. Gynaecol. 2019, 55, 109–116. [Google Scholar] [CrossRef] [PubMed]
  165. Park, J.Y.; Seong, S.J.; Kim, T.J.; Kim, J.W.; Kim, S.M.; Bae, D.S.; Nam, J.H. Pregnancy outcomes after fertility-sparing management in young women with early endometrial cancer. Obstet. Gynecol. 2013, 121, 136–142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Roberti, L.; Maggiore, U.; Martinelli, F.; Dondi, G.; Bogani, G.; Chiappa, V.; Evangelista, M.T.; Liberale, V.; Ditto, A.; Ferrero, S.; et al. Efficacy and fertility outcomes of levonorgestrel-releasing intra-uterine system treatment for patients with atypical complex hyperplasia or endometrial cancer: A retrospective study. J. Gynecol. Oncol. 2019, 30, e57. [Google Scholar]
  167. Harrison, R.F.; He, W.; Fu, S.; Zhao, H.; Sun, C.C.; Suidan, R.S.; Woodard, T.L.; Rauh-Hain, J.A.; Westin, S.N.; Giordano, S.H.; et al. National patterns of care and fertility outcomes for reproductive-aged women with endometrial cancer or atypical hyperplasia. Am. J. Obstet. Gynecol. 2019, 221, 474.e1–474.e11. [Google Scholar] [CrossRef] [Green Version]
  168. Ng, J.S.; Low, J.J.H.; Ilancheran, A. Epithelial ovarian cancer. Best Pract. Res. Clin. Obstet. Gynaecol. 2012, 26, 337–345. [Google Scholar] [CrossRef]
  169. Bentivegna, E.; Morice, P.; Uzan, C.; Gouy, S. Fertility-sparing surgery in epithelial ovarian cancer. Future Oncol. 2016, 12, 389–398. [Google Scholar] [CrossRef]
  170. Bentivegna, E.; Gouy, S.; Maulard, A.; Pautier, P.; Leary, A.; Colombo, N.; Morice, P. Fertility-sparing surgery in epithelial ovarian cancer: A systematic review of oncological issues. Ann. Oncol. 2016, 27, 1994–2004. [Google Scholar] [CrossRef]
  171. Kajiyama, H.; Shibata, K.; Mizuno, M.; Umezu, T.; Suzuki, S.; Nawa, A.; Kawai, M.; Nagasaka, T.; Kikkawa, F. Long-term survival of young women receiving fertility-sparing surgery for ovarian cancer in comparison with those undergoing radical surgery. Br. J. Cancer 2011, 105, 1288–1294. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Fruscio, R.; Ceppi, L.; Corso, S.; Galli, F.; Dell’Anna, T.; Dell’Orto, F.; Giuliani, D.; Garbi, A.; Chiari, S.; Mangioni, C.; et al. Long-term results of fertility-sparing treatment compared with standard radical surgery for early-stage epithelial ovarian cancer. Br. J. Cancer 2016, 115, 641–648. [Google Scholar] [CrossRef] [PubMed]
  173. Morice, P.; Leblanc, E.; Rey, A.; Baron, M.; Querleu, D.; Blanchot, J.; Duvillard, P.; Lhommé, C.; Castaigne, D.; Classe, J.M.; et al. Conservative treatment in epithelial ovarian cancer: Results of a multicentre study of the GCCLCC (Groupe des Chirurgiens de centre de Lutte Contre Le cancer) and SFOG (Société Francaise d’Oncologie Gynécologique). Hum. Reprod. 2005, 20, 1379–1385. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Plett, H.; Harter, P.; Ataseven, B.; Heitz, F.; Prader, S.; Schneider, S.; Heikaus, S.; Fisseler-Eckhoff, A.; Kommoss, F.; Lax, S.F.; et al. Fertility-sparing surgery and reproductive-outcomes in patients with borderline ovarian tumors. Gynecol. Oncol. 2020, 157, 411–417. [Google Scholar] [CrossRef] [PubMed]
  175. Palomba, S.; Falbo, A.; Del Negro, S.; Rocca, M.; Russo, T.; Cariati, F.; Annunziata, G.; Tolino, A.; Tagliaferri, P.; Zullo, F. Ultra-conservative fertility-sparing strategy for bilateral borderline ovarian tumours: An 11-year follow-up. Hum. Reprod. 2010, 25, 1966–1972. [Google Scholar] [CrossRef] [Green Version]
  176. Ray-Coquard, I.; Morice, P.; Lorusso, D.; Prat, J.; Oaknin, A.; Pautier, P.; Colombo, N. Non-epithelial ovarian cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2018, 29, iv1–iv18. [Google Scholar] [CrossRef]
  177. Tamauchi, S.; Kajiyama, H.; Yoshihara, M.; Ikeda, Y.; Yoshikawa, N.; Nishino, K.; Utsumi, F.; Niimi, K.; Suzuki, S.; Kikkawa, F. Reproductive outcomes of 105 malignant ovarian germ cell tumor survivors: A multicenter study. Am. J. Obstet. Gynecol. 2018, 219, 385.e1–385.e7. [Google Scholar] [CrossRef] [Green Version]
  178. Di Tucci, C.; Casorelli, A.; Morrocchi, E.; Palaia, I.; Muzii, L.; Panici, P.B. Fertility management for malignant ovarian germ cell tumors patients. Crit. Rev. Oncol. Hematol. 2017, 120, 34–42. [Google Scholar] [CrossRef]
  179. Nasioudis, D.; Frey, M.K.; Chapman-Davis, E.; Witkin, S.S.; Holcomb, K. Safety of Fertility-Sparing Surgery for Premenopausal Women with Sex Cord-Stromal Tumors Confined to the Ovary. Int. J. Gynecol. Cancer 2017, 27, 1826–1832. [Google Scholar] [CrossRef]
  180. Bergamini, A.; Fais, M.L.; Dellino, M.; Silvestri, E.; Loizzi, V.; Bocciolone, L.; Rabaiotti, E.; Cioffi, R.; Sabetta, G.; Cormio, G.; et al. Fertility sparing surgery in sex-cord stromal tumors: Oncological and reproductive outcomes. Int. J. Gynecol. Cancer 2022, 1–8. [Google Scholar] [CrossRef]
  181. Vasta, F.M.; Dellino, M.; Bergamini, A.; Gargano, G.; Paradiso, A.; Loizzi, V.; Bocciolone, L.; Silvestri, E.; Petrone, M.; Cormio, G.; et al. Reproductive Outcomes and Fertility Preservation Strategies in Women with Malignant Ovarian Germ Cell Tumors after Fertility Sparing Surgery. Biomedicines 2020, 8, 554. [Google Scholar]
  182. Bercow, A.; Nitecki, R.; Brady, P.C.; Rauh-Hain, J.A. Outcomes after Fertility-sparing Surgery for Women with Ovarian Cancer: A Systematic Review of the Literature. J. Minim. Invasive Gynecol. 2021, 28, 527–536. [Google Scholar] [CrossRef] [PubMed]
  183. Tomao, F.; Di Tucci, C.; Marchetti, C.; Perniola, G.; Bellati, F.; Benedetti Panici, P. Role of chemotherapy in the management of vulvar carcinoma. Crit. Rev. Oncol. Hematol. 2012, 82, 25–39. [Google Scholar] [CrossRef]
  184. Steiner, A.Z.; Pritchard, D.; Stanczyk, F.Z.; Kesner, J.S.; Meadows, J.W.; Herring, A.H.; Baird, D.D. Association between biomarkers of ovarian reserve and infertility among older women of reproductive age. JAMA 2017, 318, 1367–1376. [Google Scholar] [CrossRef]
  185. Lehmann, V.; Kutteh, W.H.; Sparrow, C.K.; Bjornard, K.L.; Klosky, J.L. Fertility-related services in pediatric oncology across the cancer continuum: A clinic overview. Support. Care Cancer 2020, 28, 3955–3964. [Google Scholar] [CrossRef] [PubMed]
  186. Filippi, F.; Meazza, C.; Somigliana, E.; Podda, M.; Dallagiovanna, C.; Massimino, M.; Raspagliesi, F.; Terenziani, M. Fertility preservation in childhood and adolescent female tumor survivors. Fertil. Steril. 2021, 116, 1087–1095. [Google Scholar] [CrossRef]
  187. Somigliana, E.; Costantini, M.P.; Filippi, F.; Terenziani, M.; Riccaboni, A.; Nicotra, V.; Rago, R.; Paffoni, A.; Mencaglia, L.; Magnolfi, S.; et al. Fertility counseling in women with hereditary cancer syndromes. Crit. Rev. Oncol. Hematol. 2022, 171, 103604. [Google Scholar] [CrossRef]
  188. Chen, L.; Blank, S.V.; Burton, E.; Glass, K.; Penick, E.; Woodard, T. Reproductive and hormonal considerations in women at increased risk for hereditary gynecologic cancers: Society of Gynecologic Oncology and American Society for Reproductive Medicine Evidence-Based Review. Fertil. Steril. 2019, 112, 1034–1042. [Google Scholar] [CrossRef]
Figure 1. Fertility Preservation Options.
Figure 1. Fertility Preservation Options.
Cancers 14 02500 g001
Table 1. Ovarian damage risk with chemotherapeutic agents.
Table 1. Ovarian damage risk with chemotherapeutic agents.
Damage RiskCancer Treatment
High riskCMF
Cyclophosphamide
Melphalan
Busulfan
Intermediate riskCisplatin
Carboplatin
Oxaliplatin
Doxorubicin
Low/very lowCHOP
Vinblastine
Vincristine
Methotrexate
5-fluorouracil
Notes: CHOP, cyclophosphamide/doxorubicin/vincristine/prednisone; CMF, ciclofosfamide, methotrexate e 5-fluorouracile.
Table 2. Live birth rate after fertility preservation techniques.
Table 2. Live birth rate after fertility preservation techniques.
TechniquesAuthorsLBR
Embryos-cryopreservationDolmans et al. 201520–40%
Oktay et al. 2015
Oocytes cryopreservationCobo et al. 201620–50%
Diaz-Garcia et al. 2018
Specchia et al. 2019
Ovarian tissue
cryopreservation and reimplantation
Donnez et al. 201518.2–41.6%
Van der Ven et al. 2016
Diaz-Garcia et al. 2018
Meirow et al. 2016
Donnez et al. 2017
Jensen et al. 2017
Shapira et al. 2020
In vitro maturationSilvestris et al. 20208.9%
In vitro activationSilvestris et al. 20207%
OophoropexisTerenziani et al. 2009Rare
Irten et al. 2013
Table 3. Reproductive and oncological outcomes of patients after undergoing fertility-sparing surgical procedure.
Table 3. Reproductive and oncological outcomes of patients after undergoing fertility-sparing surgical procedure.
Type of CancerAuthorTreatmentPatients Reproductive OutcomesOncological Outcomes
TotalTTC (%)N° PregnanciesART PregnanciesLBR (%)Recurrence (%)Death (%)
Cervical cancerBentivegna et al. 2016ST212NA103NA74.0NANA
VRT1355NA499NA67.0NANA
RT (lptm)735NA175NA68.0NANA
RT(MIS)314NA74NA78.0NANA
NACT161NA93NA76.0NANA
Willows et al. 2016RT (a-b: lesion </> 2 cm)1238 (a)44469NA66.74.51.7
134 (b)3010 70.011.14.2
Non-radical FSS124NA71NA67.62.70.5
NACT62NA36NA72.26.31.3
Nezhat et al. 2020ST/CKC28329.3131886.41.40.2
VRT138743.86067863.43.71.1
AbRT10604322910465.73.60.7
RT (MIS)31422.6811656.53.30.1
Schuurman et al. 2021LLETZ/CKC/ST61248.5241NA77.43.60.8
VRT153955707NA70.64.21.7
AbRT (lptm) 163548353NA583.11.5
RT (MIS) 34435.781NA71.64.51.5
Endometrial cancerPark et al. 2013Progestin therapy (PG)14149.6514466.031.90
Harrison et al. 2019PG: alone (1); followed by hysterectomy (2)421 (1)NA1316511.6NANA
397 (2)NA3421652.0NANA
Leone et al. 2019LNG-IUD4443.114823.441.5NA
Schuurman et al. 2021FSS/PG/LNG50562.6256NA7234.70.8
Ovarian cancerBentivegna et al. 2016Conservative surgery *651 (EOC)42.8323NA69.0126
Tamauchi et al. 2018Conservative surgery105 (OGCT)42.865764.610.43.8
Plett et al. 2020Conservative surgery95 (BOT)43.148NA79.013NA
Bercow et al. 2021Conservative surgery614 (EOC)50242NA76–965–188.8
992 (BOT)NA657NA23–10011.62.3
Schuurman et al. 2021Conservative surgery75044.2280NA89.415.714.7
Note: TTC: trying to conceive; ART: reproductive assisted techniques; LBR: live birth rate; NA: not available; ST: simple trachelectomy VRT—vaginal radical trachelectomy; RT: radical trachelectomy; NACT: neoadjuvant chemotherapy CK: cold knife conization; AbRT: abdominal radical trachelectomy; MIS: minimal invasive surgery (laparoscopic or robotassisted); LLETZ: large loop excision of transformation zone; lptm: laparotomy; EOC: epitelian ovarian cancer; BOT: borderline ovarian tumor; OGCT: ovarian germ cell tumor. *: considered as the preservation of at least one ovary and the uterus.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Di Tucci, C.; Galati, G.; Mattei, G.; Chinè, A.; Fracassi, A.; Muzii, L. Fertility after Cancer: Risks and Successes. Cancers 2022, 14, 2500. https://doi.org/10.3390/cancers14102500

AMA Style

Di Tucci C, Galati G, Mattei G, Chinè A, Fracassi A, Muzii L. Fertility after Cancer: Risks and Successes. Cancers. 2022; 14(10):2500. https://doi.org/10.3390/cancers14102500

Chicago/Turabian Style

Di Tucci, Chiara, Giulia Galati, Giulia Mattei, Alessandra Chinè, Alice Fracassi, and Ludovico Muzii. 2022. "Fertility after Cancer: Risks and Successes" Cancers 14, no. 10: 2500. https://doi.org/10.3390/cancers14102500

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop