The future of fertility preservation for women treated with chemotherapy

in Reproduction and Fertility
Authors:
Lauren R Alesi Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia

Search for other papers by Lauren R Alesi in
Current site
Google Scholar
PubMed
Close
,
Quynh-Nhu Nguyen Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
Paediatric Integrated Cancer Service, VIC, Australia

Search for other papers by Quynh-Nhu Nguyen in
Current site
Google Scholar
PubMed
Close
,
Jessica M Stringer Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia

Search for other papers by Jessica M Stringer in
Current site
Google Scholar
PubMed
Close
,
Amy L Winship Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia

Search for other papers by Amy L Winship in
Current site
Google Scholar
PubMed
Close
https://orcid.org/0000-0001-7848-1447
, and
Karla J Hutt Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia

Search for other papers by Karla J Hutt in
Current site
Google Scholar
PubMed
Close
https://orcid.org/0000-0002-5111-8389

Correspondence should be addressed to K J Hutt or A L Winship; Email: karla.hutt@monash.edu or amy.winship@monash.edu

*(A L Winship and K J Hutt contributed equally to this work)

Open access
Sign up for journal news

Cytotoxic chemotherapies have been a mainstay of cancer treatment but are associated with numerous systemic adverse effects, including impacts on fertility and endocrine health. Irreversible ovarian damage and follicle depletion are the side effects of chemotherapy that can lead to infertility and premature menopause, both being major concerns of young cancer patients. Notably, many women will proceed with fertility preservation, but unfortunately existing strategies do not entirely solve the problem. Most significantly, oocyte and embryo freezing do not prevent cancer treatment-induced ovarian damage from occurring, which may result in the impairment of long-term hormone production. Unfortunately, loss of endogenous endocrine function is not fully restored by hormone replacement therapy. Additionally, while GnRH agonists are standard care for patients receiving alkylating chemotherapy to lessen the risk of premature menopause, their efficacy is incomplete. The lack of more broadly effective options stems, in part, from our poor understanding of how different treatments damage the ovary. Here, we summarise the impacts of two commonly utilised chemotherapies – cyclophosphamide and cis-diamminedichloroplatinum(II) (cisplatin) – on ovarian function and fertility and discuss the mechanisms underpinning this damage. Additionally, we critically analyse current research avenues in the development of novel fertility preservation strategies, with a focus on ferto-protective agents.

Lay summary

Over the past few decades, advances in the detection and treatment of cancer have dramatically improved survival rates in young women. This means that ensuring patients have a high quality of life after cancer treatment has become a new priority. Therefore, it is important to understand and prevent any long-term negative side effects of cancer treatments, with infertility and early-onset menopause being major concerns for women receiving chemotherapy. The current fertility preservation options available to young women have significant limitations. Therefore, the identification of new approaches to protect fertility has been an intense topic of research in recent years. In this review, we provide information on the negative side effects of two commonly used chemotherapy drugs – cyclophosphamide and cis-diamminedichloroplatinum(II) (cisplatin) – on fertility, and discuss how they cause damage to the ovaries. We also critically analyse recent preclinical studies related to the development of new fertility preservation techniques.

Abstract

Cytotoxic chemotherapies have been a mainstay of cancer treatment but are associated with numerous systemic adverse effects, including impacts on fertility and endocrine health. Irreversible ovarian damage and follicle depletion are the side effects of chemotherapy that can lead to infertility and premature menopause, both being major concerns of young cancer patients. Notably, many women will proceed with fertility preservation, but unfortunately existing strategies do not entirely solve the problem. Most significantly, oocyte and embryo freezing do not prevent cancer treatment-induced ovarian damage from occurring, which may result in the impairment of long-term hormone production. Unfortunately, loss of endogenous endocrine function is not fully restored by hormone replacement therapy. Additionally, while GnRH agonists are standard care for patients receiving alkylating chemotherapy to lessen the risk of premature menopause, their efficacy is incomplete. The lack of more broadly effective options stems, in part, from our poor understanding of how different treatments damage the ovary. Here, we summarise the impacts of two commonly utilised chemotherapies – cyclophosphamide and cis-diamminedichloroplatinum(II) (cisplatin) – on ovarian function and fertility and discuss the mechanisms underpinning this damage. Additionally, we critically analyse current research avenues in the development of novel fertility preservation strategies, with a focus on ferto-protective agents.

Lay summary

Over the past few decades, advances in the detection and treatment of cancer have dramatically improved survival rates in young women. This means that ensuring patients have a high quality of life after cancer treatment has become a new priority. Therefore, it is important to understand and prevent any long-term negative side effects of cancer treatments, with infertility and early-onset menopause being major concerns for women receiving chemotherapy. The current fertility preservation options available to young women have significant limitations. Therefore, the identification of new approaches to protect fertility has been an intense topic of research in recent years. In this review, we provide information on the negative side effects of two commonly used chemotherapy drugs – cyclophosphamide and cis-diamminedichloroplatinum(II) (cisplatin) – on fertility, and discuss how they cause damage to the ovaries. We also critically analyse recent preclinical studies related to the development of new fertility preservation techniques.

Introduction

Given that approximately 5% of women diagnosed with cancer worldwide are of reproductive age and cancer mortality rates are steadily falling (Bedoschi et al. 2016), addressing the off-target effects of cancer treatment on ovarian function, fertility and endocrine health has become a prominent issue. In fact, between 40% and 80% of female cancer survivors experience infertility post-treatment (Stern et al. 2006, Knapp et al. 2011). Moreover, many patients report this risk of post-treatment infertility to be equally as distressing as the initial cancer diagnosis (Logan et al. 2019). Consequently, understanding and mitigating these long-term, off-target consequences of cancer therapy on fertility must be prioritised.

Female fertility is governed by the quantity and quality of oocytes, which are stored in the ovary within primordial follicles. Cytotoxic chemotherapy, in addition to radiotherapy and surgery, has long been the mainstay of many cancer treatment regimens and is well documented to have detrimental off-target side effects on the ovaries, fertility and endocrine health (Bedoschi et al. 2016). Primordial follicle oocytes are exquisitely sensitive to genotoxic stress (i.e. DNA damage), and their depletion is accelerated in response to many exogenous insults, including chemotherapy treatment (Kerr et al. 2012b , Winship et al. 2018).

Exposure to certain chemotherapeutic drugs may induce temporary amenorrhea (cessation of menstruation) and subfertility in the short term or cause premature ovarian insufficiency (POI) in the long term (Codacci-Pisanelli et al. 2017). Indeed, exposure to gonadotoxic cancer treatment is the leading cause of POI in young girls and women (Byrne et al. 1992). POI is characterised by reduced ovarian endocrine function, complete amenorrhea and early menopause in women younger than 40 years of age, which results in irreversible infertility (Webber et al. 2016). This not only has the potential to significantly impact a patient’s mental health but is also associated with multi-system long-term physical and psychological sequelae due to the associated ovarian endocrine failure. These sequelae can significantly impact quality of life and may include impaired sexual health, increased risk of depression and anxiety and increased risk of cardiovascular disease and osteoporosis, among others (Faubion et al. 2015, Torrealday et al. 2017). Therefore, preventing ovarian damage associated with chemotherapy is not only important for protecting the fertility of female cancer survivors but also their overall long-term health and well-being.

Unfortunately, current fertility preservation options and techniques have some significant limitations. Embryo, oocyte and ovarian cortex cryopreservation are effective for many adult women but are expensive, invasive and not available to all patients. Moreover, these treatments do not actually mitigate ovarian damage and thus do not prevent POI. Notably, treatment with gonadotropin-releasing hormone (GnRH) agonists has been shown to reduce the risk of POI in women with breast cancer (Lambertini et al. 2018). However, these have not been definitively shown to protect the ovarian reserve or preserve fertility (Lambertini et al. 2019). Therefore, the investigation of novel fertility preservation strategies is a critical area of research.

Off-target impacts of chemotherapy on ovarian function

Numerous classes of chemotherapy exist, including alkylating agents, platinum-based (alkylating-like) agents, anti-tumour antibiotics, anti-metabolites, vinca alkaloids, topoisomerase inhibitors and other miscellaneous agents. These different classes of chemotherapeutic agents have diverse mechanisms of action and, hence, varying degrees of ovarian toxicity (ovotoxicity), which have been reviewed extensively (Bedoschi et al. 2016, Spears et al. 2019, Alesi et al. 2021). Briefly, the most ovotoxic class is the alkylating agents, such as cyclophosphamide, followed by platinum-based compounds, such as cis-diamminedichloroplatinum(II) (cisplatin), and, lastly, the anthracycline antibiotic doxorubicin. However, other agents, such as the anti-metabolite 5-fluorouracil, have been demonstrated to have moderate, short-term ovotoxicity (Lambouras et al. 2018, Stringer et al. 2018). Additionally, it is important to consider that the ovotoxicity of chemotherapies is dependent on dose and frequency of treatment and is likely exacerbated in multi-dose and combination regimens.

Characterising the ovotoxicity of chemotherapies remains an active area of research, with several recent studies of previously overlooked agents revealing detrimental impacts on ovarian function (reviewed recently byAlesi et al. 2021, updates in Table 1). This new body of research highlights the requirement for more rigorous investigation of the ovotoxicity of both existing and new cancer therapies to adequately inform clinicians and patients of the risks to fertility posed by treatment. However, as cyclophosphamide and cisplatin are the most widely studied agents with respect to ovarian function, they will be the major focus of this review hereafter.

Table 1

Update on ovotoxicity of chemotherapies.

Agent Class Mechanism of action Evidence of ovotoxicity Degree of ovotoxicity Reference
Paclitaxel Taxane Induces microtubule destabilisation Single-dose (30 mg/kg) paclitaxel decreases the number of healthy antral follicles in vivo in mice, without impacting primordial follicles Mild, short term Ma et al. (2020)
5-FU Anti-metabolite Interferes with DNA replication Varies from mild, short term to high, long term, depending on the dose and frequency
Multi-dose 5-FU (50 mg/kg/day for 4 days) does not alter primordial follicle number in vivo in mice but depletes growing follicles. Additionally, oocyte maturation and early embryo development is impaired in vitro Naren et al. (2021)
Single-dose (450 mg/kg) 5-FU significantly reduces the number of follicles in all classes in vivo in mice, including primordial follicles Almeida et al. (2021)
Actinomycin D Antibiotic Interferes with DNA transcription Actinomycin D disrupts spindle assembly and chromatin condensation in mouse and human oocytes in vitro. Additionally, oocyte maturation is impaired in mouse oocytes in vitro Mild, short term Li et al. (2021)
Irinotecan (CPT-11) Topoisomerase inhibitor Interferes with DNA replication Single-dose (100 mg/kg) irinotecan significantly decreases primordial follicles and serum AMH in vivo in mice Moderate, long term Levi et al. (2022)

CPT-11, 7-ethyl-10-[4-(1-piperidino)-1-piperidino] carbonyloxycamptothecin; 5-FU, 5-fluorouracil.

Cyclophosphamide is a nitrogen mustard derivative used to treat a variety of cancers that commonly affect paediatric and reproductive-age female patients, including Hodgkin’s and non-Hodgkin’s lymphoma, breast cancer, ovarian cancer and small cell lung cancer, among others (Ogino & Prasanna 2020). Once metabolised in the liver, cyclophosphamide is converted into its active alkylating metabolites, which include phosphoramide mustard and acrolein. These form crosslinks both within and between DNA strands, resulting in double-stranded DNA breaks, which, if irreparable, culminate in apoptosis of the target cell (Chu & Rubin 2018).

Cisplatin is a platinum-based chemotherapy that is widely used and effective in the treatment of numerous cancer types. These include ovarian cancer, breast cancer, cervical cancer, lung cancer (both small-cell and non-small cell), brain cancer and neuroblastoma, all of which affect paediatric and reproductive-age females (Dasari & Tchounwou 2014, Franasiak & Scott 2016). Sometimes described as an alkylating-like agent, cisplatin is able to crosslink DNA and form both inter- and intra-strand adducts (Dasari & Tchounwou 2014). This causes DNA damage, blocks cell division, prevents induction of DNA repair and, usually, leads to apoptosis of the target cell. Another critical component of cisplatin-induced cytotoxicity is the induction of oxidative stress via the production of reactive oxygen species, predominantly within mitochondria, leading to mitochondrial dysfunction and induction of apoptosis by either the intrinsic or the extrinsic pathway, independent of DNA damage (Marullo et al. 2013, Dasari & Tchounwou 2014).

Since cyclophosphamide metabolites and cisplatin are capable of directly interacting with DNA, thereby leading to the induction of double-stranded breaks and inhibition of DNA replication, they are both considered to act in a cell cycle non-specific manner (Sun et al. 2021). However, susceptibility to damage can vary across the cell cycle, with sensitivity to cisplatin peaking at G1 just prior to the onset of DNA replication (Shah & Schwartz 2001). Therefore, even meiotically arrested cells, such as the oocytes within primordial follicles, are susceptible to cyclophosphamide- or cisplatin-induced damage.

Evidence of cyclophosphamide- and cisplatin-induced ovarian damage

The deleterious effects of cyclophosphamide and cisplatin exposure on ovarian function are well established. Despite this, studies investigating the effect of single-agent cyclophosphamide in human ovarian tissue are surprisingly limited. Similarly, given that cisplatin is rarely administered as a single agent, there is also very limited data available on the precise impact of cisplatin alone on ovarian function in humans. Thus, the vast majority of knowledge regarding the ovotoxicity of these chemotherapies has been obtained from animal studies, particularly those conducted in rodents.

The consequences of cyclophosphamide and cisplatin exposure on ovarian function fall into three major categories: (i) impacts on primordial follicles and, thus, long-term fertility; (ii) impacts on growing follicles and, thus, short-term fertility; and (iii) impacts on ovarian stroma and vasculature. Additionally, recent evidence suggests that these agents may also impact upon oocyte mitochondrial function, though the consequences of this are yet to be fully elucidated (Malott & Luderer 2021, Wang & Hutt 2021, Zhang et al. 2021).

Impacts on primordial follicles and long-term fertility

Exposure to cyclophosphamide or cisplatin is highly detrimental to primordial follicles and can permanently impact fertility in the long term. In fact, numerous animal studies conducted over the past several decades have documented a significant depletion of the primordial follicle pool in response to cyclophosphamide treatment (Mattison et al. 1981, Shiromizu et al. 1984, Kalich-Philosoph et al. 2013, Nguyen et al. 2018, Bellusci et al. 2019, Nguyen et al. 2019, Salian et al. 2020). Similarly, many in vitro and in vivo studies in mice published in recent years have demonstrated that cisplatin is also highly toxic to primordial follicles (Gonfloni et al. 2009, Chang et al. 2015, Yuksel et al. 2015, Rossi et al. 2017, Nguyen et al. 2018, Nguyen et al. 2019).

In adult mice, a single, high dose of cyclophosphamide (300 mg/kg) or cisplatin (5 mg/kg) is sufficient to eliminate the vast majority of the primordial follicle population within 24 h in vivo (Nguyen et al. 2019). Of those remaining, half are morphologically abnormal, which is indicative of imminent apoptosis (Nguyen et al. 2019). By 5 days post-treatment, approximately 5% and 25% of the primordial follicle pool remain following cyclophosphamide (300 mg/kg) and cisplatin (5 mg/kg) treatment, respectively (Nguyen et al. 2018). Additionally, lower multi-dose cyclophosphamide (100 mg/kg × 6) is also detrimental to ovarian function in mice in vivo, with very few primordial follicles remaining 4 weeks post-final treatment (Kim & You 2021). These studies clearly demonstrate the ovotoxicity of these alkylating agents.

As a consequence of the significant depletion to the primordial follicle pool, there is a dramatic reduction in the number of mature oocytes collected after superovulation, pregnancy rate, age at last litter, average number of pups per litter and total number of litters per female in cyclophosphamide or cisplatin-treated mice (Kerr et al. 2012b, Zhang et al. 2015, Nguyen et al. 2018, Roness et al. 2019, Sonigo et al. 2019, Huang et al. 2020, Salian et al. 2020, Kim & You 2021). These data indicate a significant reduction of fertility and the overall fertile life span in response to cyclophosphamide or cisplatin treatment. Interestingly, in a recent study, no gross abnormalities were reported in any offspring born from either cyclophosphamide- or cisplatin-treated mice (Nguyen et al. 2018). This data may suggest that the quality of the few remaining primordial follicles not eliminated by apoptosis is preserved or perhaps that any DNA damage was able to be repaired within the oocytes of those follicles. Supporting this hypothesis, it was recently demonstrated that primordial follicles can effectively repair DNA damage following cisplatin exposure via homologous recombination in apoptosis-resistant (Puma–/–) mice (Nguyen et al. 2021).

Although limited, there is also direct evidence of cyclophosphamide- and cisplatin-induced toxicity to primordial follicles in humans. A study using human fetal ovarian xenografts in mice demonstrated that cyclophosphamide treatment significantly depleted the primordial follicle population (Meng et al. 2014). Another study utilising cultured human ovarian tissue treated with metabolites of cyclophosphamide found similar results (Lande et al. 2017). In vitro studies utilising cultured human ovarian cortical pieces and cultured human granulosa cells have also revealed significant decreases in primordial follicles and steroid hormone production and a significant increase in granulosa cell apoptosis in response to cisplatin (Bildik et al. 2015, Yuksel et al. 2015, Bildik et al. 2018). Together, these data suggest that the negative impacts on fertility in female cancer survivors, who received cyclophosphamide and/or cisplatin, are likely to be due to a depletion in primordial follicles.

Impacts on growing follicles and short-term fertility

Cyclophosphamide or cisplatin exposure can also impair growing follicle health and survival. Although transient, these effects can still interfere with short-term fertility, pregnancy success and offspring health. Toxicity to growing follicles has been reported in humans, with diminished antral follicle count and a dramatic reduction of circulating hormones produced by growing follicles (anti-Müllerian hormone (AMH) and inhibin B) following treatment with either cyclophosphamide-containing regimens or other alkylators (Anderson et al. 2006, van den Berg et al. 2018). However, in rodent studies, evidence of toxicity to growing follicles is conflicting.

Following cyclophosphamide or cisplatin exposure, significant depletion of the growing follicle pool – primarily in the early (primary and secondary) stages – has been reported in rats (Jarrell et al. 1991, Yuksel et al. 2015), although reports in mice following cyclophosphamide or cisplatin exposure are varied. Some studies describe significant depletion of only primary and/or secondary follicles (Kalich-Philosoph et al. 2013, Nguyen et al. 2018, Nguyen et al. 2019, Bellusci et al. 2019), whereas other studies are unable to detect any depletion altogether (Mattison et al. 1981, Rossi et al. 2017). Some other papers report an increase in the early growing follicle pool following cyclophosphamide or cisplatin exposure (Kalich-Philosoph et al. 2013, Chang et al. 2015, Sonigo et al. 2019). The discrepancy in reports may be explained by differences in follicle quantification methods or by differences in the interpretation of follicle quantification. Two of the three studies reporting a significant increase in the growing follicle pool found an increased ratio of primordial to growing follicles (Chang et al. 2015, Sonigo et al. 2019) rather than looking at the follicle numbers. An increased ratio of growing follicles may be caused by the depletion of primordial follicles by cyclophosphamide or cisplatin treatment rather than an increase in the number of growing follicles. Nevertheless, there appears to be a difference in sensitivity to cyclophosphamide or cisplatin exposure between primordial and growing follicles. This may be due to growing follicles having a higher threshold for apoptosis induction compared to primordial follicles or may reflect, perhaps, a greater capacity for DNA repair.

Irrespective of whether the quantity of growing follicles is decreased, increased or unaffected, exposure to cyclophosphamide or cisplatin in the short term does appear to impact the quality of growing and mature oocytes. Early studies indicated evidence of non-disjunction and increased chromosomal aberrations in mature, metaphase-II (MII) oocytes collected from cyclophosphamide-treated mice (Hansmann 1974, Hansmann & Probeck 1979). More recently, disrupted microtubule assembly, spindle structure and chromosome alignment have been reported in mouse MII oocytes exposed to cyclophosphamide both in vivo or in vitro (Jeelani et al. 2017, Del Castillo et al. 2021). Another recent study found similar results in mouse MII oocytes exposed to carboplatin (a cisplatin analogue) in vitro (Zhou et al. 2019). Moreover, poor oocyte quality is also reflected by elevated rates of fetal malformations and resorptions in pregnancies established 1 week post-cyclophosphamide treatment (Meirow et al. 2001). This suggests that growing follicles, although appearing to have a higher threshold for apoptosis induction, may not be able to efficiently repair the DNA damage or that organelles and/or maternal factors (proteins and RNA laid down in the oocytes during folliculogenesis) were compromised. This decrease in oocyte quality could result in detrimental effects on subsequent embryos formed, negatively impacting embryo implantation, survival and offspring health in pregnancies established shortly after chemotherapy.

Impact on ovarian stroma and vasculature

The ovarian stroma and vasculature have an important role in coordinating ovarian steroid production (particularly oestradiol and testosterone) and supporting both the health of the ovarian reserve and normal follicular development (Bedoschi et al. 2016). In addition to a direct impact on ovarian follicles, cyclophosphamide and cisplatin exposure can cause stromal and vascular damage (Nicosia et al. 1985, Marcello et al. 1990, Meirow et al. 2007, Oktem & Oktay 2007b, Saleh et al. 2015, Luo et al. 2017, Pascuali et al. 2018). Moreover, a recent study examined the impact of phosphoramide mustard (a cyclophosphamide metabolite) and cisplatin on various ovarian somatic cell types in cultured prepubertal mouse ovaries, including pre-granulosa cells, pre-thecal cells and ovarian surface epithelium (Marcozzi et al. 2019). Both phosphoramide mustard and cisplatin exposure had similar effects, inducing extensive DNA damage and cell cycle arrest in these cell types (Marcozzi et al. 2019). Altogether, these effects may impact ovarian endocrine function whilst also indirectly impacting follicular development.

Mechanisms of chemotherapy-induced ovarian damage

In order to identify new targets for fertility preservation, it is important to understand the mechanisms by which chemotherapies induce ovarian damage. However, the precise molecular mechanisms by which cyclophosphamide and cisplatin exert ovarian damage and deplete the ovarian reserve are not fully understood. Several putative mechanisms have been proposed to cause primordial follicle loss, and the relative contributions of these to chemotherapy-induced ovarian reserve depletion are still under active debate. Overall, proposed mechanisms fall into two major categories: (i) direct oocyte damage and apoptosis of primordial follicles and (ii) accelerated primordial follicle activation and ‘burnout’. A third factor may be induction of oxidative stress via the production of reactive oxygen species, which is thought to mediate stromal and vascular damage (Szymanska et al. 2020).

Direct DNA damage and apoptosis

There is a large body of evidence to suggest that direct apoptosis of primordial follicles is likely the primary mechanism of cyclophosphamide- and cisplatin-induced primordial follicle loss (Plowchalk & Mattison 1992, Oktem & Oktay 2007a, Gonfloni et al. 2009, Petrillo et al. 2011, Kerr et al. 2012a, Kim et al. 2013, Morgan et al. 2013, Bellusci et al. 2019, Luan et al. 2019, Nguyen et al. 2019). For example, several recent in vivo mouse studies have demonstrated that primordial follicles sustain widespread DNA damage following cyclophosphamide or cisplatin exposure in the form of double-stranded breaks, predominantly within the oocyte (Bellusci et al. 2019, Nguyen et al. 2019, Yang et al. 2021). This occurs as early as 8 h after a single, high dose of cyclophosphamide or cisplatin (Nguyen et al. 2019) but may persist up to 24 h after low-dose cyclophosphamide treatment (Yang et al. 2021). This is evidenced by the presence of phosphorylated H2A histone family member X (γH2AX) staining in approximately half of primordial follicle oocytes (Nguyen et al. 2019). Moreover, terminal deoxynucleotidyl transferase dUTP nick-end labelling (TUNEL)-positive primordial follicle oocytes were detected at both 8- and 24-h post-treatment, which is indicative of apoptosis and imminent follicle atresia (Nguyen et al. 2019).

Shortly after the formation of double-stranded DNA breaks, ataxia–telangiectasia-mutated (ATM) kinase is activated. ATM can initiate DNA repair pathways but more commonly will phosphorylate and activate TAp63α (Suh et al. 2006, Livera et al. 2008) (Fig. 1). Subsequently, the pro-apoptotic BH3-only family members – which include PUMA and NOXA – are transcriptionally activated (Kerr et al. 2012b). Interestingly, activation of PUMA can also be mediated by other factors such as p53, p73 (another p53 homologue) and forkhead box O3a (FOXO3a), independent of transactivated p63 α (TAp63α; a p63 isoform and p53 homologue (Yu & Zhang 2008). PUMA (and to a lesser extent, NOXA) can induce apoptosis by binding and inactivating the pro-survival B-cell lymphoma 2 (BCL-2) proteins, resulting in the activation of BCL-2-associated X protein (BAX) and BCL-2 homologous antagonist killer (BAK) (Aubrey et al. 2018). PUMA is the more potent inducer of apoptosis, as it binds to all five of the pro-survival BCL-2 proteins with high affinity and can also directly interact with BAX (Youle & Strasser 2008). Upon activation of BAX and BAK, mitochondrial outer membrane permeabilisation occurs, which is regarded as the ‘point of no return’ in apoptosis. This results in cytochrome c release, caspase activation, and formation of the apoptosome, causing apoptosis of the oocyte and subsequent atresia (death) of the primordial follicle (Aubrey et al. 2018).

Figure 1
Figure 1

Direct DNA damage and apoptosis of primordial follicles following chemotherapy treatment. PUMA is the pro-apoptotic protein that mediates primordial follicle apoptosis in response to chemotherapy. In response to chemotherapy-induced DNA damage, PUMA is activated either by TAp63 or by other factors, such as p53, p73 or FOXO3a. Then, PUMA unleashes BAX and BAK either directly or indirectly by inhibiting the pro-survival BCL-2 proteins – which normally inhibit BAX and BAK. Subsequently, mitochondrial outer membrane permeabilisation occurs – the apoptotic ‘point of no return’ – causing cytochrome c release, caspase activation and formation of the apoptosome. Ultimately, this results in the apoptosis of the oocyte and atresia of the primordial follicle.

Citation: Reproduction and Fertility 4, 2; 10.1530/RAF-22-0123

Prevention of apoptosis, through either genetic knockout of apoptotic genes or small-molecule inhibitors of apoptotic proteins, prevents primordial follicle depletion in response to chemotherapy (Nguyen et al. 2018, Luan et al. 2019). In this regard, a recent study utilising apoptosis-resistant (Puma–/–) mice demonstrated that when rescued from apoptosis, primordial follicles exposed to cisplatin are able to efficiently repair DNA damage via the homologous recombination pathway (Nguyen et al. 2021). This was evidenced by positive RAD51 staining in oocytes of primordial follicles 8 h post-treatment and complete resolution of γH2AX foci by 5 days. Together, these data strongly support direct damage as a key mechanism of oocyte depletion due to cyclophosphamide and cisplatin. Thus, preventing apoptosis of primordial follicles in response to chemotherapy represents a promising avenue for fertility preservation research.

Accelerated primordial follicle activation and ‘burnout’

It is also recently been proposed that primordial follicle depletion might occur due to accelerated primordial follicle activation. Termed ‘burnout’, this theory states that cyclophosphamide and cisplatin only induce DNA damage and apoptosis of growing follicles not primordial follicles (Kalich-Philosoph et al. 2013, Chang et al. 2015, Roness et al. 2019). By rapidly depleting growing follicles, it is proposed that factors which normally regulate or suppress primordial follicle activation are impaired. These include phosphatase and tensin homolog, FOXO3a and AMH, among others (Dunlop & Anderson 2014). This results in a subsequent upregulation of factors which promote primordial follicle activation, such as the phosphoinositide 3-kinase (PI3K) and mechanistic target of rapamycin (mTOR) pathways plus their downstream regulators (Chen et al. 2020). Once these follicles are recruited from the resting pool, an increase of early growing (primary and secondary) follicles is then expected. Ultimately, since primordial follicle activation is not reversible, this increased activation is thought to result in an exhaustion, or ‘burnout’, of the remaining primordial follicle population (Fig. 2). Since chemotherapy regimens often consist of multiple doses, this could continually deplete the growing follicle population and indirectly the primordial follicle pool.

Figure 2
Figure 2

Overactivation and ‘burnout’ of primordial follicles following chemotherapy treatment. The ‘burnout’ theory states that chemotherapy treatment causes premature depletion of the primordial follicle pool via accelerated activation of primordial follicles. Chemotherapy treatment causes DNA damage in growing follicles, resulting in apoptosis and atresia of the growing follicle pool. Subsequently, this results in accelerated activation of the primordial follicle pool through increased activation of phosphoinositide 3-kinase (PI3K) and mechanistic target of rapamycin (mTOR) signalling. This may occur directly, or indirectly, via loss of inhibitory signals such as AMH (anti-Müllerian hormone) or FOXO3a (forkhead box O3a). These newly activated growing follicles are then susceptible to chemotherapy-induced damage, which leads to a cycle of accelerated activation that ultimately causes a burnout of the primordial follicle pool.

Citation: Reproduction and Fertility 4, 2; 10.1530/RAF-22-0123

However, recent literature on whether cyclophosphamide and cisplatin induce overactivation of primordial follicles is conflicting. A study utilising cultured human cortical samples treated in vitro with cyclophosphamide metabolites demonstrated significantly decreased proportions of primordial follicle and increased proportions of growing follicles, with no evidence of apoptosis found (Lande et al. 2017). It is unclear whether the changes in follicle proportions were due to increased death of primordial follicles or increased activation. However, a study examining ovaries from women treated with alkylating agents 1–6 months prior to collection reported a decrease in in primordial follicle numbers and an increase in the absolute number of growing follicles (Shai et al. 2021). Moreover, FOXO3a immunostaining indicated a possible decrease in the percentage of FOXO3a-positive primordial follicle oocytes, which could indicate increased follicle activation (Shai et al. 2021). On the other hand, continuous treatment with either 2 mg/kg or 5 mg/kg cisplatin was shown to deplete primordial follicles in vivo in mice, without evidence of accelerated primordial follicle activation (Eldani et al. 2020). Additionally, using a murine human ovarian xenograft model, single-cell RNA sequencing of laser-captured primordial follicle oocytes found no difference in the expression of Akt, rpS6 or Foxo3a 12 h following cyclophosphamide treatment (Titus et al. 2021). DNA damage and apoptosis were also detected in primordial follicles, as demonstrated by significant increases in γH2AX and active caspase-3 staining.

Overall, the current body of literature suggests that the precise contribution of accelerated primordial follicle activation to chemotherapy-induced follicle depletion remains unclear and is yet to be firmly established. Nevertheless, it is likely that this is contributing to primordial follicle loss in response to chemotherapy to some degree albeit potentially to a lesser extent than direct apoptosis.

Existing fertility preservation options

For paediatric, adolescent or reproductive-age females undergoing cancer treatment, fertility preservation methods are available but are associated with some important limitations (Table 2). These existing options fall into two categories: (i) cryopreservation (i.e. freezing), which includes cryopreservation of oocytes, embryos or ovarian tissue and (ii) suppression of ovarian function using GnRH agonists (Rodriguez-Wallberg & Oktay 2014).

Table 2

Summary of existing fertility preservation options and their respective advantages and limitations.

Category/technique Advantages Limitations
Cryopreservation
 Oocyte cryopreservation
Established technique in post-pubertal females Invasive and requires ovarian hormone stimulation
Does not require sperm and thus is more accessible to unpartnered females Can delay the commencement of cancer treatment
Unavailable to prepubertal girls
Requires subsequent IVF
Does not prevent ovarian damage from occurring and thus will not prevent POI
 Embryo cryopreservation
Established technique in post-pubertal females Invasive and requires ovarian hormone stimulation
Can delay the commencement of cancer treatment
Unavailable to prepubertal girls
Requires sperm, from either a partner or a donor
Does not prevent ovarian damage from occurring and thus will not prevent POI
 Ovarian tissue cryopreservation
Available to prepubertal girls Invasive
Does not require ovarian hormone stimulation Limited success rates
Does not significantly delay the commencement of cancer treatment Remains experimental in prepubertal patients
No longer considered experimental in adult patients Risk of cancer reintroduction upon transplant
Unable to fully compensate for endogenous ovarian function and thus will not likely prevent POI
Suppression of ovarian endocrine function Does not delay the commencement of cancer treatment Supporting evidence is conflicting and limited
Does not prevent ovarian damage from occurring and thus will not prevent POI
 GnRH agonists
 Oral contraceptives

GNRH, gonadotropin-releasing hormone; IVF, in vitro fertilisation; POI, premature ovarian insufficiency.

Oocyte and embryo cryopreservation

Oocyte and embryo cryopreservation have long been the backbone of female fertility preservation in adult cancer patients. These techniques both initially involve the retrieval of mature MII oocytes prior to commencement of chemotherapy treatment. These oocytes are either frozen after collection or undergo in vitro fertilisation (IVF) with partner or donor sperm to form embryos, which are then frozen. However, this invasive procedure first requires hormone stimulation to produce multiple mature oocytes, which in some cases will require a delay in the commencement of treatment that may not be feasible. Moreover, oocyte retrieval and IVF are inherently expensive and thus are not accessible to all patients (Quinn et al. 2008). Critically, these options do not prevent ovarian damage from occurring and thus do not prevent POI or its clinical sequelae.

Ovarian tissue cryopreservation

Ovarian tissue cryopreservation (OTCP) involves the collection of small slices of ovarian cortex usually via laparoscopic surgery prior to the commencement of cancer treatment. This tissue is then cryopreserved and retransplanted (i.e. autografted) at a later time (Quinn et al. 2008). An increasing number of livebirths have been reported following reimplantation of cryopreserved ovarian tissue, and, as such, this technique is no longer considered experimental in adult women in many countries (Donnez & Dolmans 2015). However, this technique is invasive, and, to date, success rates have been modest (Forman 2018, Andersen et al. 2019). Moreover, the transplantation of the ovarian tissue back into the patient carries a risk of reintroducing malignant cells from the primary tumour, especially for haematologic malignancies (Dolmans et al. 2010, Bittinger et al. 2011, Gook & Edgar 2019).

For prepubertal girls, fertility preservation remains a challenge. Cryopreservation of oocytes or embryos is not possible; thus, cryopreservation of ovarian tissue is the only available option. As well as the issues with OTCP already described above, to date no livebirths have been reported following the reimplantation of prepubertally harvested ovarian tissue into adult survivors of childhood cancer (Quinn et al. 2008, Forman 2018, Kim et al. 2018). Therefore, a non-invasive pharmacological ovarian protectant (i.e. ferto-protective agent) would provide extraordinary benefit to female cancer patients from childhood through reproductive age and would clearly be preferable to additional, urgent, surgical intervention around the time of cancer diagnosis.

GnRH agonists

GnRH agonists are currently the only non-invasive clinical agents aimed at preserving fertility, although their efficacy in preventing chemotherapy-induced POI has been widely debated (Partridge 2012, Loren et al. 2013, Moore et al. 2015). The use of these agents is based on the idea that by suppressing ovarian endocrine function, the ovary may be protected from the ovotoxic effects of chemotherapy (Fig. 3). This hypothesis was developed following clinical observations that post-pubertal females are more vulnerable to chemotherapy-induced POI than prepubertal girls (Meirow & Nugent 2001). However, this theory is problematic from a mechanistic standpoint, since primordial follicles do not express gonadotrophin receptors, and DNA damage can still occur to oocytes stored within primordial follicles regardless of pituitary hormone status (Oktay et al. 1997, Méduri et al. 2002). Indeed, the literature on the clinical efficacy of GnRH agonists for ovarian protection is conflicting, and many prospective randomised trials have failed to clearly demonstrate a beneficial effect for preserving fertility (Partridge 2012). However, a recent meta-analysis reported improved spontaneous pregnancy rates in breast cancer patients treated with GnRH agonists, though these findings were only applicable to hormone receptor-negative women (Li et al. 2022). Thus, despite the emergence of some prospective data, evidence of the benefit of GnRH agonists for fertility preservation remains uncertain and limited.

Figure 3
Figure 3

Rationale behind the use of GnRH agonists for fertility preservation. The rationale behind the use of GnRH agonists is that chemotherapy treatment results in a depletion of growing follicles within the ovary. Loss of these hormone-producing follicles causes a decrease in oestradiol and inhibin levels, which in turn results in an increase in follicle-stimulating hormone (FSH) levels via negative feedback. This increase in FSH is thought to accelerate recruitment of primordial follicles from the resting pool. Administration of GnRH agonists (GnRHa) prevents this increase in FSH levels which, by extension, is thought to prevent this overactivation of primordial follicles.

Citation: Reproduction and Fertility 4, 2; 10.1530/RAF-22-0123

Current avenues in oncofertility research

Considerable research has been conducted in recent years to discover new, more effective fertility preservation strategies which can prevent ovarian damage and preserve oocytes in vivo. Technologies such as whole ovarian transplantation, oocyte in vitro growth and maturation and development of artificial ovaries are currently under investigation (Kim et al. 2021, McClam & Xiao 2022). Additionally, stem cell treatment is another emerging area of research, with several studies investigating the possibility that administration of mesenchymal stem cells or stem cell-derived factors might improve ovarian function post-cyclophosphamide or cisplatin treatment in murine models (Hong et al. 2020, Çil & Mete 2021, Salvatore et al. 2021, Shin et al. 2021, Bahrehbar et al. 2022, Liu et al. 2022). However, the remainder of this review will focus on current avenues of research aimed at preventing follicle loss after damage induced by alkylating and alkylating-like agents, utilising adjuvant ferto-protective agents aimed at (i) preventing apoptosis of ovarian follicles or (ii) blocking overactivation of primordial follicles. Though, when interpreting the results of the following studies, it is important to consider the dose of the ferto-protective agent utilised as well as the ratio of this to the chemotherapy drug.

Preventing apoptosis of primordial follicles

Given that direct DNA damage and subsequent apoptosis appears to be a major mechanism behind cyclophosphamide- and cisplatin-induced primordial follicle loss, considerable research has been conducted into agents that may prevent oocyte apoptosis from occurring. These can be separated into two major categories: (i) protein kinase inhibitors and (ii) ceramide-induced death pathway inhibitors (Table 3). Additionally, the administration of luteinising hormone has shown some efficacy in preventing primordial follicle loss and preserving long-term fertility in vivo in mice following exposure to cyclophosphamide and cisplatin (Rossi et al. 2017, Del Castillo et al. 2021).

Table 3

Summary of current avenues of research into ferto-protective agents to prevent chemotherapy-induced ovarian damage.

Aim Class/agent Mechanism Target agent Evidence of primordial follicle protection
Block apoptosis of primordial follicles
 Protein kinase inhibitors
  Imatinib Prevents TAp63α activation and subsequent apoptosis via inhibition of c-ABL Cisplatin Conflicting evidence regarding efficacy and rationale behind mechanism of protection (Gonfloni et al. 2009, Kerr et al. 2012a , Kim et al. 2013, Morgan et al. 2013); Shown to actually induce primordial follicle apoptosis in mice and humans (Bildik et al. 2018, Salem et al. 2020)
  Asciminib Prevents TAp63α activation and subsequent apoptosis via inhibition of c-ABL Cyclophosphamide Evidence of efficacy in vivo in mice; however, follicle quantification method utilised may be insufficient (Mattiello et al. 2021)
  CK2II, ETP46464 Prevents TAp63α activation and subsequent apoptosis via inhibition of CHK2 and ATR Cyclophosphamide Evidence of efficacy in vitro in mice (Luan et al. 2019) but no in vivo evidence of efficacy
  KU55933 Prevents TAp63α activation and subsequent apoptosis via inhibition of ATM Cyclophosphamide Evidence of efficacy in vitro in mice (Ganesan & Keating 2016) but no in vivo evidence of efficacy
 Inhibitors of ceramide-induced apoptosis Prevents the activation of apoptosis via inhibition of ceramide Cyclophosphamide
  S1P Effective in murine human ovarian xenograft models (Meng et al. 2014, Li et al. 2017) Ineffective in rats in vivo (Kaya et al. 2008)
  C1P Evidence of efficacy in vivo in mice (Pascuali et al. 2018)
 Endogenous hormones
  LH Stimulates release of anti-apoptotic signals from somatic cells possessing LH receptors Cyclophosphamide, cisplatin Evidence of efficacy in vivo in mice following cisplatin treatment (Rossi et al. 2017); mild protection against primordial follicle loss and preservation of long-term fertility in vivo in mice following cyclophosphamide, though small sample sizes were utilised (Del Castillo et al. 2021)
Prevent overactivation of primordial follicles
 PI3K pathway inhibitors Prevent primordial follicle activation via inhibition of PI3K pathway activity
  AS101 Cyclophosphamide Evidence of efficacy in vivo in mice, although only a short-term breeding study was performed (Kalich-Philosoph et al. 2013).
  Melatonin Cisplatin Evidence of efficacy in vivo in mice; however, follicle quantification methods utilised may be insufficient and long-term fertility was not assessed (Jang et al. 2016, 2017)
  Rutin Cisplatin Evidence of efficacy in vivo in mice, although fertility was not assessed (Lins et al. 2020)
 mTOR pathway inhibitors Prevent primordial follicle activation via inhibition of mTOR pathway activity
  RAD001, INK128 Cyclophosphamide, cisplatin Evidence of efficacy in vivo in mice; however, follicle quantification method utilised may be insufficient and long-term fertility was not assessed (Goldman et al. 2017, Tanaka et al. 2018)
  Rapamycin Cyclophosphamide Evidence of efficacy in vivo in mice, although long-term fertility was not assessed (Zhou et al. 2017)
 Endogenous hormones
  rAMH Prevents primordial follicle activation via administration of AMH, a regulator of primordial follicle activation Cyclophosphamide Mixed results regarding the protection of primordial follicles in several in vivo murine studies (Kano et al. 2017, Roness et al. 2019, Sonigo et al. 2019); inconclusive efficacy in preserving long-term fertility in mice (Roness et al. 2019, Sonigo et al. 2019)
Reduce oxidative stress
 Antioxidants Prevent oxidative stress-induced stromal and vascular damage by eliminating reactive oxygen species, thus preventing primordial follicle loss indirectly
  Bilberry Cisplatin Some evidence of ovarian protection reported in one in vivo murine study, although primordial follicles were not quantified and fertility was not assessed (Pandir et al. 2014)
  Sildenafil citrate Cisplatin Evidence of efficacy in vivo in mice; however, follicle quantification methods utilised may be insufficient and fertility was not assessed (Taskin et al. 2015)
  Hydrogen-rich saline Cisplatin Some evidence of ovarian protection reported in one in vivo murine study, although primordial follicles were not quantified and fertility was not assessed (Meng et al. 2015)
  Mirtazapine, hesperidin Cyclophosphamide Higher rate of fertility reported in 6-month breeding study in mice, although no other fertility parameters were analysed (Altuner et al. 2013); evidence of efficacy in vivo in mice; however, follicle quantification methods utilised may be insufficient (Khedr 2015)
  Puerarin Cyclophosphamide Evidence of efficacy in vivo in mice, although fertility was not assessed (Chen et al. 2021)
  Resveratrol Cyclophosphamide Evidence of efficacy in vivo in mice, although fertility was not assessed (Chhabria et al. 2022)

AS101, ammonium trichloro(dioxyethylene-o,o’)tellurate; ATM, ataxia–telangiectasia mutated; ATR, ataxia–telangiectasia and Rad3 related; CHK2, checkpoint kinase 2; LH, luteinising hormone; mTOR, mammalian target of rapamycin; PI3K, phosphoinositide 3-kinase; rAMH, recombinant anti-Müllerian hormone; SIP, sphingosine-1-phosphate.

Protein kinase inhibitors

Imatinib

Imatinib is a selective inhibitor of breakpoint cluster region (BCR)-Abelson murine leukemia viral oncogene homolog 1 (ABL), a tyrosine kinase produced by the abnormal fusion of two genes – BCR and cellular-ABL (c-ABL). This chimeric gene, termed Philadelphia chromosome, is strongly associated with the development of leukaemia. Inhibition of BCR-ABL prevents apoptosis by blocking mitochondrial cytochrome c release and caspase activation (Deming et al. 2004). Additionally, imatinib is also able to inhibit c-ABL, which is associated with induction of the intrinsic apoptotic pathway via the activation of p73 (Yuan et al. 1999). This could, theoretically, downregulate TAp63-mediated apoptosis within primordial follicles.

Initially, imatinib was reported to prevent cisplatin-induced primordial follicle apoptosis in mice both in vitro and in vivo (Gonfloni et al. 2009). Similar results were obtained in two subsequent in vitro studies (Kim et al. 2013, Morgan et al. 2013). However, other studies found opposing results, showing that imatinib conferred no protection from cisplatin-induced apoptosis in vivo in wild-type mice (Kerr et al. 2012a ) or in mice bearing human ovarian cortical xenografts (Bildik et al. 2018) nor in vitro in human ovarian cortical samples (Bildik et al. 2018). However, in the latter case, it is important to consider that the imatinib dose used in vitro may equate to a higher dose than what was utilised in the aforementioned in vivo studies. Additionally, subsequent work in reply to Kerr et al. corroborated the initial findings by Gonfloni et al. (Maiani et al. 2012). The discrepancy in these two reports may be due to differences in the brands of cisplatin utilised and their respective efficacies in inducing cell death (Maiani et al. 2012). After repeating some key experiments using an equivalent cisplatin dose from Gonfloni et al. but utilising the same cisplatin brand as Kerr et al., Maiani et al. found that imatinib treatment protected oocytes from primordial and primary follicles from cisplatin both ex vivo and in vivo. It is possible that imatinib can protect oocytes at lower doses of cisplatin (Gonfloni et al. 2009, Maiani et al. 2012) but is unable to rescue damage at higher doses (Kerr et al. 2012a ). This highlights that the dose of chemotherapy, as well as the ratio of chemotherapy to the ferto-protective agent utilised, must be taken into consideration when designing and interpreting studies aimed at preventing chemotherapy-induced ovarian damage.

On the contrary, it was more recently found that c-ABL was not activated in response to cisplatin treatment, which may explain why imatinib may not in fact prevent primordial follicle apoptosis. Indeed, imatinib treatment may actually result in primordial follicle depletion and increased atresia (Bildik et al. 2018). This is the first molecular evidence that imatinib may in fact be ovotoxic to humans albeit in culture. A recent murine study corroborated this idea, showing that in vivo imatinib administration depletes primordial follicles and reduces embryo quality in mice (Salem et al. 2020). However, earlier studies have indicated that imatinib does not induce ovarian toxicity (Maiani et al. 2012). Thus, it remains unclear and contentious if imatinib is an effective fertility preservation strategy and may in fact be detrimental to ovarian function.

Asciminib

Asciminib is an allosteric inhibitor of c-ABL. A recent study showed that administration of asciminib can protect ovarian follicles from cyclophosphamide-induced apoptosis (Mattiello et al. 2021). However, this study utilised a suboptimal follicle quantification method in which the total number of follicles within the ovary was not counted and only mid-ovary sections were analysed. Furthermore, the number of primordial and primary follicles was reported together and not separately, making it difficult to determine the precise effects of asciminib on the ovarian reserve. Thus, further work is required to determine the utility of asciminib as an ovary protectant.

CK2II and ETP46464

Ataxia–telangiectasia and Rad3-related (ATR) and checkpoint kinase 2 (CHK2) are both serine–threonine kinases that have important roles in the DNA damage response. Following the induction of DNA damage, ATR activates CHK2 which, in turn, can activate TAp63 and trigger apoptosis within oocytes (Rinaldi et al. 2019). It has been shown that CHK2-deficient mice are resistant to irradiation-induced oocyte loss in mice, and in vitro inhibition of either CHK2 or CK1 is able to block oocytes from TAp63-mediated apoptosis caused by radiation, cisplatin, cyclophosphamide or doxorubicin (Kim et al. 2013, Tuppi et al. 2018, Luan et al. 2019, Rinaldi et al. 2019). CK2II (a CHK2 inhibitor) and ETP46464 (an ATR inhibitor) were both able to prevent primordial follicle apoptosis in cultured mouse ovaries in vitro from 4-hydroxyperoxycyclophophamide, a cyclophosphamide metabolite (Luan et al. 2019). Levels of phosphorylated TAp63α within exposed primordial follicle oocytes were barely detectable following CK2II and ETP46464 administration. Although promising, the in vivo efficacy of these inhibitors for fertility preservation is yet to be determined, and side effects need to be thoroughly assessed.

KU55933

ATM is a key mediator of the DNA damage response. In oocytes, when DNA damage is irreparable, ATM can induce apoptosis by activating TAp63 either directly or indirectly via CHK1/2 (Ou et al. 2005, Tuppi et al. 2018). Ganesan et al. found that KU55933, an ATM inhibitor, prevents primordial follicle loss in vitro, in cultured rat ovaries following exposure to phosphoramide mustard, a cyclophosphamide metabolite (Ganesan & Keating 2016). Currently, there is no in vivo evidence of the effectiveness of KU55933 in preventing primordial follicle apoptosis, and the important role for ATM in coordinating the DNA damage response in other cells, may preclude its use as a fertility preservation agent unless targeted delivery could be achieved.

Inhibitors of ceramide-induced apoptosis

Ceramides play key roles in both the intrinsic and extrinsic apoptotic pathways, and ceramide-induced apoptosis has been linked to the age-related decline in primordial follicles (Perez et al. 2005). Sphingosine-1-phosphate (S1P) and ceramide-1-phosphate (C1P) are endogenous sphingolipids which are natural inhibitors of ceramide-induced apoptosis.

Sphingosine-1-phosphate

S1P has previously been shown to prevent cyclophosphamide-induced primordial follicle loss in mice, in vitro and in vivo (Morita et al. 2000, Hancke et al. 2007). In a murine human ovarian xenograft model, administration of S1P was shown to prevent apoptosis in human primordial follicles in response to cyclophosphamide, as evidenced by a significantly reduced number of primordial follicles positive for activated caspase-3 (Li et al. 2014). However, an earlier study found no difference in the level of active caspase-3 or TUNEL-positive primordial follicles between cyclophosphamide-exposed S1P-treated rats compared with controls (Kaya et al. 2008). Importantly, both of these studies did not quantify the overall number of primordial follicles within the ovaries, making it difficult to fully assess the efficacy of S1P in protecting the entire primordial follicle pool. A later mouse xenograft study utilising human fetal ovaries reported that S1P treatment did significantly protect primordial follicles from cyclophosphamide (Meng et al. 2014).

Ceramide-1-phosphate

A recent mouse study demonstrated that C1P delivered intrabursally protects the ovarian reserve and prevents primordial follicle apoptosis following cyclophosphamide exposure (Pascuali et al. 2018). Additionally, in vitro maturation and IVF studies showed that C1P administration preserves oocyte quality. A short-term, two-round breeding study was also conducted, which showed fertility was also preserved. Lastly, gross uterine morphology and endometrial structure appeared normal, suggesting C1P may also protect from uterine damage. Although promising, evidence of the efficacy of C1P for fertility preservation is limited to this study only.

Blocking overactivation of primordial follicles

Although still not fully understood, primordial follicle activation is thought to be mediated by induction of PI3K and mTOR signalling – the latter of which acts downstream of PI3K – as well as loss of inhibitory signals, such as AMH (Chen et al. 2020). In addition to GnRH agonists, several other agents have been investigated in recent years with the aim of blocking overactivation of primordial follicles. These can be divided into three categories: (i) PI3K pathway inhibitors; (ii) mTOR pathway inhibitors; and (iii) glycoprotein hormones.

PI3K and mTOR pathway inhibitors

AS101

Ammonium trichloro(dioxyethylene-o,o’)tellurate (AS101) is an immunomodulator that has also been shown to inhibit PI3K pathway activity (Hayun et al. 2006). In the initial publication which proposed the ‘burnout’ theory, Kalich-Philosoph et al. also reported that AS101 is able to suppress primordial follicle activation and, thus, protect the ovarian reserve following cyclophosphamide exposure (Kalich-Philosoph et al. 2013). Co-treatment with AS101 resulted in a significant increase in primordial follicle survival when compared to mice exposed only to cyclophosphamide. A breeding study revealed that AS101 co-treated mice also appeared more fertile, producing significantly more pups per litter and cumulative pups compared to cyclophosphamide-treated mice. However, mice only underwent three breeding rounds, and thus it remains unclear whether AS101 administration can effectively preserve the fertile life span following cyclophosphamide treatment.

Melatonin

Melatonin is an endogenous hormone produced by the pineal gland, which has antioxidant properties and is able to suppress PI3K pathway activity. Two studies, from the same group, report that melatonin prevents cisplatin-induced primordial follicle depletion (Jang et al. 2016, 2017). However, follicle quantification was conducted on only three 5 µm ovarian sections per animal, which is likely insufficient to accurately estimate the number of primordial follicles within the whole ovary. Long-term fertility was also not assessed in either study. Similarly, a recent study indicated that melatonin prevents cyclophosphamide-induced POI in rats, although only total follicle numbers were reported and fertility was not assessed (Xu et al. 2022).

RAD001 and INK128

RAD001 is an mTOR complex (mTORC) 1 inhibitor, and INK128 inhibits mTORC1/2. A recent report showed that the administration of RAD001 or INK128 prevents the depletion of primordial follicles following cyclophosphamide treatment (Goldman et al. 2017). One of the limitations of this study is the use of density to quantify follicles, as this method does not account for the uneven distribution of primordial follicles within the whole ovary nor for differences in ovarian volume between groups. Follicle density may therefore not reflect actual numbers (Winship et al. 2020). Encouragingly, RAD001- and INK128-treated mice produced significantly more pups per litter after a single round of mating. In addition, another recent study reported that RAD001 administration protects from cisplatin-induced primordial follicle depletion (Tanaka et al. 2018), although a breeding study was not conducted. Thus, whether RAD001 or INK128 preserves the length of the fertile life span remains to be seen following cyclophosphamide or cisplatin exposure.

Rapamycin

Rapamycin is an allosteric inhibitor of mTOR, which has been shown to prevent cyclophosphamide-induced primordial follicle depletion in mice (Zhou et al. 2017). A breeding study was not conducted; thus, it is yet to be determined whether the administration of rapamycin can effectively preserve long-term fertility following cyclophosphamide. Additionally, PI3K/mTOR pathways have other complex roles within cells independent of primordial follicle activation, such as cell death and survival (Sarbassov et al. 2005, Steelman et al. 2011), that require further consideration when interpreting these data.

Endogenous hormones

Recombinant AMH

There is evidence to suggest that AMH is involved in suppressing primordial follicle activation and, thus, maintaining their quiescence, which has led to the hypothesis that administration of human recombinant AMH (rAMH) can prevent chemotherapy-mediated follicle loss. This concept was first investigated by Kano et al. who reported a significant increase in primordial follicles following cyclophosphamide exposure in mice treated with rAMH, when compared to controls (Kano et al. 2017). Although not discussed, follicle depletion was still extensive in cyclophosphamide exposed rAMH-treated mice compared to controls; thus, the level of protection is unclear, and long-term fertility was not assessed.

Another study reported that rAMH administration prevented cyclophosphamide-induced primordial follicle depletion; however, robust results were only obtained when primordial follicle number was expressed as a proportion of total follicles within the ovary and not in the raw follicle counts (Sonigo et al. 2019). A short breeding study was conducted, but it is not clear if fertility was improved. Interestingly, however, rAMH administration significantly increased the number of mature oocytes retrieved after superovulation, which is an exciting avenue to follow-up in future studies. A similar study by the same group reported a significant increase in primordial follicles in cyclophosphamide-exposed mice treated with rAMH (Roness et al. 2019). Fertility studies showed more promising results, with a significantly increased pregnancy rate in rAMH-treated mice over five breeding rounds. Although the breeding study ceased prior to the loss of fertility of the mice, and thus the entire fertile life span was not captured, these studies collectively suggest that rAMH may be an avenue worth investigating further.

Future perspectives

Chemotherapy may also alter uterine function

The unintended side effects of chemotherapy treatment on the ovary have been a major focus of research thus far. It is clear that chemotherapy treatment – particularly alkylating agents – can significantly impact a woman’s ovarian reserve and, by extension, fertile life span. However, the possibility of chemotherapy-induced damage to the remainder of the reproductive tract, including the uterus, has received considerably less attention.

The integrity of the uterus and, in particular, the endometrium is fundamental to the establishment and maintenance of successful pregnancy. The impact of and mechanisms behind radiotherapy-induced uterine damage on pregnancy success in female cancer survivors are incompletely characterised and have been recently reviewed (Garg et al. 2020, Griffiths et al. 2020). Additionally, there is emerging evidence to suggest that the uterus may be an additional site of chemotherapy-induced damage in women.

However, the nature and impact of chemotherapy-induced uterine injury are poorly understood. Information in the literature is sparse and restricted mostly to retrospective human studies, which are confounded by many inherent limitations and inconsistencies. These are mostly due to the lack of information available regarding treatment regimens used, including which agent(s) were used and whether combination therapies were administered. Moreover, ascertaining whether pregnancy outcomes in women have been impacted by chemotherapy-induced uterine-specific damage is often confounded by the ovarian and endocrine damage that occurs concurrently. Future studies in this area are critical to furthering our understanding of the impact of chemotherapy on the uterus and what mechanisms may underlie it. Furthermore, addressing this knowledge gap will be essential in the development of fertility preservation strategies specific to the uterus.

Novel, non-cytotoxic agents

Although chemotherapies such as cyclophosphamide and cisplatin remain a mainstay of many cancer treatment regimes, the use of more targeted therapies – such as immunotherapy and small-molecule inhibitors – is becoming more widespread. However, information on whether these newer therapies may also impact ovarian function and fertility, even in preclinical models, is somewhat limited. Recently, we and others critically reviewed the available literature and speculated on the potential impacts of immunotherapies and small-molecule inhibitors on reproductive and endocrine function in women (Alesi et al. 2021, Garutti et al. 2021, Lambertini et al. 2022, Rosario et al. 2022). We also recently demonstrated that a prominent class of immunotherapy – immune checkpoint inhibitors – causes profound ovarian dysfunction in a mouse model (Winship et al. 2022). This damage is likely to be permanent and may impact fertility, as a significant reduction of primordial follicles was found – a concerning issue as these drugs are already in widespread clinical use and reproductive endpoints are rarely assessed during clinical trials (Cui et al. 2021). A recent study looking at fertility-related prescribing information available for 32 novel non-cytotoxic cancer agents approved for use in the USA and Australia found that only four listed recommendations regarding potential human fertility risks (Volckmar et al. 2022). Clearly, investigation into the ovotoxicity of newer treatments, such as immunotherapy and other targeted therapies, must be prioritised.

Extending oncofertility research beyond animal models

Research conducted in animal models has greatly underpinned the field of oncofertility to date, given that measuring the ovotoxic effects of chemotherapy in women is difficult. This is because clinical data from women is often complicated by the fact that chemotherapies are generally administered in tailored multi-agent, multi-dose regimens, making it difficult to compare findings between different patients and to define the precise impacts of individual agents on ovarian function. Additionally, analysis of the whole ovary is generally not possible nor practical; and there are currently no methods available to directly assess the ovarian reserve of primordial follicles. Therefore, in a clinical setting, ovarian function must be investigated through alternative, indirect measures, such as assessing patient menstrual history, measuring ovarian volume or performing antral follicle counts or using surrogate endocrine markers, such as circulating AMH levels. Thus, investigation of the effects of chemotherapies and ferto-protective agents on the whole ovary utilising animal models, such as the mouse, remains imperative (Winship et al. 2020). It is also important to consider that the aforementioned clinical methods do not provide any insight into oocyte quality (Winship et al. 2020), further reinforcing the need for rigorous studies in animal models in order to fully characterise the long-term implications of chemotherapy treatment and ferto-protective agents on fertility and offspring health.

Despite these benefits and the fact that ovarian morphology, function and aging share many similarities in humans and rodents (Coxworth & Hawkes 2010, Cunha et al. 2019), rodent models do have some important limitations to consider. This includes their differences in life span, number of ovulations per cycle and the fact that they do not menstruate or experience menopause. Therefore, it is important that the field moves towards performing more studies in non-human primates and utilising human tissue more frequently, such as culturing human cortical pieces in vitro and establishing in vivo human ovarian xenograft animal models. However, access to human tissue is a significant barrier that must be improved in order to make this a reality. Additionally, the development of human ovary organoids is an exciting prospect that could also be highly beneficial. Furthermore, it is critical that further research be conducted to discover a direct biomarker of primordial follicles, to allow for more accurate measurement of the ovarian reserve in women.

Conclusions

It has been known for many years that traditional cytotoxic cancer treatments, like cyclophosphamide and cisplatin, can damage the ovary and compromise fertility. Much research now focuses on defining the cellular and molecular mechanisms that underlie this ovarian damage, as this knowledge is critical for the development of effective fertility preservation agents aimed at preventing follicle depletion to preserve endogenous endocrine function and fertility. In addition, understanding the extent to which new precision cancer treatments, such as immunotherapies, compromise ovarian function is now emerging as a priority for researchers, clinicians and patients alike. Whilst the ovary has been the focus of studies concerning cancer treatment and infertility, moving forward, assessing impacts of existing and emerging cancer treatments on the uterus may be important for maximising pregnancy outcomes in female cancer survivors.

Declaration of interest

The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the research reported.

Funding

This work was made possible through Victorian State Government Operational Infrastructure Support and Australian Government NHMRC IRIISS. Additionally, this work was supported by funding from the Australian Research Council (ARC); KJH – FT190100265 and ALW – DE21010037, the National Health and Medical Research council (NHMRC); JMS-2011299, and the Australian National Breast Cancer Foundation (NBCF) grant no. IIRS-22-092. LRA is supported by an Australian Government Research Training Program scholarship and a Monash Graduate Excellence Scholarship. Figures were created using BioRender.

Author contribution statement

LRA designed tables and figures. All authors contributed to writing and editing the manuscript.

References

  • Alesi LR, Winship AL & & Hutt KJ 2021 Evaluating the impacts of emerging cancer therapies on ovarian function. Current Opinion in Endocrine and Metabolic Research 18 1528. (https://doi.org/10.1016/j.coemr.2020.12.004)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Almeida JZ, Lima LF, Vieira LA, Maside C, Ferreira ACA, Araújo VR, Duarte ABG, Raposo RS, Báo SN, Campello CC, et al.2021 5-fluorouracil disrupts ovarian preantral follicles in young C57BL6J mice. Cancer Chemotherapy and Pharmacology 87 567578. (https://doi.org/10.1007/s00280-020-04217-7)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Altuner D, Gulaboglu M, Yapca OE & & Cetin N 2013 The effect of mirtazapine on cisplatin-induced oxidative damage and infertility in rat ovaries. TheScientificWorldJournal 2013 327240327240. (https://doi.org/10.1155/2013/327240)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Anderson RA, Themmen APN, Qahtani AA, Groome NP & & Cameron DA 2006 The effects of chemotherapy and long-term gonadotrophin suppression on the ovarian reserve in premenopausal women with breast cancer. Human Reproduction 21 25832592. (https://doi.org/10.1093/humrep/del201)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Andersen ST, Pors SE, Poulsen LC, Colmorn LB, Macklon KT, Ernst E, Humaidan P, Andersen CY & & Kristensen SG 2019 Ovarian stimulation and assisted reproductive technology outcomes in women transplanted with cryopreserved ovarian tissue: a systematic review. Fertility and Sterility 112 908921. (https://doi.org/10.1016/j.fertnstert.2019.07.008)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Aubrey BJ, Kelly GL, Janic A, Herold MJ & & Strasser A 2018 How does p53 induce apoptosis and how does this relate to p53-mediated tumour suppression? Cell Death and Differentiation 25 104113. (https://doi.org/10.1038/cdd.2017.169)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bahrehbar K, Gholami S, Nazari Z & & Malakhond MK 2022 Embryonic stem cells-derived mesenchymal stem cells do not differentiate into ovarian cells but improve ovarian function in pof mice. Biochemical and Biophysical Research Communications 635 9298. (https://doi.org/10.1016/j.bbrc.2022.10.014)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bedoschi G, Navarro PA & & Oktay K 2016 Chemotherapy-induced damage to ovary: mechanisms and clinical impact. Future Oncology 12 23332344. (https://doi.org/10.2217/fon-2016-0176)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bellusci G, Mattiello L, Iannizzotto V, Ciccone S, Maiani E, Villani V, Diederich M & & Gonfloni S 2019 Kinase-independent inhibition of cyclophosphamide-induced pathways protects the ovarian reserve and prolongs fertility. Cell Death and Disease 10 726. (https://doi.org/10.1038/s41419-019-1961-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bildik G, Akin N, Senbabaoglu F, Sahin GN, Karahuseyinoglu S, Ince U, Taskiran C, Selek U, Yakin K, Guzel Y, et al.2015 GnRH agonist leuprolide acetate does not confer any protection against ovarian damage induced by chemotherapy and radiation in vitro. Human Reproduction 30 29122925. (https://doi.org/10.1093/humrep/dev257)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bildik G, Acılan C, Sahin GN, Karahuseyinoglu S & & Oktem O 2018 C-ABL is not actıvated in DNA damage-induced and TAp63-mediated oocyte apoptosıs in human ovary. Cell Death and Disease 9 943943. (https://doi.org/10.1038/s41419-018-1026-7)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bittinger SE, Nazaretian SP, Gook DA, Parmar C, Harrup RA & & Stern CJ 2011 Detection of Hodgkin lymphoma within ovarian tissue. Fertility and Sterility 95 803.e3803.e6. (https://doi.org/10.1016/j.fertnstert.2010.07.1068)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Byrne J, Fears TR, Gail MH, Pee D, Connelly RR, Austin DF, Holmes GF, Holmes FF, Latourette HB & & Meigs JW 1992 Early menopause in long-term survivors of cancer during adolescence. American Journal of Obstetrics and Gynecology 166 788793. (https://doi.org/10.1016/0002-9378(9291335-8)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chang EM, Lim E, Yoon S, Jeong K, Bae S, Lee DR, Yoon TK, Choi Y & & Lee WS 2015 Cisplatin induces overactivation of the dormant primordial follicle through PTEN/Akt/FOXO3a pathway which leads to loss of ovarian reserve in mice. PLoS One 10 e0144245. (https://doi.org/10.1371/journal.pone.0144245)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chen Y, Yang W, Shi X, Zhang C, Song G & & Huang D 2020 The factors and pathways regulating the activation of mammalian primordial follicles in vivo. Frontiers in Cell and Developmental Biology 8 575706. (https://doi.org/10.3389/fcell.2020.575706)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chen C, Li S, Hu C, Cao W, Fu Q, Li J, Zheng L & & Huang J 2021 Protective effects of puerarin on premature ovarian failure via regulation of WNT/β-catenin signaling pathway and oxidative stress. Reproductive Sciences 28 982990. (https://doi.org/10.1007/s43032-020-00325-0)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chhabria S, Takle V, Sharma N, Kharkar P, Pansare K, Tripathi A, Tripathi A & & Bhartiya D 2022 Extremely active nano-formulation of resveratrol (xar™) attenuates and reverses chemotherapy-induced damage in mice ovaries and testes. Journal of Ovarian Research 15 115. (https://doi.org/10.1186/s13048-022-01043-8)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chu CS & & Rubin SC 2018 Basic principles of chemotherapy. In Clinical Gynecologic Oncology 9th ed. Disaia PJ, Creasman WT, Mannel RS, McMeekin DS, & Mutch DG Eds. Amsterdam: Elsevier.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Çil N & & Mete GA 2021 The effect of adipose-derived mesenchymal stem cell treatment on mTOR and p-mTOR expression in ovarian damage due to cyclophosphomide. Reproductive Toxicology 103 7178. (https://doi.org/10.1016/j.reprotox.2021.06.003)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Codacci-Pisanelli G, Del Pup L, Del Grande M & & Peccatori FA 2017 Mechanisms of chemotherapy-induced ovarian damage in breast cancer patients. Critical Reviews in Oncology/Hematology 113 9096. (https://doi.org/10.1016/j.critrevonc.2017.03.009)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Coxworth JE & & Hawkes K 2010 Ovarian follicle loss in humans and mice: lessons from statistical model comparison. Human Reproduction 25 17961805. (https://doi.org/10.1093/humrep/deq136)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cui W, Francis PA, Loi S, Hickey M, Stern C, Na L, Partridge AH, Loibl S, Anderson RA, Hutt KJ, et al.2021 Assessment of ovarian function in phase 3 (neo)adjuvant breast cancer clinical trials: a systematic evaluation. Journal of the National Cancer Institute 113 17701778. (https://doi.org/10.1093/jnci/djab111)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cunha GR, Sinclair A, Ricke WA, Robboy SJ, Cao M & & Baskin LS 2019 Reproductive tract biology: of mice and men. Differentiation; Research in Biological Diversity 110 4963. (https://doi.org/10.1016/j.diff.2019.07.004)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dasari S & & Tchounwou PB 2014 Cisplatin in cancer therapy: molecular mechanisms of action. European Journal of Pharmacology 740 364378. (https://doi.org/10.1016/j.ejphar.2014.07.025)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Del Castillo LM, Buigues A, Rossi V, Soriano MJ, Martinez J, De Felici M, Lamsira HK, Di Rella F, Klinger FG, Pellicer A, et al.2021 The cyto-protective effects of LH on ovarian reserve and female fertility during exposure to gonadotoxic alkylating agents in an adult mouse model. Human Reproduction 36 25142528. (https://doi.org/10.1093/humrep/deab165)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Deming PB, Schafer ZT, Tashker JS, Potts MB, Deshmukh M & & Kornbluth S 2004 BCR-ABL-mediated protection from apoptosis downstream of mitochondrial cytochrome C release. Molecular and Cellular Biology 24 10289–10299. (https://doi.org/10.1128/MCB.24.23.10289-10299.2004)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dolmans MM, Marinescu C, Saussoy P, Van Langendonckt A, Amorim C & & Donnez J 2010 Reimplantation of cryopreserved ovarian tissue from patients with acute lymphoblastic leukemia is potentially unsafe. Blood 116 29082914. (https://doi.org/10.1182/blood-2010-01-265751)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Donnez J & & Dolmans MM 2015 Ovarian cortex transplantation: 60 reported live births brings the success and worldwide expansion of the technique towards routine clinical practice. Journal of Assisted Reproduction and Genetics 32 11671170. (https://doi.org/10.1007/s10815-015-0544-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dunlop CE & & Anderson RA 2014 The regulation and assessment of follicular growth. Scandinavian Journal of Clinical and Laboratory Investigation. Supplementum 244 1317. (https://doi.org/10.3109/00365513.2014.936674)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Eldani M, Luan Y, Xu PC, Bargar T & & Kim SY 2020 Continuous treatment with cisplatin induces the oocyte death of primordial follicles without activation. FASEB Journal 34 1388513899. (https://doi.org/10.1096/fj.202001461RR)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Faubion SS, Kuhle CL, Shuster LT & & Rocca WA 2015 Long-term health consequences of premature or early menopause and considerations for management. Climacteric 18 483491. (https://doi.org/10.3109/13697137.2015.1020484)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Forman EJ 2018 Ovarian tissue cryopreservation: still experimental? Fertility and Sterility 109 443444. (https://doi.org/10.1016/j.fertnstert.2017.12.031)

  • Franasiak JM & & Scott RT 2016 Demographics of cancer in the reproductive age female. In Cancer & Fertility. Sabanegh JES Ed. Springer International Publishing 1119. (https://doi.org/10.1007/978-3-319-27711-0_2)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ganesan S & & Keating AF 2016 The ovarian DNA damage repair response is induced prior to phosphoramide mustard-induced follicle depletion, and ataxia telangiectasia mutated inhibition prevents PM-induced follicle depletion. Toxicology and Applied Pharmacology 292 6574. (https://doi.org/10.1016/j.taap.2015.12.010)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Garg D, Johnstone EB, Lomo L, Fair DB, Rosen MP, Taylor R, Silver B & & Letourneau JM 2020 Looking beyond the ovary for oncofertility care in women: uterine injury as a potential target for fertility-preserving treatments. Journal of Assisted Reproduction and Genetics 37 14671476. (https://doi.org/10.1007/s10815-020-01792-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Garutti M, Lambertini M & & Puglisi F 2021 Checkpoint inhibitors, fertility, pregnancy, and sexual life: a systematic review. ESMO Open 6 100276. (https://doi.org/10.1016/j.esmoop.2021.100276)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Goldman KN, Chenette D, Arju R, Duncan FE, Keefe DL, Grifo JA & & Schneider RJ 2017 Mtorc1/2 inhibition preserves ovarian function and fertility during genotoxic chemotherapy. Proceedings of the National Academy of Sciences of the United States of America 114 31863191. (https://doi.org/10.1073/pnas.1617233114)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gonfloni S, Di Tella L, Caldarola S, Cannata SM, Klinger FG, Di Bartolomeo C, Mattei M, Candi E, De Felici M, Melino G, et al.2009 Inhibition of the c-abl-tap63 pathway protects mouse oocytes from chemotherapy-induced death. Nature Medicine 15 11791185. (https://doi.org/10.1038/nm.2033)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gook DA & & Edgar DH 2019 Cryopreservation of female reproductive potential. Best Practice and Research. Clinical Obstetrics and Gynaecology 55 2336. (https://doi.org/10.1016/j.bpobgyn.2018.08.005)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Griffiths MJ, Winship AL & & Hutt KJ 2020 Do cancer therapies damage the uterus and compromise fertility? Human Reproduction Update 26 161173. (https://doi.org/10.1093/humupd/dmz041)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hancke K, Strauch O, Kissel C, Göbel H, Schäfer W & & Denschlag D 2007 Sphingosine 1-phosphate protects ovaries from chemotherapy-induced damage in vivo. Fertility and Sterility 87 172177. (https://doi.org/10.1016/j.fertnstert.2006.06.020)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hansmann I 1974 Chromosome aberrations in metaphase II-oocytes stage sensitivity in the mouse oogenesis to amethopterin and cyclophosphamide. Mutation Research 22 175191. (https://doi.org/10.1016/0027-5107(7490098-0)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hansmann I & & Probeck HD 1979 Detection of nondisjunction in mammals. Environmental Health Perspectives 31 161165. (https://doi.org/10.1289/ehp.7931161)

  • Hayun M, Naor Y, Weil M, Albeck M, Peled A, Don J, Haran-Ghera N & & Sredni B 2006 The immunomodulator AS101 induces growth arrest and apoptosis in multiple myeloma: association with the Akt/survivin pathway. Biochemical Pharmacology 72 14231431. (https://doi.org/10.1016/j.bcp.2006.06.015)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hong L, Yan L, Xin Z, Hao J, Liu W, Wang S, Liao S, Wang H & & Yang X 2020 Protective effects of human umbilical cord mesenchymal stem cell-derived conditioned medium on ovarian damage. Journal of Molecular Cell Biology 12 372385. (https://doi.org/10.1093/jmcb/mjz105)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Huang J, Shan W, Li N, Zhou B, Guo E, Xia M, Lu H, Wu Y, Chen J, Wang B, et al.2020 Melatonin provides protection against cisplatin-induced ovarian damage and loss of fertility in mice. Reproductive Biomedicine Online 6483 3053630538.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jang H, Lee OH, Lee Y, Yoon H, Chang EM, Park M, Lee JW, Hong K, Kim JO, Kim NK, et al.2016 Melatonin prevents cisplatin-induced primordial follicle loss via suppression of PTEN/Akt/FOXO3a pathway activation in the mouse ovary. Journal of Pineal Research 60 336347. (https://doi.org/10.1111/jpi.12316)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jang H, Na Y, Hong K, Lee S, Moon S, Cho M, Park M, Lee OH, Chang EM, Lee DR, et al.2017 Synergistic effect of melatonin and ghrelin in preventing cisplatin-induced ovarian damage via regulation of FOXO3a phosphorylation and binding to the p27Kip1 promoter in primordial follicles. Journal of Pineal Research 63 e12432. (https://doi.org/10.1111/jpi.12432)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jarrell JF, Bodo L, Younglai EV, Barr RD & & O'Connell GJ 1991 The short-term reproductive toxicity of cyclophosphamide in the female rat. Reproductive Toxicology 5 481485. (https://doi.org/10.1016/0890-6238(9190019-c)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jeelani R, Khan SN, Shaeib F, Kohan-Ghadr HR, Aldhaheri SR, Najafi T, Thakur M, Morris R & & Abu-Soud HM 2017 Cyclophosphamide and acrolein induced oxidative stress leading to deterioration of metaphase II mouse oocyte quality. Free Radical Biology and Medicine 110 1118. (https://doi.org/10.1016/j.freeradbiomed.2017.05.006)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kalich-Philosoph L, Roness H, Carmely A, Fishel-Bartal M, Ligumsky H, Paglin S, Wolf I, Kanety H, Sredni B & & Meirow D 2013 Cyclophosphamide triggers follicle activation and “burnout”; AS101 prevents follicle loss and preserves fertility. Science Translational Medicine 5 185ra62. (https://doi.org/10.1126/scitranslmed.3005402)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kano M, Sosulski AE, Zhang L, Saatcioglu HD, Wang D, Nagykery N, Sabatini ME, Gao G, Donahoe PK & & Pépin D 2017 AMH/MIS as a contraceptive that protects the ovarian reserve during chemotherapy. Proceedings of the National Academy of Sciences of the United States of America 114 E1688E1697. (https://doi.org/10.1073/pnas.1620729114)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kaya H, Desdicioglu R, Sezik M, Ulukaya E, Ozkaya O, Yilmaztepe A & & Demirci M 2008 Does sphingosine-1-phosphate have a protective effect on cyclophosphamide- and irradiation-induced ovarian damage in the rat model? Fertility and Sterility 89 732735. (https://doi.org/10.1016/j.fertnstert.2007.03.065)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kerr JB, Hutt KJ, Cook M, Speed TP, Strasser A, Findlay JK & & Scott CL 2012a Cisplatin-induced primordial follicle oocyte killing and loss of fertility are not prevented by imatinib. Nature Medicine 18 11701172. (https://doi.org/10.1038/nm.2889)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kerr JB, Hutt KJ, Michalak EM, Cook M, Vandenberg CJ, Liew SH, Bouillet P, Mills A, Scott CL, Findlay JK, et al.2012b DNA damage-induced primordial follicle oocyte apoptosis and loss of fertility require TAp63-mediated induction of Puma and Noxa. Molecular Cell 48 343352. (https://doi.org/10.1016/j.molcel.2012.08.017)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Khedr NF 2015 Protective effect of mirtazapine and hesperidin on cyclophosphamide-induced oxidative damage and infertility in rat ovaries. Experimental Biology and Medicine 240 16821689. (https://doi.org/10.1177/1535370215576304)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kim J & & You S 2021 Extended adverse effects of cyclophosphamide on mouse ovarian function. BMC Pharmacology and Toxicology 22 3. (https://doi.org/10.1186/s40360-020-00468-5)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kim SY, Cordeiro MH, Serna VA, Ebbert K, Butler LM, Sinha S, Mills AA, Woodruff TK & & Kurita T 2013 Rescue of platinum-damaged oocytes from programmed cell death through inactivation of the p53 family signaling network. Cell Death and Differentiation 20 987997. (https://doi.org/10.1038/cdd.2013.31)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kim S, Lee Y, Lee S & & Kim T 2018 Ovarian tissue cryopreservation and transplantation in patients with cancer. Obstetrics and Gynecology Science 61 431442. (https://doi.org/10.5468/ogs.2018.61.4.431)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kim S, Kim SW, Han SJ, Lee S, Park HT, Song JY & & Kim T 2021 Molecular mechanism and prevention strategy of chemotherapy- and radiotherapy-induced ovarian damage. International Journal of Molecular Sciences 22. (https://doi.org/10.3390/ijms22147484)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Knapp CA, Quinn GP & & Murphy D 2011 Assessing the reproductive concerns of children and adolescents with cancer: challenges and potential solutions. Journal of Adolescent and Young Adult Oncology 1 3135. (https://doi.org/10.1089/jayao.2010.0003)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lambertini M, Moore HCF, Leonard RCF, Loibl S, Munster P, Bruzzone M, Boni L, Unger JM, Anderson RA, Mehta K, et al.2018 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. Journal of Clinical Oncology 36 19811990. (https://doi.org/10.1200/JCO.2018.78.0858)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lambertini M, Richard F, Nguyen B, Viglietti G & & Villarreal-Garza C 2019 Ovarian function and fertility preservation in breast cancer: should gonadotropin-releasing hormone agonist be administered to all premenopausal patients receiving chemotherapy? Clinical Medicine Insights. Reproductive Health 13 1179558119828393. (https://doi.org/10.1177/1179558119828393)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lambertini M, Marrocco C, Spinaci S, Demeestere I & & Anderson RA 2022 Risk of gonadotoxicity with immunotherapy and targeted agents remains an unsolved but crucial issue. European Journal of Clinical Investigation 52 e13779. (https://doi.org/10.1111/eci.13779)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lambouras M, Liew SH, Horvay K, Abud HE, Stringer JM & & Hutt KJ 2018 Examination of the ovotoxicity of 5-fluorouracil in mice. Journal of Assisted Reproduction and Genetics 35 10531060. (https://doi.org/10.1007/s10815-018-1169-6)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lande Y, Fisch B, Tsur A, Farhi J, Prag-Rosenberg R, Ben-Haroush A, Kessler-Icekson G, Zahalka MA, Ludeman SM & & Abir R 2017 Short-term exposure of human ovarian follicles to cyclophosphamide metabolites seems to promote follicular activation in vitro. Reproductive Biomedicine Online 34 104114. (https://doi.org/10.1016/j.rbmo.2016.10.005)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Levi M, Ben-Aharon I & & Shalgi R 2022 Irinotecan (CPT-11) treatment induces mild gonadotoxicity. Frontiers in Reproductive Health 4 812053. (https://doi.org/10.3389/frph.2022.812053)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li F, Turan V, Lierman S, Cuvelier C, De Sutter P & & Oktay K 2014 Sphingosine-1-phosphate prevents chemotherapy-induced human primordial follicle death. Human Reproduction 29 107113. (https://doi.org/10.1093/humrep/det391)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li S, Chen J, Fang X & & Xia X 2017 Sphingosine-1-phosphate activates the akt pathway to inhibit chemotherapy induced human granulosa cell apoptosis. Gynecological Endocrinology 33 476479. (https://doi.org/10.1080/09513590.2017.1290072)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li T, Liu C, Zhen X, Yu Y & & Qiao J 2021 Actinomycin D causes oocyte maturation failure by inhibiting chromosome separation and spindle assembly†. Biology of Reproduction 104 94105. (https://doi.org/10.1093/biolre/ioaa170)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li ZY, Dong YL, Cao XZ, Ren SS & & Zhang Z 2022 Gonadotropin-releasing hormone agonists for ovarian protection during breast cancer chemotherapy: a systematic review and meta-analysis. Menopause 29 10931100. (https://doi.org/10.1097/GME.0000000000002019)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lins TLBG, Gouveia BB, Barberino RS, Silva RLS, Monte APO, Pinto JGC, Campinho DSP, Palheta RC Jr & & Matos MHT 2020 Rutin prevents cisplatin-induced ovarian damage via antioxidant activity and regulation of PTEN and FOXO3a phosphorylation in mouse model. Reproductive Toxicology 98 209217. (https://doi.org/10.1016/j.reprotox.2020.10.001)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu Q, Zhang J, Tang Y, Ma Y, Xue Z & & Wang J 2022 The effects of human umbilical cord mesenchymal stem cell transplantation on female fertility restoration in mice. Current Gene Therapy 22 319330. (https://doi.org/10.2174/1566523221666211014165341)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Livera G, Petre-Lazar B, Guerquin MJ, Trautmann E, Coffigny H & & Habert R 2008 P63 null mutation protects mouse oocytes from radio-induced apoptosis. Reproduction 135 312. (https://doi.org/10.1530/REP-07-0054)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Logan S, Perz J, Ussher JM, Peate M & & Anazodo A 2019 Systematic review of fertility-related psychological distress in cancer patients: informing on an improved model of care. Psycho-Oncology 28 2230. (https://doi.org/10.1002/pon.4927)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Loren AW, Mangu PB, Beck LN, Brennan L, Magdalinski AJ, Partridge AH, Quinn G, Wallace WH, Oktay K & American Society of Clinical Oncology 2013 Fertility preservation for patients with cancer: American Society of Clinical Oncology clinical practice guideline update. Journal of Clinical Oncology 31 25002510. (https://doi.org/10.1200/JCO.2013.49.2678)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Luan Y, Edmonds ME, Woodruff TK & & Kim SY 2019 Inhibitors of apoptosis protect the ovarian reserve from cyclophosphamide. Journal of Endocrinology 240 243256. (https://doi.org/10.1530/JOE-18-0370)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Luo Q, Yin N, Zhang L, Yuan W, Zhao W, Luan X & & Zhang H 2017 Role of SDF-1/CXCR4 and cytokines in the development of ovary injury in chemotherapy drug induced premature ovarian failure mice. Life Sciences 179 103109. (https://doi.org/10.1016/j.lfs.2017.05.001)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ma N, Chen G, Chen J, Cui M, Yin Y, Liao Q, Tang M, Feng X, Li X, Zhang S, et al.2020 Transient impact of paclitaxel on mouse fertility and protective effect of gonadotropinreleasing hormone agonist. Oncology Reports 44 1 9171 928.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Maiani E, Di Bartolomeo C, Klinger FG, Cannata SM, Bernardini S, Chateauvieux S, Mack F, Mattei M, De Felici M, Diederich M, et al.2012 Reply to: cisplatin-induced primordial follicle oocyte killing and loss of fertility are not prevented by imatinib. Nature Medicine 18 11721174. (https://doi.org/10.1038/nm.2852)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Malott KF & & Luderer U 2021 Toxicant effects on mammalian oocyte mitochondria†. Biology of Reproduction 104 784793. (https://doi.org/10.1093/biolre/ioab002)

  • Marcello MF, Nuciforo G, Romeo R, Di Dino G, Russo I, Russo A, Palumbo G & & Schilirò G 1990 Structural and ultrastructural study of the ovary in childhood leukemia after successful treatment. Cancer 66 20992104. (https://doi.org/10.1002/1097-0142(19901115)66:10<2099::aid-cncr2820661010>3.0.co;2-3)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Marcozzi S, Rossi V, Salvatore G, Di Rella F, De Felici M & & Klinger FG 2019 Distinct effects of epirubicin, cisplatin and cyclophosphamide on ovarian somatic cells of prepuberal ovaries. Aging 11 1053210556. (https://doi.org/10.18632/aging.102476)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Marullo R, Werner E, Degtyareva N, Moore B, Altavilla G, Ramalingam SS & & Doetsch PW 2013 Cisplatin induces a mitochondrial-ROS response that contributes to cytotoxicity depending on mitochondrial redox status and bioenergetic functions. PLoS One 8 e81162. (https://doi.org/10.1371/journal.pone.0081162)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mattiello L, Pucci G, Marchetti F, Diederich M & & Gonfloni S 2021 Asciminib mitigates DNA damage stress signaling induced by cyclophosphamide in the ovary. International Journal of Molecular Sciences 22 1395. (https://doi.org/10.3390/ijms22031395)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mattison DR, Chang L, Thorgeirsson SS & & Shiromizu K 1981 The effects of cyclophosphamide, azathioprine, and 6-mercaptopurine on oocyte and follicle number in C57BL/6N mice. Research Communications in Chemical Pathology and Pharmacology 31 155161.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • McClam M & & Xiao S 2022 Preserving oocytes in oncofertility†. Biology of Reproduction 106 328337. (https://doi.org/10.1093/biolre/ioac008)

  • Méduri G, Charnaux N, Driancourt MA, Combettes L, Granet P, Vannier B, Loosfelt H & & Milgrom E 2002 Follicle-stimulating hormone receptors in oocytes? Journal of Clinical Endocrinology and Metabolism 87 22662276. (https://doi.org/10.1210/jcem.87.5.8502)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Meirow D & & Nugent D 2001 The effects of radiotherapy and chemotherapy on female reproduction. Human Reproduction Update 7 535543. (https://doi.org/10.1093/humupd/7.6.535)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Meirow D, Epstein M, Lewis H, Nugent D & & Gosden RG 2001 Administration of cyclophosphamide at different stages of follicular maturation in mice: effects on reproductive performance and fetal malformations. Human Reproduction 16 632637. (https://doi.org/10.1093/humrep/16.4.632)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Meirow D, Dor J, Kaufman B, Shrim A, Rabinovici J, Schiff E, Raanani H, Levron J & & Fridman E 2007 Cortical fibrosis and blood-vessels damage in human ovaries exposed to chemotherapy. Potential mechanisms of ovarian injury. Human Reproduction 22 16261633. (https://doi.org/10.1093/humrep/dem027)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Meng Y, Xu Z, Wu F, Chen W, Xie S, Liu J, Huang X & & Zhou Y 2014 Sphingosine-1-phosphate suppresses cyclophosphamide induced follicle apoptosis in human fetal ovarian xenografts in nude mice. Fertility and Sterility 102 871877.e3. (https://doi.org/10.1016/j.fertnstert.2014.05.040)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Meng X, Chen H, Wang G, Yu Y & & Xie K 2015 Hydrogen-rich saline attenuates chemotherapy-induced ovarian injury via regulation of oxidative stress. Experimental and Therapeutic Medicine 10 22772282. (https://doi.org/10.3892/etm.2015.2787)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Moore HCF, Unger JM, Phillips KA, Boyle F, Hitre E, Porter D, Francis PA, Goldstein LJ, Gomez HL, Vallejos CS, et al.2015 Goserelin for ovarian protection during breast-cancer adjuvant chemotherapy. New England Journal of Medicine 372 923932. (https://doi.org/10.1056/NEJMoa1413204)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Morgan S, Lopes F, Gourley C, Anderson RA & & Spears N 2013 Cisplatin and doxorubicin induce distinct mechanisms of ovarian follicle loss; imatinib provides selective protection only against cisplatin. PLoS One 8 e70117. (https://doi.org/10.1371/journal.pone.0070117)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Morita Y, Perez GI, Paris F, Miranda SR, Ehleiter D, Haimovitz-Friedman A, Fuks Z, Xie Z, Reed JC, Schuchman EH, et al.2000 Oocyte apoptosis is suppressed by disruption of the acid sphingomyelinase gene or by sphingosine-1-phosphate therapy. Nature Medicine 6 11091114. (https://doi.org/10.1038/80442)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Naren G, Wang L, Zhang X, Cheng L, Yang S, Yang J, Guo J & & Nashun B 2021 The reversible reproductive toxicity of 5-fluorouracil in mice. Reproductive Toxicology 101 18. (https://doi.org/10.1016/j.reprotox.2021.02.002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Nguyen QN, Zerafa N, Liew SH, Morgan FH, Strasser A, Scott CL, Findlay JK, Hickey M & & Hutt KJ 2018 Loss of PUMA protects the ovarian reserve during DNA-damaging chemotherapy and preserves fertility. Cell Death and Disease 9 618. (https://doi.org/10.1038/s41419-018-0633-7)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Nguyen QN, Zerafa N, Liew SH, Findlay JK, Hickey M & & Hutt KJ 2019 Cisplatin- and cyclophosphamide-induced primordial follicle depletion is caused by direct damage to oocytes. Molecular Human Reproduction 25 433444. (https://doi.org/10.1093/molehr/gaz020)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Nguyen QN, Zerafa N, Findlay JK, Hickey M & & Hutt KJ 2021 DNA repair in primordial follicle oocytes following cisplatin treatment. Journal of Assisted Reproduction and Genetics 38 14051417. (https://doi.org/10.1007/s10815-021-02184-3)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Nicosia SV, Matus-Ridley M & & Meadows AT 1985 Gonadal effects of cancer therapy in girls. Cancer 55 23642372. (https://doi.org/10.1002/1097-0142(19850515)55:10<2364::aid-cncr2820551011>3.0.co;2-e)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ogino M & & Prasanna T 2020 Cyclophosphamide. Treasure Island, FL: StatPearls Publishing.

  • Oktay K, Briggs D & & Gosden RG 1997 Ontogeny of follicle-stimulating hormone receptor gene expression in isolated human ovarian follicles. Journal of Clinical Endocrinology and Metabolism 82 37483751. (https://doi.org/10.1210/jcem.82.11.4346)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Oktem O & & Oktay K 2007a A novel ovarian xenografting model to characterize the impact of chemotherapy agents on human primordial follicle reserve. Cancer Research 67 1015910162. (https://doi.org/10.1158/0008-5472.CAN-07-2042)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Oktem O & & Oktay K 2007b Quantitative assessment of the impact of chemotherapy on ovarian follicle reserve and stromal function. Cancer 110 22222229. (https://doi.org/10.1002/cncr.23071)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ou YH, Chung PH, Sun TP & & Shieh SY 2005 P53 C-terminal phosphorylation by CHK1 and CHK2 participates in the regulation of DNA-damage-induced C-terminal acetylation. Molecular Biology of the Cell 16 16841695. (https://doi.org/10.1091/mbc.e04-08-0689)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pandir D, Kara O & & Kara M 2014 Protective effect of bilberry (Vaccinium myrtillus l.) on cisplatin induced ovarian damage in rat. Cytotechnology 66 677685. (https://doi.org/10.1007/s10616-013-9621-z)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Partridge AH 2012 Ovarian suppression for prevention of premature menopause and infertility: empty promise or effective therapy? Journal of Clinical Oncology 30 479481. (https://doi.org/10.1200/JCO.2011.37.9883)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pascuali N, Scotti L, Di Pietro M, Oubiña G, Bas D, May M, Gómez Muñoz A, Cuasnicú PS, Cohen DJ, Tesone M, et al.2018 Ceramide-1-phosphate has protective properties against cyclophosphamide-induced ovarian damage in a mice model of premature ovarian failure. Human Reproduction 33 844859. (https://doi.org/10.1093/humrep/dey045)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Perez GI, Jurisicova A, Matikainen T, Moriyama T, Kim MR, Takai Y, Pru JK, Kolesnick RN & & Tilly JL 2005 A central role for ceramide in the age-related acceleration of apoptosis in the female germline. FASEB Journal 19 860862. (https://doi.org/10.1096/fj.04-2903fje)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Petrillo SK, Desmeules P, Truong TQ & & Devine PJ 2011 Detection of DNA damage in oocytes of small ovarian follicles following phosphoramide mustard exposures of cultured rodent ovaries in vitro. Toxicology and Applied Pharmacology 253 94102. (https://doi.org/10.1016/j.taap.2011.03.012)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Plowchalk DR & & Mattison DR 1992 Reproductive toxicity of cyclophosphamide in the C57BL/6N mouse: 1. Effects on ovarian structure and function. Reproductive Toxicology 6 411421. (https://doi.org/10.1016/0890-6238(9290004-d)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Quinn GP, Vadaparampil ST, Bell-Ellison BA, Gwede CK & & Albrecht TL 2008 Patient-physician communication barriers regarding fertility preservation among newly diagnosed cancer patients. Social Science and Medicine 66 784789. (https://doi.org/10.1016/j.socscimed.2007.09.013)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rinaldi VD, Bloom JC & & Schimenti JC 2019 Signaling to Trp53 and TAp63 from CHK1/CHK2 is responsible for elimination of most oocytes defective for either chromosome synapsis or recombination. bioRxiv 768150.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rodriguez-Wallberg KA & & Oktay K 2014 Fertility preservation during cancer treatment: clinical guidelines. Cancer Management and Research 6 105117. (https://doi.org/10.2147/CMAR.S32380)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Roness H, Spector I, Leichtmann-Bardoogo Y, Savino AM, Dereh-Haim S & & Meirow D 2019 Pharmacological administration of recombinant human amh rescues ovarian reserve and preserves fertility in a mouse model of chemotherapy, without interfering with anti-tumoural effects. Journal of Assisted Reproduction and Genetics 36 17931803. (https://doi.org/10.1007/s10815-019-01507-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rosario R, Cui W & & Anderson RA 2022 Potential ovarian toxicity and infertility risk following targeted anti-cancer therapies. Reproduction and Fertility 3 R147R162. (https://doi.org/10.1530/RAF-22-0020)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rossi V, Lispi M, Longobardi S, Mattei M, Di Rella F, Salustri A, De Felici M & & Klinger FG 2017 LH prevents cisplatin-induced apoptosis in oocytes and preserves female fertility in mouse. Cell Death and Differentiation 24 7282. (https://doi.org/10.1038/cdd.2016.97)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Saleh HS, Omar E, Froemming GR & & Said RM 2015 Tocotrienol preserves ovarian function in cyclophosphamide therapy. Human and Experimental Toxicology 34 946952. (https://doi.org/10.1177/0960327114564793)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Salem W, Ho JR, Woo I, Ingles SA, Chung K, Paulson RJ & & McGinnis LK 2020 Long-term imatinib diminishes ovarian reserve and impacts embryo quality. Journal of Assisted Reproduction and Genetics 37 14591466. (https://doi.org/10.1007/s10815-020-01778-7)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Salian SR, Uppangala S, Cheredath A, D'Souza F, Kalthur G, Nayak VC, Anderson RA & & Adiga SK 2020 Early prepubertal cyclophosphamide exposure in mice results in long-term loss of ovarian reserve, and impaired embryonic development and blastocyst quality. PLoS One 15 e0235140. (https://doi.org/10.1371/journal.pone.0235140)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Salvatore G, De Felici M, Dolci S, Tudisco C, Cicconi R, Campagnolo L, Camaioni A & & Klinger FG 2021 Human adipose-derived stromal cells transplantation prolongs reproductive lifespan on mouse models of mild and severe premature ovarian insufficiency. Stem Cell Research and Therapy 12 537. (https://doi.org/10.1186/s13287-021-02590-5)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sarbassov DD, Ali SM & & Sabatini DM 2005 Growing roles for the mtor pathway. Current Opinion in Cell Biology 17 596603. (https://doi.org/10.1016/j.ceb.2005.09.009)

  • Shah MA & & Schwartz GK 2001 Cell cycle-mediated drug resistance: an emerging concept in cancer therapy. Clinical Cancer Research 7 21682181.

  • Shai D, Aviel-Ronen S, Spector I, Raanani H, Shapira M, Gat I, Roness H & & Meirow D 2021 Ovaries of patients recently treated with alkylating agent chemotherapy indicate the presence of acute follicle activation, elucidating its role among other proposed mechanisms of follicle loss. Fertility and Sterility 115 12391249. (https://doi.org/10.1016/j.fertnstert.2020.11.040)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Shin EY, Kim DS, Lee MJ, Lee AR, Shim SH, Baek SW, Han DK & & Lee DR 2021 Prevention of chemotherapy-induced premature ovarian insufficiency in mice by scaffold-based local delivery of human embryonic stem cell-derived mesenchymal progenitor cells. Stem Cell Research and Therapy 12 431. (https://doi.org/10.1186/s13287-021-02479-3)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Shiromizu K, Thorgeirsson SS & & Mattison DR 1984 Effect of cyclophosphamide on oocyte and follicle number in Sprague-Dawley rats, C57BL/6N and DBA/2N mice. Pediatric Pharmacology 4 213221.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sonigo C, Beau I, Grynberg M & & Binart N 2019 AMH prevents primordial ovarian follicle loss and fertility alteration in cyclophosphamide-treated mice. FASEB Journal 33 12781287. (https://doi.org/10.1096/fj.201801089R)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Spears N, Lopes F, Stefansdottir A, Rossi V, De Felici M, Anderson RA & & Klinger FG 2019 Ovarian damage from chemotherapy and current approaches to its protection. Human Reproduction Update 25 673693. (https://doi.org/10.1093/humupd/dmz027)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Steelman LS, Chappell WH, Abrams SL, Kempf RC, Long J, Laidler P, Mijatovic S, Maksimovic-Ivanic D, Stivala F, Mazzarino MC, et al.2011 Roles of the RAF/MEK/ERK and PI3K/PTEN/Akt/mTOR pathways in controlling growth and sensitivity to therapy-implications for cancer and aging. Aging 3 192222. (https://doi.org/10.18632/aging.100296)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Stern CJ, Toledo MG, Gook DA & & Seymour JF 2006 Fertility preservation in female oncology patients. Australian and New Zealand Journal of Obstetrics and Gynaecology 46 1523. (https://doi.org/10.1111/j.1479-828X.2006.00507.x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Stringer JM, Swindells EOK, Zerafa N, Liew SH & & Hutt KJ 2018 Multidose 5-fluorouracil is highly toxic to growing ovarian follicles in mice. Toxicological Sciences 166 97107. (https://doi.org/10.1093/toxsci/kfy189)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Suh EK, Yang A, Kettenbach A, Bamberger C, Michaelis AH, Zhu Z, Elvin JA, Bronson RT, Crum CP & & McKeon F 2006 P63 protects the female germ line during meiotic arrest. Nature 444 624628. (https://doi.org/10.1038/nature05337).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sun Y, Liu Y, Ma X & & Hu H 2021 The influence of cell cycle regulation on chemotherapy. International Journal of Molecular Sciences 22. (https://doi.org/10.3390/ijms22136923)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Szymanska KJ, Tan X & & Oktay K 2020 Unraveling the mechanisms of chemotherapy-induced damage to human primordial follicle reserve: road to developing therapeutics for fertility preservation and reversing ovarian aging. Molecular Human Reproduction 26 553566. (https://doi.org/10.1093/molehr/gaaa043)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tanaka Y, Kimura F, Zheng L, Kaku S, Takebayashi A, Kasahara K, Tsuji S & & Murakami T 2018 Protective effect of a mechanistic target of rapamycin inhibitor on an in vivo model ofcisplatin-induced ovarian gonadotoxicity. Experimental Animals 67 493500. (https://doi.org/10.1538/expanim.18-0042)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Taskin MI, Yay A, Adali E, Balcioglu E & & Inceboz U 2015 Protective effects of sildenafil citrate administration on cisplatin-induced ovarian damage in rats. Gynecological Endocrinology 31 272277. (https://doi.org/10.3109/09513590.2014.984679)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Titus S, Szymanska KJ, Musul B, Turan V, Taylan E, Garcia-Milian R, Mehta S & & Oktay K 2021 Individual-oocyte transcriptomic analysis shows that genotoxic chemotherapy depletes human primordial follicle reserve in vivo by triggering proapoptotic pathways without growth activation. Scientific Reports 11 407. (https://doi.org/10.1038/s41598-020-79643-x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Torrealday S, Kodaman P & & Pal L 2017 Premature ovarian insufficiency - an update on recent advances in understanding and management. F1000Research 6 2069. (https://doi.org/10.12688/f1000research.11948.1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tuppi M, Kehrloesser S, Coutandin DW, Rossi V, Luh LM, Strubel A, Hötte K, Hoffmeister M, Schäfer B, De Oliveira T, et al.2018 Oocyte DNA damage quality control requires consecutive interplay of CHK2 and CK1 to activate p63. Nature Structural and Molecular Biology 25 261269. (https://doi.org/10.1038/s41594-018-0035-7)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • van den Berg MH, Overbeek A, Lambalk CB, Kaspers GJL, Bresters D, van den Heuvel-Eibrink MM, Kremer LC, Loonen JJ, van der Pal HJ, Ronckers CM, et al.2018 Long-term effects of childhood cancer treatment on hormonal and ultrasound markers of ovarian reserve. Human Reproduction 33 14741488. (https://doi.org/10.1093/humrep/dey229)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Volckmar X, Vallejo M, Bertoldo MJ, Nguyen QN, Handelsman DJ, Chisholm O & & Anazodo A 2022 Oncofertility information available for recently approved novel non cytotoxic and immunotherapy oncology drugs. Clinical Pharmacology and Therapeutics 111 382390. (https://doi.org/10.1002/cpt.2254)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wang Q & & Hutt KJ 2021 Evaluation of mitochondria in mouse oocytes following cisplatin exposure. Journal of Ovarian Research 14 65. (https://doi.org/10.1186/s13048-021-00817-w)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Webber L, Davies M, Anderson R, Bartlett J, Braat D, Cartwright B, Cifkova R, de Muinck Keizer-Schrama S, Hogervorst E, Janse F, et al.2016 ESHRE Guideline: Management of women with premature ovarian insufficiency. Human Reproduction 31 926937. (https://doi.org/10.1093/humrep/dew027)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Winship AL, Stringer JM, Liew SH & & Hutt KJ 2018 The importance of DNA repair for maintaining oocyte quality in response to anti-cancer treatments, environmental toxins and maternal ageing. Human Reproduction Update 24 119134. (https://doi.org/10.1093/humupd/dmy002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Winship AL, Sarma UC, Alesi LR & & Hutt KJ 2020 Accurate follicle enumeration in adult mouse ovaries. Journal of Visualized Experiments: JoVE 164 e61782. (https://doi.org/10.3791/61782)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Winship AL, Alesi LR, Sant S, Stringer JM, Cantavenera A, Hegarty T, Requesens CL, Liew SH, Sarma U, Griffiths MJ, et al.2022 Checkpoint inhibitor immunotherapy diminishes oocyte number and quality in mice. Nature Cancer 3 113. (https://doi.org/10.1038/s43018-022-00413-x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Xu H, Bao X, Kong H, Yang J, Li Y & & Sun Z 2022 Melatonin protects against cyclophosphamide-induced premature ovarian failure in rats. Human and Experimental Toxicology 41 9603271221127430. (https://doi.org/10.1177/09603271221127430)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yang W, Ma Y, Jin J, Ren P, Zhou H, Xu S, Zhang Y, Hu Z, Rong Y, Dai Y, et al.2021 Cyclophosphamide exposure causes long-term detrimental effect of oocytes developmental competence through affecting the epigenetic modification and maternal factors' transcription during oocyte growth. Frontiers in Cell and Developmental Biology 9 682060. (https://doi.org/10.3389/fcell.2021.682060)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Youle RJ & & Strasser A 2008 The BCL-2 protein family: opposing activities that mediate cell death. Nature Reviews. Molecular Cell Biology 9 4759. (https://doi.org/10.1038/nrm2308)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yu J & & Zhang L 2008 PUMA, a potent killer with or without p53. Oncogene 27(Supplement 1) S71S83. (https://doi.org/10.1038/onc.2009.45)

  • Yuan ZM, Shioya H, Ishiko T, Sun X, Gu J, Huang YY, Lu H, Kharbanda S, Weichselbaum R & & Kufe D 1999 P73 is regulated by tyrosine kinase c-ABL in the apoptotic response to DNA damage. Nature 399 814817. (https://doi.org/10.1038/21704)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yuksel A, Bildik G, Senbabaoglu F, Akin N, Arvas M, Unal F, Kilic Y, Karanfil I, Eryılmaz B, Yilmaz P, et al.2015 The magnitude of gonadotoxicity of chemotherapy drugs on ovarian follicles and granulosa cells varies depending upon the category of the drugs and the type of granulosa cells. Human Reproduction 30 29262935. (https://doi.org/10.1093/humrep/dev256)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhang Q, Xu M, Yao X, Li T, Wang Q & & Lai D 2015 Human amniotic epithelial cells inhibit granulosa cell apoptosis induced by chemotherapy and restore the fertility. Stem Cell Research and Therapy 6 152152. (https://doi.org/10.1186/s13287-015-0148-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhang N, Sun AD, Sun SM, Yang R, Shi YY, Wang QL, Li XY, Ma JH, Yue W, Xie BT, et al.2021 Mitochondrial proteome of mouse oocytes and cisplatin-induced shifts in protein profile. Acta Pharmacologica Sinica 42 21442154. (https://doi.org/10.1038/s41401-021-00687-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhou L, Xie Y, Li S, Liang Y, Qiu Q, Lin H & & Zhang Q 2017 Rapamycin prevents cyclophosphamide-induced over-activation of primordial follicle pool through PI3K/Akt/mTOR signaling pathway in vivo. Journal of Ovarian Research 10 56. (https://doi.org/10.1186/s13048-017-0350-3)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhou C, Zhang X, Zhang Y, ShiYang X, Li Y, Shi X & & Xiong B 2019 Vitamin C protects carboplatin-exposed oocytes from meiotic failure. Molecular Human Reproduction 25 601613. (https://doi.org/10.1093/molehr/gaz046)

    • PubMed
    • Search Google Scholar
    • Export Citation