Korean J Fertil Steril Search

CLOSE


Clin Exp Reprod Med > Epub ahead of print
Kim and Jee: The role of mammalian target of rapamycin inhibitors in mitigating cyclophosphamide-induced ovarian damage in a murine model

Abstract

Objective

This study aimed to determine whether pre-treatment with everolimus or rapamycin could prevent ovarian follicle damage induced by cyclophosphamide (Cp).

Methods

A total of 120 female BDF-1 mice were randomly assigned into four groups receiving specific treatments. The control group received normal saline on days 1, 3, 5, and 13. The Cp group received saline on days 1, 3, and 5, followed by Cp administration on day 13. The everolimus+Cp group was given everolimus on days 1, 3, and 5, then Cp on day 13. Similarly, the rapamycin+Cp group received rapamycin on days 1, 3, and 5, followed by Cp on day 13. On day 20, all mice were euthanized, ovaries were harvested for histological analysis, and protein expression levels of B-cell lymphoma-extra large (BCL-xL), caspase 3, and mammalian target of rapamycin were evaluated by Western blot analysis.

Results

The number of primordial follicles was lower in the Cp group than in the control group. The everolimus group and the rapamycin group also showed reduced primordial follicle counts. No significant differences were observed in the proportions of G1 primordial or G1 secondary follicles among the groups. Compared to the control group, the Cp group and the everolimus group showed lower proportions of G1 primary follicles. However, the rapamycin group had a G1 primary follicle proportion similar to that of the control group. The Cp, everolimus, and rapamycin groups exhibited lower proportions of G1 antral follicles than the control group.

Conclusion

Pre-treatment with rapamycin preserved the proportion of G1 primary follicles; however, neither everolimus nor rapamycin preserved the primordial follicle counts.

Introduction

The total number of ovarian follicles is established at birth, and a reduction in this reserve contributes to reproductive aging. Recent research has emphasized critical intracellular signaling pathways responsible for activating primordial follicles from their dormant states [1,2]. Under normal conditions, the ovary maintains a state of equilibrium. The activation of primordial follicles is mediated by the phosphatidylinositol-3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signaling pathway. Additionally, inhibitory factors secreted by developing follicles maintain most primordial follicles in dormancy.
mTOR is a conserved serine/threonine kinase integral to cell growth and proliferation. The rapamycin-sensitive mTOR complex 1 (mTORC1) promotes cellular growth and proliferation by enhancing the synthesis of proteins, lipids, and organelles while suppressing catabolic processes such as autophagy. Studies, particularly in murine models, indicate that inhibiting mTORC1 activity in oocytes is essential for preserving primordial follicle dormancy [3,4]. The synchronized roles of AKT and mTORC1 signaling pathways in managing primordial follicle dormancy and extending the female reproductive lifespan have been well-documented [5]. Therefore, dysregulated activation of the PI3K/AKT/mTOR pathway may precipitate follicular depletion and primary ovarian insufficiency (POI).
Alkylating chemotherapeutic agents, notably cyclophosphamide (Cp), are gonadotoxic and commonly associated with POI [6-9]. Cp treatment disrupts ovarian equilibrium by activating follicles through upregulation of the PI3K/AKT/mTOR pathway, potentially leading to apoptosis of developing follicles and a subsequent reduction in inhibitory factors [10]. Consequently, there is increased recruitment of primordial follicles, which develop and ultimately undergo apoptosis, causing ovarian functional loss and POI [11,12].
Theoretically, inhibition of the PI3K/AKT/mTOR signaling pathway could promote follicle dormancy, thereby reducing apoptosis, enhancing autophagy, and increasing follicle survival in patients receiving Cp treatment. Rapamycin, a macrolide derived from the bacterium Streptomyces hygroscopicus, is recognized for inducing autophagy in mammalian cells as well as in model organisms such as Saccharomyces cerevisiae and Drosophila melanogaster [13,14]. It is a general inhibitor of mTORC1 and a cell-type-specific inhibitor of mTORC2 [15]. Rapamycin is currently employed to prevent transplant rejection and has been noted to enhance glycogen synthase phosphorylation in skeletal muscles, suggesting potential therapeutic applications in glycogen storage diseases involving muscle glycogen accumulation [16].
Everolimus, another macrolide antibiotic derived from rapamycin with improved pharmacokinetic characteristics (e.g., shorter half-life and faster absorption), exhibits immunosuppressive properties and binds strongly to the intracellular receptor FK506-binding protein 12 (FKBP12). This complex acts as an mTOR inhibitor and is utilized in managing renal cell carcinoma. Furthermore, everolimus has been evaluated as targeted therapy in various malignancies, including pancreatic, breast, and ovarian cancers [17-19].
Rapamycin and everolimus inhibit mTOR signaling by binding FKBP12, forming a complex that predominantly suppresses mTORC1 activity, thereby inhibiting cellular growth and proliferation. These mTOR inhibitors potentially inhibit tumor angiogenesis and disrupt the G1/S cell cycle transition. Recently, their benefits have been explored in several age-related conditions, including neurodegenerative diseases [20]. While both drugs selectively inhibit mTORC1, prolonged exposure to rapamycin can also disrupt mTORC2 assembly, indirectly affecting Akt signaling. Such differences might influence their effectiveness in protecting ovarian follicles.
Limited animal studies have investigated the protective effects of these mTOR inhibitors against ovarian damage induced by alkylating chemotherapeutic agents. To our knowledge, only a few studies have specifically examined the efficacy of everolimus or rapamycin among mTOR inhibitors [21,22]. These investigations indicated that everolimus or rapamycin might preserve ovarian follicles in their primordial state, maintaining normal serum anti-Müllerian hormone (AMH) levels and fertility. Although these findings suggest potential protective effects of everolimus or rapamycin against chemotherapy-induced toxicity, further research is required to evaluate their full protective capacity and compare them against other agents.
The objective of this study was to determine whether pre-treatment with everolimus or rapamycin mitigates ovarian follicle damage in mice treated with Cp. This study represents the first direct comparison of the protective effects of these two mTOR inhibitor agents.

Methods

1. Study animals

This study was approved by the Institutional Animal Care and Use Committee (IACUC) of Seoul National University Bundang Hospital (IACUC number BA-2108-325-081-01). All animal handling and treatment procedures adhered strictly to the institutional protocols established by the IACUC.
Six-week-old female bromodomain factor 1 (BDF-1) mice (Orient Bio.) were maintained in an environment with a controlled temperature of 22 ℃ and a 12-hour light/dark cycle, with free access to food and water [23,24]. Observations regarding changes in weight, appearance, and behavior were conducted two to three times daily. Euthanasia was performed if a mouse’s body weight dropped by more than 20% or if the animal demonstrated no response or movement when stimulated. Euthanasia was consistently conducted via cervical dislocation.

2. Experimental design

After a 1-week acclimatization period, 120 female BDF-1 mice were randomly assigned to one of four treatment groups (30 mice per group): (1) The Cp group received 75 mg/kg of Cp (Cp monohydrate, Cat. no. 29875, Sigma-Aldrich); (2) Everolimus (LC Laboratories) was administered at 0.75 mg/kg; (3) Rapamycin (LC Laboratories) was given at 4.0 mg/kg.
The experimental design is depicted in Table 1: (1) In the control group, normal saline (0.1 mL) was injected on days 1, 3, 5, and 13; (2) In the Cp group, normal saline (0.1 mL) was injected on days 1, 3, and 5, followed by Cp on day 13; (3) In the everolimus+Cp group, everolimus was injected on days 1, 3, and 5, with Cp on day 13; (4) In the rapamycin+Cp group, rapamycin was administered on days 1, 3, and 5, followed by Cp on day 13.
On day 20, all mice were euthanized, and their ovaries were harvested. A portion of each ovary was processed for histological examination, while the remainder was used to assess the protein expression levels of B-cell lymphoma-extra large (BCL-xL; an anti-apoptotic marker), cleaved caspase-3 (an apoptotic marker), and mTOR via Western blot analysis, using α-tubulin as an internal control.

3. Histological analysis and follicle counting

Histological assessment was conducted according to methodologies reported in a prior study [25]. Ovarian tissue was fixed with 4% buffered paraformaldehyde for 1 day and embedded in paraffin. Embedded samples were serially sectioned at 4-µm thickness. Slides were stained with hematoxylin and eosin (Merck) and analyzed for follicle counting. Every section was examined under a light microscope at least twice by a single experienced technician (B.Y. Choi), who was blinded to the treatment groups to minimize observer bias. The average follicle count across sections was recorded. Only follicles with a clearly visible nucleus in the oocyte were counted.
Each follicle type was classified into the following categories [26]: Primordial: a single layer of flattened pre-granulosa cells; Primary: a single layer of granulosa cells, 1 or more being cuboidal cells; Secondary: two or more layers of cuboidal granulosa cells, with the antrum absent; Antral: multiple layers of cuboidal granulosa cells, with the antrum present.
Follicle quality was assessed based on integrity according to previously described criteria [27]: G1 (good quality) follicle: an intact spherical follicle and oocyte; G2 (fair quality) follicle: granulosa cells pulled away from the edge of follicles, but with an intact oocyte; G3 (poor quality) follicle: disruption and/or loss of granulosa-theca cells, with pyknotic nuclei and/or a missing oocyte.
Representative histological images of ovarian follicles are shown in Figure 1.

4. Western blotting

Western blot analysis involved collecting ovarian samples from five mice per group, with each sample run in technical duplicates across three independent experiments, yielding six measurements per marker per sample. Ovaries were rinsed with phosphate-buffered saline and lysed using cell lysis buffer (20 mM Tris-HCl at pH 8.0, 137 mM NaCl, 1% Nonidet P-40, and 10% glycerol), supplemented with protease inhibitors (0.5 mM phenylmethylsulfonyl fluoride, 0.025 mM N-CBZ-L-phenylalanine chloromethyl ketone, 0.025 mM N-p-tosyl-lysine chloromethyl ketone, and 0.025 mM L-1-tosylamide-2-phenyl-ethylchloromethyl ketone) for 20 minutes on ice. After centrifugation at 10,000 ×g for 15 minutes at 4 ℃, supernatants were collected, and protein concentrations were measured using a bicinchoninic acid protein assay kit (Thermo Scientific Pierce). Samples (20 µg/µL) were resolved and transferred onto polyvinylidene difluoride membranes (Millipore). Membranes were blocked and incubated overnight at 4 ℃ with primary antibodies: BCL-xL (1:100, sc-271121; Santa Cruz Biotechnology), cleaved caspase-3 (1:500, 5a1e; Cell Signaling Technology), and mTOR (1:500, orb99435; Biorbyt Ltd.). BCL-xL serves as an anti-apoptotic marker, whereas caspase-3 indicates late apoptosis. After washing three times for 15 minutes with tris-buffered saline with Tween (TBST), membranes were incubated with horseradish peroxidase (HRP)-conjugated secondary antibodies for one hour at room temperature. Following additional TBST washes, membranes were developed using ECL Prime Western Blotting detection reagent (GE Healthcare).
Representative bands from the Western blot analysis are shown in Figure 2.

5. Statistical analysis

Statistical analyses were performed using R software version 2.14.2 (R Foundation for Statistical Computing). Ovarian follicle counts were analyzed by one-way analysis of variance (ANOVA), followed by Tukey’s multiple-comparison test. Normality of data distribution was verified before performing ANOVA. Protein expression levels were compared using the Kruskal-Wallis test followed by the Mann-Whitney U test. A p-value of less than 0.05 was considered statistically significant.

Results

1. Histological analysis of follicles

Detailed counts of ovarian follicles across the four groups are presented in Table 2. The median number of primordial follicles was significantly lower in the Cp group compared to the control group (8 vs. 22, p=0.001). Similarly, the everolimus+Cp group (11.5 vs. 22, p=0.001) and the rapamycin+Cp group (8.5 vs. 22, p=0.003) also showed significantly lower numbers of primordial follicles than the control group. No significant differences in primordial follicle counts were observed among the Cp group, everolimus+Cp group, and rapamycin+Cp group. When evaluating primary, secondary, and antral follicle counts across the four groups, no significant differences were noted.
Figure 3 illustrates the proportions of G1 follicles within the four groups. There were no significant differences in the proportions of G1 primordial or G1 secondary follicles among the four groups.
The Cp group and the everolimus+Cp group had significantly lower proportions of G1 primary follicles than the control group. Conversely, the proportion of G1 primary follicles in the rapamycin+Cp group was preserved at levels comparable to the control group. The proportions of G1 antral follicles were significantly lower in the Cp group, everolimus+Cp group, and rapamycin+Cp group than in the control group.
To summarize the findings regarding ovarian follicle subtype counts and proportions of G1 follicles, pre-treatment with either everolimus or rapamycin did not preserve the number of primordial follicles. However, pre-treatment with rapamycin preserved the number of G1 primary follicles.

2. Western blot analysis of apoptosis and mTOR expression

The expression levels of BCL-xL, cleaved caspase-3, and mTOR in the four groups are displayed in Figure 4. No significant differences in the expression levels of these proteins (BCL-xL, cleaved caspase-3, and mTOR) were identified among the four groups.

Discussion

1. Fertoprotective effects of mTOR inhibitors

Pre-treatment with rapamycin prior to Cp administration was effective in preserving the proportion of G1 primary follicles. However, based on the findings of the current study, it is difficult to conclude that rapamycin has a sufficient fertoprotective effect in attenuating Cp-induced follicular damage. Specifically, our data suggest that premedication with either everolimus or rapamycin before Cp administration did not effectively preserve either the number of primordial follicles or the proportion of G1 primordial follicles.

2. Mechanism of chemotherapy-induced ovarian damage

Recent theories, particularly the ‘burn-out theory,’ provide explanations for the ovotoxic effects of chemotherapy. Agents like Cp may initiate the growth of dormant follicles, accelerating the depletion of the follicle reserve. Although this ‘burn-out theory’ has garnered significant support, the mechanisms underlying ovarian follicle depletion and their associations with infertility and early menopause remain incompletely understood. In our study, Cp treatment reduced primordial follicle numbers without impacting counts of primary, secondary, or antral follicles, consistent with our previous research [28]. This finding suggests that Cp specifically targets primordial follicles through mechanisms inducing apoptotic cell death rather than by activating or destroying growing follicles.
However, follicle activation is not the sole factor in chemotherapy-induced ovarian damage. Chemotherapy may also trigger apoptosis or cause oxidative stress. Our understanding of the mechanisms responsible for chemotherapy-induced ovarian damage remains incomplete. Further research is necessary to understand why ovarian function impairment is more severe in some women than others.

3. Dosage regimen of Cp

The specific dosage of Cp employed to induce ovarian dysfunction in mice varies depending on study objectives. Common dosages to induce such effects typically range from 75 to 150 mg/kg. A 75 mg/kg dosage is considered standard, whereas 150 mg/kg is frequently categorized as a sterilizing dose [21]. Our previous study demonstrated that administering Cp at 50 mg/kg did not reduce primordial follicle counts, whereas 75 mg/kg significantly decreased these counts [28]. Therefore, we selected 75 mg/kg as the lowest effective dose for the current experiment.

4. Dosage regimen of everolimus

Everolimus, marketed as Certican (Novartis Pharma AG), is primarily indicated for preventing organ rejection after transplantation. Tablet formulations include dosages of 0.25, 0.5, 0.75, and 1.0 mg. Specific mouse dosages vary according to study objectives.
In a previous study [21], mice receiving Cp at 75 mg/kg weekly for 3 weeks were concurrently treated with everolimus orally at 2.5 mg/kg daily. This combined treatment doubled primordial follicle counts compared to mice treated solely with Cp. Everolimus effectively preserved ovarian reserves, serum AMH levels, and fertility outcomes. In ovariectomized mice, intraperitoneal injections of everolimus at 1 mg/kg/day for 4 weeks prevented bone loss [29]. In a breast cancer study, a daily oral dose of 0.75 mg/kg was used [30].
In this study, we administered everolimus at 0.75 mg/kg on days 1, 3, and 5, with Cp injected on day 13, aiming to achieve protective effects using a minimal dosage. However, the dose and duration may have been insufficient. Future studies should explore daily dosing of everolimus at 1 to 2.5 mg/kg over extended periods (e.g., 2 to 4 weeks) or co-administration with Cp to improve follicular protection.

5. Dosage regimen of rapamycin

In a previous study [31], rapamycin at 5 mg/kg administered intraperitoneally daily for 19 days effectively prevented follicular activation and preserved ovarian reserves in mice with phosphatase and tensin homolog deleted on chromosome ten (PTEN) mutations. In another study [22], mice received daily intraperitoneal injections of rapamycin at various doses (5, 10, or 20 mg/kg) for either 15 or 30 days. A dose of 5 mg/kg significantly affected folliculogenesis. Rapamycin dosages used in lifespan extension studies for mice ranged from 2 to 4 mg/kg [32,33].
We selected a rapamycin dosage of 4 mg/kg, injected on days 1, 3, and 5, followed by Cp administration on day 13. It should be noted that the shorter duration and lower dosage might have been inadequate to achieve the desired protective effects. Future studies should consider daily dosing of rapamycin at 4 to 5 mg/kg, potentially co-administered with Cp, to assess whether prolonged exposure enhances protection. Dose-response studies and pharmacokinetic analyses could further elucidate optimal regimens.

6. Western blot analysis

Ovarian expression levels of proteins, including BCL-xL (an anti-apoptotic marker), caspase-3 (an apoptotic marker), and mTOR, were comparable across all four groups. The similar BCL-xL levels between control and Cp groups align with our previous findings [28], suggesting that BCL-xL may not play a prominent role in Cp-induced apoptosis.
Caspase-3, a primary downstream effector enzyme during late apoptosis, was previously reported to increase following Cp administration [28,34]; however, our study did not detect increased caspase-3 expression in the Cp group. This discrepancy could result from differences in experimental conditions, such as dosing, treatment duration, and analytical methods. Additionally, apoptosis is tightly regulated, and caspase-3 activation occurs specifically at the terminal phase. Our study might have captured a time point where caspase-3 activation was either not yet apparent or had already subsided. Further investigations, including time-course studies or additional apoptosis markers, could clarify the dynamics of caspase-3 activation.
Despite expectations that pre-treatment with mTOR inhibitors such as everolimus or rapamycin would increase ovarian mTOR levels, no changes in mTOR protein expression were observed after treatment, consistent with previous research [35]. In our study, Cp-treated mice exhibited increased phosphorylation of mTOR without changes in total mTOR levels.

7. Limitations

Several limitations should be acknowledged. We utilized a single fixed dose of everolimus and rapamycin over a limited duration. Future studies might benefit from co-administering mTOR inhibitors simultaneously with anticancer drugs, rather than solely as pre-treatment. Evaluating different dosages and durations of everolimus and rapamycin could offer deeper insights. Furthermore, the limited number of markers evaluated constrained the assessment of underlying drug mechanisms. Evaluating phosphorylation of proteins within the PI3K/AKT/mTOR pathway would enable a deeper understanding of our findings.

Conflict of interest

Byung Chul Jee is an editor-in-chief and Seul Ki Kim is an editorial board member of the journal, but they were not involved in the peer reviewer selection, evaluation, or decision process of this article. No other potential conflicts of interest pertinent to this article have been disclosed.

Author contributions

Conceptualization: SKK, BCJ. Methodology: SKK, BCJ. Formal analysis: SKK, BCJ. Data curation: SKK, BCJ. Investigation: SKK. Supervision: SKK, BCJ. Writing-original draft: SKK. Writing-review & editing: SKK, BCJ. Approval of final manuscript: SKK, BCJ.

Figure 1.
Representative histologic images of mouse ovarian follicles in each treatment group (Mayer’s H&E, ×100). (A) Control, 0.1 mL of normal saline; (B) cyclophosphamide (Cp) 75 mg/kg; (C) everolimus 0.75 mg/kg+Cp; and (D) rapamycin 4.0 mg/kg+Cp.
cerm-2025-07892f1.jpg
Figure 2.
The expression levels of three proteins within the whole ovaries as determined by Western blotting. Control, 0.1 mL of normal saline; cyclophosphamide (Cp) 75 mg/kg; everolimus (E) 0.75 mg/kg; rapamycin (R) 4.0 mg/kg. BCL-xL, B-cell lymphoma-extra large; mTOR, mammalian target of rapamycin.
cerm-2025-07892f2.jpg
Figure 3.
The proportion of good quality (G1) follicles in the four groups. (A) G1 primordial follicle (%). (B) G1 primary follicle (%). (C) G1 secondary follicle (%). (D) G1 antral follicle (%). Cp, cyclophosphamide. a),b)Different superscripts indicate a statistically significant difference.
cerm-2025-07892f3.jpg
Figure 4.
Western blot analysis to assess apoptosis and mammalian target of rapamycin (mTOR) expression. (A) B-cell lymphoma-extra large (BCL-xL), (B) caspase-3, and (C) mTOR. Cp, cyclophosphamide.
cerm-2025-07892f4.jpg
Table 1.
Experimental design
Time schedule Control Cp Everolimus+Cp Rapamycin+Cp
D1 Normal saline Normal saline Everolimus Rapamycin
D3 Normal saline Normal saline Everolimus Rapamycin
D5 Normal saline Normal saline Everolimus Rapamycin
D13 Normal saline Cp Cp Cp
D20 Ovariectomy Ovariectomy Ovariectomy Ovariectomy

Cp, cyclophosphamide.

Table 2.
Ovarian follicle counts by subtype
Group Control Cp Everolimus+Cp Rapamycin+Cp p-value
Primordial 22 (13–42)a) 8 (4.5–15.5)b) 11.5 (3.7–14.7)b) 8.5 (4.7–21.0)b) 0.002
Primary 19 (16–28) 23 (14–27) 22 (18–26.2) 17 (15–24.7) 0.607
Secondary 40 (31–45) 35.5 (27.7–44.2) 36 (30.7–42.0) 38.5 (32.2–42.0) 0.649
Antral 13 (9–16) 14 (12.7–17.5) 14.5 (12.7–19) 12.5 (8.5–15.5) 0.119

Values are presented as median (interquartile range).

Cp, cyclophosphamide.

a),b)Kruskal-Wallis test (different superscripts mean a statistical significance within the same row).

References

1. Adhikari D, Liu K. Molecular mechanisms underlying the activation of mammalian primordial follicles. Endocr Rev 2009;30:438-64.
crossref pmid
2. Hsueh AJ, Kawamura K, Cheng Y, Fauser BC. Intraovarian control of early folliculogenesis. Endocr Rev 2015;36:1-24.
crossref pmid
3. Adhikari D, Flohr G, Gorre N, Shen Y, Yang H, Lundin E, et al. Disruption of Tsc2 in oocytes leads to overactivation of the entire pool of primordial follicles. Mol Hum Reprod 2009;15:765-70.
crossref pmid
4. Adhikari D, Zheng W, Shen Y, Gorre N, Hamalainen T, Cooney AJ, et al. Tsc/mTORC1 signaling in oocytes governs the quiescence and activation of primordial follicles. Hum Mol Genet 2010;19:397-410.
crossref pmid
5. Adhikari D, Liu K. mTOR signaling in the control of activation of primordial follicles. Cell Cycle 2010;9:1673-4.
crossref pmid
6. Oktem O, Oktay K. Quantitative assessment of the impact of chemotherapy on ovarian follicle reserve and stromal function. Cancer 2007;110:2222-9.
crossref pmid
7. Chun EK, Jee BC, Kim JY, Kim SH, Moon SY. Effect of imatinib coadministration on in vitro oocyte acquisition and subsequent embryo development in cyclophosphamide-treated mice. Reprod Sci 2014;21:906-14.
crossref pmid pmc pdf
8. Kim SY, Kim SK, Lee JR, Woodruff TK. Toward precision medicine for preserving fertility in cancer patients: existing and emerging fertility preservation options for women. J Gynecol Oncol 2016;27:e22.
crossref pmid pdf
9. Kim J, Kim SK, Hwang KJ, Kim SH. Fertility preservation during cancer treatment: the Korean Society for Fertility Preservation clinical guidelines. Clin Exp Reprod Med 2017;44:171-4.
crossref pmid pmc pdf
10. Kalich-Philosoph L, Roness H, Carmely A, Fishel-Bartal M, Ligumsky H, Paglin S, et al. Cyclophosphamide triggers follicle activation and “burnout”; AS101 prevents follicle loss and preserves fertility. Sci Transl Med 2013;5:185ra62.
crossref pmid
11. De Vos M, Devroey P, Fauser BC. Primary ovarian insufficiency. Lancet 2010;376:911-21.
crossref pmid
12. Meirow D, Nugent D. The effects of radiotherapy and chemotherapy on female reproduction. Hum Reprod Update 2001;7:535-43.
crossref pmid
13. Kamada Y, Funakoshi T, Shintani T, Nagano K, Ohsumi M, Ohsumi Y. Tor-mediated induction of autophagy via an Apg1 protein kinase complex. J Cell Biol 2000;150:1507-13.
crossref pmid pmc pdf
14. Rusten TE, Lindmo K, Juhasz G, Sass M, Seglen PO, Brech A, et al. Programmed autophagy in the Drosophila fat body is induced by ecdysone through regulation of the PI3K pathway. Dev Cell 2004;7:179-92.
crossref pmid
15. Sarbassov DD, Ali SM, Sengupta S, Sheen JH, Hsu PP, Bagley AF, et al. Prolonged rapamycin treatment inhibits mTORC2 assembly and Akt/PKB. Mol Cell 2006;22:159-68.
crossref pmid
16. Yi H, Brooks ED, Thurberg BL, Fyfe JC, Kishnani PS, Sun B. Correction of glycogen storage disease type III with rapamycin in a canine model. J Mol Med (Berl) 2014;92:641-50.
crossref pmid pdf
17. Guo H, Zhong Y, Jackson AL, Clark LH, Kilgore J, Zhang L, et al. Everolimus exhibits anti-tumorigenic activity in obesity-induced ovarian cancer. Oncotarget 2016;7:20338-56.
crossref pmid pmc
18. Hasskarl J. Everolimus. Recent Results Cancer Res 2018;211:101-23.
crossref pmid
19. Jeong JH, Kim JE, Ahn JH, Jung KH, Koh SJ, Cheon J, et al. Leuprorelin combined with letrozole with/without everolimus in ovarian-suppressed premenopausal women with hormone receptor-positive, HER2-negative metastatic breast cancer: the LEO study. Eur J Cancer 2021;144:341-50.
crossref pmid
20. Van Skike CE, Lin AL, Roberts Burbank R, Halloran JJ, Hernandez SF, Cuvillier J, et al. mTOR drives cerebrovascular, synaptic, and cognitive dysfunction in normative aging. Aging Cell 2020;19:e13057.
crossref pmid
21. Goldman KN, Chenette D, Arju R, Duncan FE, Keefe DL, Grifo JA, et al. mTORC1/2 inhibition preserves ovarian function and fertility during genotoxic chemotherapy. Proc Natl Acad Sci U S A 2017;114:3186-91.
crossref pmid pmc
22. Chen X, Tang Z, Guan H, Xia H, Gu C, Xu Y, et al. Rapamycin maintains the primordial follicle pool and protects ovarian reserve against cyclophosphamide-induced damage. J Reprod Dev 2022;68:287-94.
crossref pmid pmc
23. Kim SK, Jee BC, Kim SH. Effects of supplementation of human endometriotic fluids on in vitro mouse preantral follicle culture. Reprod Sci 2018;25:683-9.
crossref pdf
24. Hong YH, Kim SJ, Kim SK, Lee SC, Jun JH, Jee BC, et al. Impact of imatinib or dasatinib coadministration on in vitro preantral follicle development and oocyte acquisition in cyclophosphamide-treated mice. Clin Exp Reprod Med 2020;47:269-76.
crossref pmid pmc pdf
25. Lee J, Kim SK, Youm HW, Kim HJ, Lee JR, Suh CS, et al. Effects of three different types of antifreeze proteins on mouse ovarian tissue cryopreservation and transplantation. PLoS One 2015;10:e0126252.
crossref pmid pmc
26. Lundy T, Smith P, O'Connell A, Hudson NL, McNatty KP. Populations of granulosa cells in small follicles of the sheep ovary. J Reprod Fertil 1999;115:251-62.
crossref pmid
27. Youm HW, Lee JR, Lee J, Jee BC, Suh CS, Kim SH. Optimal vitrification protocol for mouse ovarian tissue cryopreservation: effect of cryoprotective agents and in vitro culture on vitrified-warmed ovarian tissue survival. Hum Reprod 2014;29:720-30.
crossref pmid
28. Kim SJ, Kim TE, Jee BC. Impact of imatinib administration on the mouse ovarian follicle count and levels of intra-ovarian proteins related to follicular quality. Clin Exp Reprod Med 2022;49:93-100.
crossref pmid pmc pdf
29. Browne AJ, Kubasch ML, Gobel A, Hadji P, Chen D, Rauner M, et al. Concurrent antitumor and bone-protective effects of everolimus in osteotropic breast cancer. Breast Cancer Res 2017;19:92.
crossref pmid pmc pdf
30. Chen G, Ding XF, Pressley K, Bouamar H, Wang B, Zheng G, et al. Everolimus inhibits the progression of ductal carcinoma in situ to invasive breast cancer via downregulation of MMP9 expression. Clin Cancer Res 2020;26:1486-96.
crossref pmid pdf
31. Adhikari D, Risal S, Liu K, Shen Y. Pharmacological inhibition of mTORC1 prevents over-activation of the primordial follicle pool in response to elevated PI3K signaling. PLoS One 2013;8:e53810.
crossref pmid pmc
32. Chen C, Liu Y, Liu Y, Zheng P. mTOR regulation and therapeutic rejuvenation of aging hematopoietic stem cells. Sci Signal 2009;2:ra75.
crossref pmid pmc
33. Strong R, Miller RA, Bogue M, Fernandez E, Javors MA, Libert S, et al. Rapamycin-mediated mouse lifespan extension: late-life dosage regimes with sex-specific effects. Aging Cell 2020;19:e13269.
crossref pmid pmc pdf
34. Luan Y, Edmonds ME, Woodruff TK, Kim SY. Inhibitors of apoptosis protect the ovarian reserve from cyclophosphamide. J Endocrinol 2019;240:243-56.
crossref pmid pmc
35. Chen XY, Xia HX, Guan HY, Li B, Zhang W. Follicle loss and apoptosis in cyclophosphamide-treated mice: what’s the matter? Int J Mol Sci 2016;17:836.
crossref pmid pmc


ABOUT
ARTICLE CATEGORY

Browse all articles >

BROWSE ARTICLES
AUTHOR INFORMATION
Editorial Office
Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital
82 Gumi-ro 173, Bundang-gu, Seongnam 13620, Korea
Tel: +82-31-787-7254    CP: +82-10-9072-3154    E-mail: blasto@snubh.org                

Copyright © 2025 by Korean Society for Reproductive Medicine.

Developed in M2PI

Close layer
prev next