Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Dec 1.
Published in final edited form as: J Obstet Gynaecol. 2025 May 9;45(1):2502083. doi: 10.1080/01443615.2025.2502083

Simvastatin-loaded liposomal nanoparticles as treatment for adenomyosis in a patient-derived xenograft mouse model: a pilot study

Rachel Michel a,*, Kathleen L Vincent b,*, Gregory W Kirschen c, Massoud Motamedi d, Jamal Saada e, Jinping Yang d, Bulent Ozpolat f,g, Gokhan S Kilic b, Mostafa A Borahay a
PMCID: PMC12168516  NIHMSID: NIHMS2079965  PMID: 40340640

Abstract

Background:

Adenomyosis is a common gynaecological condition where ectopic endometrial glands and stroma grow within the myometrium. This condition has a high clinical burden impacting those afflicted with debilitating symptoms including heavy painful periods. Simvastatin is an oral hydroxymethylglutaryl-coenzyme A (HMG-CoA) reductase inhibitor, typically used to treat hyperlipidaemia. Simvastatin has recently shown promise for treating gynaecological conditions such as endometriosis and uterine fibroids with nanoliposomal formulations demonstrating improved efficacy. In this pilot study, we tested simvastatin-loaded liposomal nanoparticles on xenografted adenomyosis tissues in a patient-derived mouse model.

Methods:

We surgically inserted oestrogen/progesterone pellets into mice, followed by adenomyosis tissue xenografts 15 days later. Mice were then randomised into three groups: control, simvastatin, and simvastatin-loaded liposomal nanoparticles (simvastatin-NP). We quantified the changes in adenomyosis xenograft size weekly using a calliper as well as ultrasound imaging 28 days after treatment, prior to sacrifice. We also measured the proliferation of biomarker Ki67 in the xenografted tissues using immunohistochemistry after animal sacrifice.

Results:

Treatment with simvastatin-NP significantly reduced volume and weight of adenomyosis xenografts while attenuating Ki67 expression when compared to the control and simvastatin groups.

Conclusions:

This pilot study demonstrates promising improved efficacy of simvastatin delivered via liposomal nanoparticles. However, larger studies are needed to fully explore the potential of simvastatin-NP in adenomyosis.

Keywords: Adenomyosis, simvastatin, nanoparticles, animal

PLAIN LANGUAGE SUMMARY

Adenomyosis is a condition where the inner lining of the uterus exists within the muscle wall of the uterus, resulting in painful periods, heavy bleeding, and bloating. Despite adenomyosis’ debilitating symptoms, treatment options remain limited. Previous research suggests simvastatin – a medication used to treat patients with high cholesterol – is a promising treatment for gynaecological conditions. Therefore, in this study, we implanted adenomyosis tissue from humans into mice to test the efficacy of two different forms of simvastatin – regular simvastatin and simvastatin in nanoparticle form – in reducing adenomyosis lesion size. We found that simvastatin in nanoparticle form significantly reduced the adenomyosis lesion volume and weight in mice. We then examined the tissue for a marker of cellular proliferation and found that it was significantly reduced in adenomyosis tissue treated with simvastatin in nanoparticle form. Therefore, our results suggest that simvastatin in its nanoparticle form should be further investigated as a potential treatment option for adenomyosis.

Introduction

Adenomyosis is a gynaecological condition where ectopic endometrial glands and stroma grow within the underlying myometrium. Typical symptoms of adenomyosis include abnormal uterine bleeding (AUB), painful periods, and pelvic pain (Gunther and Walker 2024). Imaging studies are required for the diagnosis and characterisation of adenomyosis with ultrasound considered the gold standard (Moawad, Fruscalzo, et al. 2023). The diagnosis of adenomyosis has historically been limited to post hysterectomy, ex vivo uterine examination, thus the true prevalence and incidence of the condition are unknown (Upson and Missmer 2020). Treatment options for adenomyosis remain limited and often encompass modalities developed for contraception and other benign gynaecological conditions such as uterine fibroids or dysmenorrhoea. These modalities include non-steroidal anti-inflammatory drugs (NSAIDs); progestins (oral, injections, and levonorgestrel intrauterine system); combined oestrogen/progestins oral contraceptives; and hysterectomy which is definitive management but precludes future childbearing (Etrusco et al. 2023, Kho et al. 2021, Krentel and De Wilde 2022, Moawad, Youssef, et al. 2023, Vannuccini and Petraglia 2019). However, these treatments have limited efficacy and are not appropriate for those attempting pregnancy. Thus, there is an urgent need for non-surgical, non-hormonal treatment options.

Simvastatin is a hydroxymethylglutaryl-coenzyme A (HMG-CoA) reductase inhibitor typically used to treat hyperlipidaemia (Talreja et al. 2024) with demonstrated efficacy in preventing cardiovascular disease (Chou et al. 2022). More recently, scientists have begun to explore its potential in the treatment and prevention of gynaecological conditions, such as endometriosis and uterine fibroids (Afrin et al. 2020, 2021, Borahay et al. 2014, Bruner-Tran et al. 2009, El Sabeh et al. 2021). Furthermore, several recent studies have demonstrated improved efficacy of simvastatin-nanoparticles formulations as compared to direct delivery of simvastatin (Borahay et al. 2021, El Sabeh et al. 2022, Enazy et al. 2023, Lulseged et al. 2024).

Adenomyosis and endometriosis share several features and often coexist (Vannuccini and Petraglia 2019). Therefore, it’s plausible to infer that because simvastatin has demonstrated efficacy in treatment and prevention of endometriosis, it may produce similar results in adenomyotic tissue (Bruner-Tran et al. 2009, Duleba et al. 2014, Nasu et al. 2009). Thus, our pilot study hypothesised that simvastatin delivered via liposomal nanoparticles – as a method of drug delivery aimed at improving treatment efficacy – would lead to the reduction of adenomyotic lesion size along with lowering of Ki67 as a biomarker of proliferation.

Methods

Animal model

Following a protocol approved by the Institutional Animal Care and Use Committee at the University of Texas Medical Branch at Galveston, we purchased 6-week-old female immunodeficient NOG (NOD/Shi-scid/IL-2Rcnull) mice from Taconic (Hudson, NY). These mice are an ideal immunodeficient model as they lack mature T, B, and Natural Killer (NK) cells, allowing them to maintain growth of the human adenomyosis tissue (Borahay et al. 2015, 2021, El Sabeh et al. 2022). Due to their immunodeficient nature, these mice were housed in isolation within the animal facilities and all experimental procedures were completed under sterile conditions.

Tissue samples, processing and implantation

We used an analogous model as previously published by our group (Borahay et al. 2015, El Sabeh et al. 2022), this time with adenomyosis xenografts instead of leiomyoma xenografts. An in-vivo xenograft model was chosen as it can closely mimic human pathophysiology of adenomyosis. Adenomyosis tissue was collected from consented patients undergoing elective hysterectomy at the University of Texas Medical Branch at Galveston. Prior to patient surgery, a screening ultrasound was performed and after surgery, a histopathologic exam was performed to confirm diagnosis of adenomyosis. Samples were collected from the operating room and processed under sterile conditions. A 2-mm Keyes biopsy punch was used to create 2 × 2 × 2-mm cylinders. In order to facilitate tissue growth, each tissue xenograft was submerged in Matrigel basement membrane matrix obtained from BD Biosciences (San Jose, CA) before insertion into mouse. Two tissue xenografts were surgically implanted subcutaneously in every mouse, one per flank. Incisions were closed with sterile surgical staples. Upon completion of the procedure, mice were monitored for signs of infection and pain. The staples were removed after one week of tissue insertion and the treatment phase commenced.

Treatment phase preparation

Simvastatin was purchased from Cayman Chemical (Ann Arbour, MI) and activated as previously described (Borahay et al. 2015). Activation is necessary to convert the prodrug form to its active form. Simvastatin (25 mg) was first dissolved in 625 μL of ethanol. Next, this solution was added to 935 μL of 0.1 NaOH and heated using a water bath (50 °C) for two hours followed by dilution with water. Using HCl, the pH of the solution was adjusted to 7. Next, the solution was diluted 8-fold using water, reaching a 2 mg/mL concentration. The solution was sterile filtered and stored at 4 °C until ready for use.

Using 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) and 1,2-distearoyl-sn-glycero-3-phosphoryle-thanolamine (DSPE) purchased from Avanti Polar Lipids (Alabaster, AL), we prepared the simvastatin-loaded liposome NPs (simvastatin-NPs). Using a 10:1 mixture and 1:10 mixture, we prepared DOPC/DSPE-Peg2000 and simvastatin-DOPC/DSPE-Peg2000 respectively as previously described (El Sabeh et al. 2022). Simvastatin-DOPC/DSPE-Peg2000 was stored lyophilised at 20 °C and dissolved in saline upon use.

Animal procedures

Six mice were randomly assigned to each group: control, simvastatin, or simvastatin-NP for a total of 18 mice (Figure 1). Sample size was chosen based on feasibility and our previously published experiments (Borahay et al. 2015, El Sabeh et al. 2022). Mice were anaesthetised via isoflurane (1–2% by nosecone) for every surgical procedure. We administered postoperative buprenorphine (0.05–0.1 mg/kg) subcutaneously twice daily then as needed for pain control. Sixty-day release pellets with 17ß-estradiol (0.05 mg) + progesterone (50 mg) (Innovative Research of America, Sarasota, FL) were surgically implanted subcutaneously, and then fifteen days later, we implanted adenomyotic xenografts. The pellets were necessary to maintain tissue growth. The control, simvastatin, and simvastatin-NP groups received daily intraperitoneal (IP) injections of 0.2 mL empty vehicle and 0.2 mL of 20 mg/kg/d simvastatin during weekdays (5 days per week), and 0.2 of 20 mg/kg/d simvastatin loaded liposomes IP 3 times weekly, respectively. We based on our administration of simvastatin and simvastatin-NP, and treatment duration on previously published rodent models (Chen et al. 2002, Skoglund and Aspenberg 2007, McKay et al. 2004, Borahay et al. 2015).

Figure 1. Schematic depiction of the experiment of the experiment.

Figure 1.

E2/P4 (oestrogen/progesterone) pellets were inserted into the mice followed by adenomyosis xenografts 15 days after. The treatment phase began 7 days later, mice were randomised to injections of empty vehicle, simvastatin, or simvastatin-loaded liposome nanoparticles (simvastatin-NP). Weekly measurements of xenograft volume were made using a calliper. Mice were sacrificed after 28 days. Immunohistochemistry was then performed on xenografts for Ki67.

The mice were monitored daily after tissue implantation and the xenografts were measured weekly with a calliper for a total of 28 days. Xenograft volumes were also measured once using a high-resolution ultrasound system with a18–38 MHz probe (Vevo 2100; Visualsonics, Toronto, Canada) right before initiating sacrifice. The formula: 0.52 X length X width X height was used to calculate the volume of the xenografts. The animals were sacrificed using isoflurane overdose and cervical dislocation. We then reopened the incision sites and obtained the adenomyotic xenografts. Those initiating injections and xenograft measurements were blinded to the treatment groups. Of note, one mouse died in each of the control and simvastatin groups in the first week and two mice died in the simvastatin-NP groups, at two-week and four-week time points. Their measurements are not included in our analysis.

Immunohistochemical analysis

The adenomyotic xenografts were placed in 1 mL of a 10% buffered formalin solution for 24 hours and then saved in 70% ethanol kept at 4 °C until staining. Fixed xenografts were used to prepare paraffin sections and immunohistochemistry was performed for Ki67, a proliferation marker of adenomyosis (Matsumoto 1999). ImageJ software was used to quantify the number of Ki67 positively stained nuclei. First, each slide was divided into 4 quadrants. Using a random number generator (1–4), we then drew a 250 × 250 µm region of interest (ROI) in the selected quadrant on each slide. We then performed thresholding on this ROI to only show positively stained nuclei. We then used an automated counting feature in ImageJ to quantify the Ki67 positive nuclei, to reduce potential bias from manual counting. Scientists were blinded to each experimental group when quantifying Ki-67.

Statistical analysis

We used the robust regression and outlier removal (ROUT) method at Q = 1% to detect and remove outliers from both the adenomyosis xenograft size measurements and the Ki67 soma counts (Motulsky and Brown 2006). One outlier was removed from the xenograft measurements on day 7 in the simvastatin group for a total of n = 5 in the control group, n = 4 in the simvastatin group, and n = 4 in the simvastatin-NP group. Upon removing the outlier, normality was assessed using the Shapiro-Wilk test. Data passed normality test at alpha = 0.05. Data are presented as mean ± standard error of the mean (SEM). The Student’s t-test was utilised to compare adenomyotic xenograft measurements and Ki67 immunohistochemistry results between each treatment to the control and each treatment to each other. P values < 0.05 were considered statistically significant. All statistical analyses were performed in GraphPad Prism and Excel.

Ethics of experimentation

This study was approved by the University of Texas Medical Branch at Galveston Institutional Review Board (protocol #10–229) and Institutional Animal Care and Use Committee (protocol #1102013).

Results

Adenomyotic xenografts growth

We first examined the growth of adenomyosis implants over 28 days in the different treatment groups. As shown in Figure 2A, adenomyotic xenografts grew at similar rates until roughly day 14 when the xenografts from the simvastatin-NP group began shrinking compared to the two other groups. At this same time, the xenografts from the simvastatin group began growing at a faster rate than the control. At the final measurement on day 28 (Figure 2A), the simvastatin-NP group had shrunk to a statistically significantly smaller volume compared to the control group (p =0.042) and compared to the simvastatin group (p <0.001). On the contrary, the growth of the adenomyosis xenografts in the simvastatin group were not significantly different in this group compared to control (p =0.052) (Figure 2A). When measured by ultrasound, the volume of the adenomyotic xenografts demonstrated a statistically significant reduction in size in the simvastatin-NP group compared to the control (p =0.044) and compared to the simvastatin group (p =0.011), while the volume of the xenografts in the simvastatin group did not show a difference in volume compared to the control (p =0.236) (Figure 2B). Lastly, the weight (g) of the xenografts in the simvastatin-NP group was significantly reduced on day 28 compared to control (p =0.037) and the simvastatin group (p =0.005). The weight (g) of the xenografts in the simvastatin group did not have a statistically significant change compared to control (p =0.149) (Figure 2C).

Figure 2. Effect of simvastatin and simvastatin-loaded liposome nanoparticles (simvastatin-NP) treatment on adenomyotic xenograft volume and weight in mouse model.

Figure 2.

Schematic depiction of the experiment of the experiment. A) Xenograft volume in cubic millimetres as measured weekly by callipers. B) Xenograft volume in cubic millimetres as measured by ultrasound on day 28 prior to sacrifice. C) Xenograft weight in grams taken post sacrifice. Data are presented as mean ± SEM. Asterisk denotes statistical significance at p < 0.05.

Intratumoural proliferation

Immunohistochemistry staining was performed on the adenomyotic xenografts to visualise the proliferation marker Ki67. Cells positively stained for Ki67 appear brown in colour while the nuclei of cells negative for Ki67 appears blue. As demonstrated in Figure 3, the simvastatin-NP group significantly reduced the expression of Ki67 compared to the control (p = 0.046). The expression of Ki67 did not differ significantly between the simvastatin group and the control group (p = 0.473).

Figure 3. Effect of simvastatin and simvastatin-loaded liposome nanoparticles (simvastatin-NP) treatment on Ki67 proliferation in adenomyosis xenografted mouse model.

Figure 3.

Adenomyosis tissues collected after sacrifice were placed in 4% formalin solution for 24 hours and then transferred to 70% ethanol until staining. Ki67 expression was quantified using ImageJ. A) Representative slides of Ki67 expression in the control, simvastatin, and simvastatin-NP groups. B) Quantification of Ki67 expression in all three groups. Data are presented as mean ± SEM. Asterisk denotes statistical significance at p < 0.05.

Discussion

Our patient-derived xenograft mouse model demonstrated that 3X weekly repeated IP injections of simvastatin-NPs significantly reduced adenomyotic xenograft volume and weight. Further, treatment with simvastatin-NP for 28 days produced a significant reduction in Ki67, a proliferation marker of adenomyosis. These changes were not present in the simvastatin group, potentially demonstrating the efficacy of simvastatin delivered via nanoparticles resulting in improving simvastatin’s bioavailability.

Drug diffusion into xenografts is driven by multiple factors including the concentration of drug in mouse plasma and the xenograft microenvironment (Saggar et al. 2013). Diffusion into adenomyosis tissue likely requires time to reach therapeutic level, thus its effect is delayed from initial administration. This supports why differences in xenograft volume between treatment arms aren’t measurable until day 14. Further, while we discontinued treatment after 28 days – following the timeline of previously published rodent models – we hypothesise that, given the trend we observed in Figure 2, we would continue to see increased adenomyotic shrinkage with continued Simvastatin-NP treatment, although this has yet to be verified (Borahay et al. 2015, Neblett et al. 2025). In addition, long term experiments of pharmacotherapy in immunocomprimsed mice, especially those who have undergone surgery, is challenging given risk of infection and mortality.

Statins are primarily used to treat hyperlipidaemia through reduction of lipid levels by targeting cholesterol production. Cholesterol is biosynthesized in a multi-step pathway, with the rate limiting step involving the HMG-CoA reductase enzyme catalysing the conversion of HMG-CoA to mevalonic acid, which is then followed by subsequent reactions (Talreja et al. 2024). Simvastatin specifically acts on this rate-limiting step, serving as an HMG-CoA reductase inhibitor, resulting in reduced concentrations of cholesterol (Bansal and Cassagnol 2024).

The mechanism of action of simvastatin on the development and growth of adenomyosis in animal models is not yet clear; however, we believe that the results may be explained by the following three mechanisms in which the drug acts including A) inhibition of adenomyotic stromal growth, B) reduced angiogenesis, and C) decreased oxidative stress.

A) Firstly, adenomyosis is a condition characterised by the presence of endometrial glands and stroma within the myometrium (Petraglia et al. 2008). Previous work has demonstrated simvastatin’s ability to inhibit endometrial stromal cell growth (Piotrowski et al. 2006). Simvastatin may reduce the rate of adenomyotic stromal growth by reducing prenylation of Ras and other GTP-ases involved in cell proliferation and apoptosis, as demonstrated previously in other tissue systems (Bouterfa et al. 2000, Danesh et al. 2002, Li et al. 2002). In addition, tissue from patients with adenomyosis has been shown to have increased expression of matrix metalloproteinase-9 (MMP-9). MMPs are proteolytic enzymes hypothesised to drive stroma remodelling in adenomyotic tissue, causing them to invade surrounding uterine muscle and implant lesions in ectopic zones (Donnez et al. 2024). Simvastatin has shown efficacy in inhibiting MMP-3 in cultured human endometrial stromal cells and other cultured cells and tissues (Bruner-Tran et al. 2009, Lazzerini 2004, Wilson et al. 2005). Since MMP-3 is a known activator of MMP-9, it’s reasonable to suspect that simvastatin may also act as an inhibitor of MMP-9, thus inhibiting growth of endometrial stromal cells and reducing expression of adenomyosis (Johnson et al. 2011, Piotrowski et al. 2006, Steenport et al. 2009, Wilson et al. 2005).

B) Next, we propose that simvastatin may reduce angiogenesis in adenomyotic tissue. Simvastatin has been demonstrated to reduce vascular endothelial growth factor (VEGF) secretion – among other angiogenic growth factors – and inhibit angiogenesis in breast and colon cancer cells (Cho et al. 2008, Li et al. 2017, Wang et al. 2018). Further, studies have demonstrated statins’ potential in protecting against endometriotic implants by reducing angiogenesis, as demonstrated by reduced vascularisation of lesions (Bruner-Tran et al. 2009, Esfandiari et al. 2007). Therefore, due to angiogenic mechanistic similarities between endometriosis and adenomyosis, it’s plausible to deduce that simvastatin may act similarly in adenomyotic tissue by reducing angiogenesis (Donnez et al. 2024, Harmsen et al. 2019).

C) Lastly, simvastatin’s antioxidant properties may act mechanistically on adenomyotic tissue to reduce oxidative stress. Scientists suspect that exposure of endometrial cells to oxidative stress may contribute to excessive cell growth, creating a conducive environment for establishment of adenomyosis (Kobayashi et al. 2020). Statins have demonstrated antioxidant effects in different diseases (Mansouri et al. 2022, Verma et al. 2022). By modulating the nuclear factor erythroid 2 related factor 2/heme oxygenase-1 (Nrf2/HO-1) pathway – a pathway implicated in the pathogenesis and development of adenomyosis – simvastatin may increase the DNA-binding activity of Nfr2, inducing expression of its target genes and thus allowing protection against oxidative stress (Chen et al. 2017, Mansouri et al. 2022).

Liposomal nanoparticles are small, biocompatible carriers widely utilised for the delivery of many therapeutics, such as chemotherapeutics agents and immune modulators (Afzal et al. 2022). These nanoparticles demonstrate immense potential in the delivery of emerging therapeutics for the treatments of benign gynaecological conditions, with previous studies demonstrating that simvastatin delivered via solid lipid NPs outperformed pure drug powder in an oral rat model, as evidenced by reduced cholesterol values, indicating improved bioavailability (Padhye and Nagarsenker 2013). However, more research is needed to engineer simvastatin-NPs for optimal controlled drug release for the treatment of gynaecological conditions.

To our knowledge, this is the first study that explores the application of simvastatin-NP as a potential treatment of adenomyosis. One study examined the effect of a hypocholesterolemic agent – Probucal – on prevention of development of adenomyosis with promising results in mice. (Zhou et al. 2004). Our study lends further evidence that hypocholesterolemic agents may have a role in prevention and treatment of adenomyosis. By utilising an in vivo, xenograft animal model, we demonstrate the advantages of slow-release simvastatin via nanoparticles, offering superior therapeutic effects compared to direct simvastatin administration. However, there are several limitations that must be acknowledged. Firstly, the sample size was small, limiting conclusions drawn from this single pilot study. Further, simvastatin and simvastatin-NP concentrations in the adenomyotic xenograft tissue and mouse plasma were not measured, limiting our ability to directly compare absorption rates between the two formulations. Lastly, administering simvastatin only on weekdays, rather than using a sustained regimen, may have decreased the efficacy of naked simvastatin in inhibiting xenograft growth. However, this may better represent ‘typical use’ of the medication rather than ‘ideal use’ and may better represent actual results in real patients, who may skip doses or not take exactly as prescribed.

Also of note, statins may have potential off-target effects on reproductive tissues which may impact uterine and ovarian function. Notably, previous work has showed that statin treatment in animal models resulted in alterations in ovarian morphology, including reduced follicular development and impaired hormone production, which could have long-term implications for reproductive health (Jiao et al. 2020). Further research exploring the dose-dependent nature of off-target reproductive effects as well as the potential reversibility of statin-induced reproductive dysfunction are needed to fully elucidate the balance between therapeutic benefits versus potential adverse events when considering simvastatin as a treatment modality.

Conclusion

Despite limitations, the current study demonstrates the potential advantages of using slow-release nanoparticle to enhance the efficacy of simvastatin for the treatment of adenomyosis, as a promising therapeutic approach.

Acknowledgments

We gratefully acknowledge fruitful discussions with Dr. Dylan Faltine-Gonzalez. M.A.B. and K.L.V. conceived the project. All authors contributed to data collection. R.M., G.W.K., and M.A.B. analysed the data and drafted the manuscript. All authors reviewed, edited, and approved the final manuscript.

Funding

This work was supported, in part, by the NIH grants R01HD094380, R01HD111243, K12HD001269 and NCATS UL1TR000071 award to Mostafa Borahay.

Footnotes

Ethics statement

This study was approved by the University of Texas Medical Branch at Galveston Institutional Review Board (protocol #10–229) and Institutional Animal Care and Use Committee (protocol #1102013).

Disclosure statement

No potential conflict of interest was reported by the authors.

Data availability statement

The data that support the findings of this study are available from the corresponding author, Mostafa Borahay, upon reasonable request.

References

  1. Afrin S, et al. , 2020. Simvastatin ameliorates altered mechanotransduction in uterine leiomyoma cells. American Journal of Obstetrics and Gynecology, 223 (5), 733.e1–733.e14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Afrin S, et al. , 2021. Simvastatin modulates estrogen signaling in uterine leiomyoma via regulating receptor palmitoylation, trafficking and degradation. Pharmacological Research, 172, 105856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Afzal O, et al. , 2022. Nanoparticles in drug delivery: from history to therapeutic applications. Nanomaterials (Basel, Switzerland), 12 (24), 4494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Bansal AB and Cassagnol M, 2024. HMG-CoA reductase inhibitors. [PubMed] [Google Scholar]
  5. Borahay MA, et al. , 2014. Simvastatin potently induces calcium-dependent apoptosis of human leiomyoma cells. The Journal of Biological Chemistry, 289 (51), 35075–35086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Borahay MA, et al. , 2015. Novel effects of simvastatin on uterine fibroid tumors: in vitro and patient-derived xenograft mouse model study. American Journal of Obstetrics and Gynecology, 213 (2), 196.e1–8–196.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Borahay MA, et al. , 2021. Liposomal 2-methoxyestradiol nanoparticles for treatment of uterine leiomyoma in a patient-derived xenograft mouse model. Reproductive Sciences (Thousand Oaks, Calif.), 28 (1), 271–277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Bouterfa HL, et al. , 2000. Inhibition of Ras farnesylation by lovastatin leads to downregulation of proliferation and migration in primary cultured human glioblastoma cells. Anticancer Research, 20 (4), 2761–2771. [PubMed] [Google Scholar]
  9. Bruner-Tran KL, Osteen KG and Duleba AJ, 2009. Simvastatin protects against the development of endometriosis in a nude mouse model. The Journal of Clinical Endocrinology and Metabolism, 94 (7), 2489–2494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Chen N, et al. , 2017. Abnormal expression of Nrf2 may play an important role in the pathogenesis and development of adenomyosis. PloS One, 12 (8), e0182773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Chen Z, et al. , 2002. Simvastatin reduces neointimal thickening in low-density lipoprotein receptor–deficient mice after experimental angioplasty without changing plasma lipids. Circulation, 106 (1), 20–23. [DOI] [PubMed] [Google Scholar]
  12. Cho S, et al. , 2008. Simvastatin induces apoptosis in human colon cancer cells and in tumor xenografts, and attenuates colitis‐associated colon cancer in mice. International Journal of Cancer, 123 (4), 951–957. [DOI] [PubMed] [Google Scholar]
  13. Chou R, et al. , 2022. Statin use for the primary prevention of cardiovascular disease in adults. JAMA, 328 (8), 754–771. [DOI] [PubMed] [Google Scholar]
  14. Danesh FR, et al. , 2002. 3-Hydroxy-3-methylglutaryl CoA reductase inhibitors prevent high glucose-induced proliferation of mesangial cells via modulation of Rho GTPase/p21 signaling pathway: Implications for diabetic nephropathy. Proceedings of the National Academy of Sciences of the United States of America, 99 (12), 8301–8305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Donnez J, Stratopoulou CA and Dolmans M-M, 2024. Endometriosis and adenomyosis: Similarities and differences. Best Practice & Research. Clinical Obstetrics & Gynaecology, 92, 102432. [DOI] [PubMed] [Google Scholar]
  16. Duleba A, et al. , 2014. Simvastatin reduces volume of active endometriotic lesions in primate model of endometriosis. Fertility and Sterility, 102 (3), e47–e48. [Google Scholar]
  17. El Sabeh M, et al. , 2021. Simvastatin inhibits Wnt/β-catenin pathway in uterine leiomyoma. Endocrinology, 162 (12), 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. El Sabeh M, et al. , 2022. Simvastatin-loaded liposome nanoparticles treatment for uterine leiomyoma in a patient-derived xenograft mouse model: a pilot study. Journal of Obstetrics and Gynaecology: The Journal of the Institute of Obstetrics and Gynaecology, 42 (6), 2139–2143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Enazy SA, et al. , 2023. PEGylated polymeric nanoparticles loaded with 2-methoxyestradiol for the treatment of uterine leiomyoma in a patient-derived xenograft mouse model. Journal of Pharmaceutical Sciences, 112 (9), 2552–2560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Esfandiari N, et al. , 2007. Effect of a statin on an in vitro model of endometriosis. Fertility and Sterility, 87 (2), 257–262. [DOI] [PubMed] [Google Scholar]
  21. Etrusco A, et al. , 2023. Current medical therapy for adenomyosis: from bench to bedside. Drugs, 83 (17), 1595–1611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Gunther R and Walker C, 2024. Adenomyosis. [PubMed] [Google Scholar]
  23. Harmsen MJ, et al. , 2019. Role of angiogenesis in adenomyosis-associated abnormal uterine bleeding and subfertility: a systematic review. Human Reproduction Update, 25 (5), 647–671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Jiao X-F, et al. , 2020. Ovary and uterus-related adverse events associated with statin use: an analysis of the FDA adverse event reporting system. Scientific Reports, 10 (1), 11955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Johnson JL, et al. , 2011. Matrix metalloproteinase (MMP)-3 activates MMP-9 mediated vascular smooth muscle cell migration and neointima formation in mice. Arteriosclerosis, Thrombosis, and Vascular Biology, 31 (9), e35–e44. [DOI] [PubMed] [Google Scholar]
  26. Kho KA, Chen JS and Halvorson LM, 2021. Diagnosis, evaluation, and treatment of adenomyosis. JAMA, 326 (2), 177–178. [DOI] [PubMed] [Google Scholar]
  27. Kobayashi H, Kishi Y and Matsubara S, 2020. Mechanisms underlying adenomyosis-related fibrogenesis. Gynecologic and Obstetric Investigation, 85 (1), 1–12. [DOI] [PubMed] [Google Scholar]
  28. Krentel H and De Wilde RL, 2022. Prevalence of adenomyosis in women undergoing hysterectomy for abnormal uterine bleeding, pelvic pain or uterine prolapse - A retrospective cohort study. Annals of Medicine and Surgery (2012), 78, 103809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Lazzerini PE, et al. , 2004. Simvastatin reduces MMP-3 level in interleukin 1 stimulated human chondrocyte culture. Annals of the Rheumatic Diseases, 63 (7), 867–869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Li G, et al. , 2017. Simvastatin inhibits tumor angiogenesis in HER2-overexpressing human colorectal cancer. Biomedicine & Pharmacotherapy = Biomedecine & Pharmacotherapie, 85, 418–424. [DOI] [PubMed] [Google Scholar]
  31. Li X, et al. , 2002. Inhibition of protein geranylgeranylation and Rhoa/Rhoa kinase pathway induces apoptosis in human endothelial cells. The Journal of Biological Chemistry, 277 (18), 15309–15316. [DOI] [PubMed] [Google Scholar]
  32. Lulseged BA, et al. , 2024. The role of nanomedicine in benign gynecologic disorders. Molecules (Basel, Switzerland), 29 (9), 2095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Mansouri A, et al. , 2022. Antioxidant effects of statins by modulating Nrf2 and Nrf2/HO-1 signaling in different diseases. Journal of Clinical Medicine, 11 (5), 1313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Matsumoto Y, et al. , 1999. Apoptosis and Ki-67 expression in adenomyotic lesions and in the corresponding eutopic endometrium. Obstetrics and Gynecology, 94 (1), 71–77. [DOI] [PubMed] [Google Scholar]
  35. McKay A, et al. , 2004. A novel anti-inflammatory role of simvastatin in a murine model of allergic asthma. Journal of Immunology (Baltimore, Md.: 1950), 172 (5), 2903–2908. [DOI] [PubMed] [Google Scholar]
  36. Moawad G, Fruscalzo A, et al. , 2023. Adenomyosis: an updated review on diagnosis and classification. Journal of Clinical Medicine, 12 (14), 4828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Moawad G, Youssef Y, et al. , 2023. The present and the future of medical therapies for adenomyosis: a narrative review. Journal of Clinical Medicine, 12 (19), 6130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Motulsky HJ and Brown RE, 2006. Detecting outliers when fitting data with nonlinear regression – a new method based on robust nonlinear regression and the false discovery rate. BMC Bioinformatics, 7 (1), 123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Nasu K, et al. , 2009. Simvastatin inhibits the proliferation and the contractility of human endometriotic stromal cells: a promising agent for the treatment of endometriosis. Fertility and Sterility, 92 (6), 2097–2099. [DOI] [PubMed] [Google Scholar]
  40. Neblett MF, et al. , 2025. Evaluation of the in vivo efficacy of the JAK inhibitor AZD1480 in uterine leiomyomas using a patient-derived xenograft murine model. Reproductive Sciences (Thousand Oaks, Calif.), 32 (2), 417–427. [DOI] [PubMed] [Google Scholar]
  41. Padhye SG and Nagarsenker MS, 2013. Simvastatin solid lipid nanoparticles for oral delivery: formulation development and in vivo evaluation. Indian Journal of Pharmaceutical Sciences, 75 (5), 591–598. [PMC free article] [PubMed] [Google Scholar]
  42. Petraglia F, et al. , 2008. Hormone-dependent gynaecological disorders: a pathophysiological perspective for appropriate treatment. Best Practice & Research. Clinical Obstetrics & Gynaecology, 22 (2), 235–249. [DOI] [PubMed] [Google Scholar]
  43. Piotrowski PC, et al. , 2006. Statins inhibit growth of human endometrial stromal cells independently of cholesterol availability1. Biology of Reproduction, 75 (1), 107–111. [DOI] [PubMed] [Google Scholar]
  44. Saggar JK, et al. , 2013. The tumor microenvironment and strategies to improve drug distribution. Frontiers in Oncology, 3, 154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Skoglund B and Aspenberg P, 2007. Locally applied Simvastatin improves fracture healing in mice. BMC Musculoskeletal Disorders, 8 (1), 98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Steenport M, et al. , 2009. Matrix metalloproteinase (MMP)-1 and MMP-3 induce macrophage MMP-9: Evidence for the role of TNF-α and cyclooxygenase-2. Journal of Immunology (Baltimore, Md.: 1950), 183 (12), 8119–8127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Talreja O, Kerndt CC and Cassagnol M, 2024. Simvastatin. [PubMed] [Google Scholar]
  48. Upson K and Missmer SA, 2020. Epidemiology of adenomyosis. Seminars in Reproductive Medicine, 38 (2–03), 89–107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Vannuccini S and Petraglia F, 2019. Recent advances in understanding and managing adenomyosis. F1000Research, 8, 283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Verma K, et al. , 2022. Simvastatin ameliorates oxidative stress levels in HepG2 cells and hyperlipidemic rats. Current Research in Pharmacology and Drug Discovery, 3, 100088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Wang J, et al. , 2018. Activation of < scp > AMPK</scp > by simvastatin inhibited breast tumor angiogenesis via impeding < scp > HIF ‐1α‐ induced pro‐angiogenic factor. Cancer Science, 109 (5), 1627–1637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Wilson WRW, et al. , 2005. HMG-CoA reductase inhibitors (statins) decrease MMP-3 and MMP-9 concentrations in abdominal aortic aneurysms. European Journal of Vascular and Endovascular Surgery: The Official Journal of the European Society for Vascular Surgery, 30 (3), 259–262. [DOI] [PubMed] [Google Scholar]
  53. Zhou YF, et al. , 2004. Probucol, a hypocholesterolemic agent, prevents the development of uterine adenomyosis induced by pituitary grafting in mice. Anticancer Research, 24 (4), 2209–2212. [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

The data that support the findings of this study are available from the corresponding author, Mostafa Borahay, upon reasonable request.

RESOURCES