Novel Approaches to Possible Targeted Therapies and Prophylaxis of Uterine Fibroids
Abstract
:1. Introduction
2. Review Structure and Design
3. Mechanisms and Risk Factors for the Development of Recurrent Uterine Leiomyoma
4. Existing Methods of Medicinal Treatment for Recurrent Uterine Leiomyomata
5. Prediction of Genetic Risk and Identification of Potential Therapeutic Targets in Leiomyoma Cells Using Metagenomic, Metatranscriptomic, and Metamethylomic Methods of Analysis of Complete Genomes
- Secreted proteins that do not remain on the surface of the cells producing them;
- Proteins present exclusively in leiomyoma cells and absent in any cells of normal tissues.
- 3.
- From the work by [61]: PCP4—Purkinje cell protein 4, expressed in embryogenesis; CHRDL2—growth factor, a chordin-like antagonist of BMP; RPE65—retinol transporter from the retinal epithelium; MMP11—matrix metalloproteinase 11 (or stromelysin 3), expressed in embryogenesis, causes metastasis of breast tumors; MFAP2—microfibrillar-associated protein 2, affects the motility of cancer cells in gastric cancer, regulates the expression of a5ß1 integrin via the ERK1/2 pathway.
- 4.
- From the work by [62]:
- For HMGA2-dependent fibroid types: HMGA2, GRPR—gastrin precursor; PLAG1—proto-oncogene, the main participant of HMGA2-dependent signaling; PAPPA2—pappalysin, a protease that destroys IGFBP5, a marker of osteoblasts; MB21D2—nucleotidyltransferase, an anti-virus protection gene induced by interferon;
- For MED12-dependent fibroid types: ADAM12—membrane-bound metalloproteinase responsible for shedding—cleavage of extracellular receptor domains and proteoglycans; MMP11—matrix metalloproteinase 11 (or stromelysin 3), expressed in embryogenesis, causes metastasis of breast tumors; MMP16—matrix metalloproteinase 16, has a transmembrane domain, expressed in embryogenesis; RAD51B—protein of the DNA reparation system, similar to RecA, with ATPase activity, not expressed in normal tissues; PCP4 is a protein of 4 Purkinje cells, expressed in embryogenesis; RUNDC1 is a DNA-binding protein with a RUN-type domain at the C-terminus; THSD4 (Adamtsl6-β) matrix metalloproteinase with a thrombospondin domain associated with the cell surface; participates in the formation of ECM microfibrils, ensuring elasticity and release of deposited growth factors. Adamtsl6-β expression increases during the formation of the periodontal ligament, which consists of a fibrillin-1 microfibril.
6. Approaches to the Development of New Candidate Therapeutics for the Prevention and Treatment of Relapses of Leiomyoma, including Patients Preparing for Pregnancy or in the Process of Gestation
- (1)
- The immunobiological drugs secukinumab, ixekizumab, brodalumab, and netakimab, a recombinant humanized antibody against IL17—for all leiomyoma types;
- (2)
- Synthetic analogues of retinoids: adapalene, tretinoin, isotretinoin, and tazarotene—potential blockers of the RPE65 receptor—for all leiomyoma types;
- (3)
- Olaparib and niraparib—synthetic inhibitors of poly-ADP-ribosopolymerase (PARP1, also known as NAD+-ADP-ribosyltransferase 1 and PARP2)—for leiomyoma with biallelic inactivation of the fumarate hydratase gene as a driver mutation;
- (4)
- Resveratrol—for all leiomyoma types.
7. Discussion
8. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Sidorova, I.S.; Hunanyan, A.L.; Kogan, E.A.; Guriev, T.D. Uterine fibrosis in young patients: Clinical and pathogenetic. Obstet. Gynecol. Reprod. 2010, 4, 16–20. (In Russian) [Google Scholar]
- Krasnopolsky, I.V. Operative Gynecology; Medpress-Inform: Moscow, Russia, 2010; 319p. [Google Scholar]
- Arutyunova, E.E.; Katkova, A.S.; Buralkina, N.A. Ethnogeography of uterine fibroids: Epidemiology, age and race differences, types of surgery. Cons. Medicum 2018, 20, 26–30. [Google Scholar] [CrossRef]
- Krasnopol’skii, V.I.; Logutova, L.S.; Buianova, S.N. Reproduktivnye Problem Operirovannoi Matki; Miklosh: Moscow, Russia, 2008. [Google Scholar]
- Gupta, S.; Jose, J.; Manyonda, I. Clinical presentation of fibroids. Best. Pract. Res. Clin. Obstet. Gynaecol. 2008, 22, 615–626. [Google Scholar] [CrossRef] [PubMed]
- Vikhlyaeva, E.M.; Zheleznov, B.I.; Zaporozhan, V.N. Rukovodstvo po Endokrinnoi Ginekologii; Meditsinskoe Informatsionnoe Agenstvo: Moscow, Russia, 1997. [Google Scholar]
- Pavone, D.; Clemenza, S.; Sorbi, F.; Fambrini, M.; Petraglia, F. Epidemiology and risk factors for uterine fibroids, the best prakt. Res. Wedge. Obstetrics. Gynecol. 2018, 46, 3–11. [Google Scholar]
- Pechetov, A.A.; Lednev, A.N.; Ratnikova, N.K.; Volchansky, D.A. Benign metastatic uterine leiomyoma with lung damage: Problems of diagnosis and treatment and treatment. Surgery 2020, 9, 85–88. (In Russian) [Google Scholar]
- Torres de la Roche, L.A.; Becker, C.; Cesar, C.; Hermann, A.; Larbig, A.; Leicher, L.; Di Spiezio Sardo, A.; Thanos, B.; Vallviner, M.; Verhoeven, X.; et al. Pathobiology of uterine myomatosis: Basic knowledge necessary to support our clinical practice. Arch. Gynecol. Obstet. 2017, 296, 701–707. [Google Scholar] [CrossRef] [PubMed]
- Occhino, J.A.; Trabuco, E.C. Hysterectomy and alternative options. Obstet. Gynecol. Clin. N. Am. 2016, 43, 13–14. [Google Scholar] [CrossRef]
- Wu, J.M.; Vechter, M.E.; Geller, E.J.; Nguyen, T.V.; Visco, A.G. The frequency of hysterectomies in the United States. Obstetrician-gynecologist 2007, 110, 1091–1095. [Google Scholar]
- Dukhan, N. Modern and emerging methods of treatment of uterine fibroids. Int. J. Womens Health 2011, 3, 231–341. [Google Scholar]
- Cardozo, E.R.; Clark, A.D.; Banks, N.K.; Henne, M.B.; Stegmann, B.J.; Segars, J.H. Estimated annual cost of uterine leiomyoma treatment in the United States. Am. J. Obstet. Gynecol. 2012, 206, 211. [Google Scholar] [CrossRef]
- Edwards, T.L.; Giri, A.; Hellwege, J.N.; Hartmann, K.E.; Stewart, E.A.; Jeff, J.M.; Bray, M.J.; Pendergrass, S.A.; Torstenson, E.S.; Keaton, J.M.; et al. A Trans-Ethnic Genome-Wide Association Study of Uterine Fibroids. Font. Genet. 2019, 10, 511. [Google Scholar] [CrossRef] [PubMed]
- Gonzalez Gonzalez, V.; Erraes Moretta, A.; Mayoral Triana, A.; Riolobos Sierra, L.; Cristobal Garcia, I.; Izquierdo Mendes, N. Prolapse of cervical fibroids during pregnancy. Eur. J. Obstet. Gynecol. Reprod. Biol. 2020, 252, 150–154. [Google Scholar] [CrossRef] [PubMed]
- Stewart, E.A.; Cookson, C.L.; Gandolfo, R.A.; Schulze-Rath, R. Epidemiology of uterine fibroids: A systematic review. BJOG 2017, 124, 1501–1512. [Google Scholar] [CrossRef] [PubMed]
- Kotani, Y.; Tobiume, T.; Fujishima, R.; Shigeta, M.; Takaya, H.; Nakai, H.; Suzuki, A.; Tsuji, I.; Mandai, M.; Matsumura, N. Recurrence of uterine myoma after myomectomy: Open myomectomy versus laparoscopic myomectomy. J. Obstet. Gynaecol. Res. 2018, 44, 298–302. [Google Scholar] [CrossRef] [PubMed]
- Marugo, M.; Centonze, M.; Bernasconi, D.; Fazzuoli, L.; Berta, S.; Giordano, G. Estrogen and progesterone receptors in uterine leiomyomas. Acta Obs. Gynecol. Scand. 1989, 68, 731–735. [Google Scholar] [CrossRef] [PubMed]
- Nilbert, M.; Heim, S.; Mandahl, N.; Flodérus, U.M.; Willén, H.; Mitelman, F. Karyotypic rearrangements in 20 uterine leiomyomas. Cytogenet Cell Genet. 1988, 49, 300–304. [Google Scholar] [CrossRef] [PubMed]
- Nilbert, M.; Strömbeck, B. Independent origin of uterin leiomyomas with karyotypically identica alteratios. Gynecol. Obstet. Investig. 1992, 33, 246–248. [Google Scholar] [CrossRef] [PubMed]
- Nilbert, M.; Heim, S.; Mandahl, N.; Floderus, U.M.; Willen, H.; Mitelman, F. Trisomy 12 in uterine leiomyomas. A new cytogenetic subgroup. Cancer Genet. Cytogenet. 1990, 45, 63–66. [Google Scholar] [CrossRef]
- Hennig, Y.; Rogalla, P.; Wanschura, S.; Frey, G.; Deichert, U.; Bartnitzke, S.; Bullerdiek, J. HMGIC expressed in a uterine leiomyoma with a deletion of the long arm of chromosome 7 along with a 12q14-15 rearrangement but not in tumors showing del(7) as the sole cytogenetic abnormality. Cancer Genet. Cytogenet. 1997, 96, 129–133. [Google Scholar] [CrossRef]
- Klotzbücher, M.; Wasserfall, A.; Fuhrmann, U. Misexpression of wild-type and truncated isoforms of the high-mobility group I proteins HMGI-C and HMGI(Y) in uterine leiomyomas. Am. J. Pathol. 1999, 155, 1535–1542. [Google Scholar] [CrossRef]
- Klemke, M.; Meyer, A.; Nezhad, M.H.; Bartnitzke, S.; Drieschner, N.; Frantzen, C.; Schmidt, E.H.; Belge, G.; Bullerdiek, J. Overexpression of HMGA2 in uterine leiomyomas points to its general role for the pathogenesis of the disease. Genes Chromosomes Cancer 2009, 48, 171–178. [Google Scholar] [CrossRef] [PubMed]
- Klemke, M.; Meyer, A.; Hashemi Nezhad, M.; Belge, G.; Bartnitzke, S.; Bullerdiek, J. Loss of let-7 binding sites resulting from truncations of the 3′ untranslated region of HMGA2 mRNA in uterine leiomyomas. Cancer Genet. Cytogenet. 2010, 196, 119–123. [Google Scholar] [CrossRef] [PubMed]
- Markowski, D.N.; von Ahsen, I.; Nezhad, M.H.; Wosniok, W.; Helmke, B.M.; Bullerdiek, J. HMGA2 and the p19Arf-TP53-CDKN1A axis: A delicate balance in the growth of uterine leiomyomas. Genes Chromosomes Cancer 2010, 49, 661–668. [Google Scholar] [CrossRef] [PubMed]
- Schoenmakers, E.F.; Bunt, J.; Hermers, L.; Schepens, M.; Merkx, G.; Janssen, B.; Kersten, M.; Huys, E.; Pauwels, P.; Debiec-Rychter, M.; et al. Identification of CUX1 as the recurrent chromosomal band 7q22 target gene in human uterine leiomyoma. Genes Chromosomes Cancer 2013, 52, 11–23. [Google Scholar] [CrossRef] [PubMed]
- Holzmann, C.; Markowski, D.N.; Koczan, D.; Küpker, W.; Helmke, B.M.; Bullerdiek, J. Cytogenetically normal uterine leiomyomas without MED12-mutations—A source to identify unknown mechanisms of the development of uterine smooth muscle tumors. Mol. Cytogenet. 2014, 7, 88. [Google Scholar] [CrossRef] [PubMed]
- Pradhan, B.; Sarvilinna, N.; Matilainen, J.; Aska, E.; Sjöberg, J.; Kauppi, L. Detection and screening of chromosomal rearrangements in uterine leiomyomas by long-distance inverse PCR. Genes Chromosomes Cancer 2016, 55, 215–226. [Google Scholar] [CrossRef] [PubMed]
- Mäkinen, N.; Kämpjärvi, K.; Frizzell, N.; Bützow, R.; Vahteristo, P. Characterization of MED12, HMGA2, and FH alterations reveals molecular variability in uterine smooth muscle tumors. Mol. Cancer 2017, 16, 101. [Google Scholar] [CrossRef]
- Wu, X.; Serna, V.A.; Thomas, J.; Qiang, W.; Blumenfeld, M.L.; Kurita, T. Subtype-Specific Tumor-Associated Fibroblasts Contribute to the Pathogenesis of Uterine Leiomyoma. Cancer Res. 2017, 77, 6891–6901. [Google Scholar] [CrossRef]
- Mas, A.; Nair, S.; Laknaur, A.; Simón, C.; Diamond, M.P.; Al-Hendy, A. Stro-1/CD44 as putative human myometrial and fibroid stem cell markers. Fertil. Steril. 2015, 104, 225–234. [Google Scholar] [CrossRef]
- Mu, Y.; He, J.; Yan, R.; Hu, X.; Liu, H.; Hao, Z. IGF-1 and VEGF can be used as prognostic indicators for patients with uterine fibroids treated with uterine artery embolization. Exp. Ther. Med. 2016, 11, 645–649. [Google Scholar] [CrossRef]
- Heikkinen, T.; Äyräväinen, A.; Hänninen, J.; Ahvenainen, T.; Bützow, R.; Pasanen, A.; Vahteristo, P. MED12 mutations and fumarate hydratase inactivation in uterine adenomyomas. Hum. Reprod. Open 2018, 4, hoy020. [Google Scholar] [CrossRef] [PubMed]
- Reis, F.M.; Bloise, E.; Ortiga-Carvalho, T.M. Hormones and pathogenesis of uterine fibroids. Best. Pract. Res. Clin. Obstet. Gynaecol. 2016, 34, 13–24. [Google Scholar] [CrossRef] [PubMed]
- Sulkowski, P.L.; Sundaram, R.K.; Oeck, S.; Corso, C.D.; Liu, Y.; Noorbakhsh, S.; Niger, M.; Boeke, M.; Ueno, D.; Kalathil, A.N.; et al. Krebs-cycle-deficient hereditary cancer syndromes are defined by defects in homologous-recombination DNA repair. Nat. Genet. 2018, 50, 1086–1092. [Google Scholar] [CrossRef] [PubMed]
- Yin, P.; Ono, M.; Moravek, M.B.; Coon, J.S., 5th; Navarro, A.; Monsivais, D.; Dyson, M.T.; Druschitz, S.A.; Malpani, S.S.; Serna, V.A.; et al. Human uterine leiomyoma stem/progenitor cells expressing CD34 and CD49b initiate tumors in vivo. J. Clin. Endocrinol. Metab. 2015, 100, E601–E606. [Google Scholar] [CrossRef] [PubMed]
- Gonzalez-Barcena, D.; Alvarez, R.B.; Ochoa, E.P.; Cornejo, I.C.; Comaru-Schally, A.M.; Schally, A.V.; Engel, J.; Reissmann, T.; Riethmüller-Winzen, H. Treatment of uterine leiomyomas with luteinizing hormone-releasing hormone antagonist Cetrorelix. Hum. Reprod. 1997, 12, 2028–2035. [Google Scholar] [CrossRef] [PubMed]
- Nowicki, M.; Adamkiewicz, G.; Bryc, W.; Kokot, F. The influence of luteinizing hormone-releasing hormone analog on serum leptin and body composition in women with solitary uterine myoma. Am. J. Obstet. Gynecol. 2002, 186, 340–344. [Google Scholar] [CrossRef] [PubMed]
- Engman, M.; Granberg, S.; Williams, A.R.; Meng, C.X.; Lalitkumar, P.G.; Gemzell-Danielsson, K. Mifepristone for treatment of uterine leiomyoma. A prospective randomized placebo controlled trial. Hum. Reprod. 2009, 24, 1870–1879. [Google Scholar] [CrossRef] [PubMed]
- Bouchard, P.; Chabbert-Buffet, N.; Fauser, B.C. Selective progesterone receptor modulators in reproductive medicine: Pharmacology, clinical efficacy and safety. Fertil. Steril. 2011, 96, 1175–1189. [Google Scholar] [CrossRef]
- Malik, M.; Britten, J.; Cox, J.; Patel, A.; Catherino, W.H. Gonadotropin-releasing hormone analogues inhibit leiomyoma extracellular matrix despite presence of gonadal hormones. Fertil. Steril. 2016, 105, 214–324. [Google Scholar] [CrossRef]
- Patel, A.; Malik, M.; Britten, J.; Cox, J.; Catherino, W.H. Mifepristone inhibits extracellular matrix formation in uterine leiomyoma. Fertil. Steril. 2016, 105, 1102–1110. [Google Scholar] [CrossRef]
- Islam, M.S.; Afrin, S.; Singh, B.; Jayes, F.L.; Brennan, J.T.; Borahay, M.A.; Leppert, P.C.; Segars, J.H. Extracellular matrix and Hippo signaling as therapeutic targets of antifibrotic compounds for uterine fibroids. Clin. Transl. Med. 2021, 11, e475. [Google Scholar] [CrossRef] [PubMed]
- Dababou, S.; Garzon, S.; Laganà, A.S.; Ferrero, S.; Evangelisti, G.; Noventa, M.; D’Alterio, M.N.; Palomba, S.; Uccella, S.; Franchi, M.; et al. Linzagolix: A new GnRH-antagonist under investigation for the treatment of endometriosis and uterine myomas. Expert. Opin. Investig. Drugs 2021, 30, 903–911. [Google Scholar] [CrossRef] [PubMed]
- Middelkoop, M.A.; de Lange, M.E.; Clark, T.J.; Mol, B.W.J.; Bet, P.M.; Huirne, J.A.F.; Hehenkamp, W.J.K. Evaluation of marketing authorization and clinical implementation of ulipristal acetate for uterine fibroids. Hum. Reprod. 2022, 37, 884–894. [Google Scholar] [CrossRef] [PubMed]
- Arjona Ferreira, J.C.; Migoya, E. Development of relugolix combination therapy as a medical treatment option for women with uterine fibroids or endometriosis. F&S Rep. 2022, 4 (Suppl. S2), 73–82. [Google Scholar]
- Salas, A.; García-García, P.; Díaz-Rodríguez, P.; Évora, C.; Almeida, T.A.; Delgado, A. New local ganirelix sustained release therapy for uterine leiomyoma. Evaluation in a preclinical organ model. Biomed. Pharmacother. 2022, 156, 113909. [Google Scholar] [CrossRef] [PubMed]
- Chwalisz, K. Clinical development of the oral gonadotropin-releasing hormone antagonist elagolix. F&S Rep. 2023, 4 (Suppl. S2), 65–72. [Google Scholar]
- Hu, J.; Dong, A.; Fernandez-Ruiz, V.; Shan, J.; Kawa, M.; Martínez-Ansó, E.; Prieto, J.; Qian, C. Blockade of Wnt signaling inhibits angiogenesis and tumor growth in hepatocellular carcinoma. Cancer Res. 2009, 69, 6951–6959. [Google Scholar] [CrossRef]
- Xu, X.; Lu, Z.; Qiang, W.; Vidimar, V.; Kong, B.; Kim, J.J.; Wei, J.J. Inactivation of AKT induces cellular senescence in uterine leiomyoma. Endocrinology 2014, 155, 1510–1519. [Google Scholar] [CrossRef]
- Ye, N.; Ding, Y.; Wild, C.; Shen, Q.; Zhou, J. Small molecule inhibitors targeting activator protein 1 (AP-1). J. Med. Chem. 2014, 57, 6930–6948. [Google Scholar] [CrossRef]
- Galindo, L.J.; Hernández-Beeftink, T.; Salas, A.; Jung, Y.; Reyes, R.; de Oca, F.M.; Hernández, M.; Almeida, T.A. HMGA2 and MED12 alterations frequently co-occur in uterine leiomyomas. Gynecol. Oncol. 2018, 150, 562–568. [Google Scholar] [CrossRef]
- Xie, J.; Ubango, J.; Ban, Y.; Chakravarti, D.; Kim, J.J.; Wei, J.J. Comparative analysis of AKT and the related biomarkers in uterine leiomyomas with MED12, HMGA2, and FH mutations. Genes Chromosomes Cancer 2018, 57, 485–494. [Google Scholar] [CrossRef] [PubMed]
- Alzahrani, A.S. PI3K/Akt/mTOR inhibitors in cancer: At the bench and bedside. Semin. Cancer Biol. 2019, 59, 125–132. [Google Scholar] [CrossRef] [PubMed]
- Pilgrim, J.; Arismendi, J.; DeAngelis, A.; Lewis, T.; Britten, J.; Malik, M.; Catherino, W.H. Characterization of the role of Activator Protein 1 signaling pathway on extracellular matrix deposition in uterine leiomyoma. F&S Sci. 2020, 1, 78–89. [Google Scholar]
- Maekawa, R.; Sato, S.; Tamehisa, T.; Sakai, T.; Kajimura, T.; Sueoka, K.; Sugino, N. Different DNA methylome, transcriptome and histological features in uterine fibroids with and without MED12 mutations. Sci. Rep. 2022, 12, 8912. [Google Scholar] [CrossRef] [PubMed]
- Wang, W.; Zhang, W.; Li, D.; Qian, R.; Zhu, L.; Liu, Y.; Chen, C. Lichong decoction inhibits micro-angiogenesis by reducing the expressions of hypoxia inducible factor-1α and vascular endothelial growth factor in hysteromyoma mouse model. J. Tradit. Chin. Med. 2020, 40, 928–937. [Google Scholar] [PubMed]
- Laganà, A.S.; Vergara, D.; Favilli, A.; La Rosa, V.L.; Tinelli, A.; Gerli, S.; Noventa, M.; Vitagliano, A.; Triolo, O.; Rapisarda, A.M.C.; et al. Epigenetic and genetic landscape of uterine leiomyomas: A current view over a common gynecological disease. Arch. Gynecol. Obstet. 2017, 296, 855–867. [Google Scholar] [CrossRef] [PubMed]
- Baranov, V.S.; Osinovskaya, N.S.; Yarmolinskaya, M.I. Pathogenomics of Uterine Fibroids Development. Int. J. Mol. Sci. 2019, 24, 6151. [Google Scholar] [CrossRef] [PubMed]
- Navarro, A.; Yin, P.; Monsivais, D.; Lin, S.M.; Du, P.; Wei, J.J.; Bulun, S.E. Genome-wide DNA methylation indicates silencing of tumor suppressor genes in uterine leiomyoma. PLoS ONE 2012, 7, e33284. [Google Scholar] [CrossRef]
- Mehine, M.; Kaasinen, E.; Heinonen, H.R.; Makinen, N.; Kampjarvi, K.; Sarvilinna, N.; Aavikko, M.; Vaharautio, A.; Pasanen, A.; Butzow, R.; et al. Integrated data analysis reveals uterine leiomyoma subtypes with distinct driver pathways and biomarkers. Proc. Natl. Acad. Sci. USA 2016, 113, 1315–1320. [Google Scholar] [CrossRef]
- Anjum, S.; Sahar, T.; Nigam, A.; Wajid, S. Transcriptome Analysis of mRNA in Uterine Leiomyoma Using Next-generation RNA Sequencing. Anticancer. Agents Med. Chem. 2019, 19, 1703–1718. [Google Scholar] [CrossRef]
- Wang, T.; Zhang, X.; Obijuru, L.; Laser, J.; Aris, V.; Lee, P.; Mittal, K.; Soteropoulos, P.; Wei, J.J. A micro-RNA signature associated with race, tumor size, and target gene activity in human uterine leiomyomas. Genes Chromosomes Cancer 2007, 46, 336–347. [Google Scholar] [CrossRef] [PubMed]
- George, J.W.; Fan, H.; Johnson, B.; Carpenter, T.J.; Foy, K.K.; Chatterjee, A.; Patterson, A.L.; Koeman, J.; Adams, M.; Madaj, Z.B.; et al. Integrated Epigenome, Exome, and Transcriptome Analyzes Reveal Molecular Subtypes and Homeotic Transformation in Uterine Fibroids. Cell Rep. 2019, 29, 4069–4085. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Q.; Kanis, M.J.; Ubago, J.; Liu, D.; Scholtens, D.M.; Strohl, A.E.; Lurain, J.R.; Shahabi, S.; Kong, B.; Wei, J.J. The selected biomarker analysis in 5 types of uterine smooth muscle tumors. Hum. Pathol. 2018, 76, 17–27. [Google Scholar] [CrossRef] [PubMed]
- Ho, Y.; Yang, Y.C.; Chin, Y.T.; Chou, S.Y.; Chen, Y.R.; Shih, Y.J.; Whang-Peng, J.; Changou, C.A.; Liu, H.L.; Lin, S.J.; et al. Resveratrol inhibits human leiomyoma cell proliferation via crosstalk between integrin αvβ3 and IGF-1R. Food Chem. Toxicol. 2018, 120, 346–355. [Google Scholar] [CrossRef] [PubMed]
- Cha, P.C.; Takahashi, A.; Hosono, N.; Low, S.K.; Kamatani, N.; Kubo, M.; Nakamura, Y. A genome-wide association study identifies three loci associated with susceptibility to uterine fibroids. Nat. Genet. 2011, 43, 447–450. [Google Scholar] [CrossRef] [PubMed]
- Eggert, S.L.; Huyck, K.L.; Somasundaram, P.; Kavalla, R.; Stewart, E.A.; Lu, A.T.; Painter, J.N.; Montgomery, G.W.; Medland, S.E.; Nyholt, D.R.; et al. Genome-wide linkage and association analyses implicate FASN in predisposition to Uterine Leiomyomata. Am. J. Hum. Genet. 2012, 91, 621–628. [Google Scholar] [CrossRef] [PubMed]
- Hellwege, J.N.; Jeff, J.M.; Wise, L.A.; Gallagher, C.S.; Wellons, M.; Hartmann, K.E.; Jones, S.F.; Torstenson, E.S.; Dickinson, S.; Ruiz-Narváez, E.A.; et al. A multi-stage genome-wide association study of uterine fibroids in African Americans. Hum. Genet. 2017, 136, 1363–1373. [Google Scholar] [CrossRef]
- Välimäki, N.; Kuisma, H.; Pasanen, A.; Heikinheimo, O.; Sjöberg, J.; Bützow, R.; Sarvilinna, N.; Heinonen, H.R.; Tolvanen, J.; Bramante, S.; et al. Genetic predisposition to uterine leiomyoma is determined by loci for genitourinary development and genome stability. eLife 2018, 7, e37110. [Google Scholar] [CrossRef]
- Rafnar, T.; Gunnarsson, B.; Stefansson, O.A.; Sulem, P.; Ingason, A.; Frigge, M.L.; Stefansdottir, L.; Sigurdsson, J.K.; Tragante, V.; Steinthorsdottir, V.; et al. Variants associating with uterine leiomyoma highlight genetic background shared by various cancers and hormone-related traits. Nat. Commun. 2018, 9, 3636. [Google Scholar] [CrossRef]
- Gallagher, C.S.; Mäkinen, N.; Harris, H.R.; Rahmioglu, N.; Uimari, O.; Cook, J.P.; Shigesi, N.; Ferreira, T.; Velez-Edwards, D.R.; Edwards, T.L.; et al. Genome-wide association and epidemiological analyses reveal common genetic origins between uterine leiomyomata and endometriosis. Nat. Commun. 2019, 10, 4857. [Google Scholar] [CrossRef]
- Walker, K.J.; Nicholson, R.I.; Turkes, A.O.; Turkes, A.; Griffiths, K.; Robinson, M.; Crispin, Z.; Dris, S. Therapeutic potential of the LHRH agonist, ICI 118630, in the treatment of advanced prostatic carcinoma. Lancet 1983, 2, 413–415. [Google Scholar] [CrossRef] [PubMed]
- Hinterhuber, G.; Cauza, K.; Dingelmaier-Hovorka, R.; Diem, E.; Horvat, R.; Wolff, K.; Foedinger, D. Expression of RPE65, a putative receptor for plasma retinol-binding protein, in nonmelanocytic skin tumours. Br. J. Dermatol. 2005, 153, 785–789. [Google Scholar] [CrossRef]
- Peruzzi, D.; Mori, F.; Conforti, A.; Lazzaro, D.; De Rinaldis, E.; Ciliberto, G.; La Monica, N.; Aurisicchio, L. MMP11: A novel target antigen for cancer immunotherapy. Clin. Cancer Res. 2009, 15, 4104–4113. [Google Scholar] [CrossRef]
- Yin, P.; Lin, Z.; Reierstad, S.; Wu, J.; Ishikawa, H.; Marsh, E.E.; Innes, J.; Cheng, Y.; Pearson, K.; Coon, J.S., 5th; et al. Transcription factor KLF11 integrates progesterone receptor signaling and proliferation in uterine leiomyoma cells. Cancer Res. 2010, 70, 1722–1730. [Google Scholar] [CrossRef]
- Zheng, Y.; Tabbaa, Z.M.; Khan, Z.; Schoolmeester, J.K.; El-Nashar, S.; Famuyide, A.; Keeney, G.L.; Daftary, G.S. Epigenetic regulation of uterine biology by transcription factor KLF11 via posttranslational histone deacetylation of cytochrome p450 metabolic enzymes. Endocrinology 2014, 155, 4507–4520. [Google Scholar] [CrossRef] [PubMed]
- Grigorkevich, O.S.; Mokrov, G.V.; Kosova, L.Y. Matrix metalloproteinases and their inhibitors. Pharmacokinet. Pharmacodyn. 2019, 2, 3–16. [Google Scholar]
- Cao, T.; Jiang, Y.; Wang, Z.; Zhang, N.; Al-Hendy, A.; Mamillapalli, R.; Kallen, A.N.; Kodaman, P.; Taylor, H.S.; Li, D.; et al. H19 lncRNA has been identified as a master regulator of genes that control uterine leiomyomas. Oncogene 2019, 38, 5356–5366. [Google Scholar] [CrossRef]
- Bellinato, F.; Gisondi, P.; Girolomoni, G. Latest Advances for the Treatment of Chronic Plaque Psoriasis with Biologics and Oral Small Molecules. Biologics 2021, 15, 247–253. [Google Scholar] [CrossRef]
- Schubert, G.; Elger, W.; Kaufmann, G.; Schneider, B.; Reddersen, G.; Chwalisz, K. Discovery, Chemistry, and Reproductive Pharmacology of Asoprisnil and Related 11β-Benzaldoxime Substituted Selective Progesterone Receptor Modulators (SPRMs). Semin. Reprod. Med. 2005, 23, 58–73. [Google Scholar] [CrossRef]
Year of Publication, First Author | Reference | Key Results |
---|---|---|
Direction 1: HMGA2 gene overexpression and chromosomal rearrangements in UL | ||
Nilbert et al., 1988 | [19] | A total of 106 samples of leiomyoma biopsies were examined using classical cytogenetics methods. A normal parental karyotype was found in 57 samples (54%) capable of growing in culture, and chromosomal rearrangements were found in 20 samples (19%). In 10 cases (9%), they represented a translocation of the prethelomeric regions of chromosomes 12 and 14, and in 4 of them, there were other chromosomal rearrangements. In 10 cases where there was no translocation of the thelomeric region of chromosomes 12 and 14, minor rearrangements were observed in chromosomes 1, 2, 3, 4, 6, 8, 9, 10, 11, 13, and 19. Most often—in five cases, they affected chromosome 1. |
Nilbert et al., 1992 | [20] | It was established that with a variety of different combinations, only rearrangements of type t (1; 6) (q23;p21) and del (7) (q21.2q31.2) were systematically detected in independent samples and sufficient for the formation of a fibroid along with the most common rearrangements of type t (12; 14) (q14 - q15; q23 - q24). |
Nilbert et al., 1990 | [21] | Fibroids with complete trisomies on chromosome 12, with specific translocations t (12; 14) (q14 - 15; q23 - 24), were identified. |
Hennig et al., 1997 | [22] | It was discovered for the first time that the rearrangement involving the 12q14 - 15 prethelomeric region leads to an increase in the expression of the HMGIC gene, which later became known as HMGA2. |
Klotzbücher et al., 1999 | [23] | The expression of HMGIC (HMGA2) and HMGIY (HMGA1) genes in leiomyomas was studied using immunohistochemical staining of tissue sections. These authors reported that their expression of the genes of these non-histone proteins controlling the chromatin structure was observed in 16 of 33 samples of biopsies of the fibroids. At the same time, expression has never been observed in normal myometrium, as well as vascular endothelium and fibroblasts from the tumors. In 3 of the 16 biopsy samples showing HMGIC (HMGA2) expression, a protein product of this gene with an abnormal molecular weight was observed. |
Klemke et al., 2009 | [24] | The level of expression of the HMGA2 gene was studied on a sufficient panel of samples from 180 patients. The highest levels of HMGA2 expression were indeed observed in samples with rearrangements affecting the 12q14 - 15 region. But overexpression of HMGA2 was repeatedly found in leiomyoma samples without such aberrations, although at lower levels. |
Klemke et al., 2010 | [25] | It was found that uterine leiomyomas are characterized by chromosomal rearrangements in the 12q14 - q15 region, leading to overexpression of the HMGA2 gene. Recent studies have identified microRNAs of the let-7 family as post-transcriptional silencers of HMGA2 expression. Chromosomal rearrangements sometimes lead to the appearance of shortened or hybrid HMGA2 transcripts that lack 3’-UTR. The aim of the study was to use real-time RT-PCR to test how rearrangements of chromosomal region 12q14, leading to the appearance of shortened HMGA2 transcripts in the fibroids, affect the stability of mRNA. The presented results prove that chromosomal rearrangements involving the HMGA2 locus often lead to an increase in the mRNA lifetime, which contributes to overexpression. |
Markowski 2010 | [26] | Hormonal dependence of leiomyoma has long been known, and antitumor cytostatics are actively used in practice to suppress the growth of malignant neoplasms. The desire to use the PI3K/Akt/mTOR pathway as a therapeutic target is due to the fact that its shutdown does not cause immediate cell death, but leads to the so-called phenomenon of oncogen-induced aging, when cells under the action of proteins p16, p19, p53, and p21 gradually lose their division potential, weaken the antioxidant defense system, which weakens tumor aggressiveness with a relatively low level of side toxicity. Therefore, a balance between HMGA2 and the p19Arf-TP53-CDKN1A axis was found to be essential for the growth of uterine leiomyomas. |
Schoenmakers 2013 | [27] | It is reported that repeated genomic rearrangements: del (7) (q22), t (12; 14) (q15; q24), t (1;2) (p36; p24), transpositions involving regions 6p21 and/or 10q22 occur in about 40% of the fibroids. These authors claim that in their previous works, they identified the genes HMGA1, HMGA2, RAD51L1, MORF, and NCOA1 as the primary targets of chromosomal rearrangements that cause the appearance of a benign tumor in each of the four variants of genome rearrangement using remote PCR methods. |
Holzmann et al., 2014 | [28] | The work reports that in the foci of leiomyomas that do not have somatic mutations in the MED12 gene, chromotripsis phenomena were observed: numerous duplications and deletions of small segments were grouped mainly into five chromosomal regions: 2p14 - 2pter, 2q33.1 - 2q37.3, 5q31.3 - 5qter, 11q14.1 - 11qter and 18p11.21 - 18q2.3. Due to the small size of the rearranged fragments of genomic DNA, such rearrangements can hardly be detected by methods of classical cytogenetics. Histologically, the fibroids with chromotrypsin, as a rule, represent a cellular leiomyoma with pronounced hyperproduction of hyaluronic acid. The results of the work show that leiomyomas with a normal karyotype and without somatic mutations in the MED12 gene are a heterogeneous group of diseases characterized by chromotripsis (“firestorm”), which does not affect the sites of chromosomal rearrangements characteristic of leiomyoma, such as 12q14 - q15 and 6p21. |
Pradhan et al., 2016 | [29] | The results of using the remote reverse PCR method for the detection and screening of de novo DNA rearrangements in uterine leiomyomas are reported. The method used makes it possible to identify genome rearrangements in the leiomyoma in comparison to the normal parental myometrium without putting forward an initial hypothesis about the location of recombination points. The screening of uterine leiomyoma samples for the presence of rearrangements in genomic locations allowed them to establish that the most susceptible to rearrangements of the genome in this type of tumor are located above the coding region of the HMGA2 gene and inside the RAD51B gene. In particular, a previously undescribed point of genomic rearrangement above the HMGA2 gene was identified, which went unnoticed in a previous study performed by genome-wide sequencing, where 30 samples of uterine leiomyoma showed no rearrangements within 1107 bp and 1996 bp analyzed in the RAD51B and HMGA2 rearrangement hotspots. |
Direction 2: Mutations in MED12 | ||
Mäkinen et al. 2017 | [30] | The authors of this work were the first to express the opinion that somatic mutations in the MED12 gene, biallelic inactivation of the fumarate hydratase gene, and chromosomal aberrations leading to the overexpression of the HMGA2 gene correspond to three mutually exclusive mechanisms of leiomyoma formation. |
Wu et al., 2017 | [31] | The work is devoted to elucidating the biological features of the two most common subtypes of uterine leiomyoma, mutant by MED12 (MED12-LM) and overexpressing HMGA2 (HMGA2-LM) uterine leiomyomas. Since each tumor carries only one genetic change, both subtypes are considered monoclonal. Approximately 90% of the cells in the HMGA2 uterine leiomyoma were smooth muscle cells with overexpression of HMGA2. In contrast, MED12-LM consisted of the same number of smooth muscle cells and tumor-associated fibroblasts (TAFs). The TAFs did not carry mutations in MED12, which suggests an interaction between smooth muscle cells and fibroblasts, with different origins during the formation and growth of the fibroid. |
Yin et al., 2015 | [32] | Human uterine leiomyoma stem/progenitor cells expressing CD34 and CD49b initiate tumors in vivo. |
Mas et al., 2015 | [33] | Stro-1/CD44 as putative human myometrial and fibroid stem cell markers |
Mu et al., 2016 | [34] | IGF-1 and VEGF can be used as prognostic indicators for patients with uterine fibroids treated with uterine artery embolization. |
Heikkinen et al., 2018 | [35] | It was reported that there is a positive correlation between the increased expression of the COL3A1 gene in the fibroids and the expression of the HOXA13 gene, which is a regulator of the development of the organs of the female reproductive system, in particular, the cervix and vagina. According to these studies, a statistically significant increase in the expression of the HOXA13 gene above the level characteristic of normal myometrium was observed in both MED12-dependent and HMGA2-dependent fibroids. |
Reis et al., 2016 | [36] | Overexpression of COL4A1 and COL4A2 collagens in MED12-positive fibroids was detected. |
Direction 3: Null-mutations in FH gene | ||
Sulkowski 2018 | [37] | Krebs-cycle-deficient hereditary cancer syndromes are defined by defects in homologous recombination DNA repair |
Mäkinen 2017 | [30] | The authors of this work were the first to express the opinion that somatic mutations in the MED12 gene, biallelic inactivation of the fumarate hydratase gene, and chromosomal aberrations leading to the overexpression of the HMGA2 gene correspond to three mutually exclusive mechanisms of leiomyoma formation. |
Direction 4: Agonists and antagonists of steroid hormones | ||
Gonzalez-Barcena et al., 1997 | [38] | Treatment of uterine leiomyomas with luteinizing hormone-releasing hormone antagonist cetrorelix. |
Nowicki et al., 2002 | [39] | The influence of luteinizing hormone-releasing hormone analog on serum leptin and body composition in women with solitary uterine myoma. |
Engman et al., 2009 | [40] | Mifepristone for the treatment of uterine leiomyoma. A prospective randomized placebo-controlled trial. |
Bouchard et al., 2011 | [41] | Selective progesterone receptor modulators in reproductive medicine: pharmacology, clinical efficacy, and safety. |
Malik et al., 2016 | [42] | Gonadotropin-releasing hormone analogs inhibit leiomyoma extracellular matrix despite presence of gonadal hormones. |
Patel et al., 2016 | [43] | Mifepristone inhibits extracellular matrix formation in uterine leiomyoma. |
Islam et al., 2021 | [44] | Extracellular matrix and Hippo signaling as therapeutic targets of antifibrotic compounds for uterine fibroids. |
Dababou et al., 2021 | [45] | Linzagolix: a new GnRH antagonist under investigation for the treatment of endometriosis and uterine myomas. |
Middelkoop et al., 2022 | [46] | Evaluation of marketing authorization and clinical implementation of ulipristal acetate for uterine fibroids. |
Arjona et al., 2022 | [47] | The development of relugolix combination therapy as a medical treatment option for women with uterine fibroids or endometriosis is described. |
Salas et al., 2022 | [48] | New local ganirelix sustained release therapy for uterine leiomyoma. Evaluation in a preclinical organ model. |
Chwalisz 2023 | [49] | Clinical development of the oral gonadotropin-releasing hormone antagonist elagolix. |
Direction 5: PI3K/Akt/mTOR and other intracellular signal pathway in UL | ||
Hu et al., 2009 | [50] | Blockade of Wnt signaling inhibits angiogenesis and tumor growth in hepatocellular carcinoma. |
Xu et al., 2014 | [51] | It is hypothesized that the inhibition of AKT leads to the short-term triggering of specific mechanisms, which ultimately lead cells to cellular aging or death by the mechanism of apoptosis. It was experimentally shown that the inhibition of AKT leads to the accelerated aging of culture cells. The treatment of MK-2206 cells with an allosteric AKT inhibitor increased the content of reactive oxygen species, the level of miR-182 microRNA production, and the transcripts of several genes that are considered markers of ROS: p16, p53, p21, and β-galactosidase. The induction of ROS was associated with the hyperproduction of HMGA2, which was colocalized in the aging-related regions of heterochromatin. |
Ye et al., 2014 | [52] | Small-molecule inhibitors targeting activator protein 1 (AP-1). |
Galindo LJ et al., 2018 | [53] | Comparative analysis of AKT and the related biomarkers in uterine leiomyomas with MED12, HMGA2, and FH mutations. |
Xie et al., 2018 | [54] | The work is devoted to the study of the AKT signaling pathway and the mechanism of OIS in leiomyoma cells containing various driver mutations: MED12 mutations (n = 25), HMGA2 overexpression (n = 15), and biallelic inactivation of FH (n = 27). In each sample, the expression of genes involved in the response to sex steroids, the cell cycle, and the AKT pathway was studied by immunohistochemical method. It was found that the ER and PR genes were well expressed in all types of leiomyoma except for the FH-dependent type, which showed low ER expression and increased PR expression. HMGA2-dependent-type samples had significantly higher levels of AKT signaling and mitogenic activity than other types of the fibroids. HMGA2 activated AKT signaling by enhancing IGF2BP2 expression. The suppression of HER2 expression in leiomyoma cells led to a decrease in AKT activity and an increase in the expression of p16 and p21, which ultimately caused oncogen-induced cell aging. |
Alzahrani et al., 2019 | [55] | The application of PI3K/Akt/mTOR inhibitors in cancer is described. |
Pilgrim et al., 2020 | [56] | Characterization of the role of activator protein 1 signaling pathway on extracellular matrix deposition in uterine leiomyoma. |
Year of Publication, First Author | Reference | Key Results |
---|---|---|
Direction 1: GWAS for mapping polymorphisms associated with predisposition to UL onset | ||
Cha et al., 2011 | [68] | GWAS identifies three loci associated with susceptibility to uterine fibroids. |
Eggert et al. 2012 | [69] | Genome-wide linkage and association analyses implicate FASN in predisposition to uterine leiomyomata. |
Hellwege et al. 2017 | [70] | A multi-stage genome-wide association study of uterine fibroids in African Americans. |
Välimäki et al., 2018 | [71] | These authors used genome-wide association analysis (GWAS) to identify genetic variants that are more common in people with fibroids. Using data from the British Biobank, the genomes of more than 15,000 women with fibroids were analyzed, which were compared with a control group of more than 392,000 individuals. The analysis revealed 22 regions of the genome, the genotypes of which differed in the experimental and control groups. These regions included genes that may well contribute to the development of fibroids, such as the TP53 gene, which affects the stability of the genome, and ESR1, which encodes the estrogen receptor (it is well-known that this hormone plays an important role in stimulating the growth of fibroids). Differences in genotypes were revealed for known genes involved in the control of the development of female genital organs. |
Rafnar et al. 2018 | [72] | Variants associated with uterine leiomyoma highlight genetic background shared among various cancers and hormone-related traits. |
Edwards et al., 2019 | [14] | Trans-Ethnic Genome-Wide Association Study of Uterine Fibroids |
Gallagher et al., 2019 | [73] | Genome-wide association and epidemiological analyses reveal common genetic origins between uterine leiomyomata and endometriosis. |
Direction 2: Genome-wide transcriptome and DNA-methylome studies | ||
Wang et al., 2007 | [64] | Paired samples of leiomyomas and normal myometrium from 41 patients were examined, which were used to construct banks of micro-RNA and their subsequent sequencing. As a result of bioinformatic analysis, 45 microRNAs with significantly increased or decreased content in the fibroids compared to the corresponding myometrium were identified (p < 0.001). The five undergoing the strongest expression change are the let-7 family: miR-21, miR-23b, miR29b, and miR-197. |
Navarro et al., 2012 | [61] | The objective of the work was to identify abnormally methylated sections of the genome in UL cells in vivo using genome-wide analysis methods and to compare the data obtained with the results of the metatranscriptome analysis. Biological materials in the form of paired samples of leiomyomas and adjacent normal myometrium were selected from 18 patients of African-American origin. A total of 55 genes with differential methylation of the promoter regions were identified, which correlated with differences in the level of expression in uterine leiomyoma compared to normal myometrium. Additionally, 80% of the identified genes showed an inverse relationship between the status of DNA methylation and the content of the corresponding mRNA in uterine leiomyoma tissues, including 34 genes which demonstrated hypermethylation of the promoter region and a corresponding decrease in expression level, and 10 genes which demonstrated demethylation and an increase in expression level. |
Mehine et al., 2016 | [62] | The data of a meta-transcriptomic study of the expression profile of leiomyomas with four types of driver mutations in comparison to the expression profile of the adjacent normal myometrium, are presented. A total of 19 upregulated and a single downregulated markers of leiomyomata (in comparison to the normal myometrium) are reported. |
Anjum et al., 2019 | [63] | The levels of expression of marker transcripts in UL were measured using meta-transcriptomic analysis on the Illumina platform, followed by validation of the most significant results using real-time RT-PCR. A full transcriptome analysis showed an increase in the expression of 128 genes in the fibroids compared to normal myometrium and a decrease in the expressions of 98 genes. |
George et al., 2019 | [65] | Integrated epigenome, exome, and transcriptome analyses were carried out for revealing the molecular subtypes of fibroids. Cases of HER2 overexpression-independent chromosomal rearrangement were found. Hypomethylation of the structural region of the HMGA2 gene was found to be a reason for the overexpression. It was found that MED12 mutations and increased HMGA2 expression can coexist in the same fibroids. Increased expression of insulin receptor substrate undergoing phosphorylation when binding insulin to the receptor (IRS4) in leiomyoma cells compared to myometrium was found. |
Maekawa et al., 2022 | [57] | Differences in the DNA methylome, transcriptome, and histological features in uterine fibroids with and without MED12 mutations were studied. Genes of inflammatory response (CCL2, AOX1, ACKR1), apoptosis (ANXA1, CITED2), and metabolism associated with reactive oxygen species were reported to undergo hypermethylation in all types of leiomyoma cells. It was found that 80–90% of the fibroids of all types showed overexpression of the SATB2 and NRG1 genes compared to the myometrium, and the excess level varied from 1.5 to 20–30 times. |
Direction 3: Candidate medicines for UL treatment based on small molecules, vaccines, and RNA | ||
Walker et al., 1983 | [74] | Therapeutic potential of the LHRH agonist, ICI 118630, in the treatment of advanced prostatic carcinoma |
Hinterhuber et al., 2005 | [75] | Expression of RPE65, a putative receptor for plasma retinol-binding protein, in nonmelanocytic skin tumours |
Peruzzi et al., 2009 | [76] | MMP11 is described as a novel target antigen for cancer immunotherapy. |
Yin et al., 2010 | [77] | Transcription factor KLF11 integrates progesterone receptor signaling and proliferation in uterine leiomyoma cells |
Zheng et al., 2014 | [78] | Epigenetic regulation of uterine biology by transcription factor KLF11 via posttranslational histone deacetylation of cytochrome p450 metabolic enzymes |
Ho et al., 2018 | [67] | Resveratrol inhibits human leiomyoma cell proliferation via crosstalk between integrin avß3 and IGF-1R |
Grigorkevich et al., 2019 | [79] | Matrix metalloproteinases and their inhibitors |
Cao et al., 2019 | [80] | H19 lncRNA has been identified as a master regulator of genes that control uterine leiomyomas |
Nosological Type of the Tumor | Chemical Substances | Humanized Monoclonal Antibodies | Vaccines | RNA Delivery |
---|---|---|---|---|
MED12-dependent | ADAM12, MMP11, MMP16, KCNAB3, CACNA1C, RAD51B | ADAM12, MMP11, MMP16, RUNDC1 | ADAM12, MMP11, MMP16, RUNDC1, RAD51B | miR-200c, miR-93 |
HMGA2-dependent | PAPPA2, MB21D2, | GRPR, PLAG1, PAPPA2, MB21D2 | HMGA2, PLAG1, PAPPA2, MB21D2 | miR-21, miR-23b, miR-29b, miR-197, mir-106b |
FH-dependent | PARP (olaparib *, niraparib *), TNFRSF21, NQO1, SLC7A11, FAM46C, ABCC3 | - | - | - |
All types together | Resveratrol **, RPE65: adapalene **, tretinoin **, isotretinoin **, and tazarotene ** | IL17 *, WIF1, SFRP1, SATB2, NRG1, PCP4, CHRDL2, MFAP2 | PCP4, CHRDL2, MFAP2 | Anti-H19, miR-182 |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Kuznetsova, M.V.; Tonoyan, N.M.; Trubnikova, E.V.; Zelensky, D.V.; Svirepova, K.A.; Adamyan, L.V.; Trofimov, D.Y.; Sukhikh, G.T. Novel Approaches to Possible Targeted Therapies and Prophylaxis of Uterine Fibroids. Diseases 2023, 11, 156. https://doi.org/10.3390/diseases11040156
Kuznetsova MV, Tonoyan NM, Trubnikova EV, Zelensky DV, Svirepova KA, Adamyan LV, Trofimov DY, Sukhikh GT. Novel Approaches to Possible Targeted Therapies and Prophylaxis of Uterine Fibroids. Diseases. 2023; 11(4):156. https://doi.org/10.3390/diseases11040156
Chicago/Turabian StyleKuznetsova, Maria V., Narine M. Tonoyan, Elena V. Trubnikova, Dmitry V. Zelensky, Ksenia A. Svirepova, Leila V. Adamyan, Dmitry Y. Trofimov, and Gennady T. Sukhikh. 2023. "Novel Approaches to Possible Targeted Therapies and Prophylaxis of Uterine Fibroids" Diseases 11, no. 4: 156. https://doi.org/10.3390/diseases11040156
APA StyleKuznetsova, M. V., Tonoyan, N. M., Trubnikova, E. V., Zelensky, D. V., Svirepova, K. A., Adamyan, L. V., Trofimov, D. Y., & Sukhikh, G. T. (2023). Novel Approaches to Possible Targeted Therapies and Prophylaxis of Uterine Fibroids. Diseases, 11(4), 156. https://doi.org/10.3390/diseases11040156