Antiprogestins reduce epigenetic field cancerization in breast tissue of young healthy women

Thomas E Bartlett, Iona Evans, Allison Jones, James E Barrett, Shaun Haran, Daniel Reisel, Kiriaki Papaikonomou, Louise Jones, Chiara Herzog, Nora Pashayan, Bruno M Simões, Robert B Clarke, D Gareth Evans, Talayeh S Ghezelayagh, Sakthivignesh Ponandai-Srinivasan, Nageswara R Boggavarapu, Parameswaran G Lalitkumar, Sacha J Howell, Rosa Ana Risques, Angelique Flöter Rådestad, Louis Dubeau, Kristina Gemzell-Danielsson, Martin Widschwendter, Thomas E Bartlett, Iona Evans, Allison Jones, James E Barrett, Shaun Haran, Daniel Reisel, Kiriaki Papaikonomou, Louise Jones, Chiara Herzog, Nora Pashayan, Bruno M Simões, Robert B Clarke, D Gareth Evans, Talayeh S Ghezelayagh, Sakthivignesh Ponandai-Srinivasan, Nageswara R Boggavarapu, Parameswaran G Lalitkumar, Sacha J Howell, Rosa Ana Risques, Angelique Flöter Rådestad, Louis Dubeau, Kristina Gemzell-Danielsson, Martin Widschwendter

Abstract

Background: Breast cancer is a leading cause of death in premenopausal women. Progesterone drives expansion of luminal progenitor cells, leading to the development of poor-prognostic breast cancers. However, it is not known if antagonising progesterone can prevent breast cancers in humans. We suggest that targeting progesterone signalling could be a means of reducing features which are known to promote breast cancer formation.

Methods: In healthy premenopausal women with and without a BRCA mutation we studied (i) estrogen and progesterone levels in saliva over an entire menstrual cycle (n = 20); (ii) cancer-free normal breast-tissue from a control population who had no family or personal history of breast cancer and equivalently from BRCA1/2 mutation carriers (n = 28); triple negative breast cancer (TNBC) biopsies and healthy breast tissue taken from sites surrounding the TNBC in the same individuals (n = 14); and biopsies of ER+ve/PR+ve stage T1-T2 cancers and healthy breast tissue taken from sites surrounding the cancer in the same individuals (n = 31); and (iii) DNA methylation and DNA mutations in normal breast tissue (before and after treatment) from clinical trials that assessed the potential preventative effects of vitamins and antiprogestins (mifepristone and ulipristal acetate; n = 44).

Results: Daily levels of progesterone were higher throughout the menstrual cycle of BRCA1/2 mutation carriers, raising the prospect of targeting progesterone signalling as a means of cancer risk reduction in this population. Furthermore, breast field cancerization DNA methylation signatures reflective of (i) the mitotic age of normal breast epithelium and (ii) the proportion of luminal progenitor cells were increased in breast cancers, indicating that luminal progenitor cells with elevated replicative age are more prone to malignant transformation. The progesterone receptor antagonist mifepristone reduced both the mitotic age and the proportion of luminal progenitor cells in normal breast tissue of all control women and in 64% of BRCA1/2 mutation carriers. These findings were validated by an alternate progesterone receptor antagonist, ulipristal acetate, which yielded similar results. Importantly, mifepristone reduced both the TP53 mutation frequency as well as the number of TP53 mutations in mitotic-age-responders.

Conclusions: These data support the potential usage of antiprogestins for primary prevention of poor-prognostic breast cancers.

Trial registration: Clinical trial 1 Mifepristone treatment prior to insertion of a levonorgestrel releasing intrauterine system for improved bleeding control - a randomized controlled trial, clinicaltrialsregister.eu, 2009-009014-40 ; registered on 20 July 2009. Clinical trial 2 The effect of a progesterone receptor modulator on breast tissue in women with BRCA1 and 2 mutations, clinicaltrials.gov, NCT01898312 ; registered on 07 May 2013. Clinical trial 3 A pilot prevention study of the effects of the anti- progestin Ulipristal Acetate (UA) on surrogate markers of breast cancer risk, clinicaltrialsregister.eu, 2015-001587-19 ; registered on 15 July 2015.

Keywords: Antiprogestins; BRCA1; Breast cancer; DNA methylation; Epigenetics; Intermediate surrogate marker; Prevention.

Conflict of interest statement

RAR owns equity and serves as a scientific consultant to TwinStrand Biosciences Inc. UCLB (UCL's commercialisation company) has filed a patent on some aspects described in the paper – MW is named as inventor on this patent. JEB, CH and MW are shareholders of Sola Diagnostics GmbH, which holds an exclusive licence to the intellectual property described in this patent file. The remaining authors declare that they have no competing interests.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Summary of the rationale and design of the study. Abbreviations: mut (mutation); wt (wild-type); pcgt (polycomb-group targets); NB (normal breast); CO (control); BB (breast biopsy); BC (breast cancer)
Fig. 2
Fig. 2
Effect of the BRCA1/2 mutation carrier states on estradiol and progesterone levels during menstrual cycle progression. Estradiol and progesterone hormone levels were significantly greater over one cycle in BRCA1/2 mutation carriers (n = 12) than in controls (n = 8): a and b show the time-series (respectively) for estradiol and progesterone (normalised by cycle length) with one-week moving average lines. c One-week moving windows (as used to plot the moving average lines in a and b) were used to assess how the relative increase in hormone levels in BRCA1/2 mutation carriers (compared to controls) varies during the cycle. All significances were assessed with t-tests
Fig. 3
Fig. 3
WID-Breast29 score in triple negative breast cancer (TNBC) and adjacent normal tissues. a The WID-Breast29 score is based on 37 CpGs from 29 genes and b was significantly greater in normal breast tissue surrounding TNBC (n = 14) when compared to normal tissue from cancer-free women (n = 14); significance was assessed with the t-test. c The WID-Breast29 score also increased significantly when transitioning from normal surrounding tissue to TNBC (n = 14) or d ER+ve/PR+ve breast cancer (n = 31); gold lines indicate increasing and blue decreasing values from surround normal to the cancer tissue in individual breast cancer patients; significance was assessed with the paired-sample t-test. e The WID-Breast29 score was also significantly associated with patient survival outcome in 257 TCGA breast cancer samples (after adjusting for covariates); significance was assessed by z-tests on the Wald statistics after fitting a multivariate Cox proportional hazards model
Fig. 4
Fig. 4
Assessment of breast epithelium composition in TNBC. a Fractional composition of breast-tissue samples was assessed using a well-validated algorithm [37], based on our custom-designed DNAme reference profiles for breast epithelial cell subtypes (Materials and Methods). b There was a highly significant increase in luminal progenitor cell concentration in TNBC (compared to normal surrounding tissue in the same volunteers, n = 14), whereas c the mature luminal cell proportion was unchanged and the d basal cell fraction decreased. e Comparing normal tissues in the same volunteers (n = 31) to ER+ve/PR+ve breast cancers, there was a decrease in luminal progenitors and f a highly significant increase in mature luminal cells and g no changes were noted in basal cells; gold lines indicate increasing and blue decreasing values from surround normal to the cancer tissue in individual breast cancer patients (b-g). h RANKL and i RANK expression (respectively) were significantly correlated with mature luminal and luminal progenitor cell proportion in 38 healthy breast tissue samples from TCGA; j the mean of the normalised RANKL and RANK expression levels was significantly correlated with the WID-Breast29 index in the same 38 samples. Significances in b–g were assessed with the paired-sample t-test, and those in h–j were assessed with Pearson’s correlation test. F, Fat cells; I, Immune cells; E, Epithelial cells; S, Stromal cells; LP, Luminal Progenitors; LM, Luminal Mature; B, Basal
Fig. 5
Fig. 5
Effects of vitamins (placebo/comparator), mifepristone and ulipristal acetate treatment on intermediate cancer surrogate endpoints in normal breast tissue from healthy women. The impact of Vitamin (ac), Mifepristone (df) and Ulipristal acetate (hj) on the mitotic age index WID-Breast29 (a, d, h), luminal progenitor cell fraction (b, e, i) and basal cell fraction (c, f, j) in healthy, unaffected women at lower and higher breast cancer risk. g RANKL mRNA expression before and after treatment with mifepristone (as assessed by real-time PCR). k Correlation of the change in WID-Breast29 index with the change in breast epithelial subtype fractions in all samples in a–f, h–j. All significances in a–j were assessed with the paired-sample t-test. Significance in k was assessed with Pearson’s correlation test. Gold lines indicate increasing and blue lines decreasing values from the breast biopsy taken before to biopsy taken after treatment
Fig. 6
Fig. 6
Mifepristone treatment and the dynamics of TP53 mutations in normal breast tissue and suggested model for the prevention of triple negative breast cancer. aTP53 mutation frequency and bTP53 mutation count in normal breast tissue before and after mifepristone exposure in women who showed a decrease of the WID-Breast29 index after mifepristone (responders) and who did not show a decrease (non-responders); significance was assessed with the two-sided t-test. c Progesterone triggers release of RANKL in hormone receptor (HR) positive mature luminal cells leading to increased proliferation and thus accelerated mitotic ageing in HR negative luminal progenitor cells resulting in increased cancer risk; these effects are reduced after treatment with the progesterone antagonist mifepristone. HR, hormone receptor

References

    1. Husemann Y, Geigl JB, Schubert F, Musiani P, Meyer M, Burghart E, Forni G, Eils R, Fehm T, Riethmuller G, et al. Systemic spread is an early step in breast cancer. Cancer Cell. 2008;13(1):58–68. doi: 10.1016/j.ccr.2007.12.003.
    1. Hosseini H, Obradovic MMS, Hoffmann M, Harper KL, Sosa MS, Werner-Klein M, Nanduri LK, Werno C, Ehrl C, Maneck M, et al. Early dissemination seeds metastasis in breast cancer. Nature. 2016;540(7634):552–558. doi: 10.1038/nature20785.
    1. Sporn MB, Liby KT. Cancer chemoprevention: scientific promise, clinical uncertainty. Nat Clin Pract Oncol. 2005;2(10):518–525. doi: 10.1038/ncponc0319.
    1. Schatzkin A, Gail M. The promise and peril of surrogate end points in cancer research. Nat Rev Cancer. 2002;2(1):19–27. doi: 10.1038/nrc702.
    1. Harbeck N, Gnant M. Breast cancer. Lancet. 2017;389(10074):1134–1150. doi: 10.1016/S0140-6736(16)31891-8.
    1. Pashayan N, Antoniou AC, Ivanus U, Esserman LJ, Easton DF, French D, Sroczynski G, Hall P, Cuzick J, Evans DG, et al. Personalized early detection and prevention of breast cancer: ENVISION consensus statement. Nat Rev Clin Oncol. 2020;17:687–705. doi: 10.1038/s41571-020-0388-9.
    1. Kuchenbaecker KB, McGuffog L, Barrowdale D, Lee A, Soucy P, Dennis J, Domchek SM, Robson M, Spurdle AB, Ramus SJ, et al. Evaluation of Polygenic Risk Scores for Breast and Ovarian Cancer Risk Prediction in BRCA1 and BRCA2 Mutation Carriers. J Natl Cancer Inst. 2017;109(7):djw302. doi: 10.1093/jnci/djw302.
    1. Mavaddat N, Barrowdale D, Andrulis IL, Domchek SM, Eccles D, Nevanlinna H, Ramus SJ, Spurdle A, Robson M, Sherman M, et al. Pathology of breast and ovarian cancers among BRCA1 and BRCA2 mutation carriers: results from the Consortium of Investigators of Modifiers of BRCA1/2 (CIMBA) Cancer Epidemiol Biomark Prev. 2012;21(1):134–147. doi: 10.1158/1055-9965.EPI-11-0775.
    1. Shimelis H, LaDuca H, Hu C, Hart SN, Na J, Thomas A, Akinhanmi M, Moore RM, Brauch H, Cox A, et al. Triple-Negative Breast Cancer Risk Genes Identified by Multigene Hereditary Cancer Panel Testing. J Natl Cancer Inst. 2018;110(8):855–862. doi: 10.1093/jnci/djy106.
    1. Bianchini G, Balko JM, Mayer IA, Sanders ME, Gianni L. Triple-negative breast cancer: challenges and opportunities of a heterogeneous disease. Nat Rev Clin Oncol. 2016;13(11):674–690. doi: 10.1038/nrclinonc.2016.66.
    1. Tharmapalan P, Mahendralingam M, Berman HK, Khokha R. Mammary stem cells and progenitors: targeting the roots of breast cancer for prevention. EMBO J. 2019;38(14):e100852. doi: 10.15252/embj.2018100852.
    1. Foulkes WD, Smith IE, Reis-Filho JS. Triple-negative breast cancer. N Engl J Med. 2010;363(20):1938–1948. doi: 10.1056/NEJMra1001389.
    1. Shah SP, Roth A, Goya R, Oloumi A, Ha G, Zhao Y, Turashvili G, Ding J, Tse K, Haffari G, et al. The clonal and mutational evolution spectrum of primary triple-negative breast cancers. Nature. 2012;486(7403):395–399. doi: 10.1038/nature10933.
    1. Venkitaraman AR. Cancer suppression by the chromosome custodians, BRCA1 and BRCA2. Science. 2014;343(6178):1470–1475. doi: 10.1126/science.1252230.
    1. Moran A, O'Hara C, Khan S, Shack L, Woodward E, Maher ER, Lalloo F, Evans DG. Risk of cancer other than breast or ovarian in individuals with BRCA1 and BRCA2 mutations. Familial Cancer. 2012;11(2):235–242. doi: 10.1007/s10689-011-9506-2.
    1. Widschwendter M, Dubeau L. Non-surgical cancer risk reduction in BRCA1 mutation carriers: disabling the remote control. Cancers (Basel) 2020;12(3):547. doi: 10.3390/cancers12030547.
    1. Chodankar R, Kwang S, Sangiorgi F, Hong H, Yen HY, Deng C, Pike MC, Shuler CF, Maxson R, Dubeau L. Cell-nonautonomous induction of ovarian and uterine serous cystadenomas in mice lacking a functional Brca1 in ovarian granulosa cells. Curr Biol. 2005;15(6):561–565. doi: 10.1016/j.cub.2005.01.052.
    1. Yen HY, Gabet Y, Liu Y, Martin A, Wu NL, Pike MC, Frenkel B, Maxson R, Dubeau L. Alterations in Brca1 expression in mouse ovarian granulosa cells have short-term and long-term consequences on estrogen-responsive organs. Lab Investig. 2012;92(6):802–811. doi: 10.1038/labinvest.2012.58.
    1. Hong H, Yen HY, Brockmeyer A, Liu Y, Chodankar R, Pike MC, Stanczyk FZ, Maxson R, Dubeau L. Changes in the mouse estrus cycle in response to BRCA1 inactivation suggest a potential link between risk factors for familial and sporadic ovarian cancer. Cancer Res. 2010;70(1):221–228. doi: 10.1158/0008-5472.CAN-09-3232.
    1. Widschwendter M, Rosenthal AN, Philpott S, Rizzuto I, Fraser L, Hayward J, Intermaggio MP, Edlund CK, Ramus SJ, Gayther SA, et al. The sex hormone system in carriers of BRCA1/2 mutations: a case-control study. Lancet Oncol. 2013;14(12):1226–1232. doi: 10.1016/S1470-2045(13)70448-0.
    1. Nene NR, Reisel D, Leimbach A, Franchi D, Jones A, Evans I, Knapp S, Ryan A, Ghazali S, Timms JF, et al. Association between the cervicovaginal microbiome, BRCA1 mutation status, and risk of ovarian cancer: a case-control study. Lancet Oncol. 2019;20(8):1171–1182. doi: 10.1016/S1470-2045(19)30340-7.
    1. Widschwendter M, Burnell M, Fraser L, Rosenthal AN, Philpott S, Reisel D, Dubeau L, Cline M, Pan Y, Yi PC, et al. Osteoprotegerin (OPG), The Endogenous Inhibitor of Receptor Activator of NF-kappaB Ligand (RANKL), is Dysregulated in BRCA Mutation Carriers. EBioMedicine. 2015;2(10):1331–1339. doi: 10.1016/j.ebiom.2015.08.037.
    1. Schramek D, Leibbrandt A, Sigl V, Kenner L, Pospisilik JA, Lee HJ, Hanada R, Joshi PA, Aliprantis A, Glimcher L, et al. Osteoclast differentiation factor RANKL controls development of progestin-driven mammary cancer. Nature. 2010;468(7320):98–102. doi: 10.1038/nature09387.
    1. Joshi PA, Jackson HW, Beristain AG, Di Grappa MA, Mote PA, Clarke CL, Stingl J, Waterhouse PD, Khokha R. Progesterone induces adult mammary stem cell expansion. Nature. 2010;465(7299):803–807. doi: 10.1038/nature09091.
    1. Gonzalez-Suarez E, Jacob AP, Jones J, Miller R, Roudier-Meyer MP, Erwert R, Pinkas J, Branstetter D, Dougall WC. RANK ligand mediates progestin-induced mammary epithelial proliferation and carcinogenesis. Nature. 2010;468(7320):103–107. doi: 10.1038/nature09495.
    1. Tanos T, Sflomos G, Echeverria PC, Ayyanan A, Gutierrez M, Delaloye JF, Raffoul W, Fiche M, Dougall W, Schneider P, et al. Progesterone/RANKL is a major regulatory axis in the human breast. Sci Transl Med. 2013;5(182):182ra155. doi: 10.1126/scitranslmed.3005654.
    1. Nolan E, Vaillant F, Branstetter D, Pal B, Giner G, Whitehead L, Lok SW, Mann GB, Kathleen Cuningham Foundation Consortium for Research into Familial Breast C. Rohrbach K, et al. RANK ligand as a potential target for breast cancer prevention in BRCA1-mutation carriers. Nat Med. 2016;22(8):933–939. doi: 10.1038/nm.4118.
    1. Chlebowski RT, Anderson GL, Aragaki AK, Manson JE, Stefanick ML, Pan K, Barrington W, Kuller LH, Simon MS, Lane D, et al. Association of menopausal hormone therapy with breast cancer incidence and mortality during long-term follow-up of the women’s health initiative randomized clinical trials. JAMA. 2020;324(4):369–380. doi: 10.1001/jama.2020.9482.
    1. Poole AJ, Li Y, Kim Y, Lin SC, Lee WH, Lee EY. Prevention of Brca1-mediated mammary tumorigenesis in mice by a progesterone antagonist. Science. 2006;314(5804):1467–1470. doi: 10.1126/science.1130471.
    1. Papaikonomou K, Kopp Kallner H, Soderdahl F, Gemzell-Danielsson K. Mifepristone treatment prior to insertion of a levonorgestrel releasing intrauterine system for improved bleeding control - a randomized controlled trial. Hum Reprod. 2018;33(11):2002–2009. doi: 10.1093/humrep/dey296.
    1. Picelli S, Faridani OR, Bjorklund AK, Winberg G, Sagasser S, Sandberg R. Full-length RNA-seq from single cells using Smart-seq2. Nat Protoc. 2014;9(1):171–181. doi: 10.1038/nprot.2014.006.
    1. Kennedy SR, Schmitt MW, Fox EJ, Kohrn BF, Salk JJ, Ahn EH, Prindle MJ, Kuong KJ, Shen JC, Risques RA, et al. Detecting ultralow-frequency mutations by Duplex Sequencing. Nat Protoc. 2014;9(11):2586–2606. doi: 10.1038/nprot.2014.170.
    1. Schmitt MW, Kennedy SR, Salk JJ, Fox EJ, Hiatt JB, Loeb LA. Detection of ultra-rare mutations by next-generation sequencing. Proc Natl Acad Sci U S A. 2012;109(36):14508–14513. doi: 10.1073/pnas.1208715109.
    1. Schmitt MW, Fox EJ, Prindle MJ, Reid-Bayliss KS, True LD, Radich JP, Loeb LA. Sequencing small genomic targets with high efficiency and extreme accuracy. Nat Methods. 2015;12(5):423–425. doi: 10.1038/nmeth.3351.
    1. Cancer Genome Atlas N Comprehensive molecular portraits of human breast tumours. Nature. 2012;490(7418):61–70. doi: 10.1038/nature11412.
    1. Yang Z, Wong A, Kuh D, Paul DS, Rakyan VK, Leslie RD, Zheng SC, Widschwendter M, Beck S, Teschendorff AE. Correlation of an epigenetic mitotic clock with cancer risk. Genome Biol. 2016;17(1):205. doi: 10.1186/s13059-016-1064-3.
    1. Bartlett TE, Jia P, Chandna S, Roy S. Inference of tissue relative proportions of the breast epithelial cell types luminal progenitor, basal, and luminal mature. Sci Rep. 2021;11(1):23702. doi: 10.1038/s41598-021-03161-7.
    1. Dielen C, Fiers T, Somers S, Deschepper E, Gerris J. Correlation between saliva and serum concentrations of estradiol in women undergoing ovarian hyperstimulation with gonadotropins for IVF/ICSI. Facts Views Vis Obgyn. 2017;9(2):85–91.
    1. Brisken C. Progesterone signalling in breast cancer: a neglected hormone coming into the limelight. Nat Rev Cancer. 2013;13(6):385–396. doi: 10.1038/nrc3518.
    1. Tomasetti C, Vogelstein B. Cancer etiology. Variation in cancer risk among tissues can be explained by the number of stem cell divisions. Science. 2015;347(6217):78–81. doi: 10.1126/science.1260825.
    1. Tomasetti C, Li L, Vogelstein B. Stem cell divisions, somatic mutations, cancer etiology, and cancer prevention. Science. 2017;355(6331):1330–1334. doi: 10.1126/science.aaf9011.
    1. Widschwendter M, Fiegl H, Egle D, Mueller-Holzner E, Spizzo G, Marth C, Weisenberger DJ, Campan M, Young J, Jacobs I, et al. Epigenetic stem cell signature in cancer. Nat Genet. 2007;39(2):157–158. doi: 10.1038/ng1941.
    1. Teschendorff AE, Menon U, Gentry-Maharaj A, Ramus SJ, Weisenberger DJ, Shen H, Campan M, Noushmehr H, Bell CG, Maxwell AP, et al. Age-dependent DNA methylation of genes that are suppressed in stem cells is a hallmark of cancer. Genome Res. 2010;20(4):440–446. doi: 10.1101/gr.103606.109.
    1. Zheng SC, Webster AP, Dong D, Feber A, Graham DG, Sullivan R, Jevons S, Lovat LB, Beck S, Widschwendter M, et al. A novel cell-type deconvolution algorithm reveals substantial contamination by immune cells in saliva, buccal and cervix. Epigenomics. 2018;10(7):925–940. doi: 10.2217/epi-2018-0037.
    1. Pellacani D, Bilenky M, Kannan N, Heravi-Moussavi A, Knapp D, Gakkhar S, Moksa M, Carles A, Moore R, Mungall AJ, et al. Analysis of normal human mammary epigenomes reveals cell-specific active enhancer states and associated transcription factor networks. Cell Rep. 2016;17(8):2060–2074. doi: 10.1016/j.celrep.2016.10.058.
    1. Engman M, Skoog L, Soderqvist G, Gemzell-Danielsson K. The effect of mifepristone on breast cell proliferation in premenopausal women evaluated through fine needle aspiration cytology. Hum Reprod. 2008;23(9):2072–2079. doi: 10.1093/humrep/den228.
    1. Westhoff CL, Guo H, Wang Z, Hibshoosh H, Polaneczky M, Pike MC, Ha R. The progesterone-receptor modulator, ulipristal acetate, drastically lowers breast cell proliferation. Breast Cancer Res Treat. 2022;192:321–329. doi: 10.1007/s10549-021-06503-1.
    1. Teschendorff AE, Yang Z, Wong A, Pipinikas CP, Jiao Y, Jones A, Anjum S, Hardy R, Salvesen HB, Thirlwell C, et al. Correlation of Smoking-Associated DNA Methylation Changes in Buccal Cells With DNA Methylation Changes in Epithelial Cancer. JAMA Oncol. 2015;1(4):476–485. doi: 10.1001/jamaoncol.2015.1053.
    1. Salk JJ, Loubet-Senear K, Maritschnegg E, Valentine CC, Williams LN, Higgins JE, Horvat R, Vanderstichele A, Nachmanson D, Baker KT, et al. Ultra-Sensitive TP53 sequencing for cancer detection reveals progressive clonal selection in normal tissue over a century of human lifespan. Cell Rep. 2019;28(1):132–144. doi: 10.1016/j.celrep.2019.05.109.
    1. Moore L, Leongamornlert D, Coorens THH, Sanders MA, Ellis P, Dentro SC, Dawson KJ, Butler T, Rahbari R, Mitchell TJ, et al. The mutational landscape of normal human endometrial epithelium. Nature. 2020;580(7805):640–646. doi: 10.1038/s41586-020-2214-z.
    1. Martincorena I, Fowler JC, Wabik A, Lawson ARJ, Abascal F, Hall MWJ, Cagan A, Murai K, Mahbubani K, Stratton MR, et al. Somatic mutant clones colonize the human esophagus with age. Science. 2018;362(6417):911–917. doi: 10.1126/science.aau3879.
    1. Lee-Six H, Olafsson S, Ellis P, Osborne RJ, Sanders MA, Moore L, Georgakopoulos N, Torrente F, Noorani A, Goddard M, et al. The landscape of somatic mutation in normal colorectal epithelial cells. Nature. 2019;574(7779):532–537. doi: 10.1038/s41586-019-1672-7.
    1. Matas J, Kohrn B, Fredrickson J, Carter K, Yu M, Wang T, Gui X, Soussi T, Moreno V, Grady WM, et al. Colorectal cancer is associated with the presence of cancer driver mutations in normal colon. Cancer Res. 2022;82(8):1492–1502. doi: 10.1158/0008-5472.CAN-21-3607.
    1. Harris KL, Walia V, Gong B, McKim KL, Myers MB, Xu J, Parsons BL. Quantification of cancer driver mutations in human breast and lung DNA using targeted, error-corrected CarcSeq. Environ Mol Mutagen. 2020;61:872–889. doi: 10.1002/em.22409.
    1. Salk JJ, Schmitt MW, Loeb LA. Enhancing the accuracy of next-generation sequencing for detecting rare and subclonal mutations. Nat Rev Genet. 2018;19(5):269–285. doi: 10.1038/nrg.2017.117.
    1. Lim E, Vaillant F, Wu D, Forrest NC, Pal B, Hart AH, Asselin-Labat ML, Gyorki DE, Ward T, Partanen A, et al. Aberrant luminal progenitors as the candidate target population for basal tumor development in BRCA1 mutation carriers. Nat Med. 2009;15(8):907–913. doi: 10.1038/nm.2000.
    1. Cuzick J, Warwick J, Pinney E, Duffy SW, Cawthorn S, Howell A, Forbes JF, Warren RM. Tamoxifen-induced reduction in mammographic density and breast cancer risk reduction: a nested case-control study. J Natl Cancer Inst. 2011;103(9):744–752. doi: 10.1093/jnci/djr079.
    1. Cummings SR, San Martin J, McClung MR, Siris ES, Eastell R, Reid IR, Delmas P, Zoog HB, Austin M, Wang A, et al. Denosumab for prevention of fractures in postmenopausal women with osteoporosis. N Engl J Med. 2009;361(8):756–765. doi: 10.1056/NEJMoa0809493.
    1. Gnant M, Pfeiler G, Steger GG, Egle D, Greil R, Fitzal F, Wette V, Balic M, Haslbauer F, Melbinger-Zeinitzer E, et al. Adjuvant denosumab in postmenopausal patients with hormone receptor-positive breast cancer (ABCSG-18): disease-free survival results from a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2019;20(3):339–351. doi: 10.1016/S1470-2045(18)30862-3.
    1. Bouchard P, Chabbert-Buffet N, Fauser BC. Selective progesterone receptor modulators in reproductive medicine: pharmacology, clinical efficacy and safety. Fertil Steril. 2011;96(5):1175–1189. doi: 10.1016/j.fertnstert.2011.08.021.
    1. Greene MF, Drazen JM. A New Label for Mifepristone. N Engl J Med. 2016;374(23):2281–2282. doi: 10.1056/NEJMe1604462.
    1. Henney JE, Gayle HD. Time to Reevaluate U.S. Mifepristone Restrictions. N Engl J Med. 2019;381(7):597–598. doi: 10.1056/NEJMp1908305.
    1. Shen J, Che Y, Showell E, Chen K, Cheng L. Interventions for emergency contraception. Cochrane Database Syst Rev. 2019;1:CD001324.
    1. Singh SS, Belland L, Leyland N, von Riedemann S, Murji A. The past, present, and future of selective progesterone receptor modulators in the management of uterine fibroids. Am J Obstet Gynecol. 2018;218(6):563–572. doi: 10.1016/j.ajog.2017.12.206.
    1. Fu J, Song H, Zhou M, Zhu H, Wang Y, Chen H, Huang W. Progesterone receptor modulators for endometriosis. Cochrane Database Syst Rev. 2017;7:CD009881.
    1. Bouchard C. Selective estrogen receptor modulators and their effects on hot flashes: a dilemma. Menopause. 2011;18(5):477–479. doi: 10.1097/gme.0b013e3182176285.
    1. Gatti M, Poluzzi E, De Ponti F, Raschi E. Liver injury with ulipristal acetate: exploring the underlying pharmacological basis. Drug Saf. 2020;43:1277–1285. doi: 10.1007/s40264-020-00975-8.
    1. Gemzell-Danielsson K, Widschwendter M. Breast tissue methylation analysis. Study ID EGAS00001005070, European Genome-phenome Archive. 2022.
    1. Teschendorff A, Widschwendter M. Genome wide DNA methylation profiling of normal breast, normal adjacent and breast cancer tissue. 2022.
    1. Simões B, Howell M. A pilot prevention study of the effects of the anti- progestin Ulipristal Acetate (UA) on surrogate markers of breast cancer risk. 2022.
    1. Bogavarappu NR, Gemzell-Danielsson K. Targeting progesterone to reduce epigenetic breast cancer risk surrogates in humans. 2022.

Source: PubMed

3
Prenumerera