12

Novel Breast Cancer Treatment by Targeting Estrogen Receptor-Alpha Stability Using Proteolysis-Targeting Chimeras (PROTACs) Technology

Angeles C. Tecalco-Cruz1 Josué Orlando Ramírez-Jarquín2 Marina Macías-Silva2 Marcela Sosa-Garrocho2 César López-Camarillo1

1Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, CDMX, México; 2Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, CDMX, México

Abstract: Approximately 70% of breast cancer cases are estrogen receptor-alpha-positive (ERα+). The binding of estradiol to the ligand-binding domain activates ERα. ERα can also be activated via the phosphorylation induced by growth factors. Activated ERα functions as a transcriptional regulator with a pro-tumor activity in breast cancer cells. In recent years, it has been discovered that some proteins can stabilize ERα by inhibiting its degradation via the ubiquitin-proteasome system through several mechanisms, including ERα monoubiquitination, deubiquitination, or phosphorylation, among others. Herein, we review the proteins associated with the inhibition of ERα degradation and discuss the role of proteolysis-targeting chimeras (PROTACs) as promising therapeutic strategies for breast cancer by inducing ERα degradation. The knowledge of the multiple mechanisms that stabilize ERα protein may be central for the development of new PROTACs for novel breast cancer treatments.

Keywords: endocrine resistance in breast cancer; estrogen receptor alpha in breast cancer; novel breast cancer treatment; PROTACs; proteolysis-targeting chimeras technology

Author for Correspondence: Angeles C. Tecalco-Cruz, Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, CDMX, 03100, México. Email: angeles.tecalco@uacm.edu.mx

Cite this chapter as: Tecalco-Cruz AC, Ramírez-Jarquín JO, Macías-Silva M, Sosa-Garrocho M, López-Camarillo C. Novel Breast Cancer Treatment by Targeting Estrogen Receptor-Alpha Stability Using Proteolysis-Targeting Chimeras (PROTACs) Technology. In: Mayrovitz HN. editor. Breast Cancer. Brisbane (AU): Exon Publications. Online first 11 Jun 2022.

Doi: https://doi.org/10.36255/exon-publications-breast-cancer-protacs

In: Mayrovitz HN, editor. Breast Cancer. Brisbane (AU): Exon Publications. ISBN: 978-0-6453320-3-2. Doi: https://doi.org/10.36255/exon-publications-breast-cancer

Copyright: The Authors.

License: This open access article is licenced under Creative Commons Attribution-NonCommercial 4.0 International (CC BY-NC 4.0) https://creativecommons.org/licenses/by-nc/4.0/

INTRODUCTION

Breast cancer (BC) is a serious health problem and the leading cause of death in women worldwide (13). Estrogen receptor-alpha (ERα) is a nuclear receptor expressed in more than 70% of BC cases (14). ERα is localized and functions in the nucleus, cytoplasm, and plasma membrane, and the subcellular distribution of this receptor is highly regulated by several factors (5) (Figure 1). In the nucleus, ERα is a transcription factor that induces the expression of estradiol-dependent genes. In BC cells, ERα expression has been related to the upregulation of estradiol-target genes that are associated with mammary tumor development (68). ERα also acts as a transcriptional regulator for other transcription factors, including AP1, NF- κB, and Sp1, to modulate gene expression (6, 7). ERα can also be localized outside the nucleus, and activate the signaling pathways that contribute to BC progression and endocrine resistance (8, 9). Hence, ERα associates with diverse transmembrane receptors and cytoplasmic proteins to activate its extranuclear signaling pathways (10, 11). Moreover, ERα can be directly localized at the plasma membrane via palmitoylation to activate the signaling that influences gene expression (12, 13).

Fig 1

Figure 1. Signaling of ERα in breast cancer cells. ERα exhibit multiple localizations including the nucleus, cytoplasm, and membranes. In the cytoplasm, it is associated with cytoplasmic proteins, and in the membrane through palmitoylation or transmembrane receptors (e.g., HER2). Estradiol (E2) activates ERα, promoting its accumulation in the nucleus, where the ERα regulates the expression of E2-target genes. E2 and growth factors (EGF and IGF) can induce ERα phosphorylation and promote its transcriptional activity. Fulvestrant and tamoxifen are anti-estrogens that bind to ERα to mediate either its degradation or the repression of its target genes, respectively.

ERα contains four main domains: the activation function domain-1 (AF-1), the DNA-binding domain (DBD), the ligand-binding domain (LBD), and the activation function domain-2 (AF-2) (Figure 2). The canonical signaling pathway of estrogen hormones, such as estradiol, is initiated when the hormones are recognized by LBD (14, 15). Enhancer and promoter regions of the estradiol-target genes contain specific palindromic sequences named ERE (estrogen response elements), which bind the DBD of ERα to regulate gene expression. AF-2 recruits coregulators in the presence of E2 hormone, whereas AF-1 recruits coregulators in its absence (1619). Furthermore, AF-1 can be modulated by phosphorylation (e.g., S118, S167) in response to growth factors (1622). The recruitment of ERα to the enhancer and promoter sequences is also supported by pioneer factors (e.g., FOXA1 and GATA3) in BC cells (23, 24). Several studies have shown that ERα recruits coregulators, including coactivator and corepressor complexes. In BC, there is an increase in the levels of coactivators for ERα that open the chromatin to promote gene expression (2529).

Fig 2

Figure 2. ERα structure. ERα contains two activation-function domains (AF-1 and AF-2) that interact with coregulators, some specialized sequences (NLS, nuclear localization signal, and two NES, nuclear export sequence) necessary for nucleo-cytoplasmic transport of ERα. ERα is a target for post-translational modifications (PTMs), including palmitoylation (Palm) and phosphorylation (P). ERα phosphorylation at sites S104, S106, S118, and S167, is promoted by E2 and growth factors (GF). K302 and K303 are involved in monoubiquitination and polyubiquitination induced by E2 or fulvestrant.

The targeted drug therapy for ERα+ BC is based on endocrine therapies that include aromatase inhibitors (AIs), selective estrogen receptor degraders (SERDs), and selective estrogen receptor modulators (SERMs). The AIs inhibit estradiol production, whereas SERMs act as antagonists for ERα and induce the recruitment of corepressors, thereby inhibiting the expression of estradiol-target genes (3032). However, endocrine resistance, de novo or acquired, is displayed mainly for SERMs owing to mechanisms that are not completely understood (9, 3134). In this context, SERDs appear to be more promising for BC treatment, as is discussed later.

ESR1 MUTATIONS IN BREAST CANCER

Several mutations, including Y537S, Y537N, Y537C, D538G, and E380Q, have been detected in ESR1 gene that encodes for ERα, in both DNA from biopsies and circulating tumor DNA obtained from patients with metastatic BC, and with acquired resistance to AIs and SERMs (35, 36). These mutations are located in LBD of ERα, and are related to changes in ERα conformation that promote its estradiol-independent activity (37). Moreover, it has been reported that the gene signature modulated by ERα wild type and its mutants differ, having few common target genes. The ERα mutants can differentially interact with DNA and other proteins and modulate gene transcription, indicating their molecular complexity (36, 3840). Moreover, in the induction of acquired endocrine resistance (by long-term E2 deprivation) in BC cells, ESR1 mutations (Y537C and Y537S) are detected (39, 41).

ERα STABILITY AS A MECHANISM THAT PROMOTES PRO-TUMOR ACTIONS AND ENDOCRINE RESISTANCE

ERα is a target for polyubiquitination induced by estradiol, whereas its monoubiquitination is inhibited by estradiol (42). The polyubiquitination of ERα serves as a signal for its degradation by the ubiquitin-proteasome system (UPS); however, ERα monoubiquitination seems to confer receptor stability, and modulates its activity (4345). Furthermore, both modifications are deregulated in mammary tumors. In fact, it has been reported that some proteins interact with ERα to inhibit their polyubiquitination and degradation via UPS. These ERα-polyubiquitination inhibitor proteins (EPIPs) can act by several mechanisms leading to this receptor stability in BC (Table 1) (4664). The mechanisms that lead to the stabilization of ERα include its phosphorylation by specific kinases, association with transcription regulators, and regulation by E3-ubiquitin ligases and deubiquitinases (DUBs) (Figure 3). In recent years, new EPIPs have been discovered, demonstrating the relevance of ERα stability in pro-tumor molecular mechanisms associated with BC progression and endocrine resistance. Interestingly, several E3-ubiquitin ligases inhibit ERα degradation: RNF8, RNF31, and SHARPIN by inducing its monoubiquitination (4749), whereas Smurf1, TRIM56, and HOIL-1 by inhibiting its K48-specific polyubiquitination (50, 51). In addition, RNF181 stabilizes ERα, inducing its K63-linked ubiquitination in BC cells (52).

TABLE 1 ERα-polyubiquitination inhibitor proteins (EPIPs)

Protein Name Activity Reference
cABL Abelson tyrosine-protein kinase Kinase (53)
GSK3 Glycogen Synthase Kinase 3 Kinase (54)
LMTK3 Lemur Tyrosine Kinase 3 Kinase (55)
PIN1 Peptidyl-propyl cis-trans isomerase NIMA-interacting 1 Isomerase (56)
OTUD7B OTU Deubiquitinase 7B Deubiquitinase (57)
USP7 Ubiquitin-specific protease 7 Deubiquitinase (58)
USP15 Ubiquitin Specific Peptidase 15 Deubiquitinase (59)
USP35 Ubiquitin Specific Peptidase 35 Deubiquitinase (60)
HOIL-1 Haem-oxidised IRP2 ubiquitin ligase-1 E3-ubiquitin ligase (51)
RNF8 RING finger protein 8 E3-ubiquitin ligase (47)
RNF31 RING finger protein 31 E3-ubiquitin ligase (48)
RNF181 Ring Finger Protein 181 E3-ubiquitin ligase (52)
SHARPIN Shack-associated RH domain-interacting protein E3-ubiquitin ligase (49)
SMURF1 SMAD ubiquitination regulatory factor 1 E3-ubiquitin ligase (61)
TRIM11 Tripartite Motif Containing 11 E3-ubiquitin ligase (62)
TRIM56 Tripartite Motif Containing 56 E3-ubiquitin ligase (50)
RB Retinoblastoma Tumor suppressor (10)
MUC1 Mucin 1 Transcriptional regulator (63)
ZNF213 Zinc Finger Protein 213 Transcriptional regulator (64)

Induction of K63-linked ubiquitination: RNF181; Inhibition of K48-linked ubiquitination: HOIL-1, TRIM56, SMURF2, ZNF213; Monoubiquitination: TRIM11, RNF8, RNF31, SHARPIN.

Fig 3

Figure 3. Modulators of ERα stability in breast cancer cells. EPIPs (ERα polyubiquitination inhibitor proteins) have been reported to inhibit ERα polyubiquitination in breast cancer cells. These proteins protect ERα from degradation via the UPS. Diverse PTMs confer stability to ERα: Monoubiquitination (produced by E3 ubiquitin ligases), the inhibition of K-48 linked polyubiquitination, and deubiquitinase (DUB) enzymes, which cut off polyubiquitination tails of ERα.

Furthermore, some DUB proteins also confer stability to ERα. Nearly 100 DUBs have been identified in humans, and are classified into six families: USPs, OTUs, UCHs, MJDs, MINDYs, and JAMMs (65). The DUBs implicated in ERα stability are ubiquitin-specific proteases (USPs: USP7, USP15, and USP35), and one ovarian tumor protease (OTU: OTUD7B). In general, the proteins that stabilize ERα are upregulated in patients with mammary tumors, revealing their importance in the progression of this cancer (66). Thus, these results demonstrate the importance of understanding the mechanisms of ubiquitination (mono-and poly-ubiquitination) and de-ubiquitination in the regulation of ERα stability in BC.

SERDS and PROTACs

Fulvestrant is a SERD that induces ERα polyubiquitination and degradation via the UPS (3234). There is a complete reduction in ERα levels in BC cells after 1 h of treatment with fulvestrant (30, 6769). Fulvestrant has been also approved as the first-line endocrine therapy in BC and also in endocrine resistance to SERMs or AIs (70). Interestingly, endocrine resistance to SERDs, such as fulvestrant and AZD9496, has not been reported for the majority of ESR1 mutations (36, 39, 40). New SERDs that are being studied are AZD9496, bazedoxifene, RAD1901, GDC-0810, ZB716, and LSZ102. As fulvestrant exhibits poor bioavailability with intramuscular administration, the challenge is to develop a new generation of SERDs that can be orally administered and with better bioavailability (7176).

Proteolysis-targeting chimeras (PROTACs) technology works by inducing the degradation of specific proteins. This activity is due to the bifunctional structure of PROTACs: one part of these molecules has a ligand for a targeted protein, while the other part has the ability to recruit an E3 ubiquitin ligase, thereby stimulating the targeted protein degradation via the UPS. Hence, PROTACs have emerged as a promising alternative to control the pro-tumor effect of ERα by causing its downregulation via UPS (77, 78).

PROTAC technology

PROTACs are bimodular chimeras: they contain a binding module for the target protein and another E3 ligase recognition module; a spacer or linker sequence links these two modules (Figure 4). The PROTAC binds to its target protein, carrying a signal for its ubiquitination and subsequent degradation (79). A polyubiquitinated protein is recognized by the 26S proteasome, transported to the 20S core particle, and converted into oligopeptides by a variety of enzymes that promote their release from the proteasome; this is followed by the recycling of ubiquitin (80, 81).

Fig 4

Figure 4. PROTACs are constituted of a region that recognizes its target (ERα) and a region that recruits E3-ligase (to polyubiquitinate the receptor) for its degradation via the UPS.

Since 2001, the technology based on PROTACs is being developed by utilizing heterobifunctional molecules (82). The first ligands for recruiting E3-ubiquitin ligases in PROTAC technology were large peptides; however, they had the disadvantage of having a low level of cellular penetration owing to their size (81). PROTAC technology was developed for the first time using a chimera containing ovalicin in one domain and the phosphopeptide IκB in another domain. Thus, this chimera had the ability to bind to methionine aminopeptidase 2 (MetAP-2) (by ovalicin) and to recruit the SCFβ-TrCP E3-ubiquitin ligase complex (by phosphopeptide IκB), thereby promoting its degradation via the proteasome (81, 82). Later, other PROTACs were developed using the following E3 ubiquitin ligases: MDM2 (murine double minute 2), CRBN (cereblon), VHL (von Hippel-Lindau tumor suppressor), and IAPs (inhibitor of apoptosis proteins) (81, 83). PROTACs based on IAPs have antitumor functions; however, special care must be taken while designing them to avoid collateral effects (80, 84).

PROTACs display some disadvantages, for instance, they are generally large molecules, and their pharmacokinetic characteristics are compromised, such as bioavailability and solubility. In addition, the routes of administration of these chimeric degraders are generally intraperitoneal or subcutaneous. However, in recent years, the intravenous and oral routes are also being evaluated (79, 85, 86).

NOVEL PROTAC TECHNOLOGY FOR BREAST CANCER TREATMENT

Several PROTACs have been developed for ERα, including, SNIPER(ER)-3, PROTAC-B, PROTAC-2, compound 11, compound 24, lipophilic amino acid Boc-Trp motif, C3-linked adamantane motif, monocyclic trifluoromethyl cyclohexane motif, and ERD-148 (87). PROTACs exhibit antitumor activity and the ability to degrade ERα through the UPS in breast cancer cells (88). To date there are some limitations in the cell-penetrating and target protein degradation ability of the PROTACs designed for ERα; however, compound I-6 (a peptide-based PROTAC) contributes to cell membrane penetration. As a result, this compound can induce degradation of intracellular ERα and inhibit the ERα-dependent tumor cell proliferation and growth in in vivo studies, thereby supporting its potential use in breast cancer therapy (84).

Another molecule proposed for ERα + breast cancer based on PROTAC technology is PROTAC-2. This compound consists of estradiol that is covalently bound to the phosphopeptide of IκBα containing an E3 recognition domain, and thus recruits SCFβ-trcp, which leads to ERα ubiquitination and subsequent degradation by the 26S proteasome. The disadvantage of this PROTAC is that the IκBα peptide is susceptible to phosphatases (89). Another traditional example is PROTAC-B, which contains a pentapeptide derived from HIF-1α (hypoxia-inducible factor-1α) to be recognized by the E3-ubiquitin ligase, VHL (88, 90). Similarly, E2-octa contains a synthetic octapeptide derived from HIF-1α, which is very efficient in the degradation of ERα in human BC cells. Modifications of this PROTAC gave rise to E2-penta, which produces a reduction in ERα levels (91). It was only after 2011, when E3-ubiquitin ligase ligands were discovered, that major progress was made in the development of next-generation PROTACs. The specific and non-genetic IAP-dependent protein eraser (SNIPER), composed of a derivative of estradiol and a bestatin amide (a cIAP1 ligand), also showed a decrease in ERα levels (81, 92).

A peptide-based PROTAC design has been shown to degrade ERα through the binding of an ERα modulator peptide (TD-PERM; N-terminal aspartic acid cross-linked stabilized peptide ERα modulator) with a pentapeptide that binds to the VHL E3 ubiquitin ligase complex (77). This heterobifunctional peptide has the ability to recruit ERα and bring it to the VHL E3 ligase complex for degradation through the proteasome. Thus, this PROTAC (TD-PROTAC) induces the degradation of ERα, reduces the transcription of its target genes such as pS2, and inhibits the proliferation of cancer cells. The T47D and MCF-7 breast cancer cells that express ERα showed early apoptosis when subjected to high doses of TD-PROTAC (77).

Additionally, in a xenograft model of nude mice that received MCF-7 cells, the administration of TD-PROTAC promoted tumor regression (77). ARV-471 is another PROTAC for ERα that is well tolerated and is currently in phase 2 clinical trials. It functions by reducing the ERα levels, and ERα variants such as Y537S and D538G (80). ERD-308 is another PROTAC that displays a better ERα degradation capacity, as well as a greater cell proliferation inhibitory capacity than fulvestrant (83).

DISCUSSION

ERα stability is increased in BC cells, and it has been associated with tumor progression and the development of endocrine resistance (93). New studies are emerging, which are demonstrating that many proteins are involved in ERα stability in BC, which emphasizes the pro-tumor potential of Erα, and endocrine resistance (Table 1 and Figure 1). Therefore, the mechanisms that control ERα stability may be targeted for the development of new drugs and also the improvement of current therapies. Thereby, SERDs and PROTACs are critical therapeutic strategies for controlling breast cancer progression. Even though new SERDs are being developed, only fulvestrant has been approved thus far (32). To avoid the development of resistance to SERD treatments based on ERα degradation, it is important to consider the implications of ERα stability-associated pathways. Interestingly, although PROTACs show good promise for BC due to their abilities for causing ERα ubiquitination and degradation, the mechanisms that lead to ERα stability may also represent a limitation. This is because several USPs participate in the inhibition of polyubiquitination and degradation of ERα, and many of them are upregulated in mammary tumors. Moreover, although some E3-ubiquitin ligases can promote ERα degradation via polyubiquitination, there are others that can inhibit the degradation of this receptor by inducing its monoubiquitination (Table 1). Thus, ERα stability may also have an impact on the therapies based on the degradation of this receptor in BC cells. There is a growing need for the development of new SERDS-like molecules and PROTACs that can induce ERα degradation in BC, which can block tumor progression. Attention should be paid to the improvement of bioavailability and administration routes of SERDs and PROTACs, as well as the denominated SERD-like PROTACs (78).

PROTAC technology is a promising system that is starting to be used in patients with BC, as well as in patients with other types of cancer (80, 89). This technology shows that when suitable ligands are used to target Erα, they can promote its degradation almost entirely, and therefore, nullify its transcriptional activity and signaling, and promote the death of BC cells both in vitro and in vivo (77). Currently, some PROTACs are already in the clinical stage (80), and it is expected that they will exhibit high specificity when administered orally, with good stability and cell permeability. Thus, the PROTAC system is currently being evaluated in clinical trials conducted in patients with metastatic BC, with the interest of some pharmaceutical companies (81). At present, PROTACs are not intended to replace traditional cancer therapies, but rather, be included as co-adjuvants in such treatments. This technology could complement current cancer therapy strategies. The next step in PROTACs technology would be to improve their pharmacokinetic properties for a wider clinical application and consider the implications of the mechanisms that stabilize ERα in BC.

CONCLUSION

Several studies have suggested the existence of a wide range of proteins that confer ERα stability in BC, which is related to tumor progression and endocrine resistance. Under these conditions, the development of treatments focused on the induction of ERα degradation is required. Both SERDs and PROTACs have the ability to downregulate ERα via degradation through the UPS with the aim of reducing its pro-tumor effects. However, further studies are required to improve these drugs for use in the treatment of BC, and also to evaluate the role of ERα stability in these treatments.

Acknowledgments: This research was supported by UACM. Proyectos de investigación del Colegio de Ciencia y Tecnología (CCYT-2021-5 and CCYT-2022-8).

Conflict of Interest: The authors declare no potential conflict of interest with respect to research, authorship and/or publication of this chapter.

Copyright and Permission Statement: The authors confirm that the materials included in this chapter do not violate copyright laws. Where relevant, appropriate permissions have been obtained from the original copyright holder(s), and all original sources have been appropriately acknowledged or referenced.

REFERENCES

  1. Chen T, Zhang N, Moran MS, Su P, Haffty BG, Yang Q. Borderline ER-Positive Primary Breast Cancer Gains No Significant Survival Benefit From Endocrine Therapy: A Systematic Review and Meta-Analysis. Kd 18, Clinical Breast Cancer. 2018. lk 1–8. https://doi.org/10.1016/j.clbc.2017.06.005
  2. Collins LC, Botero ML, Schnitt SJ. Bimodal frequency distribution of estrogen receptor immunohistochemical staining results in breast cancer: An analysis of 825 cases. Kd 123, American Journal of Clinical Pathology. 2005. lk 16–20. https://doi.org/10.1309/HCF035N9WK40ETJ0
  3. Curigliano G, Burstein HJ, Winer EP, Gnant M, Dubsky P, Loibl S, et al. De-escalating and escalating treatments for early-stage breast cancer: The St. Gallen International Expert Consensus Conference on the Primary Therapy of Early Breast Cancer 2017. Ann Oncol. 2017;28(8):1700–12. https://doi.org/10.1093/annonc/mdx308
  4. Hammond MEH, Hayes DF, Dowsett M, Allred DC, Hagerty KL, Badve S, et al. American society of clinical oncology/college of american pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer. Kd 28, Journal of Clinical Oncology. 2010. lk 2784–95.
  5. Tecalco-Cruz AC, Pérez-Alvarado IA, Ramírez-Jarquín JO, Rocha-Zavaleta L. Nucleo-cytoplasmic transport of estrogen receptor alpha in breast cancer cells. Kd 34, Cellular Signalling. 2017. lk 121–32. https://doi.org/10.1016/j.cellsig.2017.03.011
  6. Björnström L, Sjöberg M. Estrogen receptor-dependent activation of AP-1 via non-genomic signalling. Nucl Recept. 2004;2. https://doi.org/10.1186/1478-1336-2-3
  7. Frasor J, Weaver A, Pradhan M, Dai Y, Miller LD, Lin CY, et al. Positive cross-talk between estrogen receptor and NF-κB in breast cancer. Cancer Res. 2009;69(23):8918–25. https://doi.org/10.1158/0008-5472.CAN-09-2608
  8. Acconcia F, Kumar R. Signaling regulation of genomic and nongenomic functions of estrogen receptors. Kd 238, Cancer Letters. 2006. lk 1–14. https://doi.org/10.1016/j.canlet.2005.06.018
  9. Alfakeeh A, Brezden-Masley C. Overcoming endocrine resistance in hormone receptor-positive breast cancer. Kd 25, Current Oncology. 2018. lk S18–27. https://doi.org/10.3747/co.25.3752
  10. Caligiuri I, Toffoli G, Giordano A, Rizzolio F. pRb controls estrogen receptor alpha protein stability and activity. Oncotarget. 2013;4(6):875–83. https://doi.org/10.18632/oncotarget.1036
  11. Kumar R, Wang RA, Mazumdar A, Talukder AH, Mandal M, Yang Z, et al. A naturally occurring MTA1 variant sequesters oestrogen receptor-α in the cytoplasm. Nature. 2002;418(6898):654–7. https://doi.org/10.1038/nature00889
  12. Acconcia F, Ascenzi P, Fabozzi G, Visca P, Marino M. S-palmitoylation modulates human estrogen receptor-α functions. Biochem Biophys Res Commun. 2004;316(3):878–83. https://doi.org/10.1016/j.bbrc.2004.02.129
  13. Pedram A, Razandi M, Deschenes RJ, Levin ER. DHHC-7 and -21 are palmitoylacyltransferases for sex steroid receptors. Mol Biol Cell. 2012;23(1):188–99. https://doi.org/10.1091/mbc.e11-07-0638
  14. Kumar R, Zakharov MN, Khan SH, Miki R, Jang H, Toraldo G, et al. The Dynamic Structure of the Estrogen Receptor. J Amino Acids. 2011;2011:1–7. https://doi.org/10.4061/2011/812540
  15. Nardulli AM, Greene GL, Shapiro DJ. Human estrogen receptor bound to an estrogen response element bends DNA. Mol Endocrinol. 1993;7(3):331–40. https://doi.org/10.1210/mend.7.3.8483477
  16. Billon-Galés A, Krust A, Fontaine C, Abot A, Flouriot G, Toutain C, et al. Activation function 2 (AF2) of estrogen receptor-α is required for the atheroprotective action of estradiol but not to accelerate endothelial healing. Proc Natl Acad Sci U S A. 2011;108(32):13311–6. https://doi.org/10.1073/pnas.1105632108
  17. Métivier R, Penot G, Flouriot G, Pakdel F. Synergism Between ERα Transactivation Function 1 (AF-1) and AF-2 Mediated by Steroid Receptor Coactivator Protein-1: Requirement for the AF-1 α-Helical Core and for a Direct Interaction Between the N- and C-Terminal Domains. Mol Endocrinol. 2001;15(11):1953–70. https://doi.org/10.1210/mend.15.11.0727
  18. Smirnova NF, Fontaine C, Buscato M, Lupieri A, Vinel A, Valera MC, et al. The activation function-1 of estrogen receptor alpha prevents arterial neointima development through a direct effect on smooth muscle cells. Circ Res. 2015;117(9):770–7. https://doi.org/10.1161/CIRCRESAHA.115.306416
  19. Zwart W, De Leeuw R, Rondaij M, Neefjes J, Mancini MA, Michalides R. The hinge region of the human estrogen receptor determines functional synergy between AF-1 and AF-2 in the quantitative response to estradiol and tamoxifen. J Cell Sci. 2010;123(8):1253–61. https://doi.org/10.1242/jcs.061135
  20. Vrtačnik P, Ostanek B, Mencej-Bedrač S, Marc J. The many faces of estrogen signaling. Kd 24, Biochemia Medica. 2014. lk 329–42. https://doi.org/10.11613/BM.2014.035
  21. Kato S. Estrogen receptor-mediated cross-talk with growth factor signaling pathways. Kd 8, Breast Cancer. 2001. lk 3–9. https://doi.org/10.1007/BF02967472
  22. Lee A V., Cui X, Oesterreich S. Cross-talk among estrogen receptor, epidermal growth factor, and insulin-like growth factor signaling in breast cancer. Clin Cancer Res [Internet]. detsember 2001;7(12 Suppl):4429s–4435s; discussion 4411s-4412s. Available at: http://www.ncbi.nlm.nih.gov/pubmed/11916236
  23. Carroll JS, Liu XS, Brodsky AS, Li W, Meyer CA, Szary AJ, et al. Chromosome-wide mapping of estrogen receptor binding reveals long-range regulation requiring the forkhead protein FoxA1. Cell. 2005;122(1):33–43. https://doi.org/10.1016/j.cell.2005.05.008
  24. Theodorou V, Stark R, Menon S, Carroll JS. GATA3 acts upstream of FOXA1 in mediating ESR1 binding by shaping enhancer accessibility. Genome Res. 2013;23(1):12–22. https://doi.org/10.1101/gr.139469.112
  25. Fleming FJ, Hill ADK, McDermott EW, O’Higgins NJ, Young LS. Differential Recruitment of Coregulator Proteins Steroid Receptor Coactivator-1 and Silencing Mediator for Retinoid and Thyroid Receptors to the Estrogen Receptor-Estrogen Response Element by β-Estradiol and 4-Hydroxytamoxifen in Human Breast Cancer. J Clin Endocrinol Metab. 2004;89(1):375–83. https://doi.org/10.1210/jc.2003-031048
  26. Hah N, Kraus WL. Hormone-regulated transcriptomes: Lessons learned from estrogen signaling pathways in breast cancer cells. Mol Cell Endocrinol. 2014;382(1):652–64. https://doi.org/10.1016/j.mce.2013.06.021
  27. Hah N, Murakami S, Nagari A, Danko CG, Lee Kraus W. Enhancer transcripts mark active estrogen receptor binding sites. Genome Res. 2013;23(8):1210–23. https://doi.org/10.1101/gr.152306.112
  28. Hervouet E, Cartron PF, Jouvenot M, Delage-Mourroux R. Epigenetic regulation of estrogen signaling in breast cancer. Kd 8, Epigenetics. 2013. lk 237–45. https://doi.org/10.4161/epi.23790
  29. Manavathi B, Samanthapudi VSK, Gajulapalli VNR. Estrogen receptor coregulators and pioneer factors: The orchestrators of mammary gland cell fate and development. Kd 2, Frontiers in Cell and Developmental Biology. 2014. https://doi.org/10.3389/fcell.2014.00034
  30. Howell SJ, Johnston SRD, Howell A. The use of selective estrogen receptor modulators and selective estrogen receptor down-regulators in breast cancer. Kd 18, Best Practice and Research: Clinical Endocrinology and Metabolism. 2004. lk 47–66. https://doi.org/10.1016/j.beem.2003.08.002
  31. Osborne CK, Schiff R. Mechanisms of endocrine resistance in breast cancer. Annu Rev Med. 2011;62:233–47. https://doi.org/10.1146/annurev-med-070909-182917
  32. Patel HK, Bihani T. Selective estrogen receptor modulators (SERMs) and selective estrogen receptor degraders (SERDs) in cancer treatment. Kd 186, Pharmacology and Therapeutics. 2018. lk 1–24. https://doi.org/10.1016/j.pharmthera.2017.12.012
  33. Osborne CK, Wakeling A, Nicholson RI. Fulvestrant: An oestrogen receptor antagonist with a novel mechanism of action. Br J Cancer. 2004;90:S2–6. https://doi.org/10.1038/sj.bjc.6601629
  34. Wardell SE, Marks JR, McDonnell DP. The turnover of estrogen receptor α by the selective estrogen receptor degrader (SERD) fulvestrant is a saturable process that is not required for antagonist efficacy. Biochem Pharmacol. 2011;82(2):122–30. https://doi.org/10.1016/j.bcp.2011.03.031
  35. Angus L, Beije N, Jager A, Martens JWM, Sleijfer S. ESR1 mutations: Moving towards guiding treatment decision-making in metastatic breast cancer patients. Kd 52, Cancer Treatment Reviews. 2017. lk 33–40. https://doi.org/10.1016/j.ctrv.2016.11.001
  36. Toy W, Weir H, Razavi P, Lawson M, Goeppert AU, Mazzola AM, et al. Activating ESR1 mutations differentially affect the efficacy of ER antagonists. Cancer Discov. 2017;7(3):277–87. https://doi.org/10.1158/2159-8290.CD-15-1523
  37. Reinert T, Gonçalves R, Bines J. Implications of ESR1 Mutations in Hormone Receptor-Positive Breast Cancer. Curr Treat Options Oncol [Internet]. 17. mai 2018;19(5):24. Available at: http://link.springer.com/10.1007/s11864-018-0542-0; https://doi.org/10.1007/s11864-018-0542-0
  38. Jeselsohn R, Bergholz JS, Pun M, Cornwell M, Liu W, Nardone A, et al. Allele-Specific Chromatin Recruitment and Therapeutic Vulnerabilities of ESR1 Activating Mutations. Cancer Cell. 2018;33(2):173–186.e5. https://doi.org/10.1016/j.ccell.2018.01.004
  39. Bahreini A, Li Z, Wang P, Levine KM, Tasdemir N, Cao L, et al. Mutation site and context dependent effects of ESR1 mutation in genome-edited breast cancer cell models. Breast Cancer Res [Internet]. 23. detsember 2017;19(1):60. Available at: https://breast-cancer-research.biomedcentral.com/articles/10.1186/s13058-017-0851-4; https://doi.org/10.1186/s13058-017-0851-4
  40. Harrod A, Fulton J, Nguyen VTM, Periyasamy M, Ramos-Garcia L, Lai CF, et al. Genomic modelling of the ESR1 Y537S mutation for evaluating function and new therapeutic approaches for metastatic breast cancer. Oncogene. 2017;36(16):2286–96. https://doi.org/10.1038/onc.2016.382
  41. Martin L-A, Ribas R, Simigdala N, Schuster E, Pancholi S, Tenev T, et al. Discovery of naturally occurring ESR1 mutations in breast cancer cell lines modelling endocrine resistance. Nat Commun [Internet]. 30. detsember 2017;8(1):1865. Available at: http://www.nature.com/articles/s41467-017-01864-y; https://doi.org/10.1038/s41467-017-01864-y
  42. La Rosa P, Marino M, Acconcia F. 17Β-Estradiol Regulates Estrogen Receptor Α Monoubiquitination. IUBMB Life. 2011;63(1):49–53. https://doi.org/10.1002/iub.414
  43. Eakin CM, MacCoss MJ, Finney GL, Klevit RE. Estrogen receptor α is a putative substrate for the BRCA1 ubiquitin ligase. Proc Natl Acad Sci U S A. 2007;104(14):5794–9. https://doi.org/10.1073/pnas.0610887104
  44. La Rosa P, Acconcia F. Signaling functions of ubiquitin in the 17β-estradiol (E2):estrogen receptor (ER) α network. Kd 127, Journal of Steroid Biochemistry and Molecular Biology. 2011. lk 223–30. https://doi.org/10.1016/j.jsbmb.2011.07.008
  45. Ma Y, Fan S, Hu C, Meng Q, Fuqua SA, Pestell RG, et al. BRCA1 regulates acetylation and ubiquitination of estrogen receptor-α. Mol Endocrinol. 2010;24(1):76–90. https://doi.org/10.1210/me.2009-0218
  46. Tecalco-Cruz AC, Ramírez-Jarquín JO, Cruz-Ramos E. Estrogen Receptor Alpha and its Ubiquitination in Breast Cancer Cells. Curr Drug Targets. 2018;20(6):690–704. https://doi.org/10.2174/1389450119666181015114041
  47. Wang S, Luo H, Wang C, Sun H, Sun G, Sun N, et al. RNF8 identified as a co-activator of estrogen receptor α promotes cell growth in breast cancer. Biochim Biophys Acta - Mol Basis Dis. 2017;1863(6):1615–28. https://doi.org/10.1016/j.bbadis.2017.02.011
  48. Zhu J, Zhao C, Kharman-Biz A, Zhuang T, Jonsson P, Liang N, et al. The atypical ubiquitin ligase RNF31 stabilizes estrogen receptor α and modulates estrogen-stimulated breast cancer cell proliferation. Oncogene. 2014;33(34):4340–51. https://doi.org/10.1038/onc.2013.573
  49. Zhuang T, Yu S, Zhang L, Yang H, Li X, Hou Y, et al. SHARPIN stabilizes estrogen receptor a and promotes breast cancer cell proliferation. Oncotarget. 2017;8(44):77137–51. https://doi.org/10.18632/oncotarget.20368
  50. Xue M, Zhang K, Mu K, Xu J, Yang H, Liu Y, et al. Regulation of estrogen signaling and breast cancer proliferation by an ubiquitin ligase TRIM56. Oncogenesis. 2019;8(5). https://doi.org/10.1038/s41389-019-0139-x
  51. Ding J, Kuang P. Regulation of ERα Stability and Estrogen Signaling in Breast Cancer by HOIL-1. Front Oncol [Internet]. 20. mai 2021;11. Available at: https://www.frontiersin.org/articles/10.3389/fonc.2021.664689/full; https://doi.org/10.3389/fonc.2021.664689
  52. Zhu J, Li X, Su P, Xue M, Zang Y, Ding Y. The ubiquitin ligase RNF181 stabilizes ERα and modulates breast cancer progression. Oncogene. 2020;39(44):6776–88. https://doi.org/10.1038/s41388-020-01464-z
  53. He X, Zheng Z, Song T, Wei C, Ma H, Ma Q, et al. C-Abl regulates estrogen receptor α transcription activity through its stabilization by phosphorylation. Oncogene. 2010;29(15):2238–51. https://doi.org/10.1038/onc.2009.513
  54. Grisouard J, Medunjanin S, Hermani A, Shukla A, Mayer D. Glycogen synthase kinase-3 protects estrogen receptor α from proteasomal degradation and is required for full transcriptional activity of the receptor. Mol Endocrinol. 2007;21(10):2427–39. https://doi.org/10.1210/me.2007-0129
  55. Giamas G, Filipović A, Jacob J, Messier W, Zhang H, Yang D, et al. Kinome screening for regulators of the estrogen receptor identifies LMTK3 as a new therapeutic target in breast cancer. Nat Med. 2011;17(6):715–9. https://doi.org/10.1038/nm.2351
  56. Rajbhandari P, Schalper KA, Solodin NM, Ellison-Zelski SJ, Ping Lu K, Rimm DL, et al. Pin1 modulates ERα levels in breast cancer through inhibition of phosphorylation-dependent ubiquitination and degradation. Oncogene. 2014;33(11):1438–47. https://doi.org/10.1038/onc.2013.78
  57. Tang J, Wu Z, Tian Z, Chen W, Wu G. OTUD7B stabilizes estrogen receptor α and promotes breast cancer cell proliferation. Cell Death Dis [Internet]. 25. juuni 2021;12(6):534. Available at: http://www.nature.com/articles/s41419-021-03785-7; https://doi.org/10.1038/s41419-021-03785-7
  58. Xia X, Liao Y, Huang C, Liu Y, He J, Shao Z, et al. Deubiquitination and stabilization of estrogen receptor α by ubiquitin-specific protease 7 promotes breast tumorigenesis. Cancer Lett [Internet]. november 2019;465:118–28. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0304383519304677; https://doi.org/10.1016/j.canlet.2019.09.003
  59. Xia X, Huang C, Liao Y, Liu Y, He J, Shao Z, et al. The deubiquitinating enzyme USP15 stabilizes ERα and promotes breast cancer progression. Cell Death Dis. 2021;12(4). https://doi.org/10.1038/s41419-021-03607-w
  60. Cao J, Wu D, Wu G, Wang Y, Ren T, Wang Y, et al. USP35, regulated by estrogen and AKT, promotes breast tumorigenesis by stabilizing and enhancing transcriptional activity of estrogen receptor α. Cell Death Dis [Internet]. 15. juuni 2021;12(6):619. Available at: http://www.nature.com/articles/s41419-021-03904-4; https://doi.org/10.1038/s41419-021-03904-4
  61. Yang H, Yu N, Xu J, Ding X, Deng W, Wu G, et al. SMURF1 facilitates estrogen receptor a signaling in breast cancer cells. J Exp Clin Cancer Res. 2018;37(1). https://doi.org/10.1186/s13046-018-0672-z
  62. Tang J, Luo Y, Tian Z, Liao X, Cui Q, Yang Q, et al. TRIM11 promotes breast cancer cell proliferation by stabilizing estrogen receptor α. Neoplasia (United States). 2020;22(9):343–51. https://doi.org/10.1016/j.neo.2020.06.003
  63. Wei X, Xu H, Kufe D. MUC1 oncoprotein stabilizes and activates estrogen receptor α. Mol Cell. 2006;21(2):295–305. https://doi.org/10.1016/j.molcel.2005.11.030
  64. Yang H, Lv X, Li X, Mao L, Niu Z, Wang T, et al. ZNF213 Facilitates ER Alpha Signaling in Breast Cancer Cells. Front Oncol. 2021;11. https://doi.org/10.3389/fonc.2021.638751
  65. Harrigan JA, Jacq X, Martin NM, Jackson SP. Deubiquitylating enzymes and drug discovery: Emerging opportunities. Kd 17, Nature Reviews Drug Discovery. 2018. lk 57–77. https://doi.org/10.1038/nrd.2017.152
  66. Tecalco-Cruz AC, Ramírez-Jarquín JO. Polyubiquitination inhibition of Estrogen receptor alpha and its implications in breast cancer. Kd 9, World Journal of Clinical Oncology. 2018. lk 60–70. https://doi.org/10.5306/wjco.v9.i4.60
  67. Berry NB, Fan M, Nephew KP. Estrogen receptor-α hinge-region lysines 302 and 303 regulate receptor degradation by the proteasome. Mol Endocrinol. 2008;22(7):1535–51. https://doi.org/10.1210/me.2007-0449
  68. Long X, Nephew KP. Fulvestrant (ICI 182,780)-dependent interacting proteins mediate immobilization and degradation of estrogen receptor-α. J Biol Chem. 2006;281(14):9607–15. https://doi.org/10.1074/jbc.M510809200
  69. Yeh WL, Shioda K, Coser KR, Rivizzigno D, McSweeney KR, Shioda T. Fulvestrant-Induced Cell Death and Proteasomal Degradation of Estrogen Receptor α Protein in MCF-7 Cells Require the CSK c-Src Tyrosine Kinase. PLoS One. 2013;8(4). https://doi.org/10.1371/journal.pone.0060889
  70. Deeks ED. Fulvestrant: A Review in Advanced Breast Cancer Not Previously Treated with Endocrine Therapy. Drugs. 2018;78(1):131–7. https://doi.org/10.1007/s40265-017-0855-5
  71. De Savi C, Bradbury RH, Rabow AA, Norman RA, De Almeida C, Andrews DM, et al. Optimization of a Novel Binding Motif to (E)-3-(3,5-Difluoro-4-((1R,3R)-2-(2-fluoro-2-methylpropyl)-3-methyl-2,3,4,9-tetrahydro-1H-pyrido[3,4-b]indol-1-yl)phenyl)acrylic Acid (AZD9496), a Potent and Orally Bioavailable Selective Estrogen Receptor Downregu. J Med Chem. 2015;58(20):8128–40. https://doi.org/10.1021/acs.jmedchem.5b00984
  72. Wardell SE, Nelson ER, Chao CA, McDonnell DP. Bazedoxifene exhibits antiestrogenic activity in animal models of tamoxifen-resistant breast cancer: Implications for treatment of advanced disease. Clin Cancer Res. 2013;19(9):2420–31. https://doi.org/10.1158/1078-0432.CCR-12-3771
  73. Garner F, Shomali M, Paquin D, Lyttle CR, Hattersley G. RAD1901: A novel, orally bioavailable selective estrogen receptor degrader that demonstrates antitumor activity in breast cancer xenograft models. Anticancer Drugs. 2015;26(9):948–56. https://doi.org/10.1097/CAD.0000000000000271
  74. Lai A, Kahraman M, Govek S, Nagasawa J, Bonnefous C, Julien J, et al. Identification of GDC-0810 (ARN-810), an Orally Bioavailable Selective Estrogen Receptor Degrader (SERD) that Demonstrates Robust Activity in Tamoxifen-Resistant Breast Cancer Xenografts. J Med Chem. 2015;58(12):4888–904. https://doi.org/10.1021/acs.jmedchem.5b00054
  75. Guo S, Zhang C, Bratton M, Mottamal M, Liu J, Ma P, et al. ZB716, a steroidal selective estrogen receptor degrader (SERD), is orally efficacious in blocking tumor growth in mouse xenograft models. Oncotarget. 2018;9(6):6924–37. https://doi.org/10.18632/oncotarget.24023
  76. Tria GS, Abrams T, Baird J, Burks HE, Firestone B, Gaither LA, et al. Discovery of LSZ102, a potent, orally bioavailable selective estrogen receptor degrader (SERD) for the treatment of estrogen receptor positive breast cancer. J Med Chem. 2018;61(7):2837–64. https://doi.org/10.1021/acs.jmedchem.7b01682
  77. Jiang Y, Deng Q, Zhao H, Xie M, Chen L, Yin F, et al. Development of Stabilized Peptide-Based PROTACs against Estrogen Receptor α. ACS Chem Biol. 2018;13(3):628–35. https://doi.org/10.1021/acschembio.7b00985
  78. Li Y, Zhang S, Zhang J, Hu Z, Xiao Y, Huang J, et al. Exploring the PROTAC degron candidates: OBHSA with different side chains as novel selective estrogen receptor degraders (SERDs). Eur J Med Chem. 2019;172:48–61. https://doi.org/10.1016/j.ejmech.2019.03.058
  79. Dragovich PS, Adhikari P, Blake RA, Blaquiere N, Chen J, Cheng YX, et al. Antibody-mediated delivery of chimeric protein degraders which target estrogen receptor alpha (ERα). Bioorganic Med Chem Lett. 2020;30(4). https://doi.org/10.1016/j.bmcl.2019.126907
  80. Qi SM, Dong J, Xu ZY, Cheng XD, Zhang WD, Qin JJ. PROTAC: An Effective Targeted Protein Degradation Strategy for Cancer Therapy. Front Pharmacol. 2021;12. https://doi.org/10.3389/fphar.2021.692574
  81. Lin X, Xiang H, Luo G. Targeting estrogen receptor α for degradation with PROTACs: A promising approach to overcome endocrine resistance. Kd 206, European Journal of Medicinal Chemistry. 2020. https://doi.org/10.1016/j.ejmech.2020.112689
  82. Sakamoto KM, Kim KB, Kumagai A, Mercurio F, Crews CM, Deshaies RJ. Protacs: Chimeric molecules that target proteins to the Skp1-Cullin-F box complex for ubiquitination and degradation. Proc Natl Acad Sci U S A. 2001;98(15):8554–9. https://doi.org/10.1073/pnas.141230798
  83. Hu J, Hu B, Wang M, Xu F, Miao B, Yang CY, et al. Discovery of ERD-308 as a Highly Potent Proteolysis Targeting Chimera (PROTAC) Degrader of Estrogen Receptor (ER). J Med Chem. 2019;62(3):1420–42. https://doi.org/10.1021/acs.jmedchem.8b01572
  84. Dai Y, Yue N, Gong J, Liu C, Li Q, Zhou J, et al. Development of cell-permeable peptide-based PROTACs targeting estrogen receptor α. Eur J Med Chem. 2020;187. https://doi.org/10.1016/j.ejmech.2019.111967
  85. Edmondson SD, Yang B, Fallan C. Proteolysis targeting chimeras (PROTACs)in ‘beyond rule-of-five’ chemical space: Recent progress and future challenges. Kd 29, Bioorganic and Medicinal Chemistry Letters. 2019. lk 1555–64. https://doi.org/10.1016/j.bmcl.2019.04.030
  86. Pesiri V, Di Muzio E, Polticelli F, Acconcia F. Selective binding of estrogen receptor α to ubiquitin chains. IUBMB Life [Internet]. juuli 2016;68(7):569–77. Available at: https://onlinelibrary.wiley.com/doi/10.1002/iub.1514; https://doi.org/10.1002/iub.1514
  87. Bafna D, Ban F, Rennie PS, Singh K, Cherkasov A. Computer-aided ligand discovery for estrogen receptor alpha. Kd 21, International Journal of Molecular Sciences. 2020. lk 1–49. https://doi.org/10.3390/ijms21124193
  88. Kargbo RB. PROTAC-Mediated Degradation of Estrogen Receptor in the Treatment of Cancer. ACS Med Chem Lett. 2019;10(10):1367–9. https://doi.org/10.1021/acsmedchemlett.9b00397
  89. Sakamoto KM, Kim KB, Verma R, Ransick A, Stein B, Crews CM, et al. Development of Protacs to target cancer-promoting proteins for ubiquitination and degradation. Mol Cell Proteomics. 2003;2(12):1350–8. https://doi.org/10.1074/mcp.T300009-MCP200
  90. Rodriguez-Gonzalez A, Cyrus K, Salcius M, Kim K, Crews CM, Deshaies RJ, et al. Targeting steroid hormone receptors for ubiquitination and degradation in breast and prostate cancer. Oncogene. 2008;27(57):7201–11. https://doi.org/10.1038/onc.2008.320
  91. Bargagna-Mohan P, Baek SH, Lee H, Kim K, Mohan R. Use of PROTACS as molecular probes of angiogenesis. Bioorganic Med Chem Lett. 2005;15(11):2724–7. https://doi.org/10.1016/j.bmcl.2005.04.008
  92. Okuhira K, Demizu Y, Hattori T, Ohoka N, Shibata N, Nishimaki-Mogami T, et al. Development of hybrid small molecules that induce degradation of estrogen receptor-alpha and necrotic cell death in breast cancer cells. Cancer Sci. 2013;104(11):1492–8. https://doi.org/10.1111/cas.12272
  93. Tecalco-Cruz AC, Ramírez-Jarquín JO. Mechanisms that Increase Stability of Estrogen Receptor Alpha in Breast Cancer. Kd 17, Clinical Breast Cancer. 2017. lk 1–10. https://doi.org/10.1016/j.clbc.2016.07.015