Issue |
Vis Cancer Med
Volume 3, 2022
|
|
---|---|---|
Article Number | 3 | |
Number of page(s) | 8 | |
DOI | https://doi.org/10.1051/vcm/2022004 | |
Published online | 12 September 2022 |
Review Article
CBP/p300 bromodomain: new promising epigenetic target
1
HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo 315000, China
2
Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo 315000, China
3
International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
4
Guangdong Provincial Key Laboratory of Biocomputing, Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510530, China
5
China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou, 510530, China
6
State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
* Corresponding author: xu_yong@gibh.ac.cn
Received:
13
July
2022
Accepted:
17
August
2022
CREB (cAMP responsive element binding protein) binding protein (CBP) and adenovirus E1A-associated 300 kDa protein (p300) are histone acetyltransferases, which are necessary for multiple cellular processes. Thus, CBP/p300 are promising potential antitumor targets. To date, despite various small molecule inhibitors of CBP/p300 bromodomain (BRD) having been reported, no specific inhibitor was approved by U.S. Food and Drug Administration (FDA). In this review, we described the discovery, optimization, binding mode evaluation, selectivity and potency evaluation, and therapeutic opportunities of our CBP/p300 bromodomain inhibitors, aiming to inspire new inhibitor design and advance drug discovery research in this field. One video presents the development of CBP/p300 bromodomain inhibitors.
Key words: CBP/p300 / Bromodomain / Inhibitor / Prostate cancer
© The Authors, published by EDP Sciences, 2022
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Introduction
CREB (cAMP responsive element binding protein) binding protein (CBP) and p300 are lysine acetyl transferases that play a key role as transcriptional coactivators in cells [1–6]. CBP and p300 show high structural similarity and functional redundancy (Figure 1) [7, 8]. Several studies have shown that CBP and p300 (hereafter together referred to as “CBP”) have been involved in masses of diverse biological functions including proliferation, cell cycle regulation, apoptosis, differentiation, and DNA damage response. Thus, overexpression or mutations of CBP have been implicated in several diseases, especially leukemia and prostate cancer [9–16].
![]() |
Figure 1 The domain structure and percent sequence identity of CBP and p300. |
CBP and p300 are large proteins with ∼2400 amino acids and share ~75% sequence similarity (Figure 1) [11, 17, 18]. CBP and p300 consist of several highly conserved domains, including a catalytic histone acetyltransferase (HAT) domain that acetylates histones and other proteins, and an adjacent bromodomain that recognizes acetylated histone tails [19]. These two domains have been recognized as promising targets for antitumor treatment [20–22].
Bromodomains (BRDs) are named after the Drosophila gene Brahma where the BRD sequence motif was first identified in the early 1990s [23, 24]. Since then, BRDs have been identified in a variety of proteins, including transcriptional coactivators [25–27], methyltransferases [28, 29], and HATs [30]. Ten years later, Zeng and Zhou defined that BRD can specifically recognize the acetylated lysine (KAc) mark on histones [31]. BRDs are made up of approximately 110 amino acids. Regardless of the diversity in their sequence, BRDs share a conserved specialized structure that consists of four alpha-helices (αZ, αA, αB, and αC), linked by variable loop regions (ZA loop and BC loop), forming the largely hydrophobic KAc binding pocket (Figure 2A and Video 1) [32]. BRDs bind the KAc N-acetyl group via a hydrogen bond between the acetyl carbonyl and the NH2 of a highly-conserved asparagine residue (Asn1168 in CBP) and a water-mediated hydrogen bond from the acetyl carbonyl to the phenolic hydroxyl group of a conserved tyrosine (Tyr1125 in CBP) [33, 34]. In addition, five conserved water molecules present at the bottom of the pocket have a significant impact on compound’s binding affinity [35]. For CBP bromodomain, it should be kept in mind that small molecules gaining interactions to “LPF shelf” (Leu1109, Pro1110, Phe1111) and Arg1173 can be used to enhance potency and selectivity [36–39].
![]() |
Figure 2 Bromodomain structure and phylogenetic tree. (A). Structure of an acetyl lysine and CBP bromodomain complex. CBP bromodomain-fold depicting the four α-helices (αZ, αA, αB, αC) and the endogenous ligand KAc (derived from PDB 5GH9). (B). Phylogenetic tree of the human bromodomain proteins. |
Video 1
New scaffolds of CBP bromodomain inhibitors, a virtual screening strategy was used as shown. |
In the human proteome, there are 61 BRDs found in 46 different proteins [40] (Figure 2B). Moreover, based on sequence and structural similarity, human BRDs can be grouped into eight subfamilies with very different functions, and several of them have been implicated in human diseases, mainly cancer, and inflammation [41]. The bromodomain present in CBP, belongs to BRD family III. CBP has a single bromodomain along with several protein interaction motifs. They are associated with a number of biological functions and act as transcriptional coactivators [42].
In recent years, the development of CBP bromodomain inhibitors has attracted great attention from research institutions and major pharmaceutical companies due to their potential therapeutic value in the treatment of cancers and other diseases [36, 37, 39, 43–55]. Known small-molecule CBP bromodomain inhibitors mainly include 3,5-dimethylisoxazole (SGC-CBP30) [36], dihydroquinoxalinone (compound 2) [43], benzoxazepine (I-CBP112) [55], benzodiazepinone (CPI-637) [47], and pyrazolopiperidine (GEN-272 and GNE-781) [39, 45], etc. (Figure 3). The first potent inhibitor for CBP bromodomain was acetyl-lysine mimetic moiety based on 3,5-dimethylisoxazole, which was later optimized as SGC-CBP30 [36, 56]. Other CBP bromodomain probes, such as 2, I-CBP112, and CPI-637, displayed good selectivity for CBP bromodomain over other bromodomains and showed significant inhibition in multiple tumor cells demonstrating the feasibility of targeting CBP bromodomain for cancer therapy. GNE-049 is a potent and selective CBP inhibitor developed by Genentech, which suppresses prostate cancer growth both in vitro and in vivo [16, 39]. CCS1477, developed by CellCentric, is the first CBP bromodomain inhibitor entering clinical trials. Recently, the phase I/IIa clinical trial was performed to evaluate its safety and pharmacokinetics in advanced drug-resistant prostate cancer and hematological tumors [53, 57]. However, there is no CBP bromodomain inhibitor approved by the U.S. Food and Drug Administration (FDA) to date. Therefore, it is an urgent need to discover novel, potent and specific CBP bromodomain inhibitors with different chemotypes to explore the full therapeutic potential of various human cancers.
![]() |
Figure 3 Structures of representative CBP bromodomain inhibitors. |
In the first part of this review, we summarize the development of our CBP bromodomain inhibitors. The structure-activity relationship studies (SARs), binding models analysis, and biochemical evaluation are also addressed. In the second part, we discuss the opportunities and challenges as well as novel approaches and strategies in the generation of chemical tools and probes for CBP bromodomain. Hopefully, the perspective will provide new insights for pharmaceutical workers to develop more drug-like CBP inhibitors.
CBP bromodomain inhibitors
To obtain new scaffolds of CBP bromodomain inhibitors, a virtual screening strategy [58–61] was used as shown in Figure 4 shown in Video 1. First, the SPECS database was filtered [50]. Then, the resulting fragments were screened through molecular docking, cluster analysis, and visual inspection. At last, 13 representative compounds were selected and purchased for biological evaluation. Among them, compound QP6 exhibited modest inhibitory activity against CBP with an IC50 value of 16.73 μM and exhibited good selectivity over other bromodomain-containing proteins. The molecular docking study indicated that compound QP6 formed a critical hydrogen bonding with Asn1168, mimicking the acetylated lysine interaction observed between CBP and its substrate. In addition, the propionic acid group formed a hydrogen bond with Arg1173. Further optimization was performed based on compound QP6.
![]() |
Figure 4 Experimental validation for compounds QP6 and QP7. Predicted binding modes of QP6 (A) and QP7 (B) in complex with CBP protein. The inhibitory curves of QP6 (C) and QP7. Half-maximum inhibitory concentration (IC50) values for QP6 (C) and QP7 (D) in AlphaScreen assay. (E) Selectivity profile of QP6 and QP7. Bromodomains’ selectivity profiles were determined by AlphaScreen. |
To ensure small molecule inhibitors can reach the LPF shelf and form further interactions with CBP, the propionic acid group was replaced with larger groups. Replacement of the propionic acid with a heterocyclic group yielded conformation constrained analog QP45, with a slight increase in inhibitory activity (IC50 = 6.8 μM). The cocrystal structure of the QP45/CBP bromodomain showed that QP45 plugged into the narrow pocket with its 1-(1H-indol-3-yl)ethanone group interacting with the conserved residues Asn1168 and Tyr1125 (Figure 5). The 1-propionylpiperidine-3-carboxylic acid group protruded out of the pocket and reached the LPF shelf, forming further interactions with CBP. Moreover, the carboxyl group was involved in a canonical hydrogen bond with Arg1173. The crystal structure also revealed that the carbonyl of the 1-propionylpiperidine-3-carboxylic acid group interacts with Arg1173 through a water-mediated hydrogen bond. To obtain more potent compounds through water-mediated hydrogen bonds, the head group 1-(1H-indol-3-yl)ethanone was replaced with 1-(1H-indol-1-yl)ethanone with an amide linker attached, which afforded compound QP82 with an IC50 value of 1.69 μM. This finding demonstrated that the carboxyl group of compound QP82 was critical for binding activity.
![]() |
Figure 5 Flowchart summarizing the design of novel 1-acetylindolederivatives. |
Then, the SAR on the benzene ring was explored. The findings suggested that small groups at the ortho-position were favorable. For the meta-position, the furan group (QP94) is a favorable moiety to improve the activity (AlphaScreen IC50 = 0.65 μM). The predicted binding mode of QP94 with CBP (Figure 5) indicated that there is some space near the BC channel and the C-6 position of the indole ring was explored. Hydroxyl (OH) and methoxy (OMe) groups indicted 4 times more potent than QP94, while longer linkers were found detrimental, likely due to the requirement of small size for the BC channel. To find a more effective CBP bromodomain inhibitor, the furan group was replaced by a 1-methyl-1-H-pyrazole motif, resulting in compound QP175 as a potent CBP bromodomain inhibitor (AlphaScreen IC50 = 0.037 μM). However, its relatively low cellular potency and unsatisfactory metabolic stability limit its further development.
To improve metabolic stability, we replaced the 1-(1H-indol-1-yl)ethan-1-one in QP175 with 1-(indolizin-3-yl)ethan-1-one, resulting in QP180 (Figure 6A) [51, 54]. In comparison to QP175, compound QP180 showed a significant potency improvement against CBP bromodomain with an IC50 value of 9.80 nM in the HTRF assay and 18-fold more potent than QP175 (HTRF, IC50 = 178.15 nM). However, QP180 did not show any cellular activity, which may be due to the limitation of cell permeability caused by highly polar carboxylic acid moiety. Therefore, further optimization was focused on improving cellular potency, leading to molecule QP210 with favorable cellular activity (IC50 = 5 μM) in C4-2B cells. Unfortunately, QP210 displayed poor oral bioavailability (F = 0.58%), which limited the use of this compound for in vivo evaluation. Further structural modifications were then conducted to improve the pharmacokinetic profiles of QP210. Thus, compound QP268 was obtained and displayed promising pharmacokinetic properties. The compound exhibited a reasonable maximum concentration (Cmax = 2037.80 μg/L), and acceptable half time (t1/2 = 3.24 h). Unfortunately, it still showed poor oral bioavailability (F = 6.18%) in SD rats at an oral dose of 25 mg/kg.
![]() |
Figure 6 Design and structural optimization strategies of 1-(indolizin-3-yl)ethan-1-ones. (A) Design strategy of coupling 3-acetylindolizine derivatives. (B-D) Crystal structure of QP268 bound to CBP bromodomain (PDB ID: 7EVJ). |
The co-crystal structure of compound QP268-CBP bromodomain is consistent with our computational prediction (Figures 6B–6D). The acetyl group of QP268 forms a hydrogen bond with the conserved residue Asn1168 and interacts with the conserved residue Tyr1125 through a water-mediated hydrogen bond. An additional hydrogen bond was also observed, the oxygen atom of the –CH2OH group formed a direct hydrogen bond with residue Leu1109. The X-ray structure analysis also revealed that the –CH2OH moiety of compound QP268 was exposed to a partially opened hydrophobic pocket. It provides an ideal position for further structural optimization to improve the pharmacokinetic profiles without obviously affecting CBP bromodomain inhibitory potency. Therefore, compound QP284 (Y08284) harboring a methyl group to –CH2OH group was designed and synthesized. QP284 displayed strong CBP bromodomain binding affinity with an IC50 value of 0.0042 μM (HTRF) and good cellular activity with an IC50 value of 6.43 μM in C4-2B cells. Additionally, molecule QP284 indicated favorable liver microsome stability, good cell permeability, and acceptable oral bioavailability (F = 25.9%). Moreover, an examination of a panel of 16 bromodomain-containing proteins indicated that compound QP284 displayed good target selectivity for CBP over other bromodomains.
Based on the strong potency, reasonable target selectivity, and good drug-like properties, QP284 was further used to explore the role of CBP bromodomain in the biology of prostate cancer [54]. The results indicated that QP284 significantly inhibited the proliferation of AR-expressing prostate cancer cell lines (C4-2B, LNCaP, and 22Rv1). Compound QP284 can inhibit colony formation and migration of prostate cancer cells. Also, compound QP284 could repress AR target gene expression, as well as c-Myc expression in prostate cancer cell lines. Furthermore, QP284 showed significant antitumor activity in a prostate cancer xenograft model. These findings indicated that CBP bromodomain is a potential target for prostate cancer treatment.
Perspective and summary
To date, much evidence has shown that CBP bromodomain plays a key role in the occurrence and development of a variety of diseases, including prostate cancer, leukemia, and breast cancer. Besides, clinical data have also indicated that the expression level of CBP bromodomain has a close relationship with the progression and prognosis of diseases [14]. Therefore, CBP is considered a potential and promising therapeutic target for human diseases. Over the past decades, researchers have made great efforts to develop novel CBP bromodomain inhibitors and explain the biological functions of CBP bromodomain. CBP bromodomain inhibitors have indicated potent inhibitory activity against some diseases. Until now, at least six representative scaffolds of CBP bromodomain inhibitors have been reported, some of which have weak affinities and nonspecific targeting. Therefore, there is an urgent need to develop more potent and selective CBP bromodomain inhibitors with novel scaffolds and drug-like properties to explore their therapeutic potential in different human diseases. Moreover, CBP-selective and p300-selective bromodomain inhibitors should be designed to study their therapeutic value in the same and/or different diseases.
Considering the current issues, many novel types of CBP inhibitors, such as PROTAC (proteolysis targeting chimeras) and allosteric inhibitors, are urgently needed. A PROTAC is a bifunctional small molecule composed of two ligands and a linker [62–64]. Rather than acting as a traditional molecule, a PROTAC works by inducing selective intracellular proteolysis. Currently, scientists have shown that degradation of target proteins could avoid drug resistance more effectively and has better anticancer effects than inhibition of target proteins. However, only one CBP bromodomain PROTAC molecule (dCBP-1) has been developed. Therefore, PROTACS deserves further investigation as a specific method for the development of novel CBP bromodomain inhibitors. Selective bromodomain inhibitors discovered all targeted the classic KAc recognition pocket, by which they could not distinguish with other bromodomains, increasing the possibility of off-target. Allosteric inhibitors could change the conformation of the bromodomain proteins to prevent the interactions between bromodomains and acetyl-lysine of histones through binding with the allosteric site. Allosteric inhibitors are common in kinases but rare in bromodomains. Recently, Liu et al. reported a BRD4(1)-selective inhibitor ZL0590 which is the first-in-class BRD4(1) inhibitor with a nonacetylated lysine binding site on BRD4(1) [65]. The result provided a structural basis for the discovery of allosteric inhibitors of CBP bromodomain.
In summary, considering the important role of CBP bromodomain in various diseases and the research status of its inhibitors, it is still necessary to develop more potent and more specific CBP bromodomain inhibitors, PROTACs, etc. In the review, we summarized the development of CBP bromodomain inhibitors in our group, including the SARs, binding modes, and pharmacological profiles. We believe that with further studies on the small molecule inhibitors, PROTACS, and crystal structures, more efficient and selective CBP inhibitors will be reported for the treatment of diseases with CBP as a potential therapeutic target.
Conflicts of interest
The authors declare that they do not have any conflict of interest.
Acknowledgments
This work was supported in part by grants from the National Natural Science Foundation of China (grant 22007088) the Guangdong Basic and Applied Basic Research Foundation (grant 2021A1515010085), the Science and Technology Program of Guangzhou (Grant 202102080016). The authors also gratefully acknowledge support from the Guangzhou Branch of the Supercomputing Center of Chinese Academy of Sciences.
References
- Bedford DC, Kasper LH, Fukuyama T, et al. Target gene context influences the transcriptional requirement for the KAT3 family of CBP and p300 histone acetyltransferases. Epigenetics. 2010;5(1):9–15. [CrossRef] [PubMed] [Google Scholar]
- Chan HM, La Thangue NB. p300/CBP proteins: HATs for transcriptional bridges and scaffolds. Journal of Cell Science. 2001;114(Pt 13):2363–2373. [CrossRef] [PubMed] [Google Scholar]
- Gerona-Navarro G, Yoel R, Mujtaba S, et al. Rational design of cyclic peptide modulators of the transcriptional coactivator CBP: a new class of p53 inhibitors. Journal of the American Chemical Society. 2011;133(7):2040–2043. [CrossRef] [PubMed] [Google Scholar]
- Goodman RH, Smolik S. CBP/p300 in cell growth, transformation, and development. Genes & Development. 2000;14(13):1553–1577. [CrossRef] [PubMed] [Google Scholar]
- Kalkhoven E. CBP and p300: HATs for different occasions. Biochemical Pharmacology. 2004;68(6):1145–1155. [CrossRef] [PubMed] [Google Scholar]
- Vo N, Goodman RH. CREB-binding protein and p300 in transcriptional regulation. Journal of Biological Chemistry. 2001;276(17):13505–13508. [CrossRef] [Google Scholar]
- Janknecht R. The versatile functions of the transcriptional coactivators p300 and CBP and their roles in disease. Histology and Histopathology. 2002;17(2):657–668. [PubMed] [Google Scholar]
- Wang L, Tang Y, Cole PA, Marmorstein R. Structure and chemistry of the p300/CBP and Rtt109 histone acetyltransferases: implications for histone acetyltransferase evolution and function. Current Opinion in Structural Biology. 2008;18(6):741–747. [CrossRef] [PubMed] [Google Scholar]
- Panagopoulos I, Fioretos T, Isaksson M, et al. Fusion of the MORF and CBP genes in acute myeloid leukemia with the t(10;16)(q22;p13). Human Molecular Genetics. 2001;10(4):395–404. [CrossRef] [PubMed] [Google Scholar]
- Bouchal J, Santer FR, Hoschele PP, et al. Transcriptional coactivators p300 and CBP stimulate estrogen receptor-beta signaling and regulate cellular events in prostate cancer. Prostate. 2011;71(4):431–437. [CrossRef] [PubMed] [Google Scholar]
- Wang F, Marshall CB, Ikura M. Transcriptional/epigenetic regulator CBP/p300 in tumorigenesis: structural and functional versatility in target recognition. Cellular and Molecular Life Sciences. 2013;70(21):3989–4008. [CrossRef] [PubMed] [Google Scholar]
- Dutta R, Tiu B, Sakamoto KM. CBP/p300 acetyltransferase activity in hematologic malignancies. Molecular Genetics and Metabolism. 2016;119(1–2):37–43. [CrossRef] [PubMed] [Google Scholar]
- Fujisawa T, Filippakopoulos P. Functions of bromodomain-containing proteins and their roles in homeostasis and cancer. Nature Reviews Molecular Cell Biology. 2017;18(4):246–262. [CrossRef] [PubMed] [Google Scholar]
- Iyer NG, Ozdag H, Caldas C. p300/CBP and cancer. Oncogene. 2004;23(24):4225–4231. [CrossRef] [PubMed] [Google Scholar]
- Ianculescu I, Wu DY, Siegmund KD, et al. Selective roles for cAMP response element-binding protein binding protein and p300 protein as coregulators for androgen-regulated gene expression in advanced prostate cancer cells. Journal of Biological Chemistry. 2012;287(6):4000–4013. [CrossRef] [Google Scholar]
- Jin L, Garcia J, Chan E, et al. Therapeutic targeting of the CBP/p300 bromodomain blocks the growth of castration-resistant prostate cancer. Cancer Research. 2017;77(20):5564–5575. [CrossRef] [PubMed] [Google Scholar]
- Schiltz RL, Mizzen CA, Vassilev A, et al. Overlapping but distinct patterns of histone acetylation by the human coactivators p300 and PCAF within nucleosomal substrates. Journal of Biological Chemistry. 1999;274(3):1189–1192. [CrossRef] [Google Scholar]
- Dancy BM, Cole PA. Protein lysine acetylation by p300/CBP. Chemical Reviews. 2015;115(6):2419–2452. [CrossRef] [PubMed] [Google Scholar]
- Ragvin A, Valvatne H, Erdal S, et al. Nucleosome binding by the bromodomain and PHD finger of the transcriptional cofactor p300. Journal of Molecular Biology. 2004;337(4):773–788. [CrossRef] [PubMed] [Google Scholar]
- Dekker FJ, Haisma HJ. Histone acetyl transferases as emerging drug targets. Drug Discovery Today. 2009;14(19–20):942–948. [CrossRef] [PubMed] [Google Scholar]
- Vidler LR, Brown N, Knapp S, et al. Druggability analysis and structural classification of bromodomain acetyl-lysine binding sites. Journal of Medicinal Chemistry. 2012;55(17):7346–7359. [CrossRef] [PubMed] [Google Scholar]
- Delvecchio M, Gaucher J, Aguilar-Gurrieri C, et al. Structure of the p300 catalytic core and implications for chromatin targeting and HAT regulation. Nature Structural & Molecular Biology. 2013;20(9):1040–1046. [CrossRef] [PubMed] [Google Scholar]
- Haynes SR, Dollard C, Winston F, et al. The bromodomain: a conserved sequence found in human, Drosophila and yeast proteins. Nucleic Acids Research. 1992;20(10):2603. [CrossRef] [PubMed] [Google Scholar]
- Tamkun JW, Deuring R, Scott MP, et al. brahma: a regulator of Drosophila homeotic genes structurally related to the yeast transcriptional activator SNF2/SWI2. Cell. 1992;68(3):561–572. [CrossRef] [PubMed] [Google Scholar]
- Venturini L, You J, Stadler M, et al. TIF1gamma, a novel member of the transcriptional intermediary factor 1 family. Oncogene. 1999;18(5):1209–1217. [CrossRef] [PubMed] [Google Scholar]
- Jacobson RH, Ladurner AG, King DS, et al. Structure and function of a human TAFII250 double bromodomain module. Science. 2000;288(5470):1422–1425. [CrossRef] [PubMed] [Google Scholar]
- Bres V, Yoh SM, Jones KA. The multi-tasking P-TEFb complex. Current Opinion in Cell Biology. 2008;20(3):334–340. [CrossRef] [PubMed] [Google Scholar]
- Gregory GD, Vakoc CR, Rozovskaia T, et al. Mammalian ASH1L is a histone methyltransferase that occupies the transcribed region of active genes. Molecular and Cellular Biology. 2007;27(24):8466–8479. [CrossRef] [PubMed] [Google Scholar]
- Malik S, Bhaumik SR. Mixed lineage leukemia: histone H3 lysine 4 methyltransferases from yeast to human. FEBS Journal. 2010;277(8):1805–1821. [CrossRef] [Google Scholar]
- Nagy Z, Tora L. Distinct GCN5/PCAF-containing complexes function as co-activators and are involved in transcription factor and global histone acetylation. Oncogene. 2007;26(37):5341–5357. [CrossRef] [PubMed] [Google Scholar]
- Zeng L, Zhou MM. Bromodomain: an acetyl-lysine binding domain. FEBS Letters. 2002;513(1):124–128. [CrossRef] [PubMed] [Google Scholar]
- Jang MK, Mochizuki K, Zhou M, et al. The bromodomain protein BRD4 is a positive regulatory component of P-TEFb and stimulates RNA polymerase II-dependent transcription. Molecular Cell. 2005;19(4):523–534. [CrossRef] [PubMed] [Google Scholar]
- Dawson MA, Prinjha RK, Dittmann A, et al. Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemia. Nature. 2011;478(7370):529–533. [CrossRef] [PubMed] [Google Scholar]
- Sanchez R, Meslamani J, Zhou MM. The bromodomain: from epigenome reader to druggable target. Biochimica et Biophysica Acta. 2014;1839(8):676–685. [CrossRef] [PubMed] [Google Scholar]
- Filippakopoulos P, Knapp S. Targeting bromodomains: epigenetic readers of lysine acetylation. Nature Reviews Drug Discovery. 2014;13(5):337–356. [CrossRef] [PubMed] [Google Scholar]
- Hay DA, Fedorov O, Martin S, et al. Discovery and optimization of small-molecule ligands for the CBP/p300 bromodomains. Journal of the American Chemical Society. 2014;136(26):9308–9319. [CrossRef] [PubMed] [Google Scholar]
- Romero FA, Taylor AM, Crawford TD, et al. Disrupting acetyl-lysine recognition: progress in the development of bromodomain inhibitors. Journal of Medicinal Chemistry. 2016;59(4):1271–1298. [CrossRef] [PubMed] [Google Scholar]
- Bronner SM, Murray J, Romero FA, et al. A unique approach to design potent and selective cyclic adenosine monophosphate response element binding protein, binding protein (CBP) inhibitors. Journal of Medicinal Chemistry. 2017;60(24):10151–10171. [CrossRef] [PubMed] [Google Scholar]
- Romero FA, Murray J, Lai KW, et al. GNE-781, a highly advanced potent and selective bromodomain inhibitor of cyclic adenosine monophosphate response element binding protein, binding protein (CBP). Journal of Medicinal Chemistry. 2017;60(22):9162–9183. [CrossRef] [PubMed] [Google Scholar]
- Filippakopoulos P, Picaud S, Mangos M, et al. Histone recognition and large-scale structural analysis of the human bromodomain family. Cell. 2012;149(1):214–231. [CrossRef] [PubMed] [Google Scholar]
- Muller S, Filippakopoulos P, Knapp S. Bromodomains as therapeutic targets. Expert Reviews in Molecular Medicine. 2011;13:e29 [CrossRef] [PubMed] [Google Scholar]
- Attar N, Kurdistani SK. Exploitation of EP300 and CREBBP lysine acetyltransferases by cancer. Cold Spring Harbor Perspectives in Medicine. 2017;7(3):a026534. [CrossRef] [PubMed] [Google Scholar]
- Rooney TP, Filippakopoulos P, Fedorov O, et al. A series of potent CREBBP bromodomain ligands reveals an induced-fit pocket stabilized by a cation-pi interaction. Angewandte Chemie International Edition in English. 2014;53(24):6126–6130. [CrossRef] [Google Scholar]
- Hammitzsch A, Tallant C, Fedorov O, et al. CBP30, a selective CBP/p300 bromodomain inhibitor, suppresses human Th17 responses. Proceedings of the National Academy of Sciences USA. 2015;112(34):10768–10773. [CrossRef] [PubMed] [Google Scholar]
- Crawford TD, Romero FA, Lai KW, et al. Discovery of a potent and selective in vivo probe (GNE-272) for the bromodomains of CBP/EP300. Journal of Medicinal Chemistry. 2016;59(23):10549–10563. [CrossRef] [PubMed] [Google Scholar]
- Popp TA, Tallant C, Rogers C, et al. Development of selective CBP/P300 benzoxazepine bromodomain inhibitors. Journal of Medicinal Chemistry. 2016;59(19):8889–8912. [CrossRef] [PubMed] [Google Scholar]
- Taylor AM, Cote A, Hewitt MC, et al. Fragment-based discovery of a selective and cell-active benzodiazepinone CBP/EP300 bromodomain inhibitor (CPI-637). ACS Medicinal Chemistry Letters. 2016;7(5):531–536. [CrossRef] [PubMed] [Google Scholar]
- Unzue A, Xu M, Dong J, et al. Fragment-based design of selective nanomolar ligands of the CREBBP bromodomain. Journal of Medicinal Chemistry. 2016;59(4):1350–1356. [CrossRef] [PubMed] [Google Scholar]
- Xu M, Unzue A, Dong J, et al. Discovery of CREBBP bromodomain inhibitors by high-throughput docking and hit optimization guided by molecular dynamics. Journal of Medicinal Chemistry. 2016;59(4):1340–1349. [CrossRef] [PubMed] [Google Scholar]
- Xiang Q, Wang C, Zhang Y, et al. Discovery and optimization of 1-(1H-indol-1-yl)ethanone derivatives as CBP/EP300 bromodomain inhibitors for the treatment of castration-resistant prostate cancer. European Journal of Medicinal Chemistry. 2018;147:238–252. [CrossRef] [PubMed] [Google Scholar]
- Zou LJ, Xiang QP, Xue XQ, et al. Y08197 is a novel and selective CBP/EP300 bromodomain inhibitor for the treatment of prostate cancer. Acta Pharmacologica Sinica. 2019;40(11):1436–1447. [CrossRef] [PubMed] [Google Scholar]
- Muthengi A, Wimalasena VK, Yosief HO, et al. Development of dimethylisoxazole-attached imidazo[1,2-a]pyridines as potent and selective CBP/P300 inhibitors. Journal of Medicinal Chemistry. 2021;64(9):5787–5801. [CrossRef] [PubMed] [Google Scholar]
- Welti J, Sharp A, Brooks N, et al. Targeting the p300/CBP axis in lethal prostate cancer. Cancer Discovery. 2021;11(5):1118–1137. [CrossRef] [PubMed] [Google Scholar]
- Xiang Q, Wang C, Wu T, et al. Design, synthesis, and biological evaluation of 1-(indolizin-3-yl)ethan-1-ones as CBP bromodomain inhibitors for the treatment of prostate cancer. Journal of Medicinal Chemistry. 2022;65(1):785–810. [CrossRef] [PubMed] [Google Scholar]
- Picaud S, Fedorov O, Thanasopoulou A, et al. Generation of a selective small molecule inhibitor of the CBP/p300 bromodomain for leukemia therapy. Cancer Research. 2015;75(23):5106–5119. [CrossRef] [PubMed] [Google Scholar]
- Hay D, Fedorov O, Filippakopoulos P, et al. The design and synthesis of 5- and 6-isoxazolylbenzimidazoles as selective inhibitors of the BET bromodomains. Medchemcomm. 2013;4(1):140–144. [CrossRef] [PubMed] [Google Scholar]
- Pegg NA, Taddei DMA, Onions ST, et al. Isoxazolyl substituted benzimidazoles. US20180127402A1. 2018. [Google Scholar]
- Zhang Y, Xue X, Jin X, et al. Discovery of 2-oxo-1,2-dihydrobenzo[cd]indole-6-sulfonamide derivatives as new RORgamma inhibitors using virtual screening, synthesis and biological evaluation. European Journal of Medicinal Chemistry. 2014;78:431–441. [CrossRef] [PubMed] [Google Scholar]
- Song Y, Xue X, Wu X et al. Identification of N-phenyl-2-(N-phenylphenylsulfonamido)acetamides as new RORgamma inverse agonists: Virtual screening, structure-based optimization, and biological evaluation. European Journal of Medicinal Chemistry. 2016;116:13–26. [CrossRef] [PubMed] [Google Scholar]
- Xue X, Zhang Y, Liu Z, et al. Discovery of benzo[cd]indol-2(1H)-ones as potent and specific bet bromodomain inhibitors: structure-based virtual screening, optimization, and biological evaluation. Journal of Medicinal Chemistry. 2016;59(4):1565–1579. [CrossRef] [PubMed] [Google Scholar]
- Zhou Y, Nie T, Zhang Y, et al. The discovery of novel and selective fatty acid binding protein 4 inhibitors by virtual screening and biological evaluation. Bioorganic & Medicinal Chemistry. 2016;24(18):4310–4317. [CrossRef] [PubMed] [Google Scholar]
- Toure M, Crews CM. Small-molecule PROTACS: New approaches to protein degradation. Angewandte Chemie International Edition in English. 2016;55(6):1966–1973. [CrossRef] [Google Scholar]
- Lai AC, Crews CM. Induced protein degradation: an emerging drug discovery paradigm. Nature Reviews Drug Discovery. 2017;16(2):101–114. [CrossRef] [PubMed] [Google Scholar]
- Pettersson M, Crews CM. PROteolysis TArgeting Chimeras (PROTACs) – Past, present and future. Drug Discovery Today: Technologies. 2019;31:15–27. [CrossRef] [Google Scholar]
- Liu Z, Li Y, Chen H, et al. Discovery, X-ray crystallography, and anti-inflammatory activity of bromodomain-containing protein 4 (BRD4) BD1 inhibitors targeting a distinct new binding site. Journal of Medicinal Chemistry. 2022;65(3):2388–2408. [CrossRef] [PubMed] [Google Scholar]
Cite this article as: Xiang Q, Zhou Y, Zhang Y & Xu Y. CBP/p300 bromodomain: new promising epigenetic target. Visualized Cancer Medicine. 2022. 3, 3.
All Figures
![]() |
Figure 1 The domain structure and percent sequence identity of CBP and p300. |
In the text |
![]() |
Figure 2 Bromodomain structure and phylogenetic tree. (A). Structure of an acetyl lysine and CBP bromodomain complex. CBP bromodomain-fold depicting the four α-helices (αZ, αA, αB, αC) and the endogenous ligand KAc (derived from PDB 5GH9). (B). Phylogenetic tree of the human bromodomain proteins. |
In the text |
![]() |
Figure 3 Structures of representative CBP bromodomain inhibitors. |
In the text |
![]() |
Figure 4 Experimental validation for compounds QP6 and QP7. Predicted binding modes of QP6 (A) and QP7 (B) in complex with CBP protein. The inhibitory curves of QP6 (C) and QP7. Half-maximum inhibitory concentration (IC50) values for QP6 (C) and QP7 (D) in AlphaScreen assay. (E) Selectivity profile of QP6 and QP7. Bromodomains’ selectivity profiles were determined by AlphaScreen. |
In the text |
![]() |
Figure 5 Flowchart summarizing the design of novel 1-acetylindolederivatives. |
In the text |
![]() |
Figure 6 Design and structural optimization strategies of 1-(indolizin-3-yl)ethan-1-ones. (A) Design strategy of coupling 3-acetylindolizine derivatives. (B-D) Crystal structure of QP268 bound to CBP bromodomain (PDB ID: 7EVJ). |
In the text |
All Movies
Video 1
New scaffolds of CBP bromodomain inhibitors, a virtual screening strategy was used as shown. |
|
In the text |
Current usage metrics show cumulative count of Article Views (full-text article views including HTML views, PDF and ePub downloads, according to the available data) and Abstracts Views on Vision4Press platform.
Data correspond to usage on the plateform after 2015. The current usage metrics is available 48-96 hours after online publication and is updated daily on week days.
Initial download of the metrics may take a while.