Europe PMC

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


Bromodomain and extraterminal (BET) proteins bind acetylated lysine residues in histones and nonhistone proteins via tandem bromodomains and regulate chromatin dynamics, cellular processes, and disease procession. Thus targeting BET proteins is a promising strategy for treating various diseases, especially malignant tumors and chronic inflammation. Many pan-BET small-molecule inhibitors have been described, and some of them are in clinical evaluation. Nevertheless, the limited clinical efficacy of the current BET inhibitors is also evident and has inspired the development of new technologies to improve their clinical outcomes and minimize unwanted side effects. In this Review, we summarize the latest protein characteristics and biological functions of BRD4 as an example of BET proteins, analyze the clinical development status and preclinical resistance mechanisms, and discuss recent advances in BRD4-selective inhibitors, dual-target BET inhibitors, proteolysis targeting chimera degraders, and protein-protein interaction inhibitors.

Free full text 


Logo of nihpaLink to Publisher's site
J Med Chem. Author manuscript; available in PMC 2021 May 8.
Published in final edited form as:
PMCID: PMC8106541
NIHMSID: NIHMS1697913
PMID: 33616410

Targeting Bromodomain and Extraterminal Proteins for Drug Discovery: From Current Progress to Technological Development

Abstract

Bromodomain and extraterminal (BET) proteins bind acetylated lysine residues in histones and nonhistone proteins via tandem bromodomains and regulate chromatin dynamics, cellular processes, and disease procession. Thus targeting BET proteins is a promising strategy for treating various diseases, especially malignant tumors and chronic inflammation. Many pan-BET small-molecule inhibitors have been described, and some of them are in clinical evaluation. Nevertheless, the limited clinical efflcacy of the current BET inhibitors is also evident and has inspired the development of new technologies to improve their clinical outcomes and minimize unwanted side effects. In this Review, we summarize the latest protein characteristics and biological functions of BRD4 as an example of BET proteins, analyze the clinical development status and preclinical resistance mechanisms, and discuss recent advances in BRD4-selective inhibitors, dual-target BET inhibitors, proteolysis targeting chimera degraders, and protein−protein interaction inhibitors.

1. INTRODUCTION

In humans, 61 bromodomains, each composed of ~110 amino acids forming four antiparallel α helices (αZ, αA, αB, and αC) and two hydrophobic (ZA and BC) loops, in 46 different proteins have been described.1 Among these, bromodomain and extraterminal (BET) proteins constitute a unique group with four family members, bromodomain-containing protein 4 (BRD4), BRD3, BRD2, and testis-specific BRDT, featuring the presence of two N-terminal tandem bromodomains (BD1 and BD2) and an ET (extraterminal) domain (Figure 1A).2,3 Notably, BRD4 and BRDT contain an extended C-terminal motif (CTM) not found in BRD2 and BRD3. BET proteins are epigenetic “readers” binding specifically to N-ε-acetyllysine (Kac) residues in histones and nonhistone proteins via their BD1 and BD2 bromodomains to engage in chromatin dynamics and transcriptional regulation.4,5 However, the molecular distinction underlying the functional redundancy and unique aspects of each BET protein, with respect to their acetylated histone and nonhistone protein recognition, remains elusive.6 Here we summarize the structural and functional characteristics of BRD4, which is the most understood member of the BET family, as an example of BET proteins.

An external file that holds a picture, illustration, etc.
Object name is nihms-1697913-f0002.jpg

(A) Domain organization of human BET proteins. (B) Conserved sequence of BRD4 BD1 and BRD4 BD2. Key varying residues are marked in different colors. (C) Recruitment of P-TEFb facilitates BRD4-enhanced Pol II elongation. BRD4 binds to Kac sites on histones and recruits BRD4 interactors and the P-TEFb complex, leading to enhanced transcriptional elongation by Ser2-phosphorylated Pol II. (D) BRD4 function in DNA damage response in both NHEJ and HR pathways.

BRD4 has three characterized protein isoforms with different C-terminal amino acid residues one long (L) and two short isoforms, S(a) and S(b) resulting from alternative mRNA splicing.7 Amino acids 1−719 are common among the three isoforms, including two highly conserved N-terminal tandem bromodomains (BD1 and BD2), the N-terminal cluster of phosphorylation sites (NPS), the basic residue-enriched interaction domain (BID), the extraterminal (ET) domain, and the C-terminal cluster of phosphorylation sites (CPS; see Figure 1A). Interestingly, the conformation of BD2 in a closed or open configuration is controlled by the downstream NPS, whose extent of phosphorylation modulated by casein kinase II (CK2)8,9 and protein phosphatase 2A (PP2A)10,11 regulates BRD4 association with different transcriptional regulators such as p53, AP-1, NF-κB, and EN1.7,10,12 Although BET bromodomains are structurally conserved in general, the ZA and BC loops between BD1 and BD2 are slightly different in sequence and length, thus contributing to the context-specific binding of acetylated lysine residues (Figure 1B). Sequence differences between BD1 and BD2 provide a structural basis for designing selective inhibitors of BD1 or BD2. Besides sequence variation, BD1 and BD2 also exhibit functional differences in which BD1 primarily serves as a chromatin-binding module and BD2 is typically used for factor recruitment,13 hence partly accounting for BD1 being indispensable for steady-state gene expression and both BD1 and BD2 being seemingly required for the acute gene response induced by inflammatory stimulation.14 In addition, the conserved BID and phospho-NPS each interact directly with p53 but with different functional outcomes in regulating p53 binding to its target sequences.8 The ET domain of BRD4 interacts with select chromatin regulators via specific structural contacts.15 A unique CTM present in BRD4-L but absent in BRD4-S(a) and BRD4-S(b) functions to recruit transcriptional regulators, such as positive transcription elongation factor b (P-TEFb)16,17 and human papillomavirus (HPV)-encoded E2 protein that is capable of inhibiting AP-1-dependent HPV transcription via specific E2-binding sites.10 Both BRD4-L and BRD4-S(a) can induce liquid−liquid phase separation (LLPS) in vitro and in the cell to mediate transcription complex assembly and regulate gene transcription.1820 Recent research reveals that the expression level of BRD4-S(a), rather than BRD4-L, is closely related to the number and size of BRD4 puncta formed in the cell.20 An oncogenic function of BRD4-S(a) and a tumor-suppressive activity of BRD4-L recently revealed in breast cancer development further indicate the importance of target selectivity in anti-BRD4 drug development.7

BRD4 interacts with both cyclin T1 (cycT1) and cyclin-dependent kinase 9 (CDK9) subunits of the dimeric P-TEFb complex8,16,21 and recruits transcription factors such as MYC,22,23 p53,8,24 AP-1,7,8,10 NF-κB,10,25 TWIST,26 SNAIL,27 EN1,7 YAP1,28,29 TEAD4,7 and NF-YA7 to facilitate RNA polymerase II (Pol II)-dependent transcription (Figure 1C).13,30,31 BRD4 is also implicated in DNA repair to control genome stability and cancer development. The role of BRD4 in the DNA damage response is not limited to nonhomologous end joining (NHEJ)32 and also involves homologous recombination (HR)33 (Figure 1D). BRD4 is associated with R-loop formation, which enhances double-strand break (DSB) formation and engages in the activation of the NHEJ pathway implicated in immunoglobulin class switch recombination32,33 and oncogenic gene rearrangement.34 In contrast, the inhibition of BRD4 induces the loss of C-terminal binding protein-interacting protein (CtIP) involved in DNA end resection and single-strand DNA (ssDNA) generation, resulting in a decrease in the HR competency.35 The involvement of BRD4 in cancer initiation and progression can be attributed to chromosomal translocation leading to the formation of oncogenic fusion drivers (e.g., BRD4-NUT)36 through epigenetic control via the recruitment of cell-type-specific transcriptional regulators37 or by modulating the activity8 or recruitment of cell-cycle-promoting factors such as CDK4/6.38 In NUT midline carcinoma (NMC), BRD4-NUT fusion oncoprotein induces the hyperacetylation of chromatin and the formation of a unique nuclear subcompartment that squelches key transcription components or chromatin regulators, leading to the inhibition of transcription initiation by RNA Pol II.39 Another study identified CDK9 as a kinase implicated in BRD4 hyperphosphorylation that contributes to its oncogenic activity,40 consistent with the finding of enhanced BRD4 phosphorylation due to the reduced PP2A activity during triple-negative breast cancer progression.11 In addition, BRD4 is also involved in blastic plasmacytoid dendritic cell neoplasm (BPDCN),41,42 Cornelia-de-Lange-like syndrome,43 and fragile X syndrome.44 Expanded research on BRD4 protein isoforms with gain- and loss-of-function approaches conducted in various breast cancer cells and mouse models, in conjunction with genome-wide transcriptome and binding profiling, unequivocally establishes the opposing functions of BRD4 isoforms in breast cancer and significantly enhances our understanding of gene targets and pathways associated with cancer progression.7

Over the past decade, numerous pan-BET inhibitors have been generated, and some of them are currently in clinical trials, such as OTX-015, CPI-0610, and I-BET762.45 Although small-molecule pan-BET inhibitors show promising effects in clinical evaluation, there are still problems and challenges to overcome, such as the moderate clinical efflcacy and the occurrence of preclinical resistance.46 Encouragingly, with recent advances in pharmaceutical chemistry and chemical biology, new technologies that facilitate the development of more selective inhibitors or other new-generation inhibitors targeting specific functional aspects of BET proteins have been used to assuage some clinical concerns.

2. CURRENT CLINICAL DEVELOPMENT STATUS AND OBSERVATIONS

In 2017, pan-BET inhibitors available at that time were reviewed in detail regarding their chemical types, activities, and therapeutic potentials.45 At present, some pan-BET inhibitors are in clinical trials for treating different cancers (Table 1). However, the clinical advancement of most pan-BET inhibitors is still in the early stages and mainly focuses on cancer treatment. The results of clinical trials published to date raise several key issues to be considered. First, some inhibitors display toxicity during clinical trials, including dose-limiting toxicity (DLT). The most common adverse effects are thrombocytopenia, diarrhea, fatigue, vomiting, anemia, and hyperbilirubinemia. A commonly observed DLT is thrombocytopenia with undamaged platelet function. Reduced platelet counts were reversible and could be restored within 1 week after drug withdrawal.46 In addition, complications and immunodeficiency are potential undesirable reactions to the use of pan-BET inhibitors.47 The phase I trial of BAY1238097 (NCT02369029) was terminated early due to severe toxic effects, and INCB057643 (NCT02711137) was also discontinued because of safety concerns. Second, several inhibitors exhibited much lower antitumor activity than those observed in preclinical trials. In a dose study of CPI-0610 given to 44 patients with relapsed or refractory lymphoma, only 2 showed a complete response, and 1 had a partial response. In addition, studies with OTX-015 (NCT02698176 and NCT02296476), as reported for I-BET151 (NCT02630251), were forced to terminate because of its limited efflcacy in solid tumors. Pan-BET inhibitors show a significant survival advantage in tumor models, which are often abrogated in transplants of drug-resistant cells.48 The main reason for the lack of clinical efflcacy is likely due to a failure to identify the right group of patients most likely to benefit from pan-BET inhibitor treatment, resulting in ineffectual dosing before DLT occurs. Moreover, modest outcomes may be partly attributed to the development of resistance, diminishing the clinical response to pan-BET inhibitors.49

Table 1.

Pan-BET Inhibitors in Cancer-Related Clinical Trialsa

drugsponsorphaseindicationNUT identifierstatus
I-BET762GlaxoSmithKlineINMC, other solid tumorsNCT01587703completed
ICRPCNCT03150056active, not recruiting
INCT02706535completed
Isolid tumors, lymphomasNCT03925428withdrawn
NMCNCT03702036no longer available
IIhematological malignanciesNCT01943851completed
IIHR+/HER2− advanced or metastatic breast cancerNCT02964507active, not recruiting
IIsolid tumorNCT03266159withdrawn
National Cancer InstituteI/IINUT carcinomaNCT04116359withdrawn
OTX015Merck Sharp & Dohme CorpINMC, TNBC, NSCLC, CRPCNCT02698176terminated
IAML, DLBCLNCT02698189active, not recruiting
Oncoethix GmbHINMC, TNBC, NSCLC, CRPCNCT02259114completed
I/IIAMLNCT02303782withdrawn
IAML, DLBCL, ALL, MMNCT01713582completed
IIGBMNCT02296476terminated
TEN-010Hoffmann-La RocheIAML, myelodysplastic syndromesNCT02308761completed
Isolid tumors, ASTNCT01987362completed
Bay1238097BayerIsolid tumors, lymphomaNCT02369029terminated
CPI-0610University of Texas Southwestern Medical CenterIImalignant peripheral nerve sheath tumorsNCT02986919withdrawn
Constellation PharmaceuticalsIpreviously treated MMNCT02157636completed
Iprogressive lymphomaNCT01949883completed
IImyelofibrosis, MDS/MPN, UNCT02158858recruiting
I-BET151GlaxoSmithKlineImetastatic, unresectable solid tumorsNCT02630251terminated
INCB054329Incyte CorporationI/IIadvanced-stage cancerNCT02431260terminated
metastatic renal cell carcinomaNCT03896815no longer available
INCB057643Incyte CorporationI/IIadvanced-stage cancerNCT02711137terminated
Iprimary myelofibrosis, secondary myelofibrosisNCT04279847not yet recruiting
I/IIsolid tumors, advanced malignancies, metastatic cancerNCT02959437active, not recruiting
ADZ5153AstraZenecaImalignant solid tumors, lymphoma, ovarian cancerNCT03205176recruiting
Acerta Pharma BV|AstraZenecaINHL, DLBCL, non-Hodgkin’s lymphomaNCT03527147recruiting
ABBV-075AbbVieIASTNCT02391480completed
ABBV-744ICRPC, AMLNCT03360006recruiting
BMS-986158Dana-Farber Cancer InstituteIsolid tumor, lymphoma, brain tumor (pediatric)NCT03936465recruiting
Bristol-Myers SquibbI/IIASTNCT02419417recruiting
PLX51107M.D. Anderson Cancer CenterIAML, myelodysplastic syndrome, MDS/MPN, UNCT04022785recruiting
PlexxikonIsolid tumors, AML, myelodysplastic syndrome, NHLNCT02683395terminated
BI 894999Boehringer IngelheimIneoplasmsNCT02516553recruiting
ZEN003694Zenith EpigeneticsI/IIMCRPCNCT04145375enrolling by invitation
ITNBCNCT03901469recruiting
IIMCRPCNCT02705469completed
I/IIMCRPCNCT02711956active, not recruiting
GS-5829Gilead SciencesI/IImetastatic CRPCNCT02607228completed
Isolid tumors, lymphomasNCT02392611completed
FT-1101Forma TherapeuticsIAML, MDSNCT02543879completed
aSourced from https://clinicaltrials.gov/ (January 2021).

3. RESISTANCE MECHANISMS

In related research of BRD4, problems with preclinical drug resistance have attracted significant attention. In drug resistance, the activation of alternative or compensatory pathways such as WNT/β-catenin and hedgehog signaling can maintain MYC expression independently of BRD4 (Figure 2).4850 In pan-BET inhibitor-resistant leukemia cells, genome-wide BRD4 binding is decreased upon BET inhibitor treatment, but alternative binding by β-catenin to key oncogenic sites or nearby WNT-responsive enhancers maintains MYC expression.48 In addition, the activation of the hedgehog signaling pathway is another mechanism to restore MYC expression through altered transcription programming.49

The phosphorylation of BRD4, which is crucial for chromatin binding and factor recruitment,8 also has a great effect on the drug resistance, as shown in triple-negative breast cancer (TNBC) cells.11,12 Significantly increased BRD4 phosphorylation (or hyperphosphorylation) due to reduced PP2A activity results in a stronger association of BRD4 with the MED1 component of the mediator complex that recruits RNA Pol II to the promoter region, thereby promoting bromodomain-independent gene activation via an alternative protein−protein (i.e., BRD4−mediator) recruitment mechanism.11,51 BRD4 hyperphosphorylation has also been observed in NMC to support its oncogenic activity.40

Furthermore, adapted apoptotic signaling through AMPK/ULK1-mediated autophagy represents another resistance mechanism to BRD4 inhibitors in certain JQ1-resistant acute myelogenous leukemia (AML) cells, where reduced apoptosis is achieved by JQ1-induced prosurvival autophagy, as the pharmacological inhibition of AMPK leads to diminished ULK1-mediated autophagy and increased apoptosis.52

Conceivably, the activity of the E3 ubiquitin ligase adaptor SPOP (speckle-type POZ protein) and deubiquitinase DUB3 that directly modulate the BRD4 protein stability may potentially account for the drug resistance, as a deficiency in SPOP binding to BRD4 results in the enhanced BRD4-dependent expression of RAC1 GTPase and the activation of cholesterol biosynthetic enzymes and AKT-mTORC1 to mediate intrinsic resistance.5355 Likewise, a mutation in NCOR2 that negatively regulates DUB3 gene transcription frequently observed in castration-resistant prostate cancer (CRPC) leads to enhanced DUB3 and BRD4 protein levels,56 thus contributing to resistance to BRD4 inhibitor treatment. Moreover, a mutation in PI3KCA has also been identified as a resistance determinant to BRD4 inhibitors based on whole-genome shRNA dropout screens performed in 77 breast cancer cell lines.57

Notably, in preclinical models, the complexity of drug resistance with diverse factors and pathways involved greatly diminishes the clinical responses. It is indeed a significant challenge to overcome resistance to BET inhibitors, and no compounds targeting specific gene mutation have thus far been reported.

4. DEVELOPMENT OF NEXT-GENERATION INHIBITORS

Poor and short-term clinical responses have become a significant issue in the use of pan-BET small-molecule inhibitors for the clinical treatment of cancer. Thus higher and stricter requirements for the development and rational use of inhibitors are essential. New technologies have been developed to facilitate the discovery of novel inhibitors, such as BD1-, BD2-, or BRD4-selective inhibitors, BET proteolysis targeting chimeras (PROTACs), dual-target inhibitors, and protein−protein interaction (PPI) inhibitors. Among these inhibitors, pan-BET inhibitors show no selectivity against specific BET protein isoforms or bromodomains, BD1- and BD2-selective inhibitors imply pan-BET activity toward BRD2/3/4/T but with selectivity for specific bromodomains, and BRD4-selective inhibitors mean target preference for BRD4 rather than BRD2/3/T but without bromodomain selectivity. A recent report depicting functional differences between BD1 and BD2 highlights the potential for developing BD1-specific inhibitors in cancer therapy and BD2-specific inhibitors in immunoinflammation that may be more favorable compared with nonselective BET bromodomain targeting.14 Dual-target inhibitors with bimolecular binding to two different proteins, such as dual BET/kinase inhibitors, display either synergistic effects or synthetic lethality that may reduce the drug resistance frequently seen with monotherapy during cancer treatment. PROTACs targeting BET proteins that allow the prompt removal of a large amount of BET proteins in the cell show strong antitumor activity in preclinical studies. Undoubtedly, PPI inhibitors blocking BRD4 interaction with other proteins offer another line of new approaches to drug discovery and cancer treatment. The development of these more selective BET inhibitors with alternative mechanisms of action is of vital importance to overcome the limited clinical efflcacy and off-target toxicities often associated with the use of pan-BET inhibitors.

4.1. BD1-Selective Inhibitors.

BD1 and BD2 each consist of four antiparallel α-helixes (αZ, αA, αB, and αC) with different lengths linked by two hydrophobic loops (ZA and BC) that together form a hydrophobic Kac recognition pocket. Amino acid differences in the binding pockets of BD1 and BD2 lead to narrower binding regions and differences in the polarity and hydrophobicity of BD2. Poor selectivity among BET proteins and between BD1 and BD2 has incurred unwanted side effects and reduced the efflcacy of BET bromodomain inhibitors in clinical trials.58 Thus selectively targeting only BD1 or BD2 in BRD4 can provide better specificity and avoid unnecessary bioreactions triggered by the other domain inhibition as well as poor clinical evaluation due to the concurrent targeting of both BD1 and BD2 with differences in functions and structures. Gilan et al. have developed respective BD1- and BD2-specific inhibitors for better disease treatment and efflcacy based on the functional and structural differences between BD1 and BD2.14 Because BD1 and BD2 within the same protein are structurally and functionally more diverse compared with the analogous BD1 or BD2 found in different BET proteins,2 it is in fact easier to develop inhibitors selective for BD1 or BD2 than for individual BET family members.

Many BD1-selective inhibitors have been discovered, including Olinone (1)59,60 and MS436 (2),60 with the 3D crystal structure of the latter with BRD4-BD1 also shown on the right (Figure 3A). ZL0580 (3) (IC50 = 163 nM), a BRD4 BD1-selective inhibitor, was discovered by structure-guided drug design.61 Overlay analysis reveals that 3 can access the Kac-binding pocket in BD1 with binding notably different from that of JQ1. Compound 3 induces the epigenetic suppression of HIV in multiple in vitro and ex vivo models. Another BD1-selective inhibitor, LT052 (4),62 with more than 100-fold selectivity for BRD4-BD1 over BRD4-BD2 and exhibiting a stable level in liver microsomal metabolic analysis inhibits monosodium urate (MSU)-induced apoptosis in human THP-1 monocytic cells and improves symptoms of acute gouty arthritis via BRD4/NF-κB/NLRP3 inflammasome signaling pathways. Chromone derivatives ZL0513 (5) and ZL0516 (6) both show high binding afflnity for BD1 with a value of 67 and 84 nM, respectively. In vivo pharmacokinetics (PK) profiles between compounds 5 and 6 are similar (half life 3 to 4 h, maximum concentration ~600 ng/mL, AUC ~5000 ng·h/mL), and the oral bioavailability of compounds 5 and 6 of around 35−38% (20 mg/kg) is generally acceptable in rats.63 GSK778 (7), identified by the functional and structural comparison between BD1 and BD2 showing 130 times higher afflnity for BD1 than BD2, inhibits not only the growth and vitality of human cancer cell lines but also the clone-forming ability of human primary AML cells.14 Further chemical modification of compound 7 leads to another potent, selective, and cell-permeable inhibitor of BD1 (GSK789 (8)), displaying more than 1000-fold selectivity for BD1 over BD2 and achieving a submicromolar inhibition of monocyte chemoattractant protein-1 (MCP-1) release in human whole blood (pIC50 = 6.5).64 Starting with I-BET151 guided by structural information leads to imidazoquinoline compound 9 with 150-fold BD1 selectivity over BD2.65 Compound 9 reduces the production of IL-6 and MCP-1 in LPS-stimulated human peripheral blood mononuclear cells (PBMCs) and whole blood and suppresses MV4–11 lymphoblast leukemia cell growth (pIC50 = 7.0), indicating that BD1-selective inhibitor compound 9 is sufflcient to maintain the anti-inflammatory and antiproliferative activity of pan-BET inhibition. Compound 10, another BD1-selective inhibitor (28 times higher afflnity for BRD4-BD1 than BRD4-BD2), displays similar selectivity against the BD2 of BRD2 and BRD3 (25 and 33 times, respectively).66 This study also reveals that the underlying BD1 selectivity lies in the flexibility of the BD1-conserved YNKP motif and the substitution of structured waters in the Kac-binding site, which, unlike typical pan-BET inhibitor-suppressed MYC expression, leads to enhanced MYC expression in compound 10-treated MM.1S multiple myeloid cells. This unexpected result needs to be further investigated.

An external file that holds a picture, illustration, etc.
Object name is nihms-1697913-f0004.jpg

Reported novel selective inhibitors 1−20. (A) Chemical structures of BRD4 BD1-selective inhibitors. The docking pose of compounds 2 and 6 with BRD4−BD1 in ribbon representation is shown on the right. Key residues, including Pro82, Gln85, Lys91, Tyr97, and Asn140, are shown in green sticks, and the amide ketone of compound 2 interacts with the critical conserved residue Asn140 to form a hydrogen bond. Similarly, in the docking pose of compound 6, key residues Asn140, Tyr97, Gln85, and Pro82 are green. The carbonyl O atom of compound 6 interacts with the conserved residue Asn140, forming a direct H bond, and with Tyr97, forming an indirect hydrogen bond mediated by a water molecule. (B) Reported BRD4 BD2-selective inhibitors. (C) Chemical structures of BRD4-selective inhibitors.

4.2. BD2-Selective Inhibitors.

Representative BD2-selective inhibitors identified to date are shown in Figure 3B. The tetrahydroquinoxaline derivative GSK340 (11) with BD2 selectivity displays good solubility in both thermodynamic and kinetic solubility assays.67 In addition, compound 11 is cell-permeable and reduces the release of the MCP-1 cytokine in human PBMCs and whole blood. BY27 (12) is a potent BD2-selective inhibitor with 7 times higher selectivity for BD2 over BD1.68 In MV4–11 cells, compound 12 reduced the c-MYC protein level and increased the p21 expression in a dose-dependent manner. Compared with I-BET762, which caused 11% weight loss at 60 mg/kg, compound 12 is less toxic (exhibiting 7% weight loss at 150 mg/kg) at high doses and has 67% inhibition of tumor growth in a MV4–11 mouse xenograft model. Currently, ABBV-744 (13),69 a highly effective inhibitor of the BD2 domain with more than 300 times higher selectivity for BD2 over BD1, is synthesized and shows moderate preclinical PK profiles, including a moderate to high volume of distribution, a 2.0−4.4 h half life, and moderate clearance.70 The structural modification of an ABBV-075 analog leads to compound 13. 2,6-Dimethylphenyl ether in compound 13 contributes to BD2 selectivity, with a tertiary alcohol on the central phenyl ring suppressing the activity against BRD4-BD1 and a fluorine atom on the phenyl ether improving the pharmacokinetic properties of compound 13. In a myelodysplastic SKM-1 cell-derived xenograft mouse model, compound 13 displays 83% tumor growth inhibition with minimal (2%) weight loss over 21 days at a dose of 18.8 mg/kg, whereas ABBV-075 has similar tumor growth inhibition with 7% weight loss at the maximally tolerated dose of 1 mg/kg, suggesting that more selective BET inhibitors may improve the tolerability, albeit modestly. Compound 13 is currently in phase I clinical evaluation. GSK046 (iBET-BD2, 14), although less effective than 8 in the human cancer cell lines examined, exhibits more than 300 times higher selectivity for BD2 over BD1 and appears to ameliorate inflammatory disease in preclinical models.14 A recent study shows that compound 14 has physicochemical and pharmacokinetic properties suitable for use in vivo, and it retains the ability to effectively inhibit the release of MCP-1 cytokines (pIC50 = 7.5) in the cellular and whole blood environment.71 Interestingly, the inhibition of BD1 or BD2 leads to the suppression of MCP-1 despite the fact that these two types of inhibitors have distinct selectivity profiles. Another study applying a template hopping approach based on the orthogonal template (compound 14) leads to the synthesis of compounds GSK620 (15) and GSK549 (16) with high selectivity and low levels of inhibition against indicators of hepatoxicity.72 SJ018 (17) and SJ432 (18), two tetrahydroquinoline analogs, inhibit the expression of MYC and downstream targets but display no rebound effects, as compared with JQ1 in pediatric tumor cell lines, suggesting that BD2-selective inhibitors are effective candidates for the design of antitumor drugs for MYC-driven pediatric malignancies.73

4.3. BRD4-Selective Inhibitors.

Besides BD1- and BD2-selective inhibitors, the specific targeting of BRD4 rather than other BET proteins is also emerging. The ubiquitously expressed BRD2, BRD3, and BRD4 have a broad range of functions and participate in housekeeping and lineage-specific gene regulation in somatic and progenitor/stem cells by interacting with distinct partner proteins. BRD2 associates with E2F transcription factors to regulate E2F-dependent cell differentiation and proliferation genes and interacts with RNA Pol II to engage in transcription elongation in embryonic kidney cells through hyperacetylated nucleosomes due to the lack of a CTM.7476 Similar to BRD2, BRD3 is mainly involved in the regulation of gene transcription related to the development of embryonic stem cells, but unlike BRD4, it interacts with RNA Pol II in a P-TEFb-independent manner.77 The germ-cell-specific BRDT associates with multiple interactors during spermatogenesis and oogenesis.78,79 Mice treated with the pan-BET inhibitor JQ1 had a reduced spermatozoa number and reduced motility, indicating that BRDT could be an ideal target for contraceptive drug development.80,81 Considering the intricate physiological functions of BET family proteins, it is important to improve the target selectivity of BRD4 inhibitors in specific biological processes and disease progression without incurring unintended side effects, especially in the treatment of cancer and inflammation.58 Two BRD4-selective inhibitors reported in recent years are shown in Figure 3C. ZL0454 (19) and ZL0420 (20) are two BRD4-selective inhibitors displaying 30−60-fold selectivity over BRD2, 50−90-fold selectivity over BRD3, and 70−120-fold selectivity over BRDT.82 Molecular docking shows that compounds 19 and 20 occupy the Kac-binding pocket of BRD4 in a similar way. In vivo experiments further found that 19 and 20 almost completely blocked the TLR3 (toll-like receptor 3)-dependent expression of innate immune genes, including ISG54, ISG56, IL-8, and Groβ in human primary small airway epithelial cells (hSAECs). Given the shared functions between BRD4 and the other BET family proteins yet to be completely defined, it is difflcult to predict what types of diseases are most suited for BRD4-selective targeting. Nevertheless, therapy with fewer unintended targets has the potential to improve the clinical outcomes.

4.4. Dual Pharmacophores.

In recent years, multitarget approaches targeting different proteins and inhibiting kinases have received increasing attention in anticancer drug development.83 A previous study that reported that the CDK inhibitor Dinaciclib could interact with the Kac-binding pocket of BRDT provided the first indication that kinase inhibitors might be used to rationally design a new generation of BRD4 inhibitors.84 Since then, on the basis of the idea that polypharmacological inhibitors with multitargets would have a more robust action, many dual BET/kinase inhibitors have been described.85,86 Some of the recently discovered dual-target inhibitors are shown in Figure 4.

An external file that holds a picture, illustration, etc.
Object name is nihms-1697913-f0005.jpg

Chemical structures of dual-target inhibitors 21−31.

Histone deacetylases (HDACs) reduce the acetylation level of lysine residues where BET proteins recognize and promote factor recruitment to targeted gene loci, thereby regulating gene transcription.2,8789 It is logical to cotarget BET proteins and HDACs for the more efflcient repression of oncogenic gene transcription and tumor cell proliferation.90,91 Compound 21, a highly potent dual BET/HDAC inhibitor, was reported with balanced activity against BRD4 BD1 and HDAC1.92 Compound 22 shows the inhibition of BRD4 at a rate of 88% at 10 μM and strong HDAC3 inhibition with an IC50 value of 5 nM.93 Compound 23, another dual BRD4/HDAC inhibitor, induces autophagic cell death in colorectal cancer (CRC) cells at nanomolar levels.90 Compound 24 induces HL-60 AML cell death with c-MYC-inhibitory activity and suppresses cell growth in BET inhibitor-resistant cells.94 Compounds 25 and 26 also exhibit potent antiproliferative activity in human leukemia cell lines.95,96

Because BRD4 plays a critical role in DNA repair (Figure 1D), the inhibition of BRD4 shows synthetic lethality with poly-(ADP-ribose) polymerase-1 (PARP1) inhibitors, resulting in HR deficiency and providing a strategy for cancer treatment.35,97 Compound 27, designed with the conception of synthetic lethality, is the first dual BRD4/PARP1 inhibitor reported that triggers breast cancer cell apoptosis and induces cell-cycle arrest in the G1 phase.98 A recent study reports that polo-like kinase 1 (PLK1) upregulates BRD4-controlled MYC transcription and protein stability, with MYC, in turn, enhancing the PLK1 gene transcription, thus forming a PLK1-MYC feed-forward circuit in double-hit lymphoma (DHL) cells.99 Via the chemical modification of the pre-existing dual BRD4/PLK1 inhibitor BI-2536 (28), the dual BET/PLK1 inhibitor WNY0824 (29) was shown to suppress CRPC cell proliferation as well as the tumor growth in an enzalutamide-resistant CRPC xenograft mouse model by blocking BRD4-dependent androgen-receptor- and MYC-driven transcription programs.100 Fine-tuning the selectivity of inhibitors can also be achieved through structural guidance. Compound 30 is identified as a well-balanced dual BRD4/PLK1 inhibitor, illustrating that the methylated amide and the cyclopentyl group of the compound 28 scaffold are important for maintaining bromodomain-binding activity but are less critical for PLK1-inhibitory activity.101 Another study also chose compound 28 as the starting compound but changed PLK1 inhibition to anaplastic lymphoma kinase (ALK) targeting with the derived inhibitor compound 31, maintaining the BRD4-inhibitory activity but shifting the kinase selectivity from diminished PLK1 inhibition to enhanced ALK inhibition, which shows the remarkable suppression of ALKF1174L- and BRD4-dependent MYCN-driven gene transcription in pediatric neuroblastoma cells.102 These strategies provide new directions for the development of novel dual-target inhibitors via the chemical modification of existing pharmacophores targeting, respectively, BET proteins and kinases. However, this new line of research also poses significant challenges in how to properly balance the selectivity between BET proteins and specific kinases in designing dual-target inhibitors with enhanced antitumor activity. A report documenting that PLK inhibition, rather than BRD4 inhibition, is the main action for the anticancer activity of compound 28 in MV4–11 cells may provide some mechanistic hints about this specific compound.103

It should be mentioned that many dual-target inhibitors were discovered through their unexpected BET-targeting activity. For instance, TG101209 and BI-2536, originally reported as JAK2 inhibitor and PLK1 inhibitor, respectively, were found to have strong inhibitory potential against BRD4 through binding to the Kac pocket, providing a rationale for the development of dual-target inhibitors.104 As the products of a new technology, dual-target inhibitors display great potential in cancer therapy, even though they are still in the preclinical stage. Combining two pharmacophores cotargeting BET proteins and HDACs based on their pharmacological activity and safety evaluation, for example, 21 synthesized by linking (+)-JQ1 to HDAC inhibitors SAHA and CI-944,92 has become a popular trend in the discovery of dual-target inhibitors.105 For dual-target inhibitors that have already been developed, structural modification may result in new inhibitors with different biological activities, such as compounds 29, 30, and 31, optimized from compound 28.96,102

Additional kinase inhibitors to be considered include those involved in the PI3K-AKT-mTOR pathway that regulates MYC expression, stability, post-translational modification, and MYC protein synthesis106 as well as CDK9, which phosphorylates the Pol II CTM at Ser2, resulting in the stimulation of transcription elongation, and is associated with BRD4 hyperphosphorylation in NMC.40,83,107 CK2, which phosphorylates BRD4, implicated in chromatin targeting and factor recruitment,8 is another promising candidate for the future development of potent dual-target inhibitors for cancer therapy.

4.5. PROTACs.

PROTACs, initially proposed by Deshaies and colleagues,108 are an effective way to inactivate protein function via compound-mediated proteolysis. PROTACs targeting BET proteins have distinct pharmacological effects compared with the corresponding small-molecule inhibitors because they can disrupt the function of the entire protein rather than just the bromodomain activity.46,109 A sustained loss of BET proteins has superior antiproliferative effects in vitro while showing better anticancer activity in vivo with no obvious toxicity in a mouse tumor regression study.110

The currently developed BET degraders use ligand binding to the cereblon (CRBN) or von Hipple-Lindau (VHL) component of an E3 ubiquitin ligase. Compounds 3238 are CRBN-based degraders, whereas compounds 3945 are based on the VHL component (Figure 5A). Among them, dBET1 (32) containing an eight-atom linker is the first degrader displaying remarkable selectivity in depleting BET proteins with more potent inhibition compared with JQ1 against MV4–11 cells, DHL-4 lymphoma cells, and primary blasts from leukemia patients.111 ARV-825 (33) with a different linker type also achieves an effective degradation of BRD4 (DC50 < 1 nM), resulting in strong and lasting c-MYC inhibition in comparison with pan-BET inhibitors JQ1 and OTX015.112 BETd-246 (34) induces BET protein degradation at low nanomolar concentrations within an hour in TNBC cells by downregulating many proliferation/apoptosis-related genes, leading to growth inhibition and apoptosis.113 Another degrader, BETd-260 (35), shows picomolar potency (IC50 = 51 pM) in inhibiting RS4–11 lymphoblastic leukemia cell growth.110

An external file that holds a picture, illustration, etc.
Object name is nihms-1697913-f0006.jpg

Reported PROTAC degraders and PPI inhibitors. (A) PROTAC degraders based on CRL4CRBN (32−38) and CRL2VHL E3 ligase (39−45). (B) Chemical structures of 46−48.

QCA570 (36) was designed and synthesized based on the 1,4-oxazepine structure with more than 1000 times higher potency compared with 33. Significantly, in both the MV4–11 and RS4–11 acute leukemia xenograft models, compound 36 shows complete and long-lasting tumor regression at 5 mg/kg without apparent toxicity.114 A recent study highlights that DP1 (37) not only achieves rapid, stable, and persistent BRD4 degradation in SU-DHL-4 lymphoma cells but also has significant inhibitory effects on immunodeficient mice with xenotransplanted lymphoma cells.115 Cellular degradation assays showed that ZXH-3–26 (38) exhibits a DC50/5h of ~5 nM and BRD4 selectivity without inducing significant inhibition or degradation of BRD2 and BRD3,116 a result similar to that observed with AT1 (39) displaying BRD4-selective depletion but negligible activity against BRD2 and BRD3 at 1 to 3 μM of 39 treatment.117 ARV-771 (40), a small-molecule pan-BET degrader, reduces the protein levels of androgen receptor (AR) signaling in a CRPC mouse xenotransplantation model, providing the first reported BET degrader activity against a solid tumor.118 Compound 40 also presents superior pharmacological properties compared with those of 33 in mantle cell lymphoma (MCL) Mino cells.119 MZ1 (41), a JQ1-based BRD4 degrader, exhibits BRD4-targeting preference in cell-based assays but comparable in vitro binding afflnities to individually purified BRD2, BRD3, and BRD4 bromodomains measured by isothermal calorimetry (ITC), suggesting that MZ1-induced proximity and accessible surface lysine residues on BRD4, versus BRD2 and BRD3, might be differentially contacted by the VHL E3 ligase module in vivo and are presumably regulated by BET interactors that are unique to each family member.120 Adding a cyclizing linker to compound 40 leads to a macrocyclic PROTAC, macroPROTAC-1 (42), with cellular activity similar to that of compound 41, thus providing a new strategy for drug design.121

The targeting effects between triazolodiazepine JQ1 and tetrahydroquinoline I-BET762 and the optimal length of linkers connecting the BET-binding ligand JQ1 or I-BET762 to the VHL ligand VH032 for degrader design were examined by synthesizing a series of I-BET762-derived degraders, including MZP-54 (43), MZP-55 (44), and MZP-61 (45),122 for comparison with MZ1-related compounds, with the results showing that JQ1-based PROTACs are more effective at inducing BET protein degradation, even though JQ1 alone is not as potent as I-BET762 in BET protein binding. This parallel comparison indicates that stronger inhibitors may not necessarily produce more effective PROTAC degraders, and the linker length is critical in PROTAC design. It is important to note that PROTAC binding to incidental or unintended targets through substrate or E3 ligase association does not guarantee substrate degradation.123 These BET degraders are effective in inhibiting cancer cell proliferation in vivo and in vitro. In mice, compounds 34, 35, and 36 show good tolerance without obvious toxicity.124

BET-targeting PROTACs are still in the preclinical stage, and thus their therapeutic potential is presently unknown. For the current degraders, the main challenge is to improve the bioavailability index through precise modification without significantly increasing the molecular size. More selective and tissue-specific degraders are also needed for clinical trials. A strategy involving antibody−PROTAC conjugation provides a proof of concept for tissue-specific BRD4 degradation.125 A trastuzumab−PROTAC conjugate (Ab-PROTAC) with blocked degradation activity by an antibody linker that can be selectively released to produce an active PROTAC demonstrates the significant potential for tissue-specific target degradation. It should be mentioned that targeting BET proteins for degradation is not limited to ubiquitination. The lysosomal/autophagy pathway provides cargo-specific degradation through a different molecular mechanism.126

4.6. Targeting the Protein−Protein Interaction Network.

The PPI forms signal nodes and hubs in normal cells and cancer cells and transmits pathophysiological cues along molecular networks to promote tumorigenesis, migration, invasion, and metastasis.127 Therefore, blocking the PPI is an effective way to inactivate network function and signal transmission. DC-1 (46) and DC-2 (47), two peptoids (i.e., N-substituted oligoglycines; Figure 5B) isolated from a combinatorial compound library of ~38 500 peptoids,128 bind specifically to phospho-NPS in human BRD4, which provides an effective strategy to inhibit the phosphorylation-dependent gene-specific function of BRD4 and factor-regulated transcription programs.10,12 The iridium(III) transition-metal-based irreversible PPI inhibitor 48 blocks BRD4-BD1 but not BRD4-BD2, binding to a tetraacetylated histone H4 peptide with IC50 = 70 nM, and suppresses melanoma cell growth in vitro and mouse xenograft tumor growth in vivo by inhibiting BRD4-driven MYC and BCL-2 gene transcription.129 Clearly, PPI inhibitors provide another promising strategy for BRD4-targeted disease treatment.

Compared with other technologies, targeting the PPI associated with BRD4 is less reported, in part due to the challenge in identifying the in vivo targets and the mechanisms of action as well as the difflculty in establishing effective screening systems.130 Encouragingly, targeting phospho-NPS in BRD4, as illustrated with compounds 46 and 47, has been shown to be a promising and effective way to inhibit BRD4 association with specific transcription factors without blocking the chromatin-binding activity of BRD4, providing a rationale for targeting nonbromodomain surfaces of BRD4 to block specific disease-associated pathways with minimal perturbation of normal BRD4 function.10 A unique interface formed between BRD4 and GATA4 interaction in cardiomyocytes that regulates the bioenergy homeostasis of the adult heart would be an interesting region for the future development of BRD4-targeting PPI inhibitors.131

5. CONCLUSIONS AND FUTURE PERSPECTIVES

In mouse tumor models, BD1-selective inhibition shows reduced proliferation, cell-cycle arrest, and apoptosis, as seen with pan-BET inhibitors, whereas BD2-selective inhibition is more effective in suppressing inflammation, metabolic diseases, and fibrosis by reducing the negative effects of enhancer reprogramming and minimizing the maladaptive transcription of disease-causing genes on potential enhancers.132 The development of BRD4-selective inhibitors is in an early stage. Considering the oncogenic role the BRD4 short isoform plays in the extracellular matrix (ECM) network during breast cancer progression7 and the potential active gene transcription through LLPS,20 selective targeting of the BRD4 short isoform without cross-inhibiting the tumor-suppressive function of the BRD4 long isoform is a critical strategy for future drug development. The advancement in BRD4 protein characterization also sheds new light on the design of selective inhibitors targeting a specific region of BRD4, such as phospho-NPS, which provides more selective interaction with cancer-associated transcription factors. Further development of inhibitors targeting other BET proteins could also be beneficial to disease treatment or birth control, such as BRDT-specific targeting for contraceptives. Conceivably, research on the structural understanding of BET proteins will contribute to the development of more selective inhibitors. A computational protein structure prediction is helpful to simulate the protein structure, with refinement making the model closer to the natural structure,133 thus allowing the exploration of whether or how small molecules bind to the protein through docking.134 All of these provide new methodologies for the further development of selective inhibitors.

At present, dual kinase/BRD4 inhibitors are still in the early stages. A rational design of dual kinase/BET inhibitors may be conceptually easier than designing bifunctional ligands targeting two different kinases due to the similarity of their catalytic pockets. It is also challenging to design dual-target inhibitors with balanced selectivity. Here computational methods are useful for predicting and identifying new ligands with dual activity by simulating the binding modes of ligands and using ligand-based and structure-based approaches. Moreover, the physical and chemical properties of dual-target inhibitors may be poor due to an increase in the relative molecular weight and structural complexity. How to balance the target selectivity of dual-target inhibitors to maximize the clinical efflcacy is another challenge. It is likely that partial coverage of the kinase and full coverage of BRD4 may achieve the maximum outcome.

The pharmacological effects of PROTACs are not limited to only protein loss but also affect the related subcellular localization and transcriptional regulation, thus improving the pharmacological efflcacy through this additional function. Another feature of PROTACs is that they require a relatively low dose to achieve pharmacological effects. This also reduces the possibility of off targets and thus improves the drug tolerance. In addition, PROTACs targeting BET proteins, in a way similar to gene knockout, can cause a more lasting loss of protein expression than other targeted inhibitors, namely, a more prolonged inhibitory effect.

The PPI is an important part of the functional exploration of BRD4. Besides the well-known bromodomain, the nonbromodomain PPI is also of great research value, especially the phospho region of BRD4 targeted by the existing peptoid inhibitors. Accordingly, PPI inhibitors, with in-depth mechanistic studies and chemical probing, have great therapeutic potential warranting further development.

All of these new technologies have shown great potential based on in vitro and in vivo bench studies. The primary challenge is to transfer the preclinical antitumor activity of compounds to clinical efflcacy. Altogether, the use of small-molecule inhibitors targeting BRD4 is expected to be effective for the treatment of a variety of cancers. Interdisciplinary cooperation among investigators is of great need to make groundbreaking drug discoveries in this exciting area of research.

ACKNOWLEDGMENTS

Funding for the authors’ research was sponsored in part by the National Natural Science Foundation of China, grant numbers 81922064 (to L.O.), 81874290 (to L.O.) and 81903502 (to J.Z.); the National Science and Technology Major Project of the Ministry of Science and Technology of China, grant number 2018ZX09735005 (to L.O.); the National Major Scientific and Technological Special Project for ‘Signficant New Drugs Development’, grant number 2018ZX09201018–021 (to L.O.); the Sichuan Science and Technology Program, grant number 2019YFS0003 (to L.O.); the Sichuan Science and Technology Program, grant number 2020YJ0091 (to J.L.); the China Postdoctoral Science Foundation, grant number 2020M673268 (to J.Z.); the U.S. National Institutes of Health (NIH), grant number 1RO1CA251698–01 (to C.-M.C.); and the Cancer Prevention and Research Institute of Texas (CPRIT), grant numbers RP180349 and RP190077 (to C.-M.C.).

ABBREVIATIONS USED

ALKanaplastic lymphoma kinase
ALLacute lymphoblastic leukemia
AMLacute myelogenous leukemia
ARandrogen receptor
ASTadvanced solid tumor
BETbromodomain and extraterminal
BIDbasic residue-enriched interaction domain
BPDCNblastic plasmacytoid dendritic cell neoplasm
BRD4bromodomain-containing protein 4
CDK9cyclin-dependent kinase 9
CK2casein kinase II
CPSC-terminal cluster of phosphorylation sites
CRBNcereblon
CRCcolorectal cancer
CRPCcastration-resistant prostate cancer
CtIPC-terminal binding protein-interacting protein
CTMC-terminal motif
DHLdouble-hit lymphoma
DLBCLdiffuse large B-cell lymphoma
DLTdose-limiting toxicity
DSBDNA double-strand break
ECMextracellular matrix
ETextraterminal
GBMglioblastoma multiform
HDAChistone deacetylase
HPVhuman papillomavirus
HRhomologous recombination
hSAEChuman primary small airway epithelial cell
ITCisothermal calorimetry
KacN-ε-acetyllysine
LLPSliquid−liquid phase separation
MCLmantle cell lymphoma
MCP-1monocyte chemoattractant protein-1
MCRPCmetastatic castration-resistant prostate cancer
MDSmyelodysplastic syndrome
MMmultiple myeloma
MPNU myeloproliferative neoplasm-unclassifiable
NHEJnonhomologous end joining
NHLnon-Hodgkin’s lymphoma
NMCNUT midline carcinoma
NPSN-terminal cluster of phosphorylation sites
NSCLCnonsmall cell lung cancer
PARP1poly(ADP-ribose) polymerase-1
PBMCperipheral blood mononuclear cell
PKpharmacokinetics
PLK1polo-like kinase 1
PP2Aprotein phosphatase 2A
PPIprotein−protein interaction
PROTACproteolysis targeting chimera
P-TEFbpositive transcription elongation factor b
SPOPspeckle-type POZ protein
TLP3toll-like receptor 3
TNBCtriple-negative breast cancer
VHLvon Hipple-Lindau

Biographies

Pan Tang received her B.S. degree in Pharmacy English from Shenyang Pharmaceutical University in 2018. She is currently a Master’s student at the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and Collaborative Innovation Center of Biotherapy, Sichuan University. Her research interest focuses on the anticancer mechanism of small-molecule compounds.

Jifa Zhang received his Ph.D. degree from Southwest Jiaotong University in 2018 and is currently a postdoctoral student at the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and Collaborative Innovation Center of Biotherapy, Sichuan University. His research interests focus on the discovery, optimization, and molecular mechanism of candidate small-molecule compounds with antitumor activity.

Jie Liu received his Ph.D. degree in Medicinal Chemistry from West China School of Pharmacy, Sichuan University in 2010. He is now a Professor at State Key Laboratory of Biotherapy, West China Hospital, Sichuan University. He has long been engaged in the application of transition-metal-mediated organic synthesis in small-molecule-targeted drugs and pharmacochemical biology.

Cheng-Ming Chiang received his Ph.D. from the Department of Biochemistry, University of Rochester in 1991. He has been an Assistant Professor in the Department of Biochemistry at the University of Illinois Urbana−Champaign (1995−2000), an Associate Professor in the Department of Biochemistry at Case Western Reserve University in Cleveland, Ohio (2000−2007), and a Professor at Simmons Comprehensive Cancer Center, Department of Biochemistry, and Department of Pharmacology at the University of Texas Southwestern Medical Center (2007−present). His research interests primarily focus on transcription mechanisms, gene regulation, epigenetic control, chromatin dynamics, posttranslational modification, human papillomavirus, cancer, and therapeutics targeting phospho-BRD4 and BET proteins.

Liang Ouyang received his Ph.D. degree in Medicinal Chemistry from West China School of Pharmacy, Sichuan University, in 2010. He is now Professor at State Key Laboratory of Biotherapy, West China Hospital, Sichuan University. He has long-time interests in the design and synthesis of drugs based on original targeting of autophagy, basic research on the application of lead compound screening, the development of new drug target recognition methods and drug discovery, and taking advantage of interdisciplinary approaches including systems biology, structural biology, computer-aided drug design, medicinal chemistry, chemical biology, and molecular biology.

Footnotes

The authors declare no competing financial interest.

Contributor Information

Pan Tang, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, China.

Jifa Zhang, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, China.

Jie Liu, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, China.

Cheng-Ming Chiang, Simmons Comprehensive Cancer Center, Department of Pharmacology, and Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States.

Liang Ouyang, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, China.

REFERENCES

(1) Filippakopoulos P; Picaud S; Mangos M; Keates T; Lambert JP; Barsyte-Lovejoy D; Felletar I; Volkmer R; Muller S; Pawson T; Gingras AC; Arrowsmith CH; Knapp S Histone recognition and large-scale structural analysis of the human bromodomain family. Cell 2012, 149, 214–231. [Europe PMC free article] [Abstract] [Google Scholar]
(2) Wu SY; Chiang CM The double bromodomain-containing chromatin adaptor BRD4 and transcriptional regulation. J. Biol. Chem 2007, 282, 13141–13145. [Abstract] [Google Scholar]
(3) Dhalluin C; Carlson JE; Zeng L; He C; Aggarwal AK; Zhou M-M; Zhou M-M Structure and ligand of a histone acetyltransferase bromodomain. Nature 1999, 399, 491–496. [Abstract] [Google Scholar]
(4) Devaiah BN; Case-Borden C; Gegonne A; Hsu CH; Chen Q; Meerzaman D; Dey A; Ozato K; Singer DS Singer DS BRD4 is a histone acetyltransferase that evicts nucleosomes from chromatin. Nat. Struct. Mol. Biol 2016, 23, 540–548. [Europe PMC free article] [Abstract] [Google Scholar]
(5) Filippakopoulos P; Knapp S Targeting bromodomains: epigenetic readers of lysine acetylation. Nat. Rev. Drug Discovery 2014, 13, 337–356. [Abstract] [Google Scholar]
(6) Spriano F; Stathis A; Bertoni F Targeting BET bromodomain proteins in cancer: The example of lymphomas. Pharmacol. Ther 2020, 215, 107631. [Abstract] [Google Scholar]
(7) Wu SY; Lee CF; Lai HT; Yu CT; Lee JE; Zuo H; Tsai SY; Tsai MJ; Ge K; Wan Y; Chiang CM Opposing functions of BRD4 isoforms in breast cancer. Mol. Cell 2020, 78, 1114–1132. [Europe PMC free article] [Abstract] [Google Scholar]
(8) Wu SY; Lee AY; Lai HT; Zhang H; Chiang CM Phospho switch triggers BRD4 chromatin binding and activator recruitment for gene-specific targeting. Mol. Cell 2013, 49, 843–857. [Europe PMC free article] [Abstract] [Google Scholar]
(9) Shu S; Wu HJ; Ge JY; Zeid R; Harris IS; Jovanović B; Murphy K; Wang B; Qiu X; Endress JE; Reyes J; Lim K; Font-Tello A; Syamala S; Xiao T; Reddy Chilamakuri CS; Papachristou EK; D’Santos C; Anand J; Hinohara K; Li W; McDonald TO; Luoma A; Modiste RJ; Nguyen QD; Michel B; Cejas P; Kadoch C; Jaffe JD; Wucherpfennig KW; Qi J; Liu XS; Long H; Brown M; Carroll JS; Brugge JS; Bradner J; Michor F; Polyak K Synthetic lethal and resistance interactions with BET bromodomain inhibitors in triple-negative breast cancer. Mol. Cell 2020, 78, 1096–1113. [Europe PMC free article] [Abstract] [Google Scholar]
(10) Wu SY; Nin DS; Lee AY; Simanski S; Kodadek T; Chiang CM BRD4 phosphorylation regulates HPV E2-mediated viral transcription, origin replication, and cellular MMP-9 expression. Cell Rep 2016, 16, 1733–1748. [Europe PMC free article] [Abstract] [Google Scholar]
(11) Shu S; Lin CY; He HH; Witwicki RM; Tabassum DP; Roberts JM; Janiszewska M; Jin Huh S; Liang Y; Ryan J; Doherty E; Mohammed H; Guo H; Stover DG; Ekram MB; Peluffo G; Brown J; D’Santos C; Krop IE; Dillon D; McKeown M; Ott C; Qi J; Ni M; Rao PK; Duarte M; Wu S-Y; Chiang C-M; Anders L; Young RA; Winer EP; Letai A; Barry WT; Carroll JS; Long HW; Brown M; Shirley Liu X; Meyer CA; Bradner JE; Polyak K Response and resistance to BET bromodomain inhibitors in triple-negative breast cancer. Nature 2016, 529, 413–417. [Europe PMC free article] [Abstract] [Google Scholar]
(12) Chiang CM Phospho-BRD4: transcription plasticity and drug targeting. Drug Discovery Today: Technol 2016, 19, 17–22. [Europe PMC free article] [Abstract] [Google Scholar]
(13) Cheung KL; Zhang F; Jaganathan A; Sharma R; Zhang Q; Konuma T; Shen T; Lee JY; Ren C; Chen CH; Lu G; Olson MR; Zhang W; Kaplan MH; Littman DR; Walsh MJ; Xiong H; Zeng L; Zhou MM Distinct roles of BRD2 and BRD4 in potentiating the transcriptional program for Th17 cell differentiation. Mol. Cell 2017, 65, 1068–1080. [Europe PMC free article] [Abstract] [Google Scholar]
(14) Gilan O; Rioja I; Knezevic K; Bell MJ; Yeung MM; Harker NR; Lam E; Chung CW; Bamborough P; Petretich M; Urh M; Atkinson SJ; Bassil AK; Roberts EJ; Vassiliadis D; Burr ML; Preston A; Wellaway C; Werner T; Gray JR; Michon AM; Gobbetti T; Kumar V; Soden PE; Haynes A; Vappiani J; Tough DF; Taylor S; Dawson SJ; Bantscheff M; Lindon M; Drewes G; Demont EH; Daniels DL; Grandi P; Prinjha RK; Dawson MA Selective targeting of BD1 and BD2 of the BET proteins in cancer and immunoinflammation. Science 2020, 368, 387–394. [Europe PMC free article] [Abstract] [Google Scholar]
(15) Crowe BL; Larue RC; Yuan C; Hess S; Kvaratskhelia M; Foster MP Structure of the BRD4 ET domain bound to a C-terminal motif from gamma-retroviral integrases reveals a conserved mechanism of interaction. Proc. Natl. Acad. Sci. U. S. A 2016, 113, 2086–2091. [Europe PMC free article] [Abstract] [Google Scholar]
(16) Bisgrove DA; Mahmoudi T; Henklein P; Verdin E Conserved P-TEFb-interacting domain of BRD4 inhibits HIV transcription. Proc. Natl. Acad. Sci. U. S. A 2007, 104, 13690–13695. [Europe PMC free article] [Abstract] [Google Scholar]
(17) Itzen F; Greifenberg AK; Bosken CA; Geyer M BRD4 activates P-TEFb for RNA polymerase II CTD phosphorylation. Nucleic Acids Res 2014, 42, 7577–7590. [Europe PMC free article] [Abstract] [Google Scholar]
(18) Sabari BR; Dall’Agnese A; Boija A; Klein IA; Coffey EL; Shrinivas K; Abraham BJ; Hannett NM; Zamudio AV; Manteiga JC; Li CH; Guo YE; Day DS; Schuijers J; Vasile E; Malik S; Hnisz D; Lee TI; Cisse II; Roeder RG; Sharp PA; Chakraborty AK; Young RA Coactivator condensation at super-enhancers links phase separation and gene control. Science 2018, 361, eaar3958. [Europe PMC free article] [Abstract] [Google Scholar]
(19) Gibson BA; Doolittle LK; Schneider M; Jensen LE; Gamarra N; Henry L; Gerlich DW; Redding S; Rosen MK Organization of chromatin by intrinsic and regulated phase separation. Cell 2019, 179, 470–484. [Europe PMC free article] [Abstract] [Google Scholar]
(20) Han X; Yu D; Gu R; Jia Y; Wang Q; Jaganathan A; Yang X; Yu M; Babault N; Zhao C; Yi H; Zhang Q; Zhou MM; Zeng L Roles of the BRD4 short isoform in phase separation and active gene transcription. Nat. Struct. Mol. Biol 2020, 27, 333–341. [Abstract] [Google Scholar]
(21) Sakurai N; Inamochi Y; Inoue T; Hariya N; Kawamura M; Yamada M; Dey A; Nishiyama A; Kubota T; Ozato K; Goda T; Mochizuki K BRD4 regulates adiponectin gene induction by recruiting the P-TEFb complex to the transcribed region of the gene. Sci. Rep 2017, 7, 11962. [Europe PMC free article] [Abstract] [Google Scholar]
(22) Sabo A; Amati B BRD4 and MYC-clarifying regulatory specificity. Science 2018, 360, 713–714. [Abstract] [Google Scholar]
(23) Devaiah BN; Mu J; Akman B; Uppal S; Weissman JD; Cheng D; Baranello L; Nie Z; Levens D; Singer DS MYC protein stability is negatively regulated by BRD4. Proc. Natl. Acad. Sci. U. S. A 2020, 117, 13457–13467. [Europe PMC free article] [Abstract] [Google Scholar]
(24) Rahnamoun H; Lee J; Sun Z; Lu H; Ramsey KM; Komives EA; Lauberth SM RNAs interact with BRD4 to promote enhanced chromatin engagement and transcription activation. Nat. Struct. Mol. Biol 2018, 25, 687–697. [Europe PMC free article] [Abstract] [Google Scholar]
(25) Huang B; Yang XD; Zhou MM; Ozato K; Chen LF BRD4 coactivates transcriptional activation of NF-kappaB via specific binding to acetylated RelA. Mol. Cell. Biol 2009, 29, 1375–1387. [Europe PMC free article] [Abstract] [Google Scholar]
(26) Shi J; Wang Y; Zeng L; Wu Y; Deng J; Zhang Q; Lin Y; Li J; Kang T; Tao M; Rusinova E; Zhang G; Wang C; Zhu H; Yao J; Zeng YX; Evers BM; Zhou MM; Zhou BP Disrupting the interaction of BRD4 with diacetylated Twist suppresses tumorigenesis in basal-like breast cancer. Cancer Cell 2014, 25, 210–225. [Europe PMC free article] [Abstract] [Google Scholar]
(27) Lu L; Chen Z; Lin X; Tian L; Su Q; An P; Li W; Wu Y; Du J; Shan H; Chiang CM; Wang H Inhibition of BRD4 suppresses the malignancy of breast cancer cells via regulation of Snail. Cell Death Differ 2020, 27, 255–268. [Europe PMC free article] [Abstract] [Google Scholar]
(28) Zanconato F; Battilana G; Forcato M; Filippi L; Azzolin L; Manfrin A; Quaranta E; Di Biagio D; Sigismondo G; Guzzardo V; Lejeune P; Haendler B; Krijgsveld J; Fassan M; Bicciato S; Cordenonsi M; Piccolo S Transcriptional addiction in cancer cells is mediated by YAP/TAZ through BRD4. Nat. Med 2018, 24, 1599–1610. [Europe PMC free article] [Abstract] [Google Scholar]
(29) Song S; Li Y; Xu Y; Ma L; Pool Pizzi M; Jin J; Scott AW; Huo L; Wang Y; Lee JH; Bhutani MS; Weston B; Shanbhag ND; Johnson RL; Ajani JA Targeting Hippo coactivator YAP1 through BET bromodomain inhibition in esophageal adenocarcinoma. Mol. Oncol 2020, 14, 1410–1426. [Europe PMC free article] [Abstract] [Google Scholar]
(30) Chiang CM BRD4 engagement from chromatin targeting to transcriptional regulation: selective contact with acetylated histone H3 and H4. F1000 Biol. Rep 2009, 1, 98. [Europe PMC free article] [Abstract] [Google Scholar]
(31) Jonkers I; Lis JT Getting up to speed with transcription elongation by RNA polymerase II. Nat. Rev. Mol. Cell Biol 2015, 16, 167–177. [Europe PMC free article] [Abstract] [Google Scholar]
(32) Stanlie A; Yousif AS; Akiyama H; Honjo T; Begum NA Chromatin reader BRD4 functions in Ig class switching as a repair complex adaptor of nonhomologous end-joining. Mol. Cell 2014, 55, 97–110. [Abstract] [Google Scholar]
(33) Kim JJ; Lee SY; Gong F; Battenhouse AM; Boutz DR; Bashyal A; Refvik ST; Chiang CM; Xhemalce B; Paull TT; Brodbelt JS; Marcotte EM; Miller KM Systematic bromodomain protein screens identify homologous recombination and R-loop suppression pathways involved in genome integrity. Genes Dev 2019, 33, 1751–1774. [Europe PMC free article] [Abstract] [Google Scholar]
(34) Li X; Baek G; Ramanand SG; Sharp A; Gao Y; Yuan W; Welti J; Rodrigues DN; Dolling D; Figueiredo I; Sumanasuriya S; Crespo M; Aslam A; Li R; Yin Y; Mukherjee B; Kanchwala M; Hughes AM; Halsey WS; Chiang CM; Xing C; Raj GV; Burma S; de Bono J; Mani RS BRD4 promotes DNA repair and mediates the formation of TMPRSS2-ERG gene rearrangements in prostate cancer. Cell Rep 2018, 22, 796–808. [Europe PMC free article] [Abstract] [Google Scholar]
(35) Sun C; Yin J; Fang Y; Chen J; Jeong KJ; Chen X; Vellano CP; Ju Z; Zhao W; Zhang D; Lu Y; Meric-Bernstam F; Yap TA; Hattersley M; O’Connor MJ; Chen H; Fawell S; Lin SY; Peng G; Mills GB BRD4 inhibition is synthetic lethal with PARP inhibitors through the induction of homologous recombination deficiency. Cancer Cell 2018, 33, 401–416. [Europe PMC free article] [Abstract] [Google Scholar]
(36) French CA; Miyoshi I; Kubonishi I; Grier HE; Perez-Atayde AR; Fletcher JA BRD4-NUT fusion oncogene: a novel mechanism in aggressive carcinoma. Cancer Res 2003, 63, 304–307. [Abstract] [Google Scholar]
(37) Lee JE; Park YK; Park S; Jang Y; Waring N; Dey A; Ozato K; Lai B; Peng W; Ge K BRD4 binds to active enhancers to control cell identity gene induction in adipogenesis and myogenesis. Nat. Commun 2017, 8, 2217. [Europe PMC free article] [Abstract] [Google Scholar]
(38) Ge JY; Shu S; Kwon M; Jovanovic B; Murphy K; Gulvady A; Fassl A; Trinh A; Kuang Y; Heavey GA; Luoma A; Paweletz C; Thorner AR; Wucherpfennig KW; Qi J; Brown M; Sicinski P; Mcdonald TO; Pellman D; Michor F; Polyak K Acquired resistance to combined BET and CDK4/6 inhibition in triple-negative breast cancer. Nat. Commun 2020, 11, 2350. [Europe PMC free article] [Abstract] [Google Scholar]
(39) Rosencrance CD; Ammouri HN; Yu Q; Ge T; Rendleman EJ; Marshall SA; Eagen KP Chromatin hyperacetylation impacts chromosome folding by forming a nuclear subcompartment. Mol. Cell 2020, 78, 112–126. [Europe PMC free article] [Abstract] [Google Scholar]
(40) Wang R; Cao XJ; Kulej K; Liu W; Ma T; Macdonald M; Chiang CM; Garcia BA; You J Uncovering BRD4 hyperphosphorylation associated with cellular transformation in NUT midline carcinoma. Proc. Natl. Acad. Sci. U. S. A 2017, 114, E5352–E5361. [Europe PMC free article] [Abstract] [Google Scholar]
(41) Ceribelli M; Hou ZE; Kelly PN; Huang DW; Wright G; Ganapathi K; Evbuomwan MO; Pittaluga S; Shaffer AL; Marcucci G; Forman SJ; Xiao W; Guha R; Zhang X; Ferrer M; Chaperot L; Plumas J; Jaffe ES; Thomas CJ; Reizis B; Staudt LM A druggable TCF4- and BRD4-dependent transcriptional network sustains malignancy in blastic plasmacytoid dendritic cell neoplasm. Cancer Cell 2016, 30, 764–778. [Europe PMC free article] [Abstract] [Google Scholar]
(42) Kleppe M; Levine RL An unexpected chink in the transcriptional armor of plasmacytoid dendritic neoplasms. Cancer Cell 2016, 30, 659–660. [Abstract] [Google Scholar]
(43) Olley G; Ansari M; Bengani H; Grimes GR; Rhodes J; von Kriegsheim A; Blatnik A; Stewart FJ; Wakeling E; Carroll N; Ross A; Park S-M; Bickmore WA; Pradeepa MM; FitzPatrick DR Fitzpatrick D. R. BRD4 interacts with NIPBL and BRD4 is mutated in a Cornelia de Lange-like syndrome. Nat. Genet 2018, 50, 329–332. [Europe PMC free article] [Abstract] [Google Scholar]
(44) Korb E; Herre M; Zucker-Scharff I; Gresack J; Allis CD; Darnell RB Excess translation of epigenetic regulators contributes to Fragile X Syndrome and is alleviated by BRD4 inhibition. Cell 2017, 170, 1209–1223. [Europe PMC free article] [Abstract] [Google Scholar]
(45) Liu Z; Wang P; Chen H; Wold EA; Tian B; Brasier AR; Zhou J Drug discovery targeting bromodomain-containing protein 4. J. Med. Chem 2017, 60, 4533–4558. [Europe PMC free article] [Abstract] [Google Scholar]
(46) Bechter O; Schoffski P Make your best BET: The emerging role of BET inhibitor treatment in malignant tumors. Pharmacol. Ther 2020, 208, 107479. [Abstract] [Google Scholar]
(47) Bolden JE; Tasdemir N; Dow LE; van Es JH; Wilkinson JE; Zhao Z; Clevers H; Lowe SW Inducible in vivo silencing of BRD4 identifies potential toxicities of sustained BET protein inhibition. Cell Rep 2014, 8, 1919–1929. [Europe PMC free article] [Abstract] [Google Scholar]
(48) Fong CY; Gilan O; Lam EY; Rubin AF; Ftouni S; Tyler D; Stanley K; Sinha D; Yeh P; Morison J; Giotopoulos G; Lugo D; Jeffrey P; Lee SC; Carpenter C; Gregory R; Ramsay RG; Lane SW; Abdel-Wahab O; Kouzarides T; Johnstone RW; Dawson SJ; Huntly BJ; Prinjha RK; Papenfuss AT; Dawson MA BET inhibitor resistance emerges from leukaemia stem cells. Nature 2015, 525, 538–542. [Europe PMC free article] [Abstract] [Google Scholar]
(49) Rathert P; Roth M; Neumann T; Muerdter F; Roe JS; Muhar M; Deswal S; Cerny-Reiterer S; Peter B; Jude J; Hoffmann T; Boryn LM; Axelsson E; Schweifer N; Tontsch-Grunt U; Dow LE; Gianni D; Pearson M; Valent P; Stark A; Kraut N; Vakoc CR; Zuber J Transcriptional plasticity promotes primary and acquired resistance to BET inhibition. Nature 2015, 525, 543–547. [Europe PMC free article] [Abstract] [Google Scholar]
(50) Kumar K; Raza SS; Knab LM; Chow CR; Kwok B; Bentrem DJ; Popovic R; Ebine K; Licht JD; Munshi HG GLI2-dependent c-MYC upregulation mediates resistance of pancreatic cancer cells to the BET bromodomain inhibitor JQ1. Sci. Rep 2015, 5, 9489. [Europe PMC free article] [Abstract] [Google Scholar]
(51) Settleman J Cancer: BET on drug resistance. Nature 2016, 529, 289–290. [Abstract] [Google Scholar]
(52) Jang JE; Eom JI; Jeung HK; Cheong JW; Lee JY; Kim JS; Min YH AMPK-ULK1-mediated autophagy confers resistance to BET Inhibitor JQ1 in acute myeloid leukemia stem cells. Clin. Cancer Res 2017, 23, 2781–2794. [Abstract] [Google Scholar]
(53) Zhang P; Wang D; Zhao Y; Ren S; Gao K; Ye Z; Wang S; Pan CW; Zhu Y; Yan Y; Yang Y; Wu D; He Y; Zhang J; Lu D; Liu X; Yu L; Zhao S; Li Y; Lin D; Wang Y; Wang L; Chen Y; Sun Y; Wang C; Huang H Intrinsic BET inhibitor resistance in SPOP-mutated prostate cancer is mediated by BET protein stabilization and AKT-mTORC1 activation. Nat. Med 2017, 23, 1055–1062. [Europe PMC free article] [Abstract] [Google Scholar]
(54) Dai X; Gan W; Li X; Wang S; Zhang W; Huang L; Liu S; Zhong Q; Guo J; Zhang J; Chen T; Shimizu K; Beca F; Blattner M; Vasudevan D; Buckley DL; Qi J; Buser L; Liu P; Inuzuka H; Beck AH; Wang L; Wild PJ; Garraway LA; Rubin MA; Barbieri CE; Wong KK; Muthuswamy SK; Huang J; Chen Y; Bradner JE; Wei W Prostate cancer-associated SPOP mutations confer resistance to BET inhibitors through stabilization of BRD4. Nat. Med 2017, 23, 1063–1071. [Europe PMC free article] [Abstract] [Google Scholar]
(55) Janouskova H; El Tekle G; Bellini E; Udeshi ND; Rinaldi A; Ulbricht A; Bernasocchi T; Civenni G; Losa M; Svinkina T; Bielski CM; Kryukov GV; Cascione L; Napoli S; Enchev RI; Mutch DG; Carney ME; Berchuck A; Winterhoff BJN; Broaddus RR; Schraml P; Moch H; Bertoni F; Catapano CV; Peter M; Carr SA; Garraway LA; Wild PJ; Theurillat J-PP Opposing effects of cancer-type-specific SPOP mutants on BET protein degradation and sensitivity to BET inhibitors. Nat. Med 2017, 23, 1046–1054. [Europe PMC free article] [Abstract] [Google Scholar]
(56) Jin X; Yan Y; Wang D; Ding D; Ma T; Ye Z; Jimenez R; Wang L; Wu H; Huang H DUB3 promotes BET inhibitor resistance and cancer progression by deubiquitinating BRD4. Mol. Cell 2018, 71, 592–605. [Europe PMC free article] [Abstract] [Google Scholar]
(57) Marcotte R; Sayad A; Brown KR; Sanchez-Garcia F; Reimand J; Haider M; Virtanen C; Bradner JE; Bader GD; Mills GB; Pe’Er D; Moffat J; Neel BG Functional genomic landscape of human breast cancer drivers, vulnerabilities, and resistance. Cell 2016, 164, 293–309. [Europe PMC free article] [Abstract] [Google Scholar]
(58) Cochran AG; Conery AR; Sims RR Bromodomains: a new target class for drug development. Nat. Rev. Drug Discovery 2019, 18, 609–628. [Abstract] [Google Scholar]
(59) Rodriguez Y; Gerona-Navarro G; Osman R; Zhou MM In silico design and molecular basis for the selectivity of Olinone toward the first over the second bromodomain of BRD4. Proteins: Struct., Funct., Genet 2020, 88, 414–430. [Europe PMC free article] [Abstract] [Google Scholar]
(60) Divakaran A; Talluri SK; Ayoub AM; Mishra NK; Cui H; Widen JC; Berndt N; Zhu JY; Carlson AS; Topczewski JJ; Schonbrunn EK; Harki DA; Pomerantz W Molecular basis for the N-terminal bromodomain-and-extra-terminal-family selectivity of a dual kinase-bromodomain inhibitor. J. Med. Chem 2018, 61, 9316–9334. [Europe PMC free article] [Abstract] [Google Scholar]
(61) Niu Q; Liu Z; Alamer E; Fan X; Chen H; Endsley J; Gelman BB; Tian B; Kim JH; Michael NL; Robb ML; Ananworanich J; Zhou J; Hu H Structure-guided drug design identifies a BRD4-selective small molecule that suppresses HIV. J. Clin. Invest 2019, 129, 3361–3373. [Europe PMC free article] [Abstract] [Google Scholar]
(62) Jiang F; Hu Q; Zhang Z; Li H; Li H; Zhang D; Li H; Ma Y; Xu J; Chen H; Cui Y; Zhi Y; Zhang Y; Xu J; Zhu J; Lu T; Chen Y Discovery of benzo[cd]indol-2(1H)-ones and pyrrolo[4,3,2-de] quinolin-2(1H)-ones as bromodomain and extra-terminal domain (BET) inhibitors with selectivity for the first bromodomain with potential high efficiency against acute gouty arthritis. J. Med. Chem 2019, 62, 11080–11107. [Abstract] [Google Scholar]
(63) Liu Z; Chen H; Wang P; Li Y; Wold EA; Leonard PG; Joseph S; Brasier AR; Tian B; Zhou J Discovery of orally bioavailable chromone derivatives as potent and selective BRD4 inhibitors: scaffold hopping, optimization, and pharmacological evaluation. J. Med. Chem 2020, 63, 5242–5256. [Europe PMC free article] [Abstract] [Google Scholar]
(64) Watson RJ; Bamborough P; Barnett H; Chung C.-w.; Davis R; Gordon L; Grandi P; Petretich M; Phillipou A; Prinjha RK; Rioja I; Soden P; Werner T; Demont EH Demont EH GSK789: a selective inhibitor of the first bromodomains (BD1) of the bromo and extra terminal domain (BET) proteins. J. Med. Chem 2020, 63, 9045–9069. [Abstract] [Google Scholar]
(65) Wellaway CR; Bamborough P; Bernard SG; Chung CW; Craggs PD; Cutler L; Demont EH; Evans JP; Gordon L; Karamshi B; Lewis AJ; Lindon MJ; Mitchell DJ; Rioja I; Soden PE; Taylor S; Watson RJ; Willis R; Woolven JM; Wyspianska BS; Kerr WJ; Prinjha RK Structure-based design of a bromodomain and extraterminal domain (BET) inhibitor selective for the N-terminal bromodomains that retains an anti-inflammatory and antiproliferative phenotype. J. Med. Chem 2020, 63, 9020–9044. [Abstract] [Google Scholar]
(66) Cui H; Divakaran A; Pandey AK; Johnson JA; Zahid H; Hoell ZJ; Ellingson MO; Shi K; Aihara H; Harki DA; Pomerantz WCK Selective N-terminal BRD4 bromodomain inhibitors by targeting non-conserved residues and structured water displacement. Angew. Chem., Int. Ed 2021, 60, 1220–1226. [Europe PMC free article] [Abstract] [Google Scholar]
(67) Law RP; Atkinson SJ; Bamborough P; Chung CW; Demont EH; Gordon LJ; Lindon M; Prinjha RK; Watson A; Hirst DJ Discovery of tetrahydroquinoxalines as bromodomain and extra-terminal domain (BET) inhibitors with selectivity for the second bromodomain. J. Med. Chem 2018, 61, 4317–4334. [Abstract] [Google Scholar]
(68) Chen D; Lu T; Yan Z; Lu W; Zhou F; Lyu X; Xu B; Jiang H; Chen K; Luo C; Zhao Y Discovery, structural insight, and bioactivities of BY27 as a selective inhibitor of the second bromodomains of BET proteins. Eur. J. Med. Chem 2019, 182, 111633. [Abstract] [Google Scholar]
(69) Faivre EJ; Mcdaniel KF; Albert DH; Mantena SR; Plotnik JP; Wilcox D; Zhang L; Bui MH; Sheppard GS; Wang L; Sehgal V; Lin X; Huang X; Lu X; Uziel T; Hessler P; Lam LT; Bellin RJ; Mehta G; Fidanze S; Pratt JK; Liu D; Hasvold LA; Sun C; Panchal SC; Nicolette JJ; Fossey SL; Park CH; Longenecker K; Bigelow L; Torrent M; Rosenberg SH; Kati WM; Shen Y Selective inhibition of the BD2 bromodomain of BET proteins in prostate cancer. Nature 2020, 578, 306–310. [Abstract] [Google Scholar]
(70) Sheppard GS; Wang L; Fidanze SD; Hasvold LA; Liu D; Pratt JK; Park CH; Longenecker K; Qiu W; Torrent M; Kovar PJ; Bui M; Faivre E; Huang X; Lin X; Wilcox D; Zhang L; Shen Y; Albert DH; Magoc TJ; Rajaraman G; Kati WM; Mcdaniel KF Discovery of N-Ethyl-4-[2-(4-fluoro-2,6-dimethyl-phenoxy)-5-(1-hydroxy-1-methyl-ethyl)phenyl]-6-methyl-7-oxo-1H-pyrrolo[2,3-c]-pyridine-2-carboxamide (ABBV-744), a BET bromodomain inhibitor with selectivity for the second bromodomain. J. Med. Chem 2020, 63, 5585–5623. [Abstract] [Google Scholar]
(71) Preston A; Atkinson S; Bamborough P; Chung CW; Craggs PD; Gordon L; Grandi P; Gray J; Jones EJ; Lindon M; Michon AM; Mitchell DJ; Prinjha RK; Rianjongdee F; Rioja I; Seal J; Taylor S; Wall I; Watson RJ; Woolven J; Demont EH Design and synthesis of a highly selective and in vivo-capable inhibitor of the second bromodomain of the bromodomain and extra terminal domain family of proteins. J. Med. Chem 2020, 63, 9070–9092. [Abstract] [Google Scholar]
(72) Seal JT; Atkinson SJ; Aylott H; Bamborough P; Chung CW; Copley R; Gordon L; Grandi P; Gray J; Harrison LA; Hayhow TG; Lindon M; Messenger C; Michon AM; Mitchell D; Preston A; Prinjha RK; Rioja I; Taylor S; Wall ID; Watson RJ; Woolven JM; Demont EH The optimization of a novel, weak bromo and extra terminal domain (BET) bromodomain fragment ligand to a potent and selective second bromodomain (BD2) inhibitor. J. Med. Chem 2020, 63, 9093–9126. [Abstract] [Google Scholar]
(73) Slavish PJ; Chi L; Yun MK; Tsurkan L; Martinez NE; Jonchere B; Chai SC; Connelly M; Waddell MB; Das S; Neale G; Li Z; Shadrick WR; Olsen RR; Freeman KW; Low JA; Price JE; Young BM; Bharatham N; Boyd VA; Yang J; Lee RE; Morfouace M; Roussel MF; Chen T; Savic D; Guy RK; White SW; Shelat AA; Potter PM Bromodomain-selective BET inhibitors are potent antitumor agents against MYC-driven pediatric cancer. Cancer Res 2020, 80, 3507–3518. [Europe PMC free article] [Abstract] [Google Scholar]
(74) Leroy G; Rickards B; Flint SJ The double bromodomain proteins BRD2 and BRD3 couple histone acetylation to transcription. Mol. Cell 2008, 30, 51–60. [Europe PMC free article] [Abstract] [Google Scholar]
(75) Loven J; Hoke HA; Lin CY; Lau A; Orlando DA; Vakoc CR; Bradner JE; Lee TI; Young RA Selective inhibition of tumor oncogenes by disruption of super-enhancers. Cell 2013, 153, 320–334. [Europe PMC free article] [Abstract] [Google Scholar]
(76) Denis GV; Mccomb ME; Faller DV; Sinha A; Romesser PB; Costello CE Identification of transcription complexes that contain the double bromodomain protein BRD2 and chromatin remodeling machines. J. Proteome Res 2006, 5, 502–511. [Europe PMC free article] [Abstract] [Google Scholar]
(77) Lambert JP; Picaud S; Fujisawa T; Hou H; Savitsky P; Uuskula-Reimand L; Gupta GD; Abdouni H; Lin ZY; Tucholska M; Knight J; Gonzalez-Badillo B; St-Denis N; Newman JA; Stucki M; Pelletier L; Bandeira N; Wilson MD; Filippakopoulos P; Gingras AC Interactome rewiring following pharmacological targeting of BET bromodomains. Mol. Cell 2019, 73, 621–638. [Europe PMC free article] [Abstract] [Google Scholar]
(78) Leroy G; Rickards B; Flint SJ The double bromodomain proteins BRD2 and BRD3 couple histone acetylation to transcription. Mol. Cell 2008, 30, 51–60. [Europe PMC free article] [Abstract] [Google Scholar]
(79) Lamonica JM; Deng W; Kadauke S; Campbell AE; Gamsjaeger R; Wang H; Cheng Y; Billin AN; Hardison RC; Mackay JP; Blobel GA Bromodomain protein BRD3 associates with acetylated GATA1 to promote its chromatin occupancy at erythroid target genes. Proc. Natl. Acad. Sci. U. S. A 2011, 108, E159–E168. [Europe PMC free article] [Abstract] [Google Scholar]
(80) Matzuk MM; Mckeown MR; Filippakopoulos P; Li Q; Ma L; Agno JE; Lemieux ME; Picaud S; Yu RN; Qi J; Knapp S; Bradner JE Small-molecule inhibition of BRDT for male contraception. Cell 2012, 150, 673–684. [Europe PMC free article] [Abstract] [Google Scholar]
(81) Bryant JM; Berger SL Low-hanging fruit: targeting BRDT in the testes. EMBO J 2012, 31, 3788–3789. [Europe PMC free article] [Abstract] [Google Scholar]
(82) Liu Z; Tian B; Chen H; Wang P; Brasier AR; Zhou J Discovery of potent and selective BRD4 inhibitors capable of blocking TLR3-induced acute airway inflammation. Eur. J. Med. Chem 2018, 151, 450–461. [Europe PMC free article] [Abstract] [Google Scholar]
(83) Lv K; Chen W; Chen D; Mou J; Zhang H; Fan T; Li Y; Cao D; Wang X; Chen L; Shen J; Pei D; Xiong B Rational design and evaluation of 6-(Pyrimidin-2-ylamino)-3,4-dihydroquinoxalin-2(1H)-ones as polypharmacological inhibitors of BET and kinases. J. Med. Chem 2020, 63, 9787–9802. [Abstract] [Google Scholar]
(84) Martin MP; Olesen SH; Georg GI; Schonbrunn E Cyclin-dependent kinase inhibitor dinaciclib interacts with the acetyl-lysine recognition site of bromodomains. ACS Chem. Biol 2013, 8, 2360–2365. [Europe PMC free article] [Abstract] [Google Scholar]
(85) Carlino L; Rastelli G Dual kinase-bromodomain inhibitors in anticancer drug discovery: a structural and pharmacological perspective. J. Med. Chem 2016, 59, 9305–9320. [Abstract] [Google Scholar]
(86) Sun D; Zhao Y; Zhang S; Zhang L; Liu B; Ouyang L Dual-target kinase drug design: current strategies and future directions in cancer therapy. Eur. J. Med. Chem 2020, 188, 112025. [Abstract] [Google Scholar]
(87) Mazur PK; Herner A; Mello SS; Wirth M; Hausmann S; Sanchez-Rivera FJ; Lofgren SM; Kuschma T; Hahn SA; Vangala D; Trajkovic-Arsic M; Gupta A; Heid I; Noel PB; Braren R; Erkan M; Kleeff J; Sipos B; Sayles LC; Heikenwalder M; Hessmann E; Ellenrieder V; Esposito I; Jacks T; Bradner JE; Khatri P; Sweet-Cordero EA; Attardi LD; Schmid RM; Schneider G; Sage J; Siveke JT Combined inhibition of BET family proteins and histone deacetylases as a potential epigenetics-based therapy for pancreatic ductal adenocarcinoma. Nat. Med 2015, 21, 1163–1171. [Europe PMC free article] [Abstract] [Google Scholar]
(88) Shi J; Vakoc CR The mechanisms behind the therapeutic activity of BET bromodomain inhibition. Mol. Cell 2014, 54, 728–736. [Europe PMC free article] [Abstract] [Google Scholar]
(89) Donati B; Lorenzini E; Ciarrocchi A Ciarrocchi A BRD4 and cancer: going beyond transcriptional regulation. Mol. Cancer 2018, 17, 164. [Europe PMC free article] [Abstract] [Google Scholar]
(90) Pan Z; Li X; Wang Y; Jiang Q; Jiang L; Zhang M; Zhang N; Wu F; Liu B; He G Discovery of thieno[2,3-d] pyrimidine-based hydroxamic acid derivatives as bromodomain-containing protein 4/histone deacetylase dual inhibitors induce autophagic cell death in colorectal carcinoma cells. J. Med. Chem 2020, 63, 3678–3700. [Abstract] [Google Scholar]
(91) Zeng H; Qu J; Jin N; Xu J; Lin C; Chen Y; Yang X; He X; Tang S; Lan X; Yang X; Chen Z; Huang M; Ding J; Geng M Feedback activation of leukemia inhibitory factor receptor limits response to histone deacetylase inhibitors in breast cancer. Cancer Cell 2016, 30, 459–473. [Abstract] [Google Scholar]
(92) He S; Dong G; Li Y; Wu S; Wang W; Sheng C Potent Dual BET/HDAC Inhibitors for Efficient Treatment of Pancreatic Cancer. Angew. Chem., Int. Ed 2020, 59, 3028–3032. [Abstract] [Google Scholar]
(93) Cheng G; Wang Z; Yang J; Bao Y; Xu Q; Zhao L; Liu D Design, synthesis and biological evaluation of novel indole derivatives as potential HDAC/BRD4 dual inhibitors and anti-leukemia agents. Bioorg. Chem 2019, 84, 410–417. [Abstract] [Google Scholar]
(94) Amemiya S; Yamaguchi T; Hashimoto Y; Noguchi-Yachide T Synthesis and evaluation of novel dual BRD4/HDAC inhibitors. Bioorg. Med. Chem 2017, 25, 3677–3684. [Abstract] [Google Scholar]
(95) Zhang Z; Hou S; Chen H; Ran T; Jiang F; Bian Y; Zhang D; Zhi Y; Wang L; Zhang L; Li H; Zhang Y; Tang W; Lu T; Chen Y Targeting epigenetic reader and eraser: Rational design, synthesis and in vitro evaluation of dimethylisoxazoles derivatives as BRD4/HDAC dual inhibitors. Bioorg. Med. Chem. Lett 2016, 26, 2931–2935. [Abstract] [Google Scholar]
(96) Shao M; He L; Zheng L; Huang L; Zhou Y; Wang T; Chen Y; Shen M; Wang F; Yang Z; Chen L Structure-based design, synthesis and in vitro antiproliferative effects studies of novel dual BRD4/HDAC inhibitors. Bioorg. Med. Chem. Lett 2017, 27, 4051–4055. [Abstract] [Google Scholar]
(97) Yang L; Zhang Y; Shan W; Hu Z; Yuan J; Pi J; Wang Y; Fan L; Tang Z; Li C; Hu X; Tanyi JL; Fan Y; Huang Q; Montone K; Dang CV; Zhang L Repression of BET activity sensitizes homologous recombination-proficient cancers to PARP inhibition. Sci. Transl. Med 2017, 9, eaal1645. [Europe PMC free article] [Abstract] [Google Scholar]
(98) Chang X; Sun D; Shi F; Wang G; Chen Y; Zhang K; Tan H; Liu J; Liu B; Ouyang L Design, synthesis, and biological evaluation of quinazolin-4(3H)-one derivatives co-targeting poly-(ADP-ribose) polymerase-1 and bromodomain containing protein 4 for breast cancer therapy. Acta Pharm. Sin. B 2021, 11, 156–180. [Europe PMC free article] [Abstract] [Google Scholar]
(99) Ren Y; Bi C; Zhao X; Lwin T; Wang C; Yuan J; Silva AS; Shah BD; Fang B; Li T; Koomen JM; Jiang H; Chavez JC; Pham LV; Sudalagunta PR; Wan L; Wang X; Dalton WS; Moscinski LC; Shain KH; Vose J; Cleveland JL; Sotomayor EM; Fu K; Tao J PLK1 stabilizes a MYC-dependent kinase network in aggressive B cell lymphomas. J. Clin. Invest 2018, 128, 5517–5530. [Europe PMC free article] [Abstract] [Google Scholar]
(100) Xu Y; Wang Q; Xiao K; Liu Z; Zhao L; Song X; Hu X; Feng Z; Gao T; Zuo W; Zeng J; Wang N; Yu L Novel dual BET and PLK1 inhibitor WNY0824 exerts potent antitumor effects in CRPC by inhibiting transcription factor function and inducing mitotic abnormality. Mol. Cancer Ther 2020, 19, 1221–1231. [Abstract] [Google Scholar]
(101) Liu S; Yosief HO; Dai L; Huang H; Dhawan G; Zhang X; Muthengi AM; Roberts J; Buckley DL; Perry JA; Wu L; Bradner JE; Qi J; Zhang W Structure-guided design and development of potent and selective dual bromodomain 4 (BRD4)/polo-like kinase 1 (PLK1) inhibitors. J. Med. Chem 2018, 61, 7785–7795. [Europe PMC free article] [Abstract] [Google Scholar]
(102) Watts E; Heidenreich D; Tucker E; Raab M; Strebhardt K; Chesler L; Knapp S; Bellenie B; Hoelder S Designing dual inhibitors of anaplastic lymphoma kinase (ALK) and bromodomain-4 (BRD4) by tuning kinase selectivity. J. Med. Chem 2019, 62, 2618–2637. [Europe PMC free article] [Abstract] [Google Scholar]
(103) Koblan LW; Buckley DL; Ott CJ; Fitzgerald ME; Ember SW; Zhu JY; Liu S; Roberts JM; Remillard D; Vittori S; Zhang W; Schonbrunn E; Bradner JE Assessment of bromodomain target engagement by a series of BI2536 analogues with miniaturized BET-BRET. ChemMedChem 2016, 11, 2575–2581. [Abstract] [Google Scholar]
(104) Ciceri P; Muller S; O’Mahony A; Fedorov O; Filippakopoulos P; Hunt JP; Lasater EA; Pallares G; Picaud S; Wells C; Martin S; Wodicka LM; Shah NP; Treiber DK; Knapp S Dual kinase-bromodomain inhibitors for rationally designed polypharmacology. Nat. Chem. Biol 2014, 10, 305–312. [Europe PMC free article] [Abstract] [Google Scholar]
(105) Liu T; Wan Y; Xiao Y; Xia C; Duan G Dual-target inhibitors based on HDACs: novel antitumor agents for cancer therapy. J. Med. Chem 2020, 63, 8977–9002. [Abstract] [Google Scholar]
(106) Carlson AS; Cui H; Divakaran A; Johnson JA; Brunner RM; Pomerantz W; Topczewski JJ Systematically mitigating the p38alpha activity of triazole-based BET inhibitors. ACS Med. Chem. Lett 2019, 10, 1296–1301. [Europe PMC free article] [Abstract] [Google Scholar]
(107) Fu J; Yoon HG; Qin J; Wong J Regulation of P-TEFb elongation complex activity by CDK9 acetylation. Mol. Cell. Biol 2007, 27, 4641–4651. [Europe PMC free article] [Abstract] [Google Scholar]
(108) 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, 8554–8559. [Europe PMC free article] [Abstract] [Google Scholar]
(109) Lai AC; Crews CM Induced protein degradation: an emerging drug discovery paradigm. Nat. Rev. Drug Discovery 2017, 16, 101–114. [Europe PMC free article] [Abstract] [Google Scholar]
(110) Zhou B; Hu J; Xu F; Chen Z; Bai L; Fernandez-Salas E; Lin M; Liu L; Yang CY; Zhao Y; Mceachern D; Przybranowski S; Wen B; Sun D; Wang S Discovery of a small-molecule degrader of bromodomain and extra-terminal (BET) proteins with picomolar cellular potencies and capable of achieving tumor regression. J. Med. Chem 2018, 61, 462–481. [Europe PMC free article] [Abstract] [Google Scholar]
(111) Winter GE; Buckley DL; Paulk J; Roberts JM; Souza A; Dhe-Paganon S; Bradner JE DRUG DEVELOPMENT. Phthali-mide conjugation as a strategy for in vivo target protein degradation. Science 2015, 348, 1376–1381. [Europe PMC free article] [Abstract] [Google Scholar]
(112) Lu J; Qian Y; Altieri M; Dong H; Wang J; Raina K; Hines J; Winkler JD; Crew AP; Coleman K; Crews CM Hijacking the E3 ubiquitin ligase cereblon to efficiently target BRD4. Chem. Biol 2015, 22, 755–763. [Europe PMC free article] [Abstract] [Google Scholar]
(113) Bai L; Zhou B; Yang CY; Ji J; Mceachern D; Przybranowski S; Jiang H; Hu J; Xu F; Zhao Y; Liu L; Fernandez-Salas E; Xu J; Dou Y; Wen B; Sun D; Meagher J; Stuckey J; Hayes DF; Li S; Ellis MJ; Wang S Targeted degradation of BET proteins in triple-negative breast cancer. Cancer Res 2017, 77, 2476–2487. [Europe PMC free article] [Abstract] [Google Scholar]
(114) Qin C; Hu Y; Zhou B; Fernandez-Salas E; Yang CY; Liu L; Mceachern D; Przybranowski S; Wang M; Stuckey J; Meagher J; Bai L; Chen Z; Lin M; Yang J; Ziazadeh DN; Xu F; Hu J; Xiang W; Huang L; Li S; Wen B; Sun D; Wang S Discovery of QCA570 as an exceptionally potent and efficacious proteolysis targeting chimera (PROTAC) degrader of the bromodomain and extra-terminal (BET)proteins capable of inducing complete and durable tumor regression. J. Med. Chem 2018, 61, 6685–6704. [Europe PMC free article] [Abstract] [Google Scholar]
(115) Li L; Mi D; Pei H; Duan Q; Wang X; Zhou W; Jin J; Li D; Liu M; Chen Y In vivo target protein degradation induced by PROTACs based on E3 ligase DCAF15. Signal Transduction Targeted Ther 2020, 5, 129. [Europe PMC free article] [Abstract] [Google Scholar]
(116) Nowak RP; Deangelo SL; Buckley D; He Z; Donovan KA; An J; Safaee N; Jedrychowski MP; Ponthier CM; Ishoey M; Zhang T; Mancias JD; Gray NS; Bradner JE; Fischer ES Plasticity in binding confers selectivity in ligand-induced protein degradation. Nat. Chem. Biol 2018, 14, 706–714. [Europe PMC free article] [Abstract] [Google Scholar]
(117) Gadd MS; Testa A; Lucas X; Chan KH; Chen W; Lamont DJ; Zengerle M; Ciulli A Structural basis of PROTAC cooperative recognition for selective protein degradation. Nat. Chem. Biol 2017, 13, 514–521. [Europe PMC free article] [Abstract] [Google Scholar]
(118) Raina K; Lu J; Qian Y; Altieri M; Gordon D; Rossi AM; Wang J; Chen X; Dong H; Siu K; Winkler JD; Crew AP; Crews CM; Coleman KG PROTAC-induced BET protein degradation as a therapy for castration-resistant prostate cancer. Proc. Natl. Acad. Sci. U. S. A 2016, 113, 7124–7129. [Europe PMC free article] [Abstract] [Google Scholar]
(119) Sun B; Fiskus W; Qian Y; Rajapakshe K; Raina K; Coleman KG; Crew AP; Shen A; Saenz DT; Mill CP; Nowak AJ; Jain N; Zhang L; Wang M; Khoury JD; Coarfa C; Crews CM; Bhalla KN BET protein proteolysis targeting chimera (PROTAC) exerts potent lethal activity against mantle cell lymphoma cells. Leukemia 2018, 32, 343–352. [Abstract] [Google Scholar]
(120) Zengerle M; Chan KH; Ciulli A Selective small molecule induced degradation of the BET bromodomain protein BRD4. ACS Chem. Biol 2015, 10, 1770–1777. [Europe PMC free article] [Abstract] [Google Scholar]
(121) Testa A; Hughes SJ; Lucas X; Wright JE; Ciulli A Structure-based design of a macrocyclic PROTAC. Angew. Chem., Int. Ed 2020, 59, 1727–1734. [Europe PMC free article] [Abstract] [Google Scholar]
(122) Chan KH; Zengerle M; Testa A; Ciulli A Impact of target warhead and linkage vector on inducing protein degradation: comparison of bromodomain and extra-terminal (BET) degraders derived from triazolodiazepine (JQ1) and tetrahydroquinoline (I-BET726) BET inhibitor scaffolds. J. Med. Chem 2018, 61, 504–513. [Europe PMC free article] [Abstract] [Google Scholar]
(123) Bondeson DP; Smith BE; Burslem GM; Buhimschi AD; Hines J; Jaime-Figueroa S; Wang J; Hamman BD; Ishchenko A; Crews CM Lessons in PROTAC design from selective degradation with a promiscuous warhead. Cell Chem. Biol 2018, 25, 78–87. [Europe PMC free article] [Abstract] [Google Scholar]
(124) Yang CY; Qin C; Bai L; Wang S Small-molecule PROTAC degraders of the Bromodomain and Extra Terminal (BET) proteins - A review. Drug Discovery Today: Technol 2019, 31, 43–51. [Abstract] [Google Scholar]
(125) Maneiro MA; Forte N; Shchepinova MM; Kounde CS; Chudasama V; Baker JR; Tate EW Antibody-PROTAC conjugates enable HER2-dependent targeted protein degradation of BRD4. ACS Chem. Biol 2020, 15, 1306–1312. [Europe PMC free article] [Abstract] [Google Scholar]
(126) Takahashi D; Moriyama J; Nakamura T; Miki E; Takahashi E; Sato A; Akaike T; Itto-Nakama K; Arimoto H AUTACs: cargo-specific degraders using selective autophagy. Mol. Cell 2019, 76, 797–810. [Abstract] [Google Scholar]
(127) Ivanov AA; Khuri FR; Fu H Targeting protein-protein interactions as an anticancer strategy. Trends Pharmacol. Sci 2013, 34, 393–400. [Europe PMC free article] [Abstract] [Google Scholar]
(128) Cai D; Lee AY; Chiang CM; Kodadek T Peptoid ligands that bind selectively to phosphoproteins. Bioorg. Med. Chem. Lett 2011, 21, 4960–4964. [Europe PMC free article] [Abstract] [Google Scholar]
(129) Zhong H-J; Lu L; Leung K-H; Wong CCL; Peng C; Yan S-C; Ma D-L; Cai Z; David Wang H-M; Leung C-H An iridium(iii)-based irreversible protein-protein interaction inhibitor of BRD4 as a potent anticancer agent. Chem. Sci 2015, 6, 5400–5408. [Europe PMC free article] [Abstract] [Google Scholar]
(130) Leung CH; Zhang JT; Yang GJ; Liu H; Han QB; Ma DL Emerging screening approaches in the development of Nrf2-Keap1 protein-protein interaction inhibitors. Int. J. Mol. Sci 2019, 20, 4445. [Europe PMC free article] [Abstract] [Google Scholar]
(131) Padmanabhan A; Alexanian M; Linares-Saldana R; Gonzalez-Teran B; Andreoletti G; Huang Y; Connolly AJ; Kim W; Hsu A; Duan Q; Winchester SAB; Felix F; Perez-Bermejo JA; Wang Q; Li L; Shah PP; Haldar SM; Jain R; Srivastava D Srivastava D BRD4 interacts with GATA4 to govern mitochondrial homeostasis in adult cardiomyocytes. Circulation 2020, 142, 2338–2355. [Europe PMC free article] [Abstract] [Google Scholar]
(132) Kulikowski E; Rakai BD; Wong N Inhibitors of bromodomain and extra-terminal proteins for treating multiple human diseases. Med. Res. Rev 2021, 41, 223–245. [Europe PMC free article] [Abstract] [Google Scholar]
(133) Feig M Computational protein structure refinement: almost there, yet still so far to go. Wiley Interdiscip Rev. Comput. Mol. Sci 2017, 7, e1307. [Europe PMC free article] [Abstract] [Google Scholar]
(134) Cerutti DS; Case DA Molecular dynamics simulations of macromolecular crystals. Wiley Interdiscip. Rev.: Comput. Mol. Sci 2019, 9, e1402. [Europe PMC free article] [Abstract] [Google Scholar]

Citations & impact 


Impact metrics

Jump to Citations
Jump to Data

Citations of article over time

Alternative metrics

Altmetric item for https://www.altmetric.com/details/100666646
Altmetric
Discover the attention surrounding your research
https://www.altmetric.com/details/100666646

Article citations


Go to all (32) article citations

Data 


Data behind the article

This data has been text mined from the article, or deposited into data resources.

Funding 


Funders who supported this work.

Cancer Prevention and Research Institute of Texas (2)

China Postdoctoral Science Foundation (1)

Department of Science and Technology of Sichuan Province (1)

NCI NIH HHS (1)

National Institutes of Health (1)

National Natural Science Foundation of China (4)