JQ1, formally known as (+)-JQ1, is a thienotriazolodiazepinesmall molecule that serves as a potent, selective, and cell-permeable inhibitor of the bromodomain and extra-terminal (BET) family of epigenetic reader proteins, including BRD2, BRD3, BRD4, and BRDT.[1] It competitively binds to the acetyl-lysine recognition pockets of these bromodomains with nanomolar affinity, typically exhibiting dissociation constants (Kd) around 50 nM for BRD4 bromodomain 1 and half-maximal inhibitory concentrations (IC50) of 77 nM and 33 nM for BRD4 bromodomains 1 and 2, respectively.[1][2]Developed through structure-based drug design, JQ1 represents the first characterized selective BETbromodomain inhibitor, synthesized as the active (+)-enantiomer from a racemic mixture to enhance potency and minimize off-target effects.[1] Its mechanism involves displacing BET proteins from chromatin-bound acetylated histones, thereby disrupting the recruitment of transcriptional complexes like P-TEFb and suppressing the expression of key oncogenes (e.g., MYC) and inflammatory mediators.[1] This selective inhibition is confined to the BET family, with no significant activity against non-BETbromodomains or other protein classes at therapeutic concentrations.[1]Initially identified for its efficacy in NUT midline carcinoma (NMC), an aggressive squamous cell malignancy driven by BRD4-NUT fusion oncoproteins, JQ1 induces squamous differentiation, growth arrest, and apoptosis in NMC cells both in vitro and in xenograft models, significantly reducing tumor burden and prolonging survival.[1] Beyond NMC, preclinical studies have demonstrated JQ1's broad anti-tumor activity across hematological malignancies (e.g., multiple myeloma, acute myeloid leukemia) and solid tumors (e.g., pancreatic ductal adenocarcinoma, triple-negative breast cancer), often by downregulating super-enhancer-driven oncogenes and sensitizing cells to DNA damage or hypoxia.[3][4][5]JQ1 has also revealed BET proteins' roles in non-oncological contexts, including suppressing neuroinflammation and tau hyperphosphorylation in Alzheimer's disease models, inhibiting inflammatory responses in macrophages, and impairing spermatogenesis in male mice, highlighting its potential as a tool for studying epigenetics in inflammation, neurodegeneration, and reproduction.[6][7][8] As a chemical probe, JQ1 paved the way for clinical BET inhibitors like OTX015 and I-BET762, though its own therapeutic development has been limited by pharmacokinetic challenges, such as rapid clearance.[5]
Chemical properties
Structure and nomenclature
JQ1 is a synthetic small molecule classified as a thienotriazolodiazepine, featuring a fused ring system that includes a thiophene, a 1,2,4-triazolo, and a 1,4-diazepine moiety. Its molecular formula is C_{23}H_{25}ClN_4O_2S, and it has a molecular weight of 457.0 g/mol.The systematic IUPAC name for JQ1 is 2-methyl-2-propanyl [(6S)-4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-f][1,2,4]triazolo[4,3-a][1,4]diazepin-6-yl]acetate (CAS 1268524-70-4), reflecting its complex polycyclic architecture.[9][10] The core scaffold consists of a thienotriazolodiazepine ring system substituted with a 4-chlorophenyl group at position 4, methyl groups at positions 2, 3, and 9, and a tert-butyl acetate ester group attached at position 6. This structural arrangement positions key functional groups for selective interactions within protein binding pockets.JQ1 possesses a single chiral center at carbon 6 (C6) of the diazepine ring, adopting the (S)-configuration in its active form.[9] The (+)-JQ1 enantiomer exhibits biological activity, whereas the (-)-JQ1 enantiomer is inactive, highlighting the importance of stereochemistry for its function. The triazolodiazepine moiety contributes to its recognition by target proteins, though detailed binding mechanisms are addressed elsewhere.
Synthesis and physical characteristics
JQ1 is synthesized through a multi-step process starting from thiophene derivatives, as originally described in patents by Mitsubishi Tanabe Pharma Corporation. The key steps include triazole ring formation via reaction of the thiophene intermediate with a cyclic ketone and sulfur in alcohol under reflux, followed by chloroacetylation and amination with liquid ammonia. Subsequent diazepine ring closure is achieved by treating the triazole intermediate with diphosphorus pentasulfide and hydrazine hydrate, then cyclizing with triethyl orthoacetate in toluene under heating. The process concludes with esterification of the carboxylic acid group using tert-butanol in the presence of sulfuric acid to yield the tert-butyl ester. This route has been adapted for scalable production using a one-pot three-step method involving thionation of a benzodiazepine precursor with Lawesson's reagent, amidrazone formation with hydrazine hydrate, and triazole installation with trimethyl orthoacetate, affording (±)-JQ1 in 60% yield over the final steps.[11][12]JQ1 appears as a white to off-white or yellowish crystalline solid. The compound exhibits moderate lipophilicity, with a calculated logP value of around 4.5, which contributes to its cell permeability but limits aqueous solubility.[13]Solubility profiles are critical for laboratory handling and formulation: JQ1 is highly soluble in dimethyl sulfoxide (DMSO) at concentrations exceeding 50 mg/mL, facilitating stock solution preparation for cell-based assays. It shows moderate solubility in ethanol (≥46.9 mg/mL with sonication) and is practically insoluble in water (<0.01 mg/mL), necessitating organic solvents or cosolvent systems for aqueous applications.[2][14][15]Under standard storage conditions (desiccated at -20°C or room temperature), JQ1 remains stable for up to 2-3 years, with no significant degradation observed.[16][2]
Mechanism of action
Bromodomain inhibition
JQ1 is a selective inhibitor of the bromodomains and extra-terminal (BET) family proteins, specifically targeting BRD2, BRD3, BRD4, and BRDT.[17] These proteins contain tandem bromodomains (BD1 and BD2) that recognize acetylated lysine residues on histones, facilitating chromatin association and transcriptional regulation. JQ1 exhibits high selectivity for BET bromodomains over other bromodomain-containing proteins, such as BRD9. Its selective inhibition is confined to the BET family, with no significant activity against non-BET bromodomains or other protein classes at therapeutic concentrations, though JQ1 has also been shown to directly activate the pregnane X receptor (PXR) in a bromodomain-independent manner, potentially affecting drug metabolism.[17][18]The binding mechanism of JQ1 involves competitive inhibition at the acetyl-lysine recognition pockets of BET bromodomains. The triazolodiazepine core of JQ1 mimics the acetyl-lysine motif, occupying the binding site and displacing histone acetylation marks. This prevents the recruitment of BET proteins to acetylated chromatin, thereby disrupting their role in transcriptional activation.[17] The inhibition is non-covalent and reversible, as demonstrated by competitive displacement assays with acetylated histone peptides.[17]Affinity data from AlphaScreen assays highlight JQ1's potency against BET bromodomains, with the following IC<sub>50</sub> values:
In contrast, JQ1 shows markedly reduced affinity for non-BET bromodomains, with an IC<sub>50</sub> >12,942 nM for BRD9.[17][15]The structural basis of JQ1 binding has been elucidated by X-ray crystallography of JQ1 in complex with BRD4 BD1 (PDB: 3MXF) and BRD2 BD2 (PDB: 3ONI). JQ1 occupies the ZA channel within the bromodomain, forming key interactions including a hydrogen bond with the conserved asparagine (Asn140 in BRD4 BD1) via its triazole nitrogen. Additional stabilization occurs through hydrophobic contacts and potential hydrogen bonding involving Tyr97 in BRD4, ensuring specific and high-affinity binding.[17][19]
Effects on cellular processes
JQ1's inhibition of bromodomains in BRD4 disrupts the interaction between BRD4 and the positive transcription elongation factor b (P-TEFb) complex, preventing the phosphorylation of the C-terminal domain of RNA polymerase II at serine 2. This interference halts the productive elongation phase of transcription, particularly affecting genes driven by super-enhancers, which are clusters of enhancers marked by high levels of histone acetylation and BRD4 occupancy. As a result, the transcription of key oncogenes is suppressed, with JQ1 treatment leading to downregulation of MYC, BCL2, and Aurora kinases in various cancer cell lines; for instance, MYC mRNA levels are reduced by more than 80% in sensitive medulloblastoma cells within hours of exposure. This occurs through eviction of BET proteins from super-enhancers.[20][21][22][23]At the chromatin level, JQ1 promotes the eviction of BRD4 from acetylated lysine residues on histones, leading to decreased histone acetylation at gene promoters and altered chromatinaccessibility. This eviction reduces the recruitment of histone acetyltransferases and other co-activators to super-enhancer regions, thereby diminishing the transcriptional output of associated genes without directly inhibiting acetyltransferase activity. In proliferating cells, these molecular changes culminate in cellular outcomes such as G1 phasecell cycle arrest, activation of caspases leading to apoptosis, and induction of senescence, with the arrest mediated by upregulation of cyclin-dependent kinase inhibitors like p21.[24][25][26][27][24]Pharmacokinetically, JQ1 exhibits rapid cellular uptake and preferential nuclear localization due to its affinity for bromodomain-containing proteins like BRD4, allowing quick engagement with nuclear targets. Although its plasmahalf-life is short, approximately 1 hour, the sustained cellular effects arise from JQ1's prolonged residence time on BRD4, which maintains inhibition even after drug clearance.[28]
JQ1 has demonstrated potent antiproliferative effects in various hematological malignancy models in vitro, particularly those dependent on BRD4 activity. In multiple myeloma cell lines, JQ1 inhibits cell growth with GI50 values in the range of 50-500 nM, primarily through downregulation of MYC expression, which is a key driver in these cancers.[29] Similarly, in NUT midline carcinoma cells harboring BRD4-NUT fusions, JQ1 exhibits selective cytotoxicity at nanomolar concentrations (IC50 typically below 1 μM), leading to rapid differentiation and growth arrest.[1] In MYC-driven lymphoma models, JQ1 shows marked selectivity, suppressing proliferation in sensitive lines while sparing non-MYC-dependent cells, highlighting its utility in targeting oncogene addiction.[29]In preclinical mouse xenograft models, JQ1 induces significant tumor regression in BRD4-dependent cancers. For instance, in MLL-fusion leukemia xenografts, once-daily dosing at 50 mg/kg IP results in over 50% tumor volume reduction and prolonged survival.[30]Efficacy extends to solid tumors, with JQ1 reducing tumor burden by approximately 60-80% in lung adenocarcinoma and prostate cancer xenografts, correlating with decreased MYC levels and impaired tumor cell proliferation.[31][32] These effects are observed across patient-derived xenografts, underscoring JQ1's broad preclinical antitumor activity in BRD4-overexpressing models.Acquired resistance to JQ1 in cancer cells arises through multiple mechanisms, including BRD4 mutations that impair inhibitor binding and activation of alternative enhancers that bypass BET dependence. Prolonged exposure in vitro leads to resistant clones in leukemia and breast cancer lines, where BRD4 phosphorylation or degradation pathways are upregulated, reducing JQ1 sensitivity. Combination strategies mitigate this, as JQ1 synergizes with HDAC inhibitors like romidepsin to enhance apoptosis in acute myeloid leukemia cells, achieving greater than additive growth inhibition.[33] Similarly, pairing JQ1 with chemotherapy agents such as gemcitabine potentiates antitumor effects in pancreatic cancer models by increasing DNA damage and cell death.[34]Despite these promising results, JQ1's clinical translation is limited by its short plasmahalf-life of approximately 1 hour in mice, necessitating frequent dosing and complicating sustained inhibition. JQ1 has not advanced to direct human trials, serving primarily as a preclinical probe, with its analogs pursued for therapeutic development. Recent studies as of 2025 continue to explore synergies, such as with selinexor in AML xenografts, demonstrating enhanced efficacy through dual MYC inhibition.[35]
Non-oncological applications
JQ1 has demonstrated preclinical efficacy in reducing inflammatory responses in non-oncological contexts, particularly by suppressing cytokine production in immune cells. In lipopolysaccharide (LPS)-stimulated mouse macrophages, such as bone marrow-derived macrophages (BMDMs) and RAW 264.7 cells, JQ1 treatment at 400 nM significantly impairs the production of proinflammatory cytokines including TNF-α and IL-6, with reductions of up to 80% observed after 24 hours of exposure (P < 0.001).[36] This effect stems from JQ1's disruption of BET protein binding to acetylated histones at cytokine gene promoters, thereby inhibiting NF-κB-mediated transcription.[36] In vivo, JQ1 administered at 50 mg/kg intraperitoneally (IP) to endotoxemic mice blunts the cytokine storm, lowering serum TNF-α and IL-6 levels and improving survival rates following lethal LPS challenge (20 mg/kg IP, P = 0.001).[37]In models of inflammatory arthritis, JQ1 attenuates disease progression and bone erosion. In the collagen-induced arthritis (CIA) mouse model, therapeutic administration of JQ1 reduces arthritis severity scores (P < 0.05), synovial inflammation, and osteoclast activation, as evidenced by decreased bone resorption pits and downregulated expression of osteoclast-related genes via inhibition of the RANKL/RANK signaling pathway.[38] Histological analyses confirm preserved joint integrity and reduced cartilage degradation in JQ1-treated CIA mice, highlighting its potential to mitigate NF-κB-driven inflammatory cascades in rheumatoid arthritis-like conditions.[38]JQ1 exhibits CNS penetration, enabling applications in neurological disorders by modulating neuroinflammation. The compound crosses the blood-brain barrier, influencing processes in brain regions such as the anterior cingulate cortex, where it impairs remote fear memory extinction by inhibiting insulin-like growth factor 2 (IGF-2) upregulation and neuroplasticity-related gene expression.[39] In experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis, BRD4 inhibition with JQ1 reduces proinflammatory cytokine release from microglia and attenuates neuroinflammatory signaling through NF-κB suppression, thereby limiting T-cell imbalance and blood-brain barrier disruption.[40] These effects underscore JQ1's role in dampening inflammasome-dependent pathways in neuroinflammatory settings.[41]In cardiovascular disease models, JQ1 prevents pathological remodeling by blocking BRD4-dependent gene expression. In mice subjected to transverse aortic constriction (TAC)-induced pressure overload, daily IP administration of JQ1 at 50 mg/kg improves left ventricular ejection fraction, reduces cardiac hypertrophy (heart weight/tibia length ratio, n=10, P < 0.05), and attenuates lung congestion, without affecting physiological hypertrophy in swimming models.[42] This protection arises from JQ1's suppression of NF-κB and TGF-β signaling networks, which drive innate inflammatory and profibrotic responses in cardiomyocytes and fibroblasts.[42] Similarly, in post-myocardial infarction models, JQ1 enhances cardiac function by decreasing left ventricular diastolic area and wall thickness (n=10 per group).[42]Beyond these areas, JQ1 shows promise in antiviral and antifibrotic applications. For HIV, JQ1 reactivates latent provirus by antagonizing BRD4's inhibition of Tat-transactivation, promoting recruitment of the super elongation complex to the HIV promoter and enhancing viral transcription without broad T-cell activation.[43] In fibrosis models, such as TGF-β-stimulated lung fibroblasts from idiopathic pulmonary fibrosis patients, JQ1 at concentrations of 100-500 nM restores redox balance by reversing NOX4 upregulation and SOD2 downregulation, while inhibiting myofibroblast differentiation and ROS production via disruption of BRD3/BRD4 binding to the NOX4 promoter.[44]Preclinical studies typically employ JQ1 dosing of 50 mg/kg IP in mice, administered daily, which achieves robust target engagement in tissues like macrophages and heart without inducing overt toxicity or mortality.[37][42]
Development and analogs
Discovery and initial studies
JQ1 was first identified in 2010 by James E. Bradner and his team at the Dana-Farber Cancer Institute, as part of efforts to develop selective inhibitors for bromodomain-containing proteins.[45] This work built directly on a 2006 patent filed by Mitsubishi Tanabe Pharma Corporation, which described thienotriazolodiazepine compounds capable of binding to bromodomains, originally explored for anti-inflammatory applications such as inhibiting CD28 co-stimulation.[46] Using molecular modeling of the BRD4 bromodomain (PDB ID: 2OSS), the researchers rationally designed JQ1—a thieno-triazolo-1,4-diazepine derivative—with a bulky t-butyl ester substituent at the C6 position to enhance specificity and minimize off-target binding to benzodiazepine receptors.[1]The key publication detailing JQ1's discovery appeared in Nature in December 2010 by Filippakopoulos et al., which demonstrated its selective inhibition of bromodomains in the BET (bromodomain and extra-terminal) family, including BRD2, BRD3, and BRD4. The study highlighted JQ1's ability to competitively displace BET proteins from acetylated chromatin, leading to anti-proliferative effects in cancer cell lines, particularly those driven by Myc oncogene dysregulation, and prompting squamous differentiation in relevant models. Initial validation involved synthesis of JQ1 enantiomers via a seven-step route, followed by biochemical assays confirming its potency.[1]High-throughput screening of JQ1 against a panel of 37 human bromodomains, using differential scanning fluorimetry (DSF), revealed high selectivity for BET proteins, with observed thermal shift values (ΔTm) ranging from 4.2°C to 10.1°C.[1] Structure-activity relationship (SAR) studies further optimized its profile, showing nanomolar binding affinities (Kd 50–90 nM) via isothermal titration calorimetry (ITC) and competitive inhibition (IC50 33–77 nM) in alpha-screen assays. Co-crystal structures with BRD4(1) and BRD2(2) bromodomains provided the structural basis for its shape complementarity in the acetyl-lysine binding pocket.[1]To accelerate global research, Bradner's team chose not to pursue patents on JQ1, instead open-sourcing the compound and its synthesis protocol, a decision rooted in promoting equitable access and rapid validation across the scientific community.[47] This ethical approach facilitated its swift adoption as a chemical probe, with the foundational Nature paper garnering over 3,900 citations by 2024 and establishing BET proteins as druggable targets in oncology and beyond.[48]
Derivatives and clinical translation
Following the discovery of JQ1 as a prototype BET bromodomain inhibitor, several derivatives were developed to address its limitations, particularly its short plasma half-life of approximately 1 hour, which precluded advancement to clinical trials.[49] Key analogs include I-BET762 (also known as molibresib or GSK525762), developed by GlaxoSmithKline, which exhibits improved pharmacokinetics with a human half-life exceeding 6 hours, enabling oral dosing in phase I/II trials for advanced solid tumors and hematologic malignancies.[50] Similarly, OTX015 (MK-8628 or birabresib) is a thienotriazolodiazepine derivative of JQ1 with enhanced stability and a prolonged half-life of 33–82 hours in humans, supporting its evaluation in phase I/II studies for nuclear protein of the testis (NUT) midline carcinoma and other cancers.[51]BMS-986158, another selective BET inhibitor from Bristol Myers Squibb, demonstrated a terminal half-life of approximately 60 hours in earlier studies, but its development was discontinued in 2024.[52]As of 2025, JQ1 itself has not progressed to clinical use due to its pharmacokinetic drawbacks, but over 20 JQ1-inspired BET inhibitors are in clinical development worldwide, primarily for oncology indications such as diffuse large B-cell lymphoma (in phase II trials) and other hematologic and solid tumors, with exploratory applications in inflammatory and neurological disorders.[53] These compounds have shown preliminary antitumor activity, including tumor regression in select patients, though efficacy has been modest and variable across trials.[54] Early-phase studies highlight common challenges, including dose-limiting thrombocytopenia (affecting platelet counts in up to 15% of patients) and gastrointestinal toxicities such as nausea and diarrhea, which often necessitate dose reductions or interruptions.[55] To mitigate these on-target effects, researchers have pursued degradation-based strategies, such as proteolysis-targeting chimeras (PROTACs) that induce ubiquitination and proteasomal degradation of BET proteins rather than mere inhibition.[56]Ongoing research emphasizes second-generation JQ1 derivatives designed for greater selectivity, such as bromodomain 2 (BD2)-specific inhibitors that spare BD1 to reduce toxicity while maintaining efficacy against oncogenic transcription.[57] Examples include PROTAC-based molecules like dBET6, a JQ1-thalidomide conjugate that potently degrades BRD4 (IC<sub>50</sub> ≈ 10 nM) and exhibits antitumor activity in preclinical models with potentially improved therapeutic windows.[58] Several analogs have received FDA orphan drug designations for rare oncology indications, including ZEN-3694 for NUT carcinoma, providing incentives for further development; however, no BET inhibitors have achieved full regulatory approval as of 2025.[59] These efforts underscore a shift toward combination therapies and tissue-specific delivery to enhance clinical translation.[60]