Aims Bromodomain-containing protein 4 (BRD4) is a member of the bromodomain and extra-terminal domain-containing (BET) family of chromatin “reader” proteins. Small molecule BET inhibitors have been developed within the context of cancer and inflammation pathologies, and more recently are of interest for central nervous system (CNS) disorders, including opioid use disorder (OUD). BRD4 has two bromodomains (BDs) that recognize specific post-translational histone variants to control downstream transcriptional outcomes. The protein-protein interaction between BRD4 and lysine residues of histones is driven through the two highly conserved N-terminal bromodomains BD1 and BD2. The aim of the present study is to pharmacologically characterize BRD4-BD1-specific inhibitors to facilitate investigations into the role of BRD4 mechanisms in preclinical models of opioid use disorder. Methods Selected potent BRD4 inhibitors were synthesized and scaled up for analyses of in vitro and in vivo drug metabolism and pharmacokinetics (DMPK), blood-brain barrier (BBB) penetration, and pharmacological profiles against a panel of CNS receptors, channels and transporters via the Psychoactive Drug Screening Program (PDSP). Based on initial profiles, we further evaluated one compound for in vivo efficacy in male Sprague-Dawley rats trained to stability on intravenous oxycodone self-administration (0.1 mg/kg/infusion). Results ZL0513, ZL0516, ZL0742, and ZL0969 exhibit selectivity for BRD4-BD1 over other BD proteins and are the subject of ongoing profiling. To date, ZL0513, ZL0516, and ZL0969 exhibit excellent in vitro DMPK profiles, including bioavailability greater than 35%, aqueous solubility, metabolic stability, and weak inhibition of CYP450 enzymes. ZL0987 (HCl salt form of ZL0516) exhibited a brain/plasma ratio (intravenous, 1 hr) of 0.08 which was improved to 0.62 for ZL0969. However, ZL0516 exhibited a cleaner PDSP profile than did ZL0969. ZL0742 exhibited a good in vivo DMPK, however its penetration of the BBB was suboptimal. Additional backup molecules ZL0513 and ZL0742 are undergoing further DMPK, BBB, and PDSP screening. Based upon our initial profiling of ZL0987, we assessed its efficacy in vivo and found that ZL0987 at the dose of 10 mg/kg suppressed oxycodone intake (p<0.05) which was sustained across the entire 180-minute self-administration session. Conclusion We have characterized several BRD4-BD1 selective inhibitors with compelling profiles of drug-like properties. Our initial in vivo data revealed that ZL0987 elicited sustained suppression of oxycodone self-administration. The development and characterization of specific BRD4 inhibitors will allow exploration of BRD4 mechanisms in preclinical models of opioid use and validate the BRD4 as a novel drug target for the development of epigenetic therapeutics for OUD.