Bromodomain and extraterminal domain (BET) proteins are epigenetic readers that regulate gene expression and are involved in cancer pathogenesis. Over the last years, several BET inhibitors have been developed and clinically tested. Results from the first clinical trials show limited single-agent activity in a small subset of patients with hematologic malignancies and in NUT carcinoma. Adverse events have been observed and may limit treatment compliance. Here, we review the preclinical rationale for targeting BET proteins in cancer and the preliminary results from clinical trials, and outline future directions for the use of BET inhibitors as antitumor agents.

Significance: BET inhibitors represent a new class of anticancer agents. Results from the first clinical trials confirm the antitumor potential of BET inhibitors, but their efficacy as single agents seems to be limited. Based on preclinical data, combination therapies with other anticancer agents and the development of a new generation of compounds may open new possibilities for targeting BET proteins as effective anticancer strategies. Cancer Discov; 8(1); 24–36. ©2017 AACR.

Human cells contain tens of thousands of active promoter regions and enhancer regions that are highly cell-type dependent, and a few hundred superenhancers, which are clusters of enhancers characterized by a very high binding of Mediator complexes and master transcription factors (1–3). For example, in a multiple myeloma cell line, Loven and colleagues have identified approximately 10,000 annotated active transcription start sites, 8,000 enhancers, and 308 superenhancers (1). Superenhancer-associated transcripts comprise the key cell identity genes and are expressed at higher levels than genes associated with normal enhancers (2–4).

Bromodomain and extraterminal domain (BET) proteins are epigenetic readers characterized by the presence of two tandem bromodomains (BD1 and BD2), an extraterminal domain (ET), and a C-terminal domain (CTD; ref. 5). They comprise the ubiquitously expressed BRD2, BRD3, and BRD4 and the testis-restricted BRDT, and mainly recognize acetylated lysine of histone 4 (5, 6). BET proteins also recognize acetylated nonhistone proteins, including different transcription factors (5–8). Examples are the binding of BRD4 to the acetylated transcription factor TWIST, involved in mesoderm formation (9); to acetylated RelA, regulating the transcriptional activity of NF-kappaB (10); or to acetylated ERG in acute myeloid leukemia (AML) cells (8). BET proteins can also bind other proteins, including transcription factors, in a bromodomain-independent manner, such as the binding of BRD4 to FLI1, MYB, SPI1 (PU.1), CEBPA, CEBPB (8), or p53 (11). Finally, BET can also have kinase activity (9), a function not yet fully understood.

The fundamental role of BET proteins is demonstrated by the fact that homozygous deletion of BRD2 or BRD4 is embryonic lethal (12–15). BET proteins, acting as scaffolds to recruit other proteins, are localized at promoters and especially at enhancers of active genes, participating with the Mediator complex, as master transcription elongation factors (refs. 4, 5, 7, 16, 17; Fig. 1). BET proteins match the chromatin acetylation status with transcriptional elongation via displacing HEXIM1/7SK snRNP from the transcription elongation factor b Cyclin T1/CDK9 complex (P-TEFb), allowing the latter to activate RNA polymerase II via phosphorylation at serine 2 (refs. 4, 7, 16, 18; Fig. 1). In addition, especially at enhancers, BET proteins also recruit the demethylase JMJD6, which induces degradation of 7SK snRNP and P-TEFb activation (7, 9, 19). However, the exact role of BET proteins in the transcription machinery has yet to be elucidated. Data obtained studying BRD4 in AML cells show that the recruitment of BET proteins at promoters and enhancers is mediated by transcription factors mainly due to the binding of BRD4 to p300/CBP acetyltransferase–mediated acetylated lysine residues (8). Moreover, a recent publication provided evidence that BRD4 is fundamental for the creation of the productive transcription elongation factor complex but not for the direct recruitment of P-TEFb (4).

Figure 1.

Schematic representation of the mechanism of action of BET inhibitors. BET proteins recognize acetylated lysine of histone 4 and act as scaffolds to recruit many other proteins to promoters and at enhancers of active genes, especially at the superenhancers of key genes, driving the transcription process. Exposure of tumor cells to BET inhibitors reduces the levels of BRD4 at enhancers and at promoters at a genome-wide level, but the reduction is more marked at superenhancers, and the genes associated with superenhancers undergo stronger and faster downregulation than genes regulated by standard enhancers. Examples of such genes are MYC, NMYC, IL7R, FOSL1, AR, ER, BCL2, BCL6, PAX5, CDK4, and CDK6. BET-i, BET inhibitor; Med, mediator; TF, transcription factor.

Figure 1.

Schematic representation of the mechanism of action of BET inhibitors. BET proteins recognize acetylated lysine of histone 4 and act as scaffolds to recruit many other proteins to promoters and at enhancers of active genes, especially at the superenhancers of key genes, driving the transcription process. Exposure of tumor cells to BET inhibitors reduces the levels of BRD4 at enhancers and at promoters at a genome-wide level, but the reduction is more marked at superenhancers, and the genes associated with superenhancers undergo stronger and faster downregulation than genes regulated by standard enhancers. Examples of such genes are MYC, NMYC, IL7R, FOSL1, AR, ER, BCL2, BCL6, PAX5, CDK4, and CDK6. BET-i, BET inhibitor; Med, mediator; TF, transcription factor.

Close modal

Preclinical Rationale for Targeting BET Proteins in Human Cancer

Several lines of evidence coming from preclinical studies indicate a role of BET proteins in human cancer and have provided the rationale for targeting BET proteins as a strategy for the development of new anticancer drugs. Genetic screening programs performed in different tumor types have recurrently identified the genes encoding BET proteins as genes on which neoplastic cells depend for their survival (20–23). Overexpression of BRD2/BRD4 genes also occurs in human cancers (23, 24). Finally, the most direct and clear evidence of the involvement of BET proteins in the pathogenesis of human cancer derives from NUT carcinoma, a rare and aggressive form of undifferentiated squamous cell carcinoma that mainly affects midline structures. Initially thought of as a disease of children and adolescents, NUT carcinoma can actually occur at any age. NUT carcinoma is genetically defined by chromosomal rearrangements involving the NUT gene on chromosome 15q14 fused to the BET gene BRD4 on chromosome 19p13.1, or less commonly to other genes, including BRD3 and NSD3BRD (25, 26). The translocation creates an in-frame BRD4–NUT oncogene driven by the BRD4 promoter that is considered a major pathogenetic driver of cellular transformation (26). Silencing of the BRD4–NUT fusion gene results in differentiation and growth arrest of NUT carcinoma cells (26). More importantly, the displacement of the BRD4 oncoprotein from chromatin using the BET inhibitor JQ1 has antiproliferative activity with squamous differentiation in BRD4-dependent cell lines and patient-derived xenograft models (27).

Exposure of tumor cells to BET inhibitors results in a genome-wide reduction of the levels of BRD4 at enhancers and at promoters with important changes at the gene expression level, largely represented by a downregulation of transcripts (refs. 1, 4, 17, 27–30; Fig. 1). Downregulation of key oncogenes that are associated with superenhancers, such as MYC, represents a possible mechanism of the antitumor activity of BET inhibitors (1). Biomarkers of exposure to BET inhibitors have been proposed, such as transcriptional modulation of MYC and HEXIM1 (31, 32) and CCR1 and IL1RN (33).

Signatures of exposure to BET inhibitors are relatively similar among different tumor types (31, 34, 35), but specific changes predominate in different cellular contexts in which BET inhibitors directly affect key tissue- or cancer-specific genes, mainly transcription factors (1, 4, 8, 28, 29, 31, 36–42). Examples of key genes are MYC (4, 20, 22, 24, 35–38, 43–47), AR and TMPRSS2ETS fusion genes (48, 49), FOSL1 (50, 51), E2F2 (38), ITK (31), IL7R (35, 38, 44, 50), TERT (24, 46), BCL2 (22, 24, 43, 46), CDK6 (17, 22, 38, 43), IRF4 (24, 37, 45), and IKZF1 (24, 45). There are data indicating that the reduction in BRD4 binding is more marked at superenhancers and that their corresponding genes undergo stronger and faster downregulation than genes regulated by standard enhancers (1, 17, 37, 52).

BET Inhibitors

After the initial description done by Yoshitomi Pharmaceuticals (then Mitsubishi Tanabe Pharma) of thienotriazolodiazepines with antitumor activity and the ability to inhibit the binding between acetylated histone and bromodomain-containing proteins (patent PCT/JP2008/073864), in 2010 there were two seminal papers demonstrating that BET inhibitors can induce terminal differentiation and apoptosis in preclinical NUT carcinoma models (27) and can have important anti-inflammatory activity (53). Many other publications followed describing novel BET inhibitors and, more importantly, demonstrating that pharmacologic BET inhibition has clear preclinical antitumor activity in a variety of solid tumors and hematologic cancers (54, 55). BRD2 and BRD4 have 80% homology, and their different specificities are believed to mainly depend on their ET and CTD domains (5). Because all of the currently available BET inhibitors target the bromodomains, they are to be considered pan-BET inhibitors, although they can differ in their capacity to bind both BD1 and BD2 or preferentially one of the two bromodomains (56–58).

Several compounds have entered clinical development in phase I or II studies for patients with solid tumors and hematologic malignancies (Table 1). Although results of most of these studies are only preliminary, some first signals of clinical activity have emerged and together with the adverse events observed may direct their further clinical development (Fig. 2). In the following sections, we have summarized information on the preliminary clinical activity of BET inhibitors and their toxicity profiles.

Table 1.

BET inhibitors in clinical trials, from ClinicalTrials.gov (https://clinicaltrials.gov/), last accessed on September 10, 2017

Drug (administration)ClinicalTrials.gov # (start date)PhaseTumor typeStatusResults
GSK525762 (oral) NCT01587703 (March 2012) Solid tumors, NUT carcinoma Ongoing/recruiting patients Abstract (67) 
 NCT01943851 (May 2014) Hematologic malignancies Ongoing/recruiting patients Not reported 
 NCT02706535 (May 2016) Advanced malignancies, effects of itraconazole and rifampicin on the pharmacokinetics of GSK525762 Completed Not reported 
 NCT03150056 (May 2017) Prostate cancer, in combination with androgen deprivation Ongoing/recruiting patients Not reported 
 NCT02964507 (February 2017) II Estrogen receptor–positive breast cancer, in combination with fulvestrant Ongoing/recruiting patients Not reported 
OTX015/MK-8628 (oral) NCT01713582 (December 2012) Hematologic malignancies Completed Reported (60, 61) 
 NCT02259114 (October 2014) Solid tumors Completed Abstract (65) 
 NCT02296476 (October 2014) Glioblastoma Terminated Abstract (69) 
 NCT02698176 (May 2016) Solid tumors Terminated Not reported 
CPI-0610 (oral) NCT01949883 (September 2013) Non-Hodgkin lymphomas Ongoing/recruiting patients Abstract (62) 
 NCT02157636 (July 2014) Multiple myeloma Ongoing/recruiting patients Not reported 
 NCT02158858 (June 2014) Myelodysplastic syndrome, leukemia Ongoing/recruiting patients Not reported 
RO6870810/TEN-010 (subcutaneous injection) NCT01987362 (October 2013) Solid tumors Ongoing/not recruiting patients Abstract (66) 
 NCT02308761 (November 2014) Myelodysplastic syndrome, leukemia Ongoing/recruiting patients Not reported 
 NCT03068351 (June 2017) Multiple myeloma, single agent and in combination with daratumumab Ongoing/recruiting patients Not reported 
 NCT03255096 (August 2017) Lymphomas, in combination with venetoclax and rituximab Ongoing/recruiting patients Not reported 
BAY1238097 (oral) NCT02369029 (February 2015) Solid tumors, non-Hodgkin lymphomas Terminated Abstract (70) 
GS-5829 (oral) NCT02392611 (March 2015) Solid tumors and lymphomas; expansion cohort in breast cancer in combination with exemestane or fulvestrant Ongoing/not recruiting patients Not reported 
 NCT02607228 (December 2015) I/II Prostate cancer, single agent and in combination with enzalutamide Ongoing/recruiting patients Not reported 
 NCT02983604(January 2017) I/II Breast cancer, in combination with exemestane or fulvestarant Ongoing/recruiting patients Not reported 
ABBV-075 (oral) NCT02391480 (April 2015) Solid tumors and hematologic malignancies; expansion cohorts in combination with venetoclax Ongoing/recruiting patients Not reported 
FT-1101 (oral) NCT02543879 (September 2015) Hematologic malignancies Ongoing/recruiting patients Not reported 
INCB054329 (oral) NCT02431260 (May 2015) I/II Solid tumors, non-Hodgkin lymphomas, acute leukemia, chronic myeloid leukemia, multiple myeloma Ongoing/not recruiting patients Not reported 
BMS-986158 (oral) NCT02419417 (June 2015) I/II Solid tumors Ongoing/recruiting patients Not reported 
BI 894999 (oral) NCT02516553 (July 2015) Solid tumors, non-Hodgkin lymphomas Ongoing/not recruiting patients Not reported 
N-methyl pyrrolidone (oral) NCT02468687 (August 2015) Multiple myeloma Ongoing/recruiting patients Not reported 
PLX51107 (oral) NCT02683395 (March 2016) Solid tumors, hematologic malignancies Ongoing/recruiting patients Not reported 
GSK2820151 (oral) NCT02630251 (April 2016) Solid tumors Ongoing/recruiting patients Not reported 
ZEN-3694 (oral) NCT02705469 (April 2016) Prostate cancer Ongoing/recruiting patients Not reported 
 NCT02711956 (December 2016) Prostate cancer, in combination with enzalutamide Ongoing/not recruiting patients Not reported 
INCB057643 (oral) CT02711137 (May 2016) I/II Solid tumors, hematologic malignancies Ongoing/recruiting patients Not reported 
ODM-207 (oral) NCT03035591 (December 2016) I/II Solid tumors Ongoing/recruiting patients Not reported 
Drug (administration)ClinicalTrials.gov # (start date)PhaseTumor typeStatusResults
GSK525762 (oral) NCT01587703 (March 2012) Solid tumors, NUT carcinoma Ongoing/recruiting patients Abstract (67) 
 NCT01943851 (May 2014) Hematologic malignancies Ongoing/recruiting patients Not reported 
 NCT02706535 (May 2016) Advanced malignancies, effects of itraconazole and rifampicin on the pharmacokinetics of GSK525762 Completed Not reported 
 NCT03150056 (May 2017) Prostate cancer, in combination with androgen deprivation Ongoing/recruiting patients Not reported 
 NCT02964507 (February 2017) II Estrogen receptor–positive breast cancer, in combination with fulvestrant Ongoing/recruiting patients Not reported 
OTX015/MK-8628 (oral) NCT01713582 (December 2012) Hematologic malignancies Completed Reported (60, 61) 
 NCT02259114 (October 2014) Solid tumors Completed Abstract (65) 
 NCT02296476 (October 2014) Glioblastoma Terminated Abstract (69) 
 NCT02698176 (May 2016) Solid tumors Terminated Not reported 
CPI-0610 (oral) NCT01949883 (September 2013) Non-Hodgkin lymphomas Ongoing/recruiting patients Abstract (62) 
 NCT02157636 (July 2014) Multiple myeloma Ongoing/recruiting patients Not reported 
 NCT02158858 (June 2014) Myelodysplastic syndrome, leukemia Ongoing/recruiting patients Not reported 
RO6870810/TEN-010 (subcutaneous injection) NCT01987362 (October 2013) Solid tumors Ongoing/not recruiting patients Abstract (66) 
 NCT02308761 (November 2014) Myelodysplastic syndrome, leukemia Ongoing/recruiting patients Not reported 
 NCT03068351 (June 2017) Multiple myeloma, single agent and in combination with daratumumab Ongoing/recruiting patients Not reported 
 NCT03255096 (August 2017) Lymphomas, in combination with venetoclax and rituximab Ongoing/recruiting patients Not reported 
BAY1238097 (oral) NCT02369029 (February 2015) Solid tumors, non-Hodgkin lymphomas Terminated Abstract (70) 
GS-5829 (oral) NCT02392611 (March 2015) Solid tumors and lymphomas; expansion cohort in breast cancer in combination with exemestane or fulvestrant Ongoing/not recruiting patients Not reported 
 NCT02607228 (December 2015) I/II Prostate cancer, single agent and in combination with enzalutamide Ongoing/recruiting patients Not reported 
 NCT02983604(January 2017) I/II Breast cancer, in combination with exemestane or fulvestarant Ongoing/recruiting patients Not reported 
ABBV-075 (oral) NCT02391480 (April 2015) Solid tumors and hematologic malignancies; expansion cohorts in combination with venetoclax Ongoing/recruiting patients Not reported 
FT-1101 (oral) NCT02543879 (September 2015) Hematologic malignancies Ongoing/recruiting patients Not reported 
INCB054329 (oral) NCT02431260 (May 2015) I/II Solid tumors, non-Hodgkin lymphomas, acute leukemia, chronic myeloid leukemia, multiple myeloma Ongoing/not recruiting patients Not reported 
BMS-986158 (oral) NCT02419417 (June 2015) I/II Solid tumors Ongoing/recruiting patients Not reported 
BI 894999 (oral) NCT02516553 (July 2015) Solid tumors, non-Hodgkin lymphomas Ongoing/not recruiting patients Not reported 
N-methyl pyrrolidone (oral) NCT02468687 (August 2015) Multiple myeloma Ongoing/recruiting patients Not reported 
PLX51107 (oral) NCT02683395 (March 2016) Solid tumors, hematologic malignancies Ongoing/recruiting patients Not reported 
GSK2820151 (oral) NCT02630251 (April 2016) Solid tumors Ongoing/recruiting patients Not reported 
ZEN-3694 (oral) NCT02705469 (April 2016) Prostate cancer Ongoing/recruiting patients Not reported 
 NCT02711956 (December 2016) Prostate cancer, in combination with enzalutamide Ongoing/not recruiting patients Not reported 
INCB057643 (oral) CT02711137 (May 2016) I/II Solid tumors, hematologic malignancies Ongoing/recruiting patients Not reported 
ODM-207 (oral) NCT03035591 (December 2016) I/II Solid tumors Ongoing/recruiting patients Not reported 
Figure 2.

Schematic representation of available clinical data including preliminary antitumor activity and main toxicities observed with BET inhibitors. AL, acute leukemia; NHL, non-Hodgkin lymphoma; MM, multiple myeloma; NUT ca, NUT carcinoma; GLB, glioblastoma; GI, gastrointestinal; n, number of patients included; CR, complete remission; PR, partial remission; NR, no response. *This study included 70 patients at the time of reporting, and preliminary activity was reported in 10 patients with NUT carcinoma (67). Cited references: Berthon, et al. (60); Amorim, et al. (61); Stathis, et al. (64); Massard, et al. (65); Hottinger, et al. (69); Abramson, et al. (62); O'Dwyer, et al. (67); Shapiro, et al. (66); Postel-Vinay, et al. (70).

Figure 2.

Schematic representation of available clinical data including preliminary antitumor activity and main toxicities observed with BET inhibitors. AL, acute leukemia; NHL, non-Hodgkin lymphoma; MM, multiple myeloma; NUT ca, NUT carcinoma; GLB, glioblastoma; GI, gastrointestinal; n, number of patients included; CR, complete remission; PR, partial remission; NR, no response. *This study included 70 patients at the time of reporting, and preliminary activity was reported in 10 patients with NUT carcinoma (67). Cited references: Berthon, et al. (60); Amorim, et al. (61); Stathis, et al. (64); Massard, et al. (65); Hottinger, et al. (69); Abramson, et al. (62); O'Dwyer, et al. (67); Shapiro, et al. (66); Postel-Vinay, et al. (70).

Close modal

Leukemia and Lymphoma

The first published clinical results of a BET inhibitor derive from the thienotriazolodiazepine OTX015 (MK-8682). In preclinical experiments, OTX015 resulted in cell growth inhibition, cell-cycle arrest, and apoptosis in acute leukemia cell lines in which it also decreased the expression of BRD2, BRD4, and MYC and increased the expression of the MYC-negative regulator HEXIM1 (59).

OTX015 was also evaluated in a large panel of cell lines derived from mature B-cell lymphoid malignancies, showing antiproliferative activity among all the different histologic subtypes. The activity of OTX015 in lymphoma cell lines is mainly cytostatic, with the exception of a subgroup of cell lines derived from non–germinal center B cell–like diffuse large B-cell lymphoma (DLBCL) bearing wild-type TP53 and mutations in MYD88, CD79B, or CARD11 in which it induces apoptosis (36). Both in vitro and in vivo, OTX015 inhibits NFkB/TLR/JAK/STAT signaling pathways and MYC- and E2F1-regulated genes (36).

Based on its preclinical activity, OTX015 was investigated in a phase I dose-finding study in two parallel cohorts of patients with advanced hematologic malignancies: one cohort of patients with acute leukemia and one cohort of patients with nonleukemic hematologic malignancies (including lymphoma and multiple myeloma). Doses from 10 to 160 mg daily in either continuous or different intermittent schedules were evaluated, establishing the recommended phase II dose at 80 mg once daily for 14 days every 21 days in both cohorts.

In the acute leukemia cohort, among 41 patients (36 with AML, 3 with acute lymphoblastic leukemia, 1 with acute undifferentiated leukemia, and 1 with refractory anemia with excess of blasts) previously failing a median of 2 systemic treatments, preliminary activity was observed in 5 patients: 2 (1 with acute leukemia treated at 40 mg once a day and 1 with refractory anemia with excess of blasts treated at 160 mg once a day) achieved complete remission lasting 5 and 3 months, respectively, and 1 with acute leukemia treated at 80 mg achieved a complete remission with incomplete recovery of platelets. In addition, 2 patients (1 with AML secondary to polycythemia vera treated at 10 mg and 1 with myelodysplastic syndrome treated at 80 mg) had partial blast clearance. No correlation was found between somatic mutations in 42 genes (including NPM1, IDH2, FLT3, EV11, and MLL) and response to OTX015 comparing 5 responders versus 28 nonresponders (60).

In the nonleukemic cohort, among 45 patients (33 with lymphoma and 12 with multiple myeloma) previously treated with a median of three systemic lines, 2 patients with DLBCL, both initially treated at 120 mg once a day, achieved complete remissions lasting 4.5 and 13.7 months, respectively, and another patient with DLBCL treated at 80 mg once a day achieved a partial remission lasting 6 months. Additionally, 6 patients (2 with DLBCL, 2 with follicular lymphoma, 1 with extranodal marginal zone lymphoma, and 1 with lymphoplasmacytic lymphoma) had tumor reductions not meeting the criteria for objective response. In a retrospective subgroup analysis, out of the 10 patients with non–germinal center B cell–like DLBCL, 4 (40%) had evidence of clinical activity versus 2 of 12 (17%) patients with germinal center B cell–like DLBCL. With regard to MYC protein expression, among 5 patients with MYC-positive DLBCL, only 1 responded to treatment. No activity was detected in any of the 12 patients with multiple myeloma (61).

Antitumor activity has also been reported with CPI-0610, a benzoisoxazoloazepine compound currently in evaluation in a phase I study in patients with relapsed or refractory lymphomas. Preliminary results in 44 patients who received CPI-0610 orally once daily on days 1 to 14 in 21-day cycles at doses ranging from 6 to 230 mg have been reported (62). Two patients with DLBCL achieved a complete response (1 maintained after 7 cycles and 1 followed by allogeneic transplantation after 6 cycles), and 1 patient with follicular lymphoma achieved a partial response (lasting at least 6 cycles). Five patients experienced smaller decreases in tumor volume not meeting the criteria for objective response. Expression of the BET target gene CCR1 was suppressed at the 170-mg once-daily and 230-mg dose levels.

The above-reported clinical results, albeit coming from a small number of patients, provide evidence that BET inhibitors used in tolerable doses can result in clinical activity in patients with acute leukemia and lymphoma, in particular in DLBCL. Results from the OTX015 phase I trial might suggest an association between the non–germinal center B cell–like phenotype and treatment response, in line with preclinical data reporting strong capacity of BET inhibitors to downregulate the NF-kB, TLR, and JAK/STAT3 signaling pathways (28, 36), crucial in this subtype of DLBCL (63). However, this finding will need confirmation in additional studies. On the other hand, no association was found between MYC expression and sensitivity to OTX015. Future studies should aim to identify molecularly defined subsets of patients with DLBCL that are most likely to benefit from BET inhibitors.

NUT Carcinoma

NUT carcinoma represents a disease prototype for the clinical testing of BET inhibitors due to the strong preclinical rationale, as reported above, and to the need for novel therapeutic approaches. Indeed, NUT carcinoma is one of the most lethal solid tumors, characterized by a very aggressive course, lack of benefit from chemotherapy or radiotherapy, and an overall survival of 6 to 9 months (26).

Our institution was one of the centers participating in the dose-escalation phase I study of OTX015 in patients with hematologic malignancies. Four patients were referred to our center with advanced, previously treated NUT carcinoma with confirmed BRD4–NUT fusions and received OTX015 on a compassionate basis (64). The schedule consisted of 80 mg once daily, administered orally in 3-week cycles, based on the dose that was already tested and declared safe in the phase I hematologic study (61). Among the 4 treated patients, 2 responded and 1 had a meaningful disease stabilization with a minor metabolic response (64). Responses were rapid with symptomatic relief (including a clinical response after 1 week of treatment in 1 of the patients presenting at treatment start with a bulky tumor involving the left maxillary and mandibular region) and were confirmed by PET-CT after two cycles of treatment. The duration of response was 13 cycles in 1 patient and 3 cycles in the second patient (64).

Following the phase I study in hematologic malignancies, OTX015 was subsequently evaluated in a phase I study in patients with selected solid tumors, including patients with NUT carcinoma. In a preliminary report of this study, among 10 patients with NUT carcinoma, 3 patients achieved a partial response (65).

At least two other BET inhibitors that are currently in clinical development have been evaluated in small numbers of patients with NUT carcinoma. Similar to OTX015, evidence of clinical activity was observed. TEN-010, a BET inhibitor structurally related to JQ1, is under clinical evaluation in solid tumors, including NUT carcinoma, and hematologic malignancies. Preliminary data were reported for the 3 patients with NUT carcinoma treated with subcutaneous daily dosing of TEN-010 for 3 weeks in 4-week cycles. One patient received TEN-010 at 0.1 mg/kg and 2 received 0.45 mg/kg. Although the patient treated at the low dose had a rapid tumor progression, both patients treated at the higher dose had clinical responses, 1 with a 30% and 1 with a 50% tumor regression after 1 and 2 cycles of treatment, respectively. A symptomatic improvement was rapidly obtained in both patients. At the time of reporting of these results, 1 of the responding patients had experienced disease progression after 2 cycles of treatment and treatment of the second patient was ongoing in cycle 3 (66).

GSK525762 is another pan-BET inhibitor that is being evaluated in patients with hematologic malignancies and solid tumors (67). Preliminary results after the inclusion of 70 patients (including 17 patients with NUT carcinoma) showed good tolerability at the dose of 80 mg once daily, which was the dose selected for expansion cohorts. The most common adverse events of any grade included thrombocytopenia, gastrointestinal adverse events (nausea, vomiting, decreased appetite, diarrhea, and dysgeusia), anemia, and fatigue. At the time of reporting of these results, among 10 response-evaluable patients with NUT carcinoma, 2 patients achieved partial response and 4 had stable disease.

Results from this small number of patients provide clinical evidence of the activity of BET inhibition in NUT carcinoma and represent an important example of therapy with small molecules resulting in antitumor activity by targeting the causative oncoprotein.

Based on the available data, it is not possible to estimate what the clinical impact of BET inhibitors used as monotherapy could be in NUT carcinoma. Not all patients responded; in fact, only 30% of patients with NUT carcinoma responded in the phase I trial of OTX015 in solid tumors, and 20% in the phase I trial of GSK525762. On the other hand, among those patients who achieved a response and for which follow-up data were available, all patients relapsed during treatment. Genetic data from tumor biopsies before treatment start and at time of disease progression are available from only one of the first 4 patients treated with OTX015, but they did not detect any genetic alterations at the time of progression that could possibly explain the development of resistance to treatment (64). Future studies should aim to molecularly characterize patients with NUT carcinoma who are treated with BET inhibitors. In addition, although FISH confirmation of the chromosomal translocation is not necessary for diagnostic purposes (68), it should be implemented in clinical trials to assess whether the partner gene (BRD4 vs. other genes) fused to NUT may predict response to BET inhibition. Finally, combination therapies may be necessary to overcome the resistance that can develop to BET inhibition.

Other Solid Tumors

The currently available clinical data in other solid tumors are very scant. Preliminary results of the phase I study of OTX015 in patients with NUT carcinoma, castration-resistant prostate cancer (CRPC), or KRAS-mutated or ALK-positive non–small cell lung cancer (NSCLC) showed evidence of clinical activity in NUT carcinoma as reported above and in CRPC. Among 46 patients treated (26 CRPC, 10 NUT carcinoma, 9 KRAS-mutated, and 1 ALK-positive NSCLC), 4 patients had a partial response (including the 3 patients with NUT carcinoma and 1 patient with CRPC). In addition, prolonged stable disease was observed in 5 patients with CRPC (4–8 months) and 2 patients with KRAS-mutated NSCLC (65).

On the other hand, no clinical activity was seen in a small phase IIa study in 12 patients with glioblastoma, and the trial was closed due to lack of clinical activity (69).

Adverse Events

Similar to the data reporting on their clinical activity, the adverse events profile of BET inhibitors must be better defined, because the results of most of the compounds currently in phase I trials are preliminary. The main toxicities seen with OTX015 in the phase I study in patients with hematologic malignancies were represented by reversible thrombocytopenia (which indeed required a schedule of administration of 2 weeks on/1 week off to permit platelet recovery), anemia, neutropenia, gastrointestinal symptoms (including nausea, diarrhea, and dysgeusia), fatigue, and bilirubin elevation. Overall, OTX015 was well tolerated, and the toxicities were reversible with treatment interruptions. The favorable safety profile of OTX015 was confirmed in the phase I trial in patients with solid tumors (65). Thrombocytopenia, fatigue, gastrointestinal symptoms, and hyperbilirubinemia are among side effects also reported in patients treated with other BET inhibitors (62, 66, 67), although the results of these trials are currently preliminary and reported only in abstract form. Additional results from the ongoing studies will add information on the safety of these compounds, and the impact that these adverse events could have in treatment compliance. On the other hand, the development of another BET inhibitor, BAY 1238097, was prematurely interrupted due to severe adverse events (mainly headaches) that occurred at doses below the predicted therapeutic dose (70).

Further side effects might be predicted based on preclinical data. Human BET proteins, mainly BRD4, interact with viral proteins and are involved in viral life cycles (18, 71–81). There is preclinical evidence that BET inhibitors activate DNA replication of human immunodeficiency virus (HIV; refs. 76, 77, 79), human papillomavirus (HPV) 16 (80), human herpes simplex virus 1 and 2 (HSV-1 and HSV-2; ref. 78), and hepatitis B virus (HBV; ref. 73). Although this can be therapeutically exploited to eliminate latent viral infections (76, 77, 82), it advises for a close monitoring of patients with cancer receiving BET inhibitors for reactivation of viral infections.

The complete lack of either BRD2 or BRD4 is lethal (12–15) and, in general, low BET protein level is associated with reduced cell growth (12, 13). Mice with reduced BRD4 levels present different reversible phenotypes with a decrease in the number of hematopoietic cells, skin hyperplasia with abnormal hair follicles, and disruption of the intestinal crypts with loss of the secretory cells and increased intestinal toxicity after exposure to radiation or doxorubicin (83). Mice with reduced levels of BRD2 show important neuronal defects and obesity with hyperinsulinemia in the presence of a lowered blood glucose (13–15). Impaired long-term memory (84), reduced explorative motor activity, and heightened anxiety-like behavior in the open field (85) have been observed in mice exposed to the BET inhibitors JQ1 (84) and I-BET858 (85), further suggesting that neurologic symptoms might be expected. Finally, a tumor-suppressor role for BET proteins has also been reported in which their inhibition would result in a reduced immune surveillance (86) and reduced capacity of healthy cells to counteract the neoplastic transformation process (87). These observations suggest monitoring for second tumors in patients exposed to BET inhibitors.

Resistance

Mechanisms of resistance to BET inhibitors so far derive from preclinical models. Resistance does not appear to derive from somatic mutations or copy-number changes affecting BET bromodomain genes (42, 88). Increased WNT signaling with β-catenin–mediated MYC expression in AML (88, 89), activation of the Hedgehog pathway with GLI2-mediated MYC expression in pancreatic cancer (90), hyperphosphorylation of BRD4 that leads to a bromodomain-independent binding to MED1 in triple-negative breast cancer (42), kinome reprogramming in ovarian cancer (91), activation of the MAPK pathway in colorectal cancer (45), AMPK–ULK1-mediated autophagy (92) or MCL1 upregulation in AML (29), and RAS pathway activation with BCL2 upregulation in lymphoma (93) all represent possible mechanisms of resistance to BET inhibitors. The presence of concomitant KRAS and LKB1 mutations in NSCLC (38, 46) and PIK3CA mutations in breast cancer (23) has been also associated with resistance to BET inhibitors. Mutations of the SPOP gene, coding for a ubiquitin ligase adaptor protein, are associated with high sensitivity to BET inhibition in endometrial cancer cell lines but to resistance in prostate cancer cell lines due to an increased or decreased, respectively, degradation of the BET proteins in cases bearing SPOP mutations (94).

Gene expression signatures associated with sensitivity to BET inhibitors have been identified in different tumor models, but they still need to be further validated in the clinical context (36, 39, 41).

Future Directions

The above-reported preliminary information on BET inhibitors in clinical trials provides evidence of their antitumor potential in a subset of patients with hematologic malignancies and in NUT carcinoma. Although results of ongoing trials are awaited and may add information on activity in other solid tumors, two areas of preclinical research are being actively pursued and may guide future development of BET inhibitors. Below we report the preclinical rationale for combination treatments and outline the current research aiming at developing a new generation of BET inhibitors.

Combinations

BET inhibitors have shown preclinical synergism with different classes of compounds and in almost all the different tumor types that have been investigated (Table 2). Synergism largely appears to be due to the ability of BET inhibitors to block protective feedback mechanisms that would lead to the upregulation of additional kinases by a BRD-mediated mechanism (35, 95).

Table 2.

Combination partners that have shown synergism with BET inhibitors in preclinical tumor models

ClassesSecond compoundExperimental disease model
Small molecules   
 ALK inhibitors Lymphoma (31) 
 BTK inhibitors Lymphoma (28, 31, 35, 36, 100, 101, 114) 
 CDK inhibitors Lymphoma (100, 101), osteosarcoma (51) 
 BCL2/MCL1 inhibitors ALL (115), AML (29, 116), LC (117), lymphoma (99–101) 
 EGFR/ERBB2 inhibitors BC (97) 
 FLT3 inhibitors AML (118) 
 Hedgehog inhibitors Lymphoma (31) 
 JAK inhibitors AML (119) 
 MEK/ERK inhibitors Lymphoma (120), OC (91) 
 mTOR inhibitors BC (40), glioblastoma (121), lymphoma (28, 35, 36, 114), osteosarcoma (122) 
 PARP inhibitors BC (96), OC (96) 
 PI3K inhibitors, pan or selective BC (95), CC (95), lymphoma (28, 36, 102), glioblastoma (95), OC (90, 95) 
 Proteasome inhibitors MM (116, 123) 
Antibodies   
 Anti-CD20 monoclonal antibodies Lymphoma (36, 114, 124) 
Immune modulators   
 Immunomodulatory drugs (IMiD) Lymphoma (36, 125, 126), MM (45) 
 Anti–PD-1 monoclonal antibodies Lymphoma (30) 
 Anti–4-1BB monoclonal antibodies Lymphoma (30) 
 Chimeric antigen receptor (CAR) T cells ALL (105) 
Epigenetic drugs   
 EZH2 inhibitors Lymphoma (35, 127) 
 HDAC inhibitors AML (128), BC (129), LC (130), lymphoma (31, 36, 101, 114, 131), melanoma (132), neuroblastoma (133), PC (134) 
 Azacytidine AML (116) 
 Decitabine Lymphoma (36) 
Chemotherapy   
 Temozolomide Glioblastoma (121) 
Hormone therapy   
 Antiandrogen PrC (49) 
 Estrogen receptor degrader BC (135) 
ClassesSecond compoundExperimental disease model
Small molecules   
 ALK inhibitors Lymphoma (31) 
 BTK inhibitors Lymphoma (28, 31, 35, 36, 100, 101, 114) 
 CDK inhibitors Lymphoma (100, 101), osteosarcoma (51) 
 BCL2/MCL1 inhibitors ALL (115), AML (29, 116), LC (117), lymphoma (99–101) 
 EGFR/ERBB2 inhibitors BC (97) 
 FLT3 inhibitors AML (118) 
 Hedgehog inhibitors Lymphoma (31) 
 JAK inhibitors AML (119) 
 MEK/ERK inhibitors Lymphoma (120), OC (91) 
 mTOR inhibitors BC (40), glioblastoma (121), lymphoma (28, 35, 36, 114), osteosarcoma (122) 
 PARP inhibitors BC (96), OC (96) 
 PI3K inhibitors, pan or selective BC (95), CC (95), lymphoma (28, 36, 102), glioblastoma (95), OC (90, 95) 
 Proteasome inhibitors MM (116, 123) 
Antibodies   
 Anti-CD20 monoclonal antibodies Lymphoma (36, 114, 124) 
Immune modulators   
 Immunomodulatory drugs (IMiD) Lymphoma (36, 125, 126), MM (45) 
 Anti–PD-1 monoclonal antibodies Lymphoma (30) 
 Anti–4-1BB monoclonal antibodies Lymphoma (30) 
 Chimeric antigen receptor (CAR) T cells ALL (105) 
Epigenetic drugs   
 EZH2 inhibitors Lymphoma (35, 127) 
 HDAC inhibitors AML (128), BC (129), LC (130), lymphoma (31, 36, 101, 114, 131), melanoma (132), neuroblastoma (133), PC (134) 
 Azacytidine AML (116) 
 Decitabine Lymphoma (36) 
Chemotherapy   
 Temozolomide Glioblastoma (121) 
Hormone therapy   
 Antiandrogen PrC (49) 
 Estrogen receptor degrader BC (135) 

Abbreviations: ALL, acute lymphoblastic leukemia; AML, acute myeloid leukemia; BC, breast cancer; CC, colorectal cancer; LC, lung cancer; MM, multiple myeloma; OC, ovarian cancer; PC, pancreatic cancer, PrC; prostate cancer.

Several preclinical studies have explored BET inhibitors in combination with molecularly targeted agents in solid tumors and hematologic malignancies (Table 2). A benefit has been observed when combining BET inhibitors with PI3K inhibitors in breast, ovarian, and colorectal cancers (95), ERK inhibitors in ovarian cancer (91), PARP inhibitors in ovarian and breast cancers (96), and the ERBB2 inhibitor lapatinib in breast cancer (97). In lymphomas, synergistic activity has been observed in combinations with small molecules that have established single-agent clinical activity such as BTK, PI3K, or BCL2 inhibitors (28, 35, 36, 98–102). At least two clinical trials are currently testing BET inhibitors in combination with BCL2 inhibitors in patients with relapsed lymphoma (NCT02391480 and NCT03255096).

Interestingly, some kinase inhibitors, such as the PLK1 inhibitor BI-2536 or the JAK2/FLT3 inhibitor TG-101348, also inhibit BET proteins (103, 104). Considering the observed synergisms of combinations containing BET inhibitors and kinase inhibitors, the possibility of targeting both classes of proteins is under active investigation (103).

Due to the diffuse use of immune checkpoint modulators in solid tumors and in some hematologic cancers, it is important to highlight that BET inhibitors have shown synergism with this class of compounds in preclinical models (30, 105). BET inhibitors decrease BRD4 binding to the CD274 locus with downregulation of PD-L1 expression on tumor cells and improvements of the response to anti–PD-1 or anti–4-1BB (30). BET inhibitors also increase the response to chimeric antigen receptor–transduced T cells via supporting the maintenance of CD8+ T cells with a phenotype of central memory T cells or stem cell–like memory T cells (105). Finally, BET inhibitors downregulate ITK expression in T-cell lymphomas (31), and this, if confirmed in normal T cells, could favor the generation of Th1 cells, IFNγ production, and increased antitumor immunity as observed with the BTK inhibitor ibrutinib that also inhibits ITK (106).

Finally, BET inhibitors have been tested in CRPC and tamoxifen-resistant breast cancer in combination with enzalutamide and fulvestrant, respectively, showing synergistic activity. At least five clinical trials (NCT02392611, NCT02964507, NCT02607228, NCT02983604, and NCT02711956) are currently evaluating combination therapies of BET inhibitors with hormone therapy in patients with prostate or breast cancer (Table 1).

Next Generation of Compounds

Different experimental models show that tumor cells resistant to BET inhibitors are still dependent on BET proteins (42, 88) and, in general, exposure to BET inhibitors results in downregulation of BET proteins that is reversible and often followed by an upregulation of the BET proteins themselves (36, 107). Thus, there are ongoing efforts to obtain stronger and more sustained suppression of the BET protein activity, which would lead to increased antitumor activity. Compounds such as “biBET (6)” (108), MT1 (109), and AZD5153 (31), which engage both bromodomains simultaneously in a bivalent mode, have shown promising in vitro results. Another extremely active field is the creation of BET degraders, chimeric compounds that merge a BET inhibitor that allows the binding to BET proteins, linked to an additional small molecule that mediates the binding to an E3 ubiquitin ligase complex, thus degrading the BET proteins via the proteasome. These compounds are based on the idea of the proteolysis targeting chimeras (PROTAC), first described in 2001 (110). Currently available BET degraders, which are still at the preclinical level, differ both in the use of the BET inhibitor component and in the exploited E3 ligase complex (Table 3).

Table 3.

BET degraders (PROTACs) with their components and mechanism of action

CompoundBET inhibitor componentSmall-molecule componentE3 ubiquitin ligase complexReference
ARV-771 JQ1 VHL ligand Von Hippel–Lindau-containing complex (111) 
ARV-825 OTX015 Thalidomide Cereblon E3 ubiquitin ligase complex (107) 
AT1 JQ1 VHL ligand Von Hippel–Lindau-containing complex (136) 
BETd-246 RX-37 Thalidomide Cereblon E3 ubiquitin ligase complex (113) 
BETd-260/ZBC260 HJB97/Beti-211 Lenalidomide Cereblon E3 ubiquitin ligase complex (113, 137) 
dBET1 JQ1 Thalidomide Cereblon E3 ubiquitin ligase complex (138) 
dBET6 JQ1 Thalidomide Cereblon E3 ubiquitin ligase complex (4) 
MZ1 JQ1 VHL ligand Von Hippel–Lindau-containing complex (139) 
CompoundBET inhibitor componentSmall-molecule componentE3 ubiquitin ligase complexReference
ARV-771 JQ1 VHL ligand Von Hippel–Lindau-containing complex (111) 
ARV-825 OTX015 Thalidomide Cereblon E3 ubiquitin ligase complex (107) 
AT1 JQ1 VHL ligand Von Hippel–Lindau-containing complex (136) 
BETd-246 RX-37 Thalidomide Cereblon E3 ubiquitin ligase complex (113) 
BETd-260/ZBC260 HJB97/Beti-211 Lenalidomide Cereblon E3 ubiquitin ligase complex (113, 137) 
dBET1 JQ1 Thalidomide Cereblon E3 ubiquitin ligase complex (138) 
dBET6 JQ1 Thalidomide Cereblon E3 ubiquitin ligase complex (4) 
MZ1 JQ1 VHL ligand Von Hippel–Lindau-containing complex (139) 

BET degraders appear to have a different mechanism of action from BET inhibitors, due to the loss of both bromodomain-dependent and bromodomain-independent functions of the BET proteins and to a collapse of the core transcriptional machinery, with wider changes at the gene expression level (4, 100). Accordingly, degradation of BET proteins induced by BET degraders results in a higher cytotoxic effect than that achieved with BET inhibitors (4, 107, 111–113). ARV-825, a heterobifunctional PROTAC that exploits the E3 ubiquitin ligase cereblon and leads to rapid BRD4 degradation, induces increased apoptosis in Burkitt lymphoma cell lines in comparison to either JQ1 or OTX015, and a more effective suppression of MYC levels and downstream genes (107). ARV-771, a von Hippel–Landau (VHL) E3 ligase–based BET PROTAC, has superior in vitro and in vivo activity in CRPC in comparison to BET inhibitors and results in attenuation of androgen receptor transcript levels (111). BETd-246 and its further optimized analogue BETd-260, both using cereblon as E3 ubiquitin ligase, show potent in vitro and in vivo antitumor activity, superior to the parental BET inhibitor BETi-211, in triple-negative breast cancer models (113).

Seven years following the first description of the preclinical activity of JQ1 in models of NUT carcinoma, a tumor type driven by an oncogenic form of BRD4, several BET inhibitors have entered clinical evaluation, while many others are currently in preclinical development. Clinical activity has been observed in NUT carcinoma and in hematologic malignancies. Regarding their safety, with the exception of one compound whose development was stopped due to the emergence of adverse events, preliminary information from other BET inhibitors shows a favorable safety profile. Hematologic (mainly thrombocytopenia) and nonhematologic adverse events are reversible with treatment interruption but, alongside potential side effects foreseen based on experimental models, will need to be taken into consideration in the planning of future trials, especially if in combination with other agents. Additional data from ongoing clinical trials will be able to determine the impact of these adverse events in treatment compliance. Although final results of most of the ongoing studies are still awaited, current evidence supports further clinical development of BET inhibitors in hematologic malignancies and in solid tumors. Combination strategies alongside the development of new generations of compounds, not limited to targeting the bromodomains, will open new possibilities for future clinical development of BET inhibitors as anticancer agents.

F. Bertoni reports receiving commercial research grants from Oncology Therapeutic Development and Bayer. No potential conflicts of interest were disclosed by the other author.

A. Stathis served as principal investigator and received institutional funding from Oncoethix and Merck for the clinical trials with OTX015/MK-8628. This work was partially supported by the Nelia et Amadeo Barletta Foundation and the Gelu Foundation.

1.
Loven
J
,
Hoke
HA
,
Lin
CY
,
Lau
A
,
Orlando
DA
,
Vakoc
CR
, et al
Selective inhibition of tumor oncogenes by disruption of super-enhancers
.
Cell
2013
;
153
:
320
34
.
2.
Whyte
WA
,
Orlando
DA
,
Hnisz
D
,
Abraham
BJ
,
Lin
CY
,
Kagey
MH
, et al
Master transcription factors and mediator establish super-enhancers at key cell identity genes
.
Cell
2013
;
153
:
307
19
.
3.
Hnisz
D
,
Abraham
BJ
,
Lee
TI
,
Lau
A
,
Saint-Andre
V
,
Sigova
AA
, et al
Super-enhancers in the control of cell identity and disease
.
Cell
2013
;
155
:
934
47
.
4.
Winter
GE
,
Mayer
A
,
Buckley
DL
,
Erb
MA
,
Roderick
JE
,
Vittori
S
, et al
BET bromodomain proteins function as master transcription elongation factors independent of CDK9 recruitment
.
Mol Cell
2017
;
67
:
5
18.e19
.
5.
Belkina
AC
,
Denis
GV
. 
BET domain co-regulators in obesity, inflammation and cancer
.
Nat Rev Cancer
2012
;
12
:
465
77
.
6.
Dawson
MA
,
Kouzarides
T
,
Huntly
BJ
. 
Targeting epigenetic readers in cancer
.
N Engl J Med
2012
;
367
:
647
57
.
7.
Shi
J
,
Vakoc
CR
. 
The mechanisms behind the therapeutic activity of BET bromodomain inhibition
.
Mol Cell
2014
;
54
:
728
36
.
8.
Roe
JS
,
Mercan
F
,
Rivera
K
,
Pappin
DJ
,
Vakoc
CR
. 
BET bromodomain inhibition suppresses the function of hematopoietic transcription factors in acute myeloid leukemia
.
Mol Cell
2015
;
58
:
1028
39
.
9.
Shi
J
,
Wang
Y
,
Zeng
L
,
Wu
Y
,
Deng
J
,
Zhang
Q
, et al
Disrupting the interaction of BRD4 with diacetylated Twist suppresses tumorigenesis in basal-like breast cancer
.
Cancer Cell
2014
;
25
:
210
25
.
10.
Zou
Z
,
Huang
B
,
Wu
X
,
Zhang
H
,
Qi
J
,
Bradner
J
, et al
Brd4 maintains constitutively active NF-kappaB in cancer cells by binding to acetylated RelA
.
Oncogene
2014
;
33
:
2395
404
.
11.
Wu
S-Y
,
Lee
AY
,
Lai
H-T
,
Zhang
H
,
Chiang
C-M
. 
Phospho switch triggers Brd4 chromatin binding and activator recruitment for gene-specific targeting
.
Molecular cell
2013
;
49
:
843
57
.
12.
Houzelstein
D
,
Bullock
SL
,
Lynch
DE
,
Grigorieva
EF
,
Wilson
VA
,
Beddington
RS
. 
Growth and early postimplantation defects in mice deficient for the bromodomain-containing protein Brd4
.
Mol Cell Biol
2002
;
22
:
3794
802
.
13.
Shang
E
,
Wang
X
,
Wen
D
,
Greenberg
DA
,
Wolgemuth
DJ
. 
Double bromodomain-containing gene Brd2 is essential for embryonic development in mouse
.
Dev Dyn
2009
;
238
:
908
17
.
14.
Gyuris
A
,
Donovan
DJ
,
Seymour
KA
,
Lovasco
LA
,
Smilowitz
NR
,
Halperin
AL
, et al
The chromatin-targeting protein Brd2 is required for neural tube closure and embryogenesis
.
Biochim Biophys Acta
2009
;
1789
:
413
21
.
15.
Wang
F
,
Liu
H
,
Blanton
WP
,
Belkina
A
,
Lebrasseur
NK
,
Denis
GV
. 
Brd2 disruption in mice causes severe obesity without Type 2 diabetes
.
Biochem J
2010
;
425
:
71
83
.
16.
Jang
MK
,
Mochizuki
K
,
Zhou
M
,
Jeong
HS
,
Brady
JN
,
Ozato
K
. 
The bromodomain protein Brd4 is a positive regulatory component of P-TEFb and stimulates RNA polymerase II-dependent transcription
.
Mol Cell
2005
;
19
:
523
34
.
17.
Bhagwat
AS
,
Roe
JS
,
Mok
BY
,
Hohmann
AF
,
Shi
J
,
Vakoc
CR
. 
BET Bromodomain inhibition releases the mediator complex from select cis-regulatory elements
.
Cell Rep
2016
;
15
:
519
30
.
18.
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
5
.
19.
Liu
W
,
Ma
Q
,
Wong
K
,
Li
W
,
Ohgi
K
,
Zhang
J
, et al
Brd4 and JMJD6-associated anti-pause enhancers in regulation of transcriptional pause release
.
Cell
2013
;
155
:
1581
95
.
20.
Baratta
MG
,
Schinzel
AC
,
Zwang
Y
,
Bandopadhayay
P
,
Bowman-Colin
C
,
Kutt
J
, et al
An in-tumor genetic screen reveals that the BET bromodomain protein, BRD4, is a potential therapeutic target in ovarian carcinoma
.
Proc Natl Acad Sci U S A
2015
;
112
:
232
7
.
21.
Toyoshima
M
,
Howie
HL
,
Imakura
M
,
Walsh
RM
,
Annis
JE
,
Chang
AN
, et al
Functional genomics identifies therapeutic targets for MYC-driven cancer
.
Proc Natl Acad Sci U S A
2012
;
109
:
9545
50
.
22.
Zuber
J
,
Shi
J
,
Wang
E
,
Rappaport
AR
,
Herrmann
H
,
Sison
EA
, et al
RNAi screen identifies Brd4 as a therapeutic target in acute myeloid leukaemia
.
Nature
2011
;
478
:
524
8
.
23.
Marcotte
R
,
Sayad
A
,
Brown
KR
,
Sanchez-Garcia
F
,
Reimand
J
,
Haider
M
, et al
Functional genomic landscape of human breast cancer drivers, vulnerabilities, and resistance
.
Cell
2016
;
164
:
293
309
.
24.
Delmore
JE
,
Issa
GC
,
Lemieux
ME
,
Rahl
PB
,
Shi
J
,
Jacobs
HM
, et al
BET bromodomain inhibition as a therapeutic strategy to target c-Myc
.
Cell
2011
;
146
:
904
17
.
25.
French
CA
,
Rahman
S
,
Walsh
EM
,
Kuhnle
S
,
Grayson
AR
,
Lemieux
ME
, et al
NSD3-NUT fusion oncoprotein in NUT midline carcinoma: implications for a novel oncogenic mechanism
.
Cancer Discov
2014
;
4
:
928
41
.
26.
French
CA
,
Ramirez
CL
,
Kolmakova
J
,
Hickman
TT
,
Cameron
MJ
,
Thyne
ME
, et al
BRD-NUT oncoproteins: a family of closely related nuclear proteins that block epithelial differentiation and maintain the growth of carcinoma cells
.
Oncogene
2008
;
27
:
2237
42
.
27.
Filippakopoulos
P
,
Qi
J
,
Picaud
S
,
Shen
Y
,
Smith
WB
,
Fedorov
O
, et al
Selective inhibition of BET bromodomains
.
Nature
2010
;
468
:
1067
73
.
28.
Ceribelli
M
,
Kelly
PN
,
Shaffer
AL
,
Wright
GW
,
Xiao
W
,
Yang
Y
, et al
Blockade of oncogenic IkappaB kinase activity in diffuse large B-cell lymphoma by bromodomain and extraterminal domain protein inhibitors
.
Proc Natl Acad Sci U S A
2014
;
111
:
11365
70
.
29.
Zhao
Y
,
Liu
Q
,
Acharya
P
,
Stengel
KR
,
Sheng
Q
,
Zhou
X
, et al
High-resolution mapping of RNA polymerases identifies mechanisms of sensitivity and resistance to BET inhibitors in t(8;21) AML
.
Cell Rep
2016
;
16
:
2003
16
.
30.
Hogg
SJ
,
Vervoort
SJ
,
Deswal
S
,
Ott
CJ
,
Li
J
,
Cluse
LA
, et al
BET-bromodomain inhibitors engage the host immune system and regulate expression of the immune checkpoint ligand PD-L1
.
Cell Rep
2017
;
18
:
2162
74
.
31.
Rhyasen
GW
,
Hattersley
MM
,
Yao
Y
,
Dulak
A
,
Wang
W
,
Petteruti
P
, et al
AZD5153: a novel bivalent BET bromodomain inhibitor highly active against hematologic malignancies
.
Mol Cancer Ther
2016
;
15
:
2563
74
.
32.
Lin
X
,
Huang
X
,
Uziel
T
,
Hessler
P
,
Albert
DH
,
Roberts-Rapp
LA
, et al
HEXIM1 as a robust pharmacodynamic marker for monitoring target engagement of BET family bromodomain inhibitors in tumors and surrogate tissues
.
Mol Cancer Ther
2017
;
16
:
388
96
.
33.
Tsujikawa
L
,
Norek
K
,
Calosing
C
,
Attwell
S
,
Gilham
D
,
Sharma
N
, et al
LB-038/1: Preclinical development and clinical validation of a whole blood pharmacodynamic marker assay for the BET bromodomain inhibitor ZEN-3694 in metastatic castration-resistant prostate cancer (mCRPC) patients
.
Proceedings of the 107th Annual Meeting of the American Association for Cancer Research
2017
.
34.
Puissant
A
,
Frumm
SM
,
Alexe
G
,
Bassil
CF
,
Qi
J
,
Chanthery
YH
, et al
Targeting MYCN in neuroblastoma by BET bromodomain inhibition
.
Cancer Discov
2013
;
3
:
308
23
.
35.
Bernasconi
E
,
Gaudio
E
,
Lejeune
P
,
Tarantelli
C
,
Cascione
L
,
Kwee
I
, et al
Preclinical evaluation of the BET bromodomain inhibitor BAY 1238097 for the treatment of lymphoma
.
Br J Haematol
2017
;
178
:
936
48
.
36.
Boi
M
,
Gaudio
E
,
Bonetti
P
,
Kwee
I
,
Bernasconi
E
,
Tarantelli
C
, et al
The BET bromodomain inhibitor OTX015 affects pathogenetic pathways in preclinical B-cell tumor models and synergizes with targeted drugs
.
Clin Cancer Res
2015
;
21
:
1628
38
.
37.
Chapuy
B
,
McKeown
MR
,
Lin
CY
,
Monti
S
,
Roemer
MG
,
Qi
J
, et al
Discovery and characterization of super-enhancer-associated dependencies in diffuse large B cell lymphoma
.
Cancer Cell
2013
;
24
:
777
90
.
38.
Riveiro
ME
,
Astorgues-Xerri
L
,
Vazquez
R
,
Frapolli
R
,
Kwee
I
,
Rinaldi
A
, et al
OTX015 (MK-8628), a novel BET inhibitor, exhibits antitumor activity in non-small cell and small cell lung cancer models harboring different oncogenic mutations
.
Oncotarget
2016
;
7
:
84675
87
.
39.
Urbanucci
A
,
Barfeld
SJ
,
Kytola
V
,
Itkonen
HM
,
Coleman
IM
,
Vodak
D
, et al
Androgen receptor deregulation drives bromodomain-mediated chromatin alterations in prostate cancer
.
Cell Rep
2017
;
19
:
2045
59
.
40.
Vazquez
R
,
Riveiro
ME
,
Astorgues-Xerri
L
,
Odore
E
,
Rezai
K
,
Erba
E
, et al
The bromodomain inhibitor OTX015 (MK-8628) exerts anti-tumor activity in triple-negative breast cancer models as single agent and in combination with everolimus
.
Oncotarget
2017
;
8
:
7598
613
.
41.
Bian
B
,
Bigonnet
M
,
Gayet
O
,
Loncle
C
,
Maignan
A
,
Gilabert
M
, et al
Gene expression profiling of patient-derived pancreatic cancer xenografts predicts sensitivity to the BET bromodomain inhibitor JQ1: implications for individualized medicine efforts
.
EMBO Mol Med
2017
;
9
:
482
97
.
42.
Shu
S
,
Lin
CY
,
He
HH
,
Witwicki
RM
,
Tabassum
DP
,
Roberts
JM
, et al
Response and resistance to BET bromodomain inhibitors in triple-negative breast cancer
.
Nature
2016
;
529
:
413
17
.
43.
Dawson
MA
,
Prinjha
RK
,
Dittmann
A
,
Giotopoulos
G
,
Bantscheff
M
,
Chan
WI
, et al
Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemia
.
Nature
2011
;
478
:
529
33
.
44.
Ott
CJ
,
Kopp
N
,
Bird
L
,
Paranal
RM
,
Qi
J
,
Bowman
T
, et al
BET bromodomain inhibition targets both c-Myc and IL7R in high-risk acute lymphoblastic leukemia
.
Blood
2012
;
120
:
2843
52
.
45.
Siu
KT
,
Ramachandran
J
,
Yee
AJ
,
Eda
H
,
Santo
L
,
Panaroni
C
, et al
Preclinical activity of CPI-0610, a novel small-molecule bromodomain and extra-terminal protein inhibitor in the therapy of multiple myeloma
.
Leukemia
2017
;
31
:
1760
69
.
46.
Shimamura
T
,
Chen
Z
,
Soucheray
M
,
Carretero
J
,
Kikuchi
E
,
Tchaicha
JH
, et al
Efficacy of BET bromodomain inhibition in Kras-mutant non-small cell lung cancer
.
Clin Cancer Res
2013
;
19
:
6183
92
.
47.
Bandopadhayay
P
,
Bergthold
G
,
Nguyen
B
,
Schubert
S
,
Gholamin
S
,
Tang
Y
, et al
BET bromodomain inhibition of MYC-amplified medulloblastoma
.
Clin Cancer Res
2014
;
20
:
912
25
.
48.
Faivre
EJ
,
Wilcox
D
,
Lin
X
,
Hessler
P
,
Torrent
M
,
He
W
, et al
Exploitation of castration-resistant prostate cancer transcription factor dependencies by the novel BET inhibitor ABBV-075
.
Mol Cancer Res
2017
;
15
:
35
44
.
49.
Asangani
IA
,
Wilder-Romans
K
,
Dommeti
VL
,
Krishnamurthy
PM
,
Apel
IJ
,
Escara-Wilke
J
, et al
BET bromodomain inhibitors enhance efficacy and disrupt resistance to AR antagonists in the treatment of prostate cancer
.
Mol Cancer Res
2016
;
14
:
324
31
.
50.
Lockwood
WW
,
Zejnullahu
K
,
Bradner
JE
,
Varmus
H
. 
Sensitivity of human lung adenocarcinoma cell lines to targeted inhibition of BET epigenetic signaling proteins
.
Proc Natl Acad Sci U S A
2012
;
109
:
19408
13
.
51.
Baker
EK
,
Taylor
S
,
Gupte
A
,
Sharp
PP
,
Walia
M
,
Walsh
NC
, et al
BET inhibitors induce apoptosis through a MYC independent mechanism and synergise with CDK inhibitors to kill osteosarcoma cells
.
Sci Rep
2015
;
5
:
10120
.
52.
Suzuki
HI
,
Young
RA
,
Sharp
PA
. 
Super-enhancer-mediated RNA processing revealed by integrative MicroRNA network analysis
.
Cell
2017
;
168
:
1000
14.e15
.
53.
Nicodeme
E
,
Jeffrey
KL
,
Schaefer
U
,
Beinke
S
,
Dewell
S
,
Chung
CW
, et al
Suppression of inflammation by a synthetic histone mimic
.
Nature
2010
;
468
:
1119
23
.
54.
Ali
I
,
Choi
G
,
Lee
K
. 
BET inhibitors as anticancer agents: a patent review
.
Recent Pat Anticancer Drug Discov
2017
;
12
:
340
64
.
55.
Liu
Z
,
Wang
P
,
Chen
H
,
Wold
EA
,
Tian
B
,
Brasier
AR
, et al
Drug discovery targeting bromodomain-containing protein 4
.
J Med Chem
2017
;
60
:
4533
58
.
56.
Kharenko
OA
,
Gesner
EM
,
Patel
RG
,
Norek
K
,
White
A
,
Fontano
E
, et al
RVX-297- a novel BD2 selective inhibitor of BET bromodomains
.
Biochem Biophys Res Commun
2016
;
477
:
62
7
.
57.
Picaud
S
,
Wells
C
,
Felletar
I
,
Brotherton
D
,
Martin
S
,
Savitsky
P
, et al
RVX-208, an inhibitor of BET transcriptional regulators with selectivity for the second bromodomain
.
Proc Natl Acad Sci U S A
2013
;
110
:
19754
9
.
58.
Zhang
G
,
Plotnikov
AN
,
Rusinova
E
,
Shen
T
,
Morohashi
K
,
Joshua
J
, et al
Structure-guided design of potent diazobenzene inhibitors for the BET bromodomains
.
J Med Chem
2013
;
56
:
9251
64
.
59.
Coude
MM
,
Braun
T
,
Berrou
J
,
Dupont
M
,
Bertrand
S
,
Masse
A
, et al
BET inhibitor OTX015 targets BRD2 and BRD4 and decreases c-MYC in acute leukemia cells
.
Oncotarget
2015
;
6
:
17698
712
.
60.
Berthon
C
,
Raffoux
E
,
Thomas
X
,
Vey
N
,
Gomez-Roca
C
,
Yee
K
, et al
Bromodomain inhibitor OTX015 in patients with acute leukaemia: a dose-escalation, phase 1 study
.
Lancet Haematol
2016
;
3
:
e186
95
.
61.
Amorim
S
,
Stathis
A
,
Gleeson
M
,
Iyengar
S
,
Magarotto
V
,
Leleu
X
, et al
Bromodomain inhibitor OTX015 in patients with lymphoma or multiple myeloma: a dose-escalation, open-label, pharmacokinetic, phase 1 study
.
Lancet Haematol
2016
;
3
:
e196
204
.
62.
Abramson
J
,
Blum
K
,
Flinn
I
,
Gutierrez
M
,
Goy
A
,
Maris
M
, et al
BET Inhibitor CPI-0610 is well tolerated and induces responses in diffuse large B-Cell lymphoma and follicular lymphoma: preliminary analysis of an ongoing phase 1 study
.
Blood
2015
;
126
:
1491
.
63.
Dalla-Favera
R
. 
Molecular genetics of aggressive B-cell lymphoma
.
Hematol Oncol
2017
;
35
:
76
79
.
64.
Stathis
A
,
Zucca
E
,
Bekradda
M
,
Gomez-Roca
C
,
Delord
JP
,
de La Motte Rouge
T
, et al
Clinical response of carcinomas harboring the BRD4-NUT oncoprotein to the targeted bromodomain inhibitor OTX015/MK-8628
.
Cancer Discov
2016
;
6
:
492
500
.
65.
Massard
C
,
Soria
J
,
Stathis
A
,
Delord
J
,
Awada
A
,
Peters
S
, et al
A phase Ib trial with MK-8628/OTX015, a small molecule inhibitor of bromodomain (BRD) and extra-terminal (BET) proteins, in patients with selected advanced solid tumors
.
Eur J Cancer
2016
;
69
:
S2
S3
.
66.
Shapiro
G
,
Dowlati
A
,
LoRusso
P
,
Eder
J
,
Anderson
A
,
Do
K
, et al
Clinically efficacy of the BET bromodomain inhibitor TEN-010 in an open-label substudy with patients with documented NUT-midline carcinoma (NMC)
.
Mol Cancer Ther
2015
;
14
:
Abstract nr A49
.
67.
O'Dwyer
P
,
Piha-Paul
S
,
French
C
,
Harward
S
,
Ferron-Brady
G
,
Wu
Y
, et al
GSK525762, a selective bromodomain (BRD) and extra terminal protein (BET) inhibitor: results from part 1 of a phase I/II open-label single-agent study in patients with NUT midline carcinoma (NMC) and other cancers
.
Cancer Res
2016
;
76
:
Abstract nr CT014
.
68.
Haack
H
,
Johnson
LA
,
Fry
CJ
,
Crosby
K
,
Polakiewicz
RD
,
Stelow
EB
, et al
Diagnosis of NUT midline carcinoma using a NUT-specific monoclonal antibody
.
Am J Surg Pathol
2009
;
33
:
984
91
.
69.
Hottinger
A
,
Sanson
M
,
Moyal
E
,
Delord
J
,
Rezai
K
,
Leung
A
, et al
Dose optimization of MK-8628 (OTX015), a small molecule inhibitor of bromodomain and extra-terminal (BET) proteins, in patients with recurrent glioblastoma
.
Neuro Oncol
2016
;
18
:
iv56
.
70.
Postel-Vinay
S
,
Herbschleb
K
,
Massard
C
,
Woodcock
V
,
Ocker
M
,
Wilkinson
G
, et al
First-in-human phase I dose escalation study of the Bromodomain and Extra-Terminal motif (BET) inhibitor BAY 1238097 in subjects with advanced malignancies
.
Eur J Cancer
2016
;
69
:
S7
S8
.
71.
Wu
SY
,
Lee
AY
,
Hou
SY
,
Kemper
JK
,
Erdjument-Bromage
H
,
Tempst
P
, et al
Brd4 links chromatin targeting to HPV transcriptional silencing
.
Genes Dev
2006
;
20
:
2383
96
.
72.
Keck
KM
,
Moquin
SA
,
He
A
,
Fernandez
SG
,
Somberg
JJ
,
Liu
SM
, et al
Bromodomain and extraterminal inhibitors block the Epstein–Barr virus lytic cycle at two distinct steps
.
J Biol Chem
2017
;
292
:
13284
95
.
73.
Francisco
JC
,
Dai
Q
,
Luo
Z
,
Wang
Y
,
Chong
RH
,
Tan
YJ
, et al
Transcriptional elongation control of hepatitis B virus covalently closed circular DNA transcription by super elongation complex and BRD4
.
Mol Cell Biol
2017
;
37
:
e00040
17
.
74.
Lin
A
,
Wang
S
,
Nguyen
T
,
Shire
K
,
Frappier
L
. 
The EBNA1 protein of Epstein–Barr virus functionally interacts with Brd4
.
J Virol
2008
;
82
:
12009
19
.
75.
You
J
,
Srinivasan
V
,
Denis
GV
,
Harrington
WJ
 Jr.
,
Ballestas
ME
,
Kaye
KM
, et al
Kaposi's sarcoma-associated herpesvirus latency-associated nuclear antigen interacts with bromodomain protein Brd4 on host mitotic chromosomes
.
J Virol
2006
;
80
:
8909
19
.
76.
Banerjee
C
,
Archin
N
,
Michaels
D
,
Belkina
AC
,
Denis
GV
,
Bradner
J
, et al
BET bromodomain inhibition as a novel strategy for reactivation of HIV-1
.
J Leukoc Biol
2012
;
92
:
1147
54
.
77.
Lu
P
,
Qu
X
,
Shen
Y
,
Jiang
Z
,
Wang
P
,
Zeng
H
, et al
The BET inhibitor OTX015 reactivates latent HIV-1 through P-TEFb
.
Sci Rep
2016
;
6
:
24100
.
78.
Ren
K
,
Zhang
W
,
Chen
X
,
Ma
Y
,
Dai
Y
,
Fan
Y
, et al
An epigenetic compound library screen identifies BET inhibitors that promote HSV-1 and -2 replication by bridging P-TEFb to viral gene promoters through BRD4
.
PLoS Pathog
2016
;
12
:
e1005950
.
79.
Boehm
D
,
Calvanese
V
,
Dar
RD
,
Xing
S
,
Schroeder
S
,
Martins
L
, et al
BET bromodomain-targeting compounds reactivate HIV from latency via a Tat-independent mechanism
.
Cell Cycle
2013
;
12
:
452
62
.
80.
Wang
X
,
Helfer
CM
,
Pancholi
N
,
Bradner
JE
,
You
J
. 
Recruitment of Brd4 to the human papillomavirus type 16 DNA replication complex is essential for replication of viral DNA
.
J Virol
2013
;
87
:
3871
84
.
81.
Wang
X
,
Li
J
,
Schowalter
RM
,
Jiao
J
,
Buck
CB
,
You
J
. 
Bromodomain protein Brd4 plays a key role in Merkel cell polyomavirus DNA replication
.
PLoS Pathog
2012
;
8
:
e1003021
.
82.
Zhu
J
,
Gaiha
GD
,
John
SP
,
Pertel
T
,
Chin
CR
,
Gao
G
, et al
Reactivation of latent HIV-1 by inhibition of BRD4
.
Cell Rep
2012
;
2
:
807
16
.
83.
Bolden
JE
,
Tasdemir
N
,
Dow
LE
,
van Es
JH
,
Wilkinson
JE
,
Zhao
Z
, et al
Inducible in vivo silencing of Brd4 identifies potential toxicities of sustained BET protein inhibition
.
Cell Rep
2014
;
8
:
1919
29
.
84.
Korb
E
,
Herre
M
,
Zucker-Scharff
I
,
Darnell
RB
,
Allis
CD
. 
BET protein Brd4 activates transcription in neurons and BET inhibitor Jq1 blocks memory in mice
.
Nat Neurosci
2015
;
18
:
1464
73
.
85.
Sullivan
JM
,
Badimon
A
,
Schaefer
U
,
Ayata
P
,
Gray
J
,
Chung
CW
, et al
Autism-like syndrome is induced by pharmacological suppression of BET proteins in young mice
.
J Exp Med
2015
;
212
:
1771
81
.
86.
Tasdemir
N
,
Banito
A
,
Roe
JS
,
Alonso-Curbelo
D
,
Camiolo
M
,
Tschaharganeh
DF
, et al
BRD4 connects enhancer remodeling to senescence immune surveillance
.
Cancer Discov
2016
;
6
:
612
29
.
87.
Fernandez
P
,
Scaffidi
P
,
Markert
E
,
Lee
JH
,
Rane
S
,
Misteli
T
. 
Transformation resistance in a premature aging disorder identifies a tumor-protective function of BRD4
.
Cell Rep
2014
;
9
:
248
60
.
88.
Fong
CY
,
Gilan
O
,
Lam
EYN
,
Rubin
AF
,
Ftouni
S
,
Tyler
D
, et al
BET inhibitor resistance emerges from leukaemia stem cells
.
Nature
2015
;
525
:
538
42
.
89.
Rathert
P
,
Roth
M
,
Neumann
T
,
Muerdter
F
,
Roe
JS
,
Muhar
M
, et al
Transcriptional plasticity promotes primary and acquired resistance to BET inhibition
.
Nature
2015
;
525
:
543
7
.
90.
Kumar
K
,
Raza
SS
,
Knab
LM
,
Chow
CR
,
Kwok
B
,
Bentrem
DJ
, et al
GLI2-dependent c-MYC upregulation mediates resistance of pancreatic cancer cells to the BET bromodomain inhibitor JQ1
.
Sci Rep
2015
;
5
:
9489
.
91.
Kurimchak
AM
,
Shelton
C
,
Duncan
KE
,
Johnson
KJ
,
Brown
J
,
O'Brien
S
, et al
Resistance to BET bromodomain inhibitors is mediated by kinome reprogramming in ovarian cancer
.
Cell Rep
2016
;
16
:
1273
86
.
92.
Jang
JE
,
Eom
JI
,
Jeung
HK
,
Cheong
JW
,
Lee
JY
,
Kim
JS
, et al
AMPK-ULK1-mediated autophagy confers resistance to BET inhibitor JQ1 in acute myeloid leukemia stem cells
.
Clin Cancer Res
2017
;
23
:
2781
94
.
93.
Hogg
SJ
,
Newbold
A
,
Vervoort
SJ
,
Cluse
LA
,
Martin
BP
,
Gregory
GP
, et al
BET inhibition induces apoptosis in aggressive B-Cell Lymphoma via epigenetic regulation of BCL-2 family members
.
Mol Cancer Ther
2016
;
15
:
2030
41
.
94.
Janouskova
H
,
El Tekle
G
,
Bellini
E
,
Udeshi
ND
,
Rinaldi
A
,
Ulbricht
A
, et al
Opposing effects of cancer-type-specific SPOP mutants on BET protein degradation and sensitivity to BET inhibitors
.
Nat Med
2017
;
23
:
1046
54
.
95.
Stratikopoulos
EE
,
Dendy
M
,
Szabolcs
M
,
Khaykin
AJ
,
Lefebvre
C
,
Zhou
MM
, et al
Kinase and BET inhibitors together clamp inhibition of PI3K signaling and overcome resistance to therapy
.
Cancer Cell
2015
;
27
:
837
51
.
96.
Yang
L
,
Zhang
Y
,
Shan
W
,
Hu
Z
,
Yuan
J
,
Pi
J
, et al
Repression of BET activity sensitizes homologous recombination-proficient cancers to PARP inhibition
.
Sci Transl Med
2017
;
9
:
eaal1645
.
97.
Stuhlmiller
TJ
,
Miller
SM
,
Zawistowski
JS
,
Nakamura
K
,
Beltran
AS
,
Duncan
JS
, et al
Inhibition of lapatinib-induced kinome reprogramming in ERBB2-positive breast cancer by targeting BET family bromodomains
.
Cell Rep
2015
;
11
:
390
404
.
98.
Boi
M
,
Todaro
M
,
Vurchio
V
,
Yang
SN
,
Moon
J
,
Kwee
I
, et al
Therapeutic efficacy of the bromodomain inhibitor OTX015/MK-8628 in ALK-positive anaplastic large cell lymphoma: an alternative modality to overcome resistant phenotypes
.
Oncotarget
2016
;
7
:
79637
53
.
99.
Lasorsa
E
,
Smonksey
M
,
Kirk
JS
,
Rosario
S
,
Hernandez-Ilizaliturri
FJ
,
Ellis
L
. 
Mitochondrial protection impairs BET bromodomain inhibitor-mediated cell death and provides rationale for combination therapeutic strategies
.
Cell Death Dis
2015
;
6
:
e2014
.
100.
Sun
B
,
Fiskus
W
,
Qian
Y
,
Rajapakshe
K
,
Raina
K
,
Coleman
KG
, et al
BET protein proteolysis targeting chimera (PROTAC) exerts potent lethal activity against mantle cell lymphoma cells
.
Leukemia
2017
Jun 30 [Epub ahead of print].
101.
Sun
B
,
Shah
B
,
Fiskus
W
,
Qi
J
,
Rajapakshe
K
,
Coarfa
C
, et al
Synergistic activity of BET protein antagonist-based combinations in mantle cell lymphoma cells sensitive or resistant to ibrutinib
.
Blood
2015
;
126
:
1565
74
.
102.
Tinsley
S
,
Meja
K
,
Shepherd
C
,
Khwaja
A
. 
Synergistic induction of cell death in haematological malignancies by combined phosphoinositide-3-kinase and BET bromodomain inhibition
.
Br J Haematol
2015
;
170
:
275
8
.
103.
Ciceri
P
,
Muller
S
,
O'Mahony
A
,
Fedorov
O
,
Filippakopoulos
P
,
Hunt
JP
, et al
Dual kinase-bromodomain inhibitors for rationally designed polypharmacology
.
Nat Chem Biol
2014
;
10
:
305
12
.
104.
Ember
SW
,
Lambert
QT
,
Berndt
N
,
Gunawan
S
,
Ayaz
M
,
Tauro
M
, et al
Potent Dual BET bromodomain-kinase inhibitors as value-added multitargeted chemical probes and cancer therapeutics
.
Mol Cancer Ther
2017
;
16
:
1054
67
.
105.
Kagoya
Y
,
Nakatsugawa
M
,
Yamashita
Y
,
Ochi
T
,
Guo
T
,
Anczurowski
M
, et al
BET bromodomain inhibition enhances T cell persistence and function in adoptive immunotherapy models
.
J Clin Invest
2016
;
126
:
3479
94
.
106.
Gotwals
P
,
Cameron
S
,
Cipolletta
D
,
Cremasco
V
,
Crystal
A
,
Hewes
B
, et al
Prospects for combining targeted and conventional cancer therapy with immunotherapy
.
Nat Rev Cancer
2017
;
17
:
286
301
.
107.
Lu
J
,
Qian
Y
,
Altieri
M
,
Dong
H
,
Wang
J
,
Raina
K
, et al
Hijacking the E3 ubiquitin ligase cereblon to efficiently target BRD4
.
Chem Biol
2015
;
22
:
755
63
.
108.
Waring
MJ
,
Chen
H
,
Rabow
AA
,
Walker
G
,
Bobby
R
,
Boiko
S
, et al
Potent and selective bivalent inhibitors of BET bromodomains
.
Nat Chem Biol
2016
;
12
:
1097
104
.
109.
Tanaka
M
,
Roberts
JM
,
Seo
HS
,
Souza
A
,
Paulk
J
,
Scott
TG
, et al
Design and characterization of bivalent BET inhibitors
.
Nat Chem Biol
2016
;
12
:
1089
96
.
110.
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
9
.
111.
Raina
K
,
Lu
J
,
Qian
Y
,
Altieri
M
,
Gordon
D
,
Rossi
AM
, et al
PROTAC-induced BET protein degradation as a therapy for castration-resistant prostate cancer
.
Proc Natl Acad Sci U S A
2016
;
113
:
7124
9
.
112.
Tarantelli
C
,
Elkeh
H
,
Moscatello
C
,
Gaudio
E
,
Testa
A
,
Zucca
E
, et al
The BRD4 degrader MZ1 exhibits potent anti-tumoral activity in diffuse large B cell lymphoma of the activated B cell-like type
.
2017 AACR-NCI-EORTC Molecular Targets and Cancer Therapeutics Conference 2017
:
A179
.
113.
Bai
L
,
Zhou
B
,
Yang
CY
,
Ji
J
,
McEachern
D
,
Przybranowski
S
, et al
Targeted degradation of BET proteins in triple-negative breast cancer
.
Cancer Res
2017
;
77
:
2476
87
.
114.
Gaudio
E
,
Tarantelli
C
,
Ponzoni
M
,
Odore
E
,
Rezai
K
,
Bernasconi
E
, et al
Bromodomain inhibitor OTX015 (MK-8628) combined with targeted agents shows strong in vivo antitumor activity in lymphoma
.
Oncotarget
2016
;
7
:
58142
47
.
115.
Peirs
S
,
Frismantas
V
,
Matthijssens
F
,
Van Loocke
W
,
Pieters
T
,
Vandamme
N
, et al
Targeting BET proteins improves the therapeutic efficacy of BCL-2 inhibition in T-cell acute lymphoblastic leukemia
.
Leukemia
2017
;
31
:
2037
47
.
116.
Bui
MH
,
Lin
X
,
Albert
DH
,
Li
L
,
Lam
LT
,
Faivre
EJ
, et al
Preclinical characterization of BET family bromodomain inhibitor ABBV-075 suggests combination therapeutic strategies
.
Cancer Res
2017
;
77
:
2976
89
.
117.
Lam
LT
,
Lin
X
,
Faivre
EJ
,
Yang
Z
,
Huang
X
,
Wilcox
DM
, et al
Vulnerability of small-cell lung cancer to apoptosis induced by the combination of BET bromodomain proteins and BCL2 inhibitors
.
Mol Cancer Ther
2017
;
16
:
1511
20
.
118.
Fiskus
W
,
Sharma
S
,
Qi
J
,
Shah
B
,
Devaraj
SG
,
Leveque
C
, et al
BET protein antagonist JQ1 is synergistically lethal with FLT3 tyrosine kinase inhibitor (TKI) and overcomes resistance to FLT3-TKI in AML cells expressing FLT-ITD
.
Mol Cancer Ther
2014
;
13
:
2315
27
.
119.
Saenz
DT
,
Fiskus
W
,
Qian
Y
,
Manshouri
T
,
Rajapakshe
K
,
Raina
K
, et al
Novel BET protein proteolysis-targeting chimera exerts superior lethal activity than bromodomain inhibitor (BETi) against post-myeloproliferative neoplasm secondary (s) AML cells
.
Leukemia
2017
;
31
:
1951
61
.
120.
Gaudio
E
,
Bernasconi
E
,
Tarantelli
C
,
Cascione
L
,
Kwee
I
,
Rinaldi
A
, et al
Abstract B77: The BET bromodomain inhibitor OTX015 (MK-8628) in mantle cell lymphoma (MCL): in vivo activity and identification of novel combinations to overcome adaptive resistance
.
Mol Cancer Ther
2016
;
14
:
B77
B77
.
121.
Berenguer-Daize
C
,
Astorgues-Xerri
L
,
Odore
E
,
Cayol
M
,
Cvitkovic
E
,
Noel
K
, et al
OTX015 (MK-8628), a novel BET inhibitor, displays in vitro and in vivo antitumor effects alone and in combination with conventional therapies in glioblastoma models
.
Int J Cancer
2016
;
139
:
2047
55
.
122.
Lee
DH
,
Qi
J
,
Bradner
JE
,
Said
JW
,
Doan
NB
,
Forscher
C
, et al
Synergistic effect of JQ1 and rapamycin for treatment of human osteosarcoma
.
Int J Cancer
2015
;
136
:
2055
64
.
123.
Siegel
MB
,
Liu
SQ
,
Davare
MA
,
Spurgeon
SE
,
Loriaux
MM
,
Druker
BJ
, et al
Small molecule inhibitor screen identifies synergistic activity of the bromodomain inhibitor CPI203 and bortezomib in drug resistant myeloma
.
Oncotarget
2015
;
6
:
18921
32
.
124.
Emadali
A
,
Rousseaux
S
,
Bruder-Costa
J
,
Rome
C
,
Duley
S
,
Hamaidia
S
, et al
Identification of a novel BET bromodomain inhibitor-sensitive, gene regulatory circuit that controls Rituximab response and tumour growth in aggressive lymphoid cancers
.
EMBO Mol Med
2013
;
5
:
1180
95
.
125.
Gopalakrishnan
R
,
Matta
H
,
Tolani
B
,
Triche
T
 Jr.
,
Chaudhary
PM
. 
Immunomodulatory drugs target IKZF1-IRF4-MYC axis in primary effusion lymphoma in a cereblon-dependent manner and display synergistic cytotoxicity with BRD4 inhibitors
.
Oncogene
2016
;
35
:
1797
810
.
126.
Moros
A
,
Rodriguez
V
,
Saborit-Villarroya
I
,
Montraveta
A
,
Balsas
P
,
Sandy
P
, et al
Synergistic antitumor activity of lenalidomide with the BET bromodomain inhibitor CPI203 in bortezomib-resistant mantle cell lymphoma
.
Leukemia
2014
;
28
:
2049
59
.
127.
Zhao
X
,
Lwin
T
,
Zhang
X
,
Huang
A
,
Wang
J
,
Marquez
VE
, et al
Disruption of the MYC-miRNA-EZH2 loop to suppress aggressive B-cell lymphoma survival and clonogenicity
.
Leukemia
2013
;
27
:
2341
50
.
128.
Fiskus
W
,
Sharma
S
,
Qi
J
,
Valenta
JA
,
Schaub
LJ
,
Shah
B
, et al
Highly active combination of BRD4 antagonist and histone deacetylase inhibitor against human acute myelogenous leukemia cells
.
Mol Cancer Ther
2014
;
13
:
1142
54
.
129.
Borbely
G
,
Haldosen
LA
,
Dahlman-Wright
K
,
Zhao
C
. 
Induction of USP17 by combining BET and HDAC inhibitors in breast cancer cells
.
Oncotarget
2015
;
6
:
33623
35
.
130.
Adeegbe
DO
,
Liu
Y
,
Lizotte
PH
,
Kamihara
Y
,
Aref
AR
,
Almonte
C
, et al
Synergistic immunostimulatory effects and therapeutic benefit of combined histone deacetylase and bromodomain inhibition in non-small cell lung cancer
.
Cancer Discov
2017
;
7
:
852
67
.
131.
Bhadury
J
,
Nilsson
LM
,
Muralidharan
SV
,
Green
LC
,
Li
Z
,
Gesner
EM
, et al
BET and HDAC inhibitors induce similar genes and biological effects and synergize to kill in Myc-induced murine lymphoma
.
Proc Natl Acad Sci U S A
2014
;
111
:
E2721
30
.
132.
Heinemann
A
,
Cullinane
C
,
De Paoli-Iseppi
R
,
Wilmott
JS
,
Gunatilake
D
,
Madore
J
, et al
Combining BET and HDAC inhibitors synergistically induces apoptosis of melanoma and suppresses AKT and YAP signaling
.
Oncotarget
2015
;
6
:
21507
21
.
133.
Shahbazi
J
,
Liu
PY
,
Atmadibrata
B
,
Bradner
JE
,
Marshall
GM
,
Lock
RB
, et al
The bromodomain inhibitor JQ1 and the histone deacetylase inhibitor panobinostat synergistically reduce N-Myc expression and induce anticancer effects
.
Clin Cancer Res
2016
;
22
:
2534
44
.
134.
Mazur
PK
,
Herner
A
,
Mello
SS
,
Wirth
M
,
Hausmann
S
,
Sanchez-Rivera
FJ
, et al
Combined inhibition of BET family proteins and histone deacetylases as a potential epigenetics-based therapy for pancreatic ductal adenocarcinoma
.
Nat Med
2015
;
21
:
1163
71
.
135.
Feng
Q
,
Zhang
Z
,
Shea
MJ
,
Creighton
CJ
,
Coarfa
C
,
Hilsenbeck
SG
, et al
An epigenomic approach to therapy for tamoxifen-resistant breast cancer
.
Cell Res
2014
;
24
:
809
19
.
136.
Gadd
MS
,
Testa
A
,
Lucas
X
,
Chan
KH
,
Chen
W
,
Lamont
DJ
, et al
Structural basis of PROTAC cooperative recognition for selective protein degradation
.
Nat Chem Biol
2017
;
13
:
514
21
.
137.
Zhou
B
,
Hu
J
,
Xu
F
,
Chen
Z
,
Bai
L
,
Fernandez-Salas
E
, et al
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
2017
Mar 24 [Epub ahead of print].
138.
Winter
GE
,
Buckley
DL
,
Paulk
J
,
Roberts
JM
,
Souza
A
,
Dhe-Paganon
S
, et al
DRUG DEVELOPMENT. Phthalimide conjugation as a strategy for in vivo target protein degradation
.
Science
2015
;
348
:
1376
81
.
139.
Zengerle
M
,
Chan
KH
,
Ciulli
A
. 
Selective small molecule induced degradation of the BET bromodomain protein BRD4
.
ACS Chem Biol
2015
;
10
:
1770
7
.