Open Access

Targeting BRD4: Potential therapeutic strategy for head and neck squamous cell carcinoma (Review)

  • Authors:
    • Voraporn Yongprayoon
    • Napasporn Wattanakul
    • Winnada Khomate
    • Nathakrit Apithanangsiri
    • Tarathip Kasitipradit
    • Danupon Nantajit
    • Mahvash Tavassoli
  • View Affiliations

  • Published online on: April 12, 2024     https://doi.org/10.3892/or.2024.8733
  • Article Number: 74
  • Copyright: © Yongprayoon et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

As a member of BET (bromodomain and extra-terminal) protein family, BRD4 (bromodomain‑containing protein 4) is a chromatin‑associated protein that interacts with acetylated histones and actively recruits regulatory proteins, leading to the modulation of gene expression and chromatin remodeling. The cellular and epigenetic functions of BRD4 implicate normal development, fibrosis and inflammation. BRD4 has been suggested as a potential therapeutic target as it is often overexpressed and plays a critical role in regulating gene expression programs that drive tumor cell proliferation, survival, migration and drug resistance. To address the roles of BRD4 in cancer, several drugs that specifically target BRD4 have been developed. Inhibition of BRD4 has shown promising results in preclinical models, with several BRD4 inhibitors undergoing clinical trials for the treatment of various cancers. Head and neck squamous cell carcinoma (HNSCC), a heterogeneous group of cancers, remains a health challenge with a high incidence rate and poor prognosis. Conventional therapies for HNSCC often cause adverse effects to the patients. Targeting BRD4, therefore, represents a promising strategy to sensitize HNSCC to chemo‑ and radiotherapy allowing de‑intensification of the current therapeutic regime and subsequent reduced side effects. However, further studies are required to fully understand the underlying mechanisms of action of BRD4 in HNSCC in order to determine the optimal dosing and administration of BRD4‑targeted drugs for the treatment of patients with HNSCC.

Introduction

Head and neck squamous cell carcinoma (HNSCC) is one of the most prevalent cancer globally, which arises from stratified mucosa of the mouth, trachea and larynx. Despite improved treatments, overall survival remains low, with more than 450,000 deaths in 2018 (1). There is a rise in annual incidence of oropharynx squamous cell carcinoma, a subtype of HNSCC, over the past decade, within the non-smokers, non-alcoholics and aged <50 years white male demographic group. This occurrence is associated with the human papillomavirus (HPV) infection, particularly HPV16, with the risk factor being an increase in sexual partners for oral or vaginal sex at a younger age (2).

Bromodomain protein 4 (BRD4) is one of the members of the bromodomain and extra-terminal (BET) family and a dual bromodomain protein consisting of two N-terminal bromodomains and an extra-terminal (ET) domain. BRD4 binds and acetylates lysine residues on target proteins including histones as a transcriptional and epigenetic regulator (3). BRD4 plays an important role in transcription, replication and DNA repair. It also binds to non-histone proteins, DNA and RNA, contributing to development, tissue growth and various physiological processes (4). BRD4 is a crucial element in the regulation of cell cycle and mitosis which ensures the integrity of cell differentiation and development. The protein is also a predominant component of super-enhancers (SEs) associated with all active promoters and a significant proportion of active promoters in the genome including activation of genes involved in cell growth and cell cycle progression (5,6). The critical roles of BRD4 regarding transcriptional regulation are growth and cell division, metabolic processes, immune responses and embryo development regulation. In normal and transformed cells, BRD4 dysfunction results in pathogenesis and disease development in humans, such as prolonged inflammation, as the protein directly regulates the activity of NF-κB including inflammation, fibrosis, viral infections and neoplasia (7,8). Several studies have addressed the involvement of BRD4 in development of various tumors, where BET can promote aberrant expression of oncogenes such as c-Myc in acute lymphocytic leukemia and acute myeloid leukemia (AML) (9). The diverse role of BRD4 depends in several contexts on its interaction partners. It is considered that interfering with BET activity can reduce cancer cell proliferation and induce apoptosis (10). The most significant evidence for involvement of BRD4 in HNSCC carcinogenesis came from NUT carcinoma. Inhibition of the BRD4-NUT fusion gene on the BRD4 section using BETi (BET inhibitors) stalled the growth of NUT carcinoma cells (11). Therefore, targeting BRD4 through inhibition of BET protein has been explored as a therapeutic option for various cancers including HNSCC.

The normal functions of BRD4

BRD4 is an indispensable protein for cellular development. For example, bone marrow stem cells cannot differentiate into lymphoid stem cells without the presence of BRD4 (12). Moreover, the full expression of BRD4 is also essential for embryogenesis. A previous study conducted in mice reported that one allele of BRD4 was only enough to allow the embryonic stem cells to differentiate but insufficient for complete mouse development (13).

Post-translational modification of histone alters gene expression by regulating the chromatin landscape through changing the overall charge of the chromatin which recruits chromatin modifier enzymes (14). The epigenetic phenomenon is partly operated by BRD4 as the protein has histone acetyltransferase (HAT) and kinase activities phosphorylating serine2 residue of the RNA polymerase II carboxy-terminal domain. The binding of bromodomains to acetylated histone and lysine residues at the histone H3 site and H4 on chromatin regulates downstream gene expression. As BRD4 regulates chromatin remodeling by acetylating histone H3 Lys122, it causes instability and ejection of nucleosomes from chromatin as well as chromatin structural detachment; this leads to an increase in transcription. The resulting chromatin fragmentation permits DNA accessibility and allows access to transcriptional machinery (1517). The perturbed chromatin structure and nucleosome remodeling at the promoters allow transcription factors as well as RNA Polymerase II to enter and start the transcription process (18). Furthermore, BRD4 coupling with RNA Polymerase II complex assists the complex to elongate through hyperacetylated nucleosomes by interacting with acetylated histones using bromodomains (19).

The HAT activity of BRD4 is responsible for a smooth transition from G1 to M phase of the cell cycle as it mediates transcription and pause-release. Similarly, the G2 to M phase transition has been known to be under the control of BRD4 via its interaction with a GAP protein, SPA-1. This again, relieves the block to cell cycle progression (20). Likewise, BRD4 controls the levels of Aurora B which is concentrated around the sites of attachment of chromosomes to spindle microtubules such as the centromeres or kinetochores and allows for chromosome segregation to occur appropriately (21). With low levels of BRD4, mitosis may become abnormal leading to increased incidence of lagging chromosomes, micronuclei and bridging chromosomes, eventually resulting in failed cytokinesis and multilobulated nuclei (16). As numerous genes regulated by BRD4 are involved in the processes of cell differentiation and development, dysregulation of BRD4 could become oncogenic which leads to pathogenesis of a wide variety of cancers (22).

Roles of BRD4 in tumor development

Aberrant expression or function of BRD4 is well-connected to oncogenic processes which includes HNSCC tumorigenesis (23). BRD4 has two well-structured N-terminal bromodomains (BD1 and BD2); in addition to BD1 and BD2, the molecular actions of BRD4 depend on the CK2-phosphorylated region, conserved ET domain and the distinct C-terminal motif. The regions are the interactive platform for recruiting chromatin and transcriptional regulators (24). BRD4 has three isoforms of different lengths but there are two main isoforms, BRD4 long (BRD4-L) and BRD4 short (BRD4-S). Evidence suggests that a disruption of the balance between the two BRD4 isoforms occurs in certain cancer types leading to substantial biological consequences (25). BRD4 and other BET proteins are often overexpressed in cancer and this leads to abnormal chromatin remodeling and tumorigenesis-mediated gene transcription. BRD4-mediated histone modifications regulate gene expression and maintain normal cellular homeostasis, which are vital for the cells (26). Studies conducted in human cancer types have shown that BRD4 overexpression is one of the reasons for oncogene amplification such as Myc, Notch3 and NRG1 leading to cancer progression (25,27). The progression of triple-negative breast cancer (TNBC) is also linked to increased phosphorylation of BRD4 in the acidic region due to decreased protein phosphatase 2A (PP2A) activity (28). These studies all point to BRD4 as a central protein for tumor development, specifically by inducing and maintaining the pool of cancer stem cells in squamous cell carcinoma including HNSCC (29).

Oncogenic mechanisms resulting from changes in genome structure may include mutations, copy number changes, or genome rearrangements. ‘Oncogene addiction’ is a mechanism used by cancer cells to maintain their unchecked proliferative needs (30). This is largely due to the functions of BET proteins, in addition to their role in transcriptional regulation by forming the Twist/BRD4/P-TEFb/RNA-Pol II complex which lead to stem cell-like properties and tumorigenicity (31). BRD4 is a key protein in numerous cancer hallmarks, it can stimulate cancer cell proliferation through the functions of Jagged1 and Notch1 in breast cancer (32). It also controls oncogenic network gene expression by interacting with acetylated transcription factors including RELA, ER, p53 and twist (33). BET inhibitors have demonstrated remarkable anticancer effects for treatment by interfering with BRD4 expression or activity and effectively inhibit the progression of the cell cycle and induce apoptosis which reduces tumor cell proliferation and subsequent cancer development (3436). Inhibition of BRD4 has revealed significant effect on sensitizing various tumor cell types to therapeutic agents including diffuse large B-cell lymphoma, neuroblastoma, lung cancer, NUT midline cancer and HNSCC (3741). BRD4 has been linked with poor prognosis in a wide range of cancer patients including HNSCC (23,27). In addition to solid tumors, BRD4 inhibition has also been identified to be effective against hematological malignancies (42). The anticancer efficacy of BRD4 has been reported and clinical trials are ongoing (10,43). Incoming data from these studies will further validate the use of BRD4 inhibitor in antitumor therapy. However, it is critical to investigate whether targeting BRD4 is feasible for HNSCC treatment as there is a currently unsolved dilemma for the cancer as discussed below.

DNA damage repair and therapy resistance

Genetic mutations resulting from unrepaired DNA damage may increase the risk of precipitating genetic disorders and cancers. Although an isoform of BRD4 functions as an internal inhibitor of DNA damage response by remodeling the chromatin complex (44), the protein transcriptionally regulates DNA damage repair-related genes such as RAD51AP1 and TopBP1 as well as engages in double strand breaks (DSBs) through both non-homologous end joining (NHEJ) and homologous recombination (HR) pathways (4548). BRD4 particularly involves in HR through direct contact with the SWI/SNF chromatin remodeling complex (49). It is worth noting that BET proteins inhibition itself can induce DNA damage potentially through deposition of R-loops leading to transcription-replication collision events (46). Additionally, BRD4 assists in maintaining genome stability through non-transcriptional functions such as DNA damage repair, checkpoint activation and telomere homeostasis (27). The use of BET inhibitors, namely JQ1 and AZD5153, has been revealed to prolong DNA DSBs and repress NHEJ-related genes XRCC4 and SHLD1 (50). JQ1 treatment leads to the substitution of BET proteins and transcription regulatory complexes from acetylated chromatin (51). JQ1 not only increases the damage level of DNA, but also attenuates DNA damage repair, particularly double strand break repair, which consequently sensitize the tumor cells to PARP inhibitor Olaparib (52). Additionally, JQ1 can inhibit the growth of ARID2-deficient hepatocellular carcinoma cells as well as induce apoptosis when ARID2-depleted through the aggravated DNA damage of DSBs (53).

BRD4 amplification has been shown as a prognostic factor in various cancer types such as ovarian, esophageal, non-small cell lung cancer and HNSCC (23,25,54,55). This could be due to numerous pro-survival functions of the protein including acetylation of histone H4 by DNA damage recruits BRD4 to stabilize the DNA repair complex (47). The multiple underlying roles of BRD4 in DNA damage repair is conceivably the major contributor in tumor cell resistance to therapy. For HNSCC, mutations in the DNA repair genes have enabled HNSCC to become resistant to therapy (56). Additionally, certain DNA damage repair genes appear to be upregulated including Ku80 and APEX1 and linked with patient prognosis (57,58). However, a recent study has indicated that a change in the expression of individual DNA repair proteins may not necessarily cause resistance to therapy. Rather, a balanced expression and coordination within the DNA repair signaling cascade is rather the actual cause of the resistance (59). Thus, targeting BRD4 protein which is upstream of DNA damage response may hypothetically benefit cancer patients, especially those with therapy-resistant HNSCC. The use of BRD4 inhibitor has been revealed to enhance the radiosensitivity of HNSCC as well as other tumors in pre-clinical models potentially through the upregulation of p21 and suppression of RAD51AP1 and Mcl-1 (41,6063). Targeting the protein is also suggested to attenuate YAP, Myc-AP4 and E2F2 signaling which are often upregulated in various tumors (6466). Degradation of BRD4 could cause a genome wide pausing of Pol II as BRD4-PTEFb is the main driving partner for phosphorylation of Pol II C-terminal domain and Pol II transcription (67,68). Similarly, BRD4 inhibition has been shown to sensitize colorectal tumor to doxorubicin as the protein is the cause of cisplatin resistance in bladder tumor through the Sonic hedgehog pathway (69,70). These accumulating data further have supported targeting the bromodomain protein as a therapeutic option to enhance the efficacy of current conventional therapies.

Epithelial mesenchymal transition (EMT) and cancer progression/aggressiveness

EMT remains a challenge in cancer treatment as the phenomenon provides not only an escape route with resistant features for cancer cells under therapeutic stress but also an opportunity for tumor expansion and metastasis. BRD4 is regarded as a key regulator of EMT as it governs key transcription factors that drive EMT particularly through the transcription of snail, both SNAI1 and SNAI2, as well as involves in TGF-β mediated EMT (71,72). Coupling between BRD4 and di-acetylated Twist was also shown to enhance downstream transcriptional targets of Twist for EMT (31,73). Overexpression of BRD4 enhances EMT and EMT is inhibited with reduced expression of BRD4 (74). There is a controversy whether the other BET proteins are involved in activation of EMT, namely BRD2 and BRD3; essentially, they are demonstrated to have a degree of control over EMT activation (75). Inhibition of BRD4 has frequently been demonstrated to suppress EMT through various mechanisms; for example, through activation of the NF-κB-NLRP3-caspase-1 pyroptosis signaling pathway in renal cell carcinoma (51), and inhibition of RelA-initiated TGF-β induced EMT via inflammatory tissue remodeling (76). BRD4 also regulates Jagged1 expression and Notch1 signaling for cancer cell dissemination (32). Treatment with BRD4 inhibitors has been identified to effectively suppress EMT-associated tumor invasion and metastasis through the regulation of key EMT proteins as well as attenuate the expression of MMP-2 and MMP-9, thus reducing HNSCC metastatic potential (7780). Likewise, the activity of BRD4 in transitioning HNSCC cells to mesenchymal phenotype has equipped the cells to become cisplatin-resistant (81). These studies further emphasize the roles of BRD4 in tumor proliferation and expansion. The multiple functions of BRD4 on the development and expansion of HNSCC described are demonstrated in Fig. 1.

BRD4 in inflammation

Research involving BRD4 has evolved greatly as a result of its role in inflammation, which is associated with cancer through genomic instability, a cancer hallmark. Various studies have established that cancer is a disease that develops and progresses within inflammatory diseases including HNSCC (82). BRD4 enhances acetylation of RelA-K310ac which activates cyclin-dependent kinase 9 (CDK9), leading to the phosphorylation of RNA polymerase II to promote NF-κB gene transcription, thereby initiating the production of proteins responsible for inflammatory stimuli (83). Additionally, BRD4 regulates the expression of inflammatory genes through activation of enhancer RNAs or the Mnk2-eIF4E pathway-dependent translational regulation of IκBα synthesis in modulating inflammatory gene expression (84). There is also a direct association between BRD4 and the acetylated p65 subunit of NF-κB as well as the transcription factor of Nrf2, a key regulator of inflammation (85). Studies have investigated the efficiency of BRD4 inhibition of the inflammatory process. In primary human umbilical cord-derived vascular endothelial cells treated with TNF-α, JQ1 lessens the overexpression of FN1 induced by TNFα and could possibly slow down the progression of atherosclerosis (86). JQ1 has been shown to effectively protect colon-tight junctions from endotoxemia-induced inflammatory injury (87). In rat kidney triggered by Cadmium for inflammatory response, BRD4 inhibition reduced NF-κB nuclear translocation and its subsequent transcriptional activity (83). The use of I-BET, a BET inhibitor, can effectively inhibit pro-inflammatory protein production in lipopolysaccharide-activated macrophages (88). In primary and human bronchial epithelial cell lines, oxidative stress induced by IL-1β was significantly reduced by BRD4 inhibition (89). These studies have implicated that targeting BRD4 can subside inflammation in HNSCC and maybe beneficial to the patients as various studies have reported that inflammatory markers are prognostic factors for these patients (9093). Thus, alleviating inflammation as a result of targeting BRD4 could prove to be useful in the treatment of HNSCC.

The relationship between BRD4 and HPV

Most common in the United States and other high-income countries, HPV-related HNSCC is becoming far more common than HPV-associated cervical cancer (94). Oncogenic HPV can be latent and cause malignant transformation years later; however, infection of high-risk HPV types can lead to pre-cancerous, in certain tissue, and cancer (95). A total of ~25% of all HNSCCs were positive for HPV-DNA with HPV-16 being the most prevalent subtype (96). There are 15 high-risk types of HPV, but the two most common ones, HPV16 and HPV18 accounting for ~72% of the total (97). The life cycle of HPV is highly dependent on the host cellular differentiation program. Although a receptor for HPV infection has not been recognized, it has been postulated to be heparan sulfate proteoglycan on the basal membrane (98). The role of viral protein E1 is unclear, whereas E2 is responsible for the transcription of E6 and E7 viral genes. In addition, the binding of HPV E2 protein to DNA is involved in viral DNA replication, transcription, genome maintenance and isolation (99). HPV E6/E7 expression is required for the binding of viral genome to DNA in the regions of genomic instability. This is followed by disruption of the E2 coding region and abnormal regulation of E6/E7 itself. Because of this, HPV can produce persistent infection (100). Degradation of p53 and pRb, by E6 and E7, respectively, contribute to cancer induced by this virus (101).

In HPV-associated tumors, BRD4 plays an important role in replication of HPV (17). The viral genome is attached to the mitotic chromosomes for segregation; BRD4 is used as a cellular adapter, where BRD4 typically interacts with the virus-encoded E2 protein to facilitate viral genome segregation (102,103). BRD4 and E2 form a complex between the viral genome and the host chromosomes to allow the viral genome insertion at fragile sites of the host genome (104). BRD4 is recognized as an atypical chromatin binding factor that binds to chromosomes throughout mitosis, known as MCAP (mitotic chromosome-associated protein) and is expressed as a mitotic bookmark. As transcriptional regulation is a fundamental role of BRD4, it is vital for several E2 functions and stability. Disruption of the interaction between BRD4 and E2 inhibits E2-mediated transactivation (105). In conjunction with E2, BRD4 is required to suppress the transcription process in early viral promoter, an essential process during the early gene expression in order to maintain the infection in the basal cells, in which the copy number of the viral genome remains very low (106). Phosphorylation of BRD4 regulated by Casein kinase II and PP2A is essential for the binding of BRD4 to acetylated chromatin and recruiting major transcription factors including p53, AP-1 and NF-kB to control the viral transcription program (107). The role of BRD4 is evidently important in regulating HPV transcription particularly in the early stage of viral transcription. Treatment with BETi has been shown to reduce the viral transcription in a HPV11 infected model (108). A combination of BET inhibitor and HDAC6 inhibitor has demonstrated significant synergistic effects against HPV-positive and HPV-negative in HNSCC cells (77). Similarly, it has beem reported by the authors that BRD4 inhibitor is effective in reducing HPV E6/E7 transcription in HPV-associated HNSCC cell lines (37). It is also worth noting that inhibition of BRD4 also offers antiviral activities by decompacting chromatin structure and activating DNA damage-dependent immune responses which attenuates viral attachment to the host chromosome and subsequently improves host resistance to viral infection (109). Thus, BRD4 inhibition is potentially an effective approach against viruses-associated malignancies (110).

BRD4 roles and therapeutic approaches for HNSCC

For primary HNSCC, surgical resection of the tumor and lymph node followed by radiotherapy with or without platinum-based chemotherapy or definitive concurrent chemoradiation therapy is the main modality for treating the patients (111). Cisplatin is often the chemo-reagent for the course; however, significant acute and late toxicity is often observed (112). Thus, deintensification approach has been trialed using cetuximab to target epidermal growth factor receptor (EGFR), replacing cisplatin for those with HPV-positive HNSCC. However, the patients receiving cetuximab appear to be at a higher risk of death and relapse of the disease than those receiving cisplatin (113,114). Despite EGFR upregulation is an acknowledged biomarker suggesting treatment resistance and aggressiveness in HNSCC (115), targeting EGFR has shown significant benefits for HPV-negative HNSCC (116). However, inhibition of the receptor in HPV-associated HNSCC leads to lesser therapeutic outcomes suggesting that EGFR plays opposing roles in the two HNSCC subtypes (117). Increasing evidence has demonstrated that cetuximab may not be the best course for HPV-positive HNSCC therapy (118,119). In HPV-positive HNSCC cells, overexpression of EGFR suppresses cellular proliferation and increases radiosensitivity through inhibition of BRD4 via miR-9-5p and subsequently reduced HPV E6/E7 transcription (37). Therefore, targeting EGFR may not be the best course of therapy for HPV-positive HNSCC, but targeting specific signaling pathways such as BRD4 could provide a preferable new treatment to improve the therapeutic outcome of HNSCC (120).

BRD4 has been clinically linked to several oncogenes, such as activating Myc in leukemia and lymphoma (121). It has also been observed that BRD4 protein and its mRNA levels are abnormally regulated in HNSCC samples, correlated with tumor features such as size, proliferation and advanced disease degree (23). A previous study in HNSCC reported that BRD4 overexpression decreases the mRNA stability of cyclin-dependent kinase inhibitor 1B (p27), and the protein p27 is responsible for inhibiting tumor progression (122). The protein can also act as a pro-oncogene that accelerates tumor growth and metastasis as a critical part of SEs (123). BRD4 has thus been identified as a prognostic biomarker of HNSCC (23).

Inhibition of BRD4 using JQ1 has been demonstrated to induce senescence in head and neck tumor cells through downregulation of acetylated histone H4 and phosphorylated SIRT1(ser47) leading to p21 and p16ink4 accumulation (124). Treatment with the inhibitor also blocks SEs, decreases TP63 expression in HNSCC, and effectively eliminates both cancer stem cells and lymph node metastasis (125). Additionally, BRD4 is a regulator of JOSD1, a protein linked to poor prognosis in patients with HNSCC. JQ1 treatment has been identified to downregulate both the JOSD1 protein and mRNA expression. Overexpression of the protein indicates a poor clinical prognosis for patients with HNSCC (126). Similarly, cooperation between YAP1 and BRD4, which can be attenuated by JQ1 treatment, enhances HNSCC tumorigenesis (127). Treatment with the inhibitor has been also shown to overcome cetuximab resistance in HPV-negative subtype of HNSCC (128). These studies have accumulated evidence in favor of the use of BRD4 inhibitor as a part of HNSCC therapy in the near future to resolve the dilemma of targeting EGFR in HPV-associated HNSCC, as demonstrated in Fig. 2.

BRD4 and immune response

Tumor cells often modulate the expression of genes or immune signaling pathways to avoid immune recognition and promote tumor growth and metastasis (129). Therefore, it is critically important to recognize their interaction with immunologic cells in order to stratify toward immunotherapy. Immune cell infiltration into tumor tissue, particularly for cytotoxic T lymphocytes and natural killer cells have been closely investigated in recent years. The roles of BRD4 regarding immune response to tumor have been investigated with BRD4 expression being associated with levels of infiltrating monocytes, tumor-associated macrophages, M1/M2 macrophages and T cells (Th1/Th2/Treg) in breast cancer (130). In hepatocellular carcinoma, expression of BRD4 mRNA is elevated and correlated with immune infiltrating levels of B cells, CD8+ T cells, CD4+ T cells, macrophages, neutrophils and dendritic cells (131). BRD4 expression is also connected with low infiltration of T-bet+ tumor-infiltrating T lymphocytes leading to poorer prognosis potentially through activation of Jagged1 signaling pathways (132). BRD4 regulates programmed death-ligand 1 (PD-L1) expression through c-Myc implicating that targeting BRD4 can influence immune system against tumor cells (133). Suppression of BRD4 leads to downregulation of PD-L1 in TNBC, thus potentially permitting an improved outcome with immunotherapy approach (134). BRD4 expression, above the other BET proteins, is the most negatively correlated with immune checkpoint as well as abundance of macrophage, neutrophil and CD8+ T-cell in glioblastoma multiforme (135). These studies have all designated BRD4 as a prognostic marker for patient survival further highlighting the bromodomain protein as a therapeutic target. Furthermore, for HNSCC, the expression of PD-L1 in HNSCC cell lines could be reduced by JQ1 or MZ1 treatment (136). Likewise, suppression of the BET protein could enhance antitumor immunity through the induction of MHC class I expression and consequently improve the efficacy of anti-PD-1 immunotherapy in an in vivo model (137).

Although the exact mechanism on how BRD4 mediates tumor microenvironment and immune infiltration needs further elucidation, BRD4 is involved in the acetylation of lysine-310 of the RelA NF-κB subunit which activates the transcription factor and modulates proinflammatory cytokines as well as Th17 immune response (138). The epigenetic regulator has been shown to be responsible for the expression of a cohort of immunosuppressive genes including PD-L1, PD-L2, HVEM, GAL9, IL6, IL8, CSF2, BIRC3, IDO1 and IL1B (139). BRD4 is also suggested to be the protein responsible for immunosuppressive M2 macrophage polarization (140,141). Collectively, these studies have provided evidence that targeting BRD4 may shift the landscape of tumor microenvironment for immunotherapy and antitumor immune response which could be useful for the treatment of HNSCC as well as other solid tumors. It is enticing to explore the interaction between BETi and immunotherapeutic agents such as nivolumab and pembrolizumab which have been approved for HNSCC therapy. The co-administration between BETi and immunotherapy could lead to an effective therapy against HNSCC.

Resistance to BET inhibition

Despite the promise of BRD4 inhibition in cancer therapy mentioned, it has been suggested that the efficacy of BRD4 inhibition as a monotherapy could be transient and moderate (123). Several studies have demonstrated that tumor cells may develop resistance after a prolonged use of JQ1 due to the rewiring of proteins involved in transcriptional regulation which also affect other chromatin-targeted therapies. For example, in AML, WNT/β-catenin signaling pathway is shown as the primary and acquired driver for resistance to BETi (142,143). In lung adenocarcinoma, BET inhibition is effective in blocking cell growth through FOS-like 1 (FOSL1) suppression; however, resistance to JQ1 occurs independently of its effect on FOSL1 or Myc expression. Phosphorylation of BRD4 by casein kinase 2 (CK2) is suggested as a cause of BETi resistance (144). DUB3, which is upregulated by JQ1 treatment, deubiquitinates and stabilizes BRD4 causing prostate cancer to become resistant to BETi (145). Similarly, JQ1 resistance was demonstrated to be due to the loss of BRD4/FOXD3/miR-548d-3p axis which is compensated by JunD/RSK3 signaling which essentially builds up BETi resistance in basal-like breast cancer (146). For TNBC, the cells can rapidly develop resistance due to various mechanisms, including changes in signaling pathways involving ZNF33A upregulation, deletion of SNF/SWI complex components as well as ubiquitination-related genes such as SPOP, UBE2M, CUL3 and USP14 (147,148). In ovarian cancer cell lines, autophagy (shown by increased expression of ATG5 and Beclin1) induced by inactivation of Akt (Ser473)/mTOR (Ser2448) pathway, is linked with resistance to BETi possibly as a way to bypass BET inhibition (149). Thus, it is essential to note that resistance to JQ1 has distinct mechanisms depending on cancer types; however, increase in Myc expression has been pinpointed as common cause of resistance to BETi (147,150,151). BRD4 stabilization and its subsequent activation of AKT-mTORC1 activation has also been described as another route of BETi resistance (152). Therefore, the issue of resistance to BET inhibition should be closely investigated especially when BET inhibition is applied particularly as a monotherapy. The use of BRD4 inhibition as a part of combination therapy could be a more viable option considering this matter.

Clinical response to BRD4 targeting and adverse events

Currently, no BETi has been approved by the US FDA for the use of cancer; however, there are a number of phase I clinical studies which have provided initial information regarding the safety of BETi in patients. As shown in Table I, BETi appears to be safe in most patients with the prevalent treatment emergent adverse events (TEAEs) including thrombocytopenia, diarrhea, nausea, anorexia, vomiting, fatigue and anemia. The most significant dose-limiting toxicities are thrombocytopenia and fatigue. All studies have concluded that BETi was largely tolerable by the patients; thus, applying BETi for deintensification of HNSCC therapy could be viable although more data should be collected from further clinical studies especially for patients with HNSCC. Regarding the potential efficacy against HNSCC, it is premature to conclude the antitumor efficacy of BETi as the majority of these clinical studies have been conducted in patients with hematologic malignancies. However, a few studies in solid cancers have mentioned that the patients had achieved longer progression-free survival with 95% confidence intervals of [1.8–1.9] and [4.6–12.9] (153,154). Despite the promising safety of BETi in patients, future clinical studies should proceed with care and prepare to address the TEAEs which are likely to emerge.

Table I.

Clinical studies of BET inhibitors and the observed adverse events in the patients.

Table I.

Clinical studies of BET inhibitors and the observed adverse events in the patients.

BET inhibitorTargetDiseasePhaseAdverse effects(Refs.)
ABBV-075BRD2/3/4,Uveal melanoma, breast cancer,I- TEAEs were dysgeusia, thrombocytopenia, fatigue, and nausea(153)
BRDTpancreatic, HNSCC, CRPC, and others - TEAEs (Grade 3/4) were thrombocytopenia and anemia
- DLT included thrombocytopenia, gastrointestinal bleed, hypertension, fatigue,
decreased appetite, and aspartate aminotransferase elevation
FT-1101BRD2/3/4,AML, MDS, NHLI- TEAEs (all grades) were diarrhea, fatigue, dyspnea, nausea, anemia,
BRDT and platelet count decreased
- AML/MDS pts had diarrhea, nausea or pleural effusion, cough, decreased
appetite, or dyspnea
- Severe (≥ grade 3) TEAEs were anemia, decreased platelets, pneumonia, sepsis,
febrile neutropenia, and disease progression(160)
GSK525762/BRD2/3/4,CRC, NUT carcinoma, and other solidI- TEAEs (all grades) were thrombocytopenia, diarrhea, nausea, vomiting, anemia,(161)
I-BET762BRDTtumors decreased appetite, dysgeusia, and fatigue
R/R AML, AML after MDS, new AMLI/II- TEAEs were dysgeusia, diarrhea, nausea, and elevated bilirubin(162)
- DLTs at 100 mg: grade 3 diarrhea
- DLTs at 120 mg: grade 3 ejection fraction decrease
- TEAEs (Grade 3/4) were diarrhea, febrile neutropenia, thrombocytopenia,
hyperglycemia, and fatigue/asthenia
INCB057643BRD2/3/4,Any advanced/recurrent malignancyI/II- TEAEs (All grades) were Nausea, thrombocytopenia, fatigue, decreased(163)
BRDTProstate cancer, colorectal cancer, appetite, diarrhea, dysgeusia, anemia, hyperglycemia
breast cancer, ovarian cancer,
lymphoma, AML, pancreatic cancer,
glioblastoma, MDS, and others
R/R MF, other advanced myeloidI- TEAEs were thrombocytopenia, dysgeusia, nausea, anemia, blood bilirubin(164)
neoplasms increased, ejection fraction decreased
ZEN003694BRD2/3/4,Metastatic CRPCIb/IIa- Toxicities grade ≥ 3 included: nausea, thrombocytopenia, anemia, fatigue, and(154)
BRDT hypophosphatemia
- TEAEs were visual symptoms
ODM207BRD2/3/4,CRPC, melanoma, NMC, ER+BC, andI- DLT was intolerable fatigue
BRDTSolid tumors - Common TEAEs were thrombocytopenia, asthenia, nausea, anorexia, diarrhea,(165)
fatigue, and vomiting
OTX015BRD2/3/4AML, ALL, RA with a large numberI- Non-cumulative grade 1–2 gastrointestinal events (diarrhea, dysgueusia,(166)
of blasts abdominal pain, nausea, anorexia), hyperglycemia, coagulation factor VII
decrease, and direct bilirubin increase
- DLT was diarrhea, anorexia/fatigue
Acute leukemia, MDSI- DLT was G3 diarrhea, G3 fatigue. Diarrhea, fatigue, and rash limited compliance(167)
Lymphoma, myelomaI- DLTs: G4 thrombocytopenia, G4 neutropenia, G3 hyponatremia(167)
- Other toxicity: Grade 1–2 events: gastrointestinal/diarrhea, nausea, fatigue,
dysgeusia, anemia
NMC, CRPC, NSCLCIb- DLTs at 80 mg once daily: grade 3 thrombocytopenia, ALT/hyperbilirubinemia(168)
- DLTs at 100 mg once daily: grade 2 anorexia and nausea, grade 4
thrombocytopenia
- TEAEs were diarrhea, nausea, anorexia, vomiting, thrombocytopenia
CC-90010BRD2/4Solid tumors, NHLI- TRAEs (Grade 3/4) were thrombocytopenia, anemia, and fatigue(169)
- TRAEs (Grade 4) thrombocytopenia associated with grade 3 skin hemorrhage
- TEAEs (Grades ≤3) thrombocytopenia, hyperglycemia, and asthenia
- TEAEs were nausea/vomiting, fatigue/asthenia, and thrombocytopenia
CPI-0610BRD4R/R LymphomaI- Primary toxicity: Thrombocytopenia, Grade 3 diarrhea, rash, neutropenia(170)
MyelofibrosisII- TEAE (All grade): diarrhea, nausea, cough, and upper respiratory tract infection(171)
- TEAEs (≥3 Grade) were anemia and thrombocytopenia
PLX51107BRD4Solid tumors, lymphoma, AML, MDSI- Nonhematologic toxicities were febrile neutropenia and pneumonia in(172)
12 patients each; 6 patients had severe hyperbilirubinemia
- TEAEs (Grade 1–2) were fatigue, vomiting, diarrhea, nausea, bilirubin
increase, and INR increase
- TEAEs were G3 nausea, G2 vomiting, and G2 kidney injury
- Uveal melanoma, sarcoma, NSCLC, breast cancer and CRPC, and other
solid tumors
II- The most common toxicity was grade1-2 fatigue, vomiting, diarrhea, nausea,(173)
bilirubin increase, and INR increase
- TEAEs: G3 nausea, and G2 kidney injury
- DLTs were G3 thrombocytopenia, G3 nausea, G2 kidney injury
RO6870810/BRD4MDS, AMLI- TEAEs were fatigue, decreased appetite and injection-site erythema,(174)
TEN-010 injection-site pain, nausea, fatigue, thrombocytopenia, anemia, and
hyperbilirubinemia
NC, DLBCLI- TEAEs (All grades) were fatigue, decreased appetite and injection-site(175)
erythema, injection-site pain and nausea
- TEAEs (grade 3/4) were fatigue, thrombocytopenia, anaemia, and
hyperbilirubinemia
NMCI- Nonhematologic toxicities were Irritation at injection site,(170)
hyperbilirubinemia, and anorexia
AZD5153BRD4R/R malignant solid tumor andI- TEAEs (Grade ≥3) were thrombocytopenia, fatigue, anemia, diarrhea,(176)
lymphoma and platelet count decreased.
- DLTs were thrombocytopenia and diarrhea with herpetic rash
BMS986158BRD4Advanced cancerI/IIa- TEAEs (Grade 3–4) were diarrhea, thrombocytopenia, fatigue, nausea,(177)
anemia, and vomiting
- DLT was Grade 4 thrombocytopenia
GSK3358699Pan-BETHealthy subjectsN/A- Headache, non-sustained ventricular tachycardia, tachycardia, ventricular(178)
extrasystoles, and atrial fibrillation

[i] R/R, relapsed/refractory; AML, acute myeloid leukemia; ALL, acute lymphoblastic leukemia; MDS, myelodysplastic syndrome; NHL, non-Hodgkin's lymphoma; NSCLC, non-small cell lung cancer; CRPC, castration-resistant prostate cancer; RA, refractory anemia; CRC, colorectal cancer; NUT, Nuclear protein of the testis; NMC, NUT midline carcinoma; ER + BC, estrogen receptor-positive breast cancer, Head and neck squamous cell carcinoma; MF, Myelofibrosis; NC, nuclear protein of the testis carcinoma; DLBCL, diffuse large B-cell lymphoma; TEAEs, Treatment emergent adverse events; DLTs, Dose-limiting toxicities.

Toxicity of BET inhibition

BET inhibitors have been tested and assessed in both in vivo models and human clinical trials for their safety and efficacy in cancer therapy. As BET family proteins play critical roles in regulating multiple cellular functions, it is expected that BET inhibition would have adverse effects. These side effects have been observed in various occasions in animal models following the tests to the animals, as listed in Table II. Additionally, the widely used in vitro inhibitor JQ1 has failed to advance to human clinical trial due to its poor pharmacokinetic profile (34). Another adverse effect which has been observed in the animal models is thrombocytopenia (155). Consistently, a systematic review of various BET inhibitors administered to treat hematological malignancies and solid tumors indicated that all BET inhibitor leads to exposure-dependent thrombocytopenia (43). Thus, the issue of adverse effects should be closely monitored for patients receiving BET inhibitor as a part of their cancer therapy courses particularly for those with HNSCC which could be affected by toxicities to the surrounding organs in the head and neck region.

Table II.

Summary of the results from pre-clinical models inhibiting BRD4 using three different BET inhibitors, JQ1, I-BET-151 and ABBV-075.

Table II.

Summary of the results from pre-clinical models inhibiting BRD4 using three different BET inhibitors, JQ1, I-BET-151 and ABBV-075.

BET inhibitorBET targetsDiseaseModelToxicity in clinical or model studies
JQ1BRD2/3/4Acute myeloid leukemia (143)Mouse modelModel studies
Triple-negative breast cancer (148)Patient-derived xenografts- Dosage-dependent toxicity and long-term JQ1 treatment
Multiple myeloma (180)Mouse xenograft modelhave been shown to affect resistant cells (143)
Normal cells and mesenchymal stemIn vitroClinical studies
cells: Neuronal derivatives (181) - JQ1 has not been tested in clinical trials due to its poor pharmacokinetic profile,
Pancreatic cancer (182)In vitro and in vivolow oral bioavailability, and the need for the drug to be administered twice per day (34)
- High doses of JQ1 have been associated with potential toxicity (179)
I-BET-151BRD2/3/4/9CardiacIn vivo (mouse and rat)- I-BET-151 exhibits a dose-dependent reduction in the respiratory activity of
cardiac mitochondria, related cardiotoxicity (183)
- Cytotoxicity arises from disrupting fundamental cell processes such as cell growth
and the progression of the cell cycle that occurs in all dividing cells (184)
ABBV-075BRD4Prostate cancerRat modelModel studies
- Reduction in platelets and loss of goblet cells (155)

[i] BRD4, bromodomain-containing protein 4; BET, bromodomain and extra-terminal.

In order to limit the toxicity of BETi, an alternative approach for precise delivery of BRD4 inhibitors or other targeting molecules is engineered exosome. The idea of exploiting exosome as a drug delivery system has become popular as it can surpass barrier created by tumor microenvironment and can be equipped with targeting properties. Small molecule drugs such as paclitaxel and curcumin have been delivered to specific target cells (156). Delivery of microRNAs targeting BRD4 could perhaps further alleviate the adverse effects shown by several BETi as the formation and delivery of exosomal microRNAs is becoming more practical (157). The precision in drug delivery will surely offer a more endurable therapy for the patients.

Future perspective

A number of BET inhibitors have shown great potential to be effective for cancer therapy which could enhance the efficacy of chemo-, radio- and immunotherapy against HNSCC. As a chromatin-targeted therapy, BET/BRD4 inhibitor could be a viable candidate for replacing the EGFR inhibitor knowing that it could be effective against HNSCC regardless of its HPV association, as cetuximab may not provide the best outcome for the HPV-associated subtype of head and neck cancer. Another potential role which targeting BRD4 may come into play is the de-escalation of the current HNSCC therapy regimens which are facing a challenge in terms of the side effects. This could allow lesser adverse effects to the patients which typically affect the patients' quality of life. A clinical trial proving the efficacy of BRD4/BET inhibitor for the treatment of HNSCC is also desirable in order to demonstrate its clinical application in addition to its potential shown in vitro and in vivo models. In addition, the combination between BRD4 and other inhibitors should be considered. For example, BRD4 inhibitor in combination with suberoylanilide hydroxamic acid as a histone deacetylase inhibitor have been tested and exhibited promising results in certain tumors (158,159). This approach could further expand therapeutic options but may also need to proceed with caution due to adverse effects of such inhibitions. The investigation concerning immunological effects of BET inhibition should also be considered to evaluate the applicability of targeting BRD4 in head and neck cancer therapy.

Acknowledgements

Not applicable.

Funding

The present study was supported by Chulabhorn Royal Academy (grant no. ISF06-001/2566).

Availability of data and materials

Not applicable.

Authors' contributions

NW, WK, NA and TK wrote the first draft of the manuscript. VY revised the manuscript and generated all figures and tables. DN conceptualized the study and critically revised the manuscript. MT provided guidance and edited the manuscript prior to submission. All authors read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

References

1 

Johnson DE, Burtness B, Leemans CR, Lui VWY, Bauman JE and Grandis JR: Head and neck squamous cell carcinoma. Nat Rev Dis Primers. 6:922020. View Article : Google Scholar : PubMed/NCBI

2 

Marur S, D'Souza G, Westra WH and Forastiere AA: HPV-associated head and neck cancer: A virus-related cancer epidemic. Lancet Oncol. 11:781–789. 2010. View Article : Google Scholar

3 

Cipriano A, Milite C, Feoli A, Viviano M, Pepe G, Campiglia P, Sarno G, Picaud S, Imaide S, Makukhin N, et al: Discovery of Benzo[d]imidazole-6-sulfonamides as Bromodomain and Extra-Terminal Domain (BET) Inhibitors with Selectivity for the First Bromodomain. ChemMedChem. 17:e2022003432022. View Article : Google Scholar : PubMed/NCBI

4 

Liang Y, Tian J and Wu T: BRD4 in physiology and pathology: ‘BET’ on its partners. Bioessays. 43:e21001802021. View Article : Google Scholar : PubMed/NCBI

5 

Liu B, Liu X, Han L, Chen X, Wu X, Wu J, Yan D, Wang Y, Liu S, Shan L, et al: BRD4-directed super-enhancer organization of transcription repression programs links to chemotherapeutic efficacy in breast cancer. Proc Natl Acad Sci USA. 119:e21091331192022. View Article : Google Scholar : PubMed/NCBI

6 

Sakamaki JI, Wilkinson S, Hahn M, Tasdemir N, O'Prey J, Clark W, Hedley A, Nixon C, Long JS, New M, et al: Bromodomain protein BRD4 is a transcriptional repressor of autophagy and lysosomal function. Mol Cell. 66:517–532.e9. 2017. View Article : Google Scholar

7 

Hu J, Pan D, Li G, Chen K and Hu X: Regulation of programmed cell death by Brd4. Cell Death Dis. 13:10592022. View Article : Google Scholar : PubMed/NCBI

8 

Liu N, Ling R, Tang X, Yu Y, Zhou Y and Chen D: Post-Translational modifications of BRD4: Therapeutic targets for tumor. Front Oncol. 12:8477012022. View Article : Google Scholar

9 

Abedin SM, Boddy CS and Munshi HG: BET inhibitors in the treatment of hematologic malignancies: Current insights and future prospects. Onco Targets Ther. 9:5943–5953. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Lu T, Lu W and Luo C: A patent review of BRD4 inhibitors (2013–2019). Expert Opin Ther Pat. 30:57–81. 2020. View Article : Google Scholar

11 

French CA: NUT Carcinoma: Clinicopathologic features, pathogenesis, and treatment. Pathol Int. 68:583–595. 2018. View Article : Google Scholar

12 

Dey A, Yang W, Gegonne A, Nishiyama A, Pan R, Yagi R, Grinberg A, Finkelman FD, Pfeifer K, Zhu J, et al: BRD4 directs hematopoietic stem cell development and modulates macrophage inflammatory responses. EMBO J. 38:e1002932019. View Article : Google Scholar : PubMed/NCBI

13 

Houzelstein D, Bullock SL, Lynch DE, Grigorieva EF, Wilson VA and Beddington RS: Growth and early postimplantation defects in mice deficient for the bromodomain-containing protein Brd4. Mol Cell Biol. 22:3794–3802. 2002. View Article : Google Scholar

14 

Gonzales-Cope M, Sidoli S, Bhanu NV, Won KJ and Garcia BA: Histone H4 acetylation and the epigenetic reader Brd4 are critical regulators of pluripotency in embryonic stem cells. BMC Genomics. 17:952016. View Article : Google Scholar : PubMed/NCBI

15 

Devaiah BN, Case-Borden C, Gegonne A, Hsu CH, Chen Q, Meerzaman D, Dey A, Ozato K and Singer DS: BRD4 is a histone acetyltransferase that evicts nucleosomes from chromatin. Nat Struct Mol Biol. 23:540–548. 2016. View Article : Google Scholar

16 

Devaiah BN, Gegonne A and Singer DS: Bromodomain 4: A cellular swiss army knife. J Leukoc Biol. 100:679–686. 2016. View Article : Google Scholar

17 

Devaiah BN, Lewis BA, Cherman N, Hewitt MC, Albrecht BK, Robey PG, Ozato K, Sims RJ III and Singer DS: BRD4 is an atypical kinase that phosphorylates serine2 of the RNA polymerase II carboxy-terminal domain. Proc Natl Acad Sci USA. 109:6927–6932. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Jha RK, Levens D and Kouzine F: Mechanical determinants of chromatin topology and gene expression. Nucleus. 13:94–115. 2022. View Article : Google Scholar : PubMed/NCBI

19 

Kanno T, Kanno Y, LeRoy G, Campos E, Sun HW, Brooks SR, Vahedi G, Heightman TD, Garcia BA, Reinberg D, et al: BRD4 assists elongation of both coding and enhancer RNAs by interacting with acetylated histones. Nat Struct Mol Biol. 21:1047–1057. 2014. View Article : Google Scholar

20 

Farina A, Hattori M, Qin J, Nakatani Y, Minato N and Ozato K: Bromodomain protein Brd4 binds to GTPase-activating SPA-1, modulating its activity and subcellular localization. Mol Cell Biol. 24:9059–9069. 2004. View Article : Google Scholar

21 

You J, Li Q, Wu C, Kim J, Ottinger M and Howley PM: Regulation of aurora B expression by the bromodomain protein Brd4. Mol Cell Biol. 29:5094–5103. 2009. View Article : Google Scholar

22 

Wang R, Cao XJ, Kulej K, Liu W, Ma T, MacDonald M, Chiang CM, Garcia BA and You J: Uncovering BRD4 hyperphosphorylation associated with cellular transformation in NUT midline carcinoma. Proc Natl Acad Sci USA. 114:E5352–E5361. 2017.PubMed/NCBI

23 

Wu Y, Wang Y, Diao P, Zhang W, Li J, Ge H, Song Y, Li Z, Wang D, Liu L, et al: Therapeutic targeting of BRD4 in head neck squamous cell carcinoma. Theranostics. 9:1777–1793. 2019. View Article : Google Scholar : PubMed/NCBI

24 

Wu SY, Lee CF, Lai HT, Yu CT, Lee JE, Zuo H, Tsai SY, Tsai MJ, Ge K, Wan Y, et al: Opposing functions of BRD4 isoforms in breast cancer. Mol Cell. 78:1114–1132.e10. 2020. View Article : Google Scholar

25 

Drumond-Bock AL and Bieniasz M: The role of distinct BRD4 isoforms and their contribution to high-grade serous ovarian carcinoma pathogenesis. Mol Cancer. 20:1452021. View Article : Google Scholar : PubMed/NCBI

26 

White ME, Fenger JM and Carson WE III: Emerging roles of and therapeutic strategies targeting BRD4 in cancer. Cell Immunol. 337:48–53. 2019. View Article : Google Scholar

27 

Donati B, Lorenzini E and Ciarrocchi A: BRD4 and Cancer: Going beyond transcriptional regulation. Mol Cancer. 17:1642018. View Article : Google Scholar : PubMed/NCBI

28 

Shu S, Lin CY, He HH, Witwicki RM, Tabassum DP, Roberts JM, Janiszewska M, Huh SJ, Liang Y, Ryan J, et al: Response and resistance to BET bromodomain inhibitors in triple-negative breast cancer. Nature. 529:413–417. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Hamad M, Ali A and Muhammad JS: BRD4 regulates the induction and maintenance of cancer stem cells in squamous cell carcinoma. Stem Cell Investig. 9:62022. View Article : Google Scholar : PubMed/NCBI

30 

Pagliarini R, Shao W and Sellers WR: Oncogene addiction: Pathways of therapeutic response, resistance, and road maps toward a cure. EMBO Rep. 16:280–296. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Shi J, Wang Y, Zeng L, Wu Y, Deng J, Zhang Q, Lin Y, Li J, Kang T, Tao M, et al: Disrupting the interaction of BRD4 with diacetylated Twist suppresses tumorigenesis in basal-like breast cancer. Cancer Cell. 25:210–225. 2014. View Article : Google Scholar

32 

Andrieu G, Tran AH, Strissel KJ and Denis GV: BRD4 regulates breast cancer dissemination through Jagged1/Notch1 signaling. Cancer Res. 76:6555–6567. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Samani K, Raj Sharma U, Raj Sharma A, Pm M and V S: Role of BRD4 in cancer-A review. J Diagnostic Pathol Oncolo. 5:128–134. 2020. View Article : Google Scholar

34 

Shorstova T, Foulkes WD and Witcher M: Achieving clinical success with BET inhibitors as anti-cancer agents. Br J Cancer. 124:1478–1490. 2021. View Article : Google Scholar : PubMed/NCBI

35 

Tan Y, Wang L, Du Y, Liu X, Chen Z, Weng X, Guo J, Chen H, Wang M and Wang X: Inhibition of BRD4 suppresses tumor growth in prostate cancer via the enhancement of FOXO1 expression. Int J Oncol. 53:2503–2517. 2018.

36 

Wang J, Quan Y, Lv J, Gong S and Dong D: BRD4 promotes glioma cell stemness via enhancing miR-142-5p-mediated activation of Wnt/β-catenin signaling. Environ Toxicol. 35:368–376. 2020. View Article : Google Scholar

37 

Nantajit D, Presta L, Sauter T and Tavassoli M: EGFR-induced suppression of HPV E6/E7 is mediated by microRNA-9-5p silencing of BRD4 protein in HPV-positive head and neck squamous cell carcinoma. Cell Death Dis. 13:9212022. View Article : Google Scholar : PubMed/NCBI

38 

Schmitt A, Grimm M, Kreienkamp N, Junge H, Labisch J, Schuhknecht L, Schonfeld C, Gorsch ES, Tibello A, Menck K, et al: BRD4 inhibition sensitizes diffuse large B-cell lymphoma cells to ferroptosis. Blood. 142:1143–1155. 2023. View Article : Google Scholar : PubMed/NCBI

39 

Stathis A, Zucca E, Bekradda M, Gomez-Roca C, Delord JP, de La Motte Rouge T, Uro-Coste E, de Braud F, Pelosi G and French CA: Clinical response of carcinomas harboring the BRD4-NUT oncoprotein to the targeted bromodomain inhibitor OTX015/MK-8628. Cancer Discov. 6:492–500. 2016. View Article : Google Scholar : PubMed/NCBI

40 

Zhang X, Guo X, Zhuo R, Tao Y, Liang W, Yang R, Chen Y, Cao H, Jia S, Yu J, et al: BRD4 inhibitor MZ1 exerts anti-cancer effects by targeting MYCN and MAPK signaling in neuroblastoma. Biochem Biophys Res Commun. 604:63–69. 2022. View Article : Google Scholar : PubMed/NCBI

41 

Zong D, Gu J, Cavalcante GC, Yao W, Zhang G, Wang S, Owonikoko TK, He X and Sun SY: BRD4 levels determine the response of human lung cancer cells to BET degraders that potently induce apoptosis through suppression of Mcl-1. Cancer Res. 80:2380–2393. 2020. View Article : Google Scholar : PubMed/NCBI

42 

Bauer K, Berghoff AS, Preusser M, Heller G, Zielinski CC, Valent P and Grunt TW: Degradation of BRD4-a promising treatment approach not only for hematologic but also for solid cancer. Am J Cancer Res. 11:530–545. 2021.PubMed/NCBI

43 

Sun Y, Han J, Wang Z, Li X, Sun Y and Hu Z: Safety and efficacy of bromodomain and Extra-Terminal inhibitors for the treatment of hematological malignancies and solid tumors: A systematic study of clinical trials. Front Pharmacol. 11:6210932020. View Article : Google Scholar

44 

Floyd SR, Pacold ME, Huang Q, Clarke SM, Lam FC, Cannell IG, Bryson BD, Rameseder J, Lee MJ, Blake EJ, et al: The bromodomain protein Brd4 insulates chromatin from DNA damage signalling. Nature. 498:246–250. 2013. View Article : Google Scholar : PubMed/NCBI

45 

Ni M, Li J, Zhao H, Xu F, Cheng J, Yu M, Ke G and Wu X: BRD4 inhibition sensitizes cervical cancer to radiotherapy by attenuating DNA repair. Oncogene. 40:2711–2724. 2021. View Article : Google Scholar : PubMed/NCBI

46 

Lam FC, Kong YW, Huang Q, Vu Han TL, Maffa AD, Kasper EM and Yaffe MB: BRD4 prevents the accumulation of R-loops and protects against transcription-replication collision events and DNA damage. Nat Commun. 11:40832020. View Article : Google Scholar : PubMed/NCBI

47 

Li X, Baek G, Ramanand SG, Sharp A, Gao Y, Yuan W, Welti J, Rodrigues DN, Dolling D, Figueiredo I, et al: BRD4 promotes DNA repair and mediates the formation of TMPRSS2-ERG gene rearrangements in prostate cancer. Cell Rep. 22:796–808. 2018. View Article : Google Scholar : PubMed/NCBI

48 

Sun C, Yin J, Fang Y, Chen J, Jeong KJ, Chen X, Vellano CP, Ju Z, Zhao W, Zhang D, et al: BRD4 inhibition is synthetic lethal with PARP inhibitors through the induction of homologous recombination deficiency. Cancer Cell. 33:401–416.e8. 2018. View Article : Google Scholar

49 

Barrows JK, Lin B, Quaas CE, Fullbright G, Wallace EN and Long DT: BRD4 promotes resection and homology-directed repair of DNA double-strand breaks. Nat Commun. 13:30162022. View Article : Google Scholar : PubMed/NCBI

50 

Takashima Y, Kikuchi E, Kikuchi J, Suzuki M, Kikuchi H, Maeda M, Shoji T, Furuta M, Kinoshita I, Dosaka-Akita H, et al: Bromodomain and extraterminal domain inhibition synergizes with WEE1-inhibitor AZD1775 effect by impairing nonhomologous end joining and enhancing DNA damage in nonsmall cell lung cancer. Int J Cancer. 146:1114–1124. 2020. View Article : Google Scholar : PubMed/NCBI

51 

Tan YF, Wang M, Chen ZY, Wang L and Liu XH: Inhibition of BRD4 prevents proliferation and epithelial-mesenchymal transition in renal cell carcinoma via NLRP3 inflammasome-induced pyroptosis. Cell Death Dis. 11:2392020. View Article : Google Scholar : PubMed/NCBI

52 

Miller AL, Fehling SC, Garcia PL, Gamblin TL, Council LN, van Waardenburg R, Yang ES, Bradner JE and Yoon KJ: The BET inhibitor JQ1 attenuates double-strand break repair and sensitizes models of pancreatic ductal adenocarcinoma to PARP inhibitors. EBioMedicine. 44:419–430. 2019. View Article : Google Scholar : PubMed/NCBI

53 

He DD, Shang XY, Wang N, Wang GX, He KY, Wang L and Han ZG: BRD4 inhibition induces synthetic lethality in ARID2-deficient hepatocellular carcinoma by increasing DNA damage. Oncogene. 41:1397–1409. 2022. View Article : Google Scholar : PubMed/NCBI

54 

Li L, Gao L, Zhou H, Shi C, Zhang X, Zhang D and Liu H: High expression level of BRD4 is associated with a poor prognosis and immune infiltration in esophageal squamous cell carcinoma. Dig Dis Sci. 68:2997–3008. 2023. View Article : Google Scholar

55 

Liao YF, Wu YB, Long X, Zhu SQ, Jin C, Xu JJ and Ding JY: High level of BRD4 promotes non-small cell lung cancer progression. Oncotarget. 7:9491–9500. 2016. View Article : Google Scholar

56 

Burcher KM, Faucheux AT, Lantz JW, Wilson HL, Abreu A, Salafian K, Patel MJ, Song AH, Petro RM, Lycan T Jr, et al: Prevalence of DNA repair gene mutations in blood and tumor tissue and impact on prognosis and treatment in HNSCC. Cancers (Basel). 13:31182021. View Article : Google Scholar : PubMed/NCBI

57 

Moeller BJ, Yordy JS, Williams MD, Giri U, Raju U, Molkentine DP, Byers LA, Heymach JV, Story MD, Lee JJ, et al: DNA repair biomarker profiling of head and neck cancer: Ku80 expression predicts locoregional failure and death following radiotherapy. Clin Cancer Res. 17:2035–2043. 2011. View Article : Google Scholar : PubMed/NCBI

58 

Mahjabeen I, Ali K, Zhou X and Kayani MA: Deregulation of base excision repair gene expression and enhanced proliferation in head and neck squamous cell carcinoma. Tumour Biol. 35:5971–5983. 2014. View Article : Google Scholar

59 

Bold IT, Specht AK, Droste CF, Zielinski A, Meyer F, Clauditz TS, Munscher A, Werner S, Rothkamm K, Petersen C, et al: DNA damage response during replication correlates with CIN70 score and determines survival in HNSCC patients. Cancers (Basel). 13:11942021. View Article : Google Scholar : PubMed/NCBI

60 

Zhang S, Lai Y, Pan J, Saeed M, Li S, Zhou H, Jiang X, Gao J, Zhu Y, Yu H, et al: PROTAC Prodrug-Integrated nanosensitizer for potentiating radiation therapy of cancer. Adv Mater. e23141322024.doi: 10.1002/adma.202314132 (Epub ahead of print). View Article : Google Scholar

61 

Wang J, Wang Y, Mei H, Yin Z, Geng Y, Zhang T, Wu G and Lin Z: The BET bromodomain inhibitor JQ1 radiosensitizes non-small cell lung cancer cells by upregulating p21. Cancer Lett. 391:141–151. 2017. View Article : Google Scholar

62 

Garcia PL, Miller AL, Zeng L, van Waardenburg R, Yang ES and Yoon KJ: The BET inhibitor JQ1 potentiates the anticlonogenic effect of radiation in pancreatic cancer Cells. Front Oncol. 12:9257182022. View Article : Google Scholar

63 

Kim S, Jeon SH, Han MG, Kang MH and Kim IA: BRD4 inhibition enhances the antitumor effects of radiation therapy in a murine breast cancer model. Int J Mol Sci. 24:130622023. View Article : Google Scholar

64 

Santos-de-Frutos K, Segrelles C and Lorz C: Hippo pathway and YAP signaling alterations in squamous cancer of the head and neck. J Clin Med. 8:21312019. View Article : Google Scholar : PubMed/NCBI

65 

Choi SK, Hong SH, Kim HS, Shin CY, Nam SW, Choi WS, Han JW and You JS: JQ1, an inhibitor of the epigenetic reader BRD4, suppresses the bidirectional MYC-AP4 axis via multiple mechanisms. Oncol Rep. 35:1186–1194. 2016. View Article : Google Scholar : PubMed/NCBI

66 

Hong SH, Eun JW, Choi SK, Shen Q, Choi WS, Han JW, Nam SW and You JS: Epigenetic reader BRD4 inhibition as a therapeutic strategy to suppress E2F2-cell cycle regulation circuit in liver cancer. Oncotarget. 7:32628–32640. 2016. View Article : Google Scholar

67 

Zheng B, Gold S, Iwanaszko M, Howard BC, Wang L and Shilatifard A: Distinct layers of BRD4-PTEFb reveal bromodomain-independent function in transcriptional regulation. Mol Cell. 83:2896–2910.e4. 2023. View Article : Google Scholar

68 

Itzen F, Greifenberg AK, Bosken CA and Geyer M: Brd4 activates P-TEFb for RNA polymerase II CTD phosphorylation. Nucleic Acids Res. 42:7577–7590. 2014. View Article : Google Scholar : PubMed/NCBI

69 

He Y, Ju Y, Hu Y, Wang B, Che S, Jian Y, Zhuo W, Fu X, Cheng Y, Zheng S, et al: Brd4 proteolysis-targeting chimera nanoparticles sensitized colorectal cancer chemotherapy. J Control Release. 354:155–166. 2023. View Article : Google Scholar : PubMed/NCBI

70 

Liu T, Zhang Z, Wang C, Huang H and Li Y: BRD4 promotes the migration and invasion of bladder cancer cells through the Sonic hedgehog signaling pathway and enhances cisplatin resistance. Biochem Cell Biol. 100:179–187. 2022. View Article : Google Scholar

71 

Lu L, Chen Z, Lin X, Tian L, Su Q, An P, Li W, Wu Y, Du J, Shan H, et al: Inhibition of BRD4 suppresses the malignancy of breast cancer cells via regulation of Snail. Cell Death Differ. 27:255–268. 2020. View Article : Google Scholar : PubMed/NCBI

72 

Shafran JS, Jafari N, Casey AN, Gyorffy B and Denis GV: BRD4 regulates key transcription factors that drive epithelial-mesenchymal transition in castration-resistant prostate cancer. Prostate Cancer Prostatic Dis. 24:268–277. 2021. View Article : Google Scholar : PubMed/NCBI

73 

Shi J, Cao J and Zhou BP: Twist-BRD4 complex: Potential drug target for basal-like breast cancer. Curr Pharm Des. 21:1256–1261. 2015. View Article : Google Scholar : PubMed/NCBI

74 

Zhang P, Dong Z, Cai J, Zhang C, Shen Z, Ke A, Gao D, Fan J and Shi G: BRD4 promotes tumor growth and epithelial-mesenchymal transition in hepatocellular carcinoma. Int J Immunopathol Pharmacol. 28:36–44. 2015. View Article : Google Scholar

75 

Andrieu GP and Denis GV: BET proteins exhibit transcriptional and functional opposition in the Epithelial-to-Mesenchymal transition. Mol Cancer Res. 16:580–586. 2018. View Article : Google Scholar : PubMed/NCBI

76 

Tian B, Zhao Y, Sun H, Zhang Y, Yang J and Brasier AR: BRD4 mediates NF-κB-dependent epithelial-mesenchymal transition and pulmonary fibrosis via transcriptional elongation. Am J Physiol Lung Cell Mol Physiol. 311:L1183–L1201. 2016. View Article : Google Scholar

77 

Cho HY, Lee SW, Jeon YH, Lee DH, Kim GW, Yoo J, Kim SY and Kwon SH: Combination of ACY-241 and JQ1 synergistically suppresses metastasis of HNSCC via regulation of MMP-2 and MMP-9. Int J Mol Sci. 21:68732020. View Article : Google Scholar

78 

Hu Y, Zhou J, Ye F, Xiong H, Peng L, Zheng Z, Xu F, Cui M, Wei C, Wang X, et al: BRD4 inhibitor inhibits colorectal cancer growth and metastasis. Int J Mol Sci. 16:1928–1948. 2015. View Article : Google Scholar

79 

Wang L, Wu X, Wang R, Yang C, Li Z, Wang C, Zhang F and Yang P: BRD4 inhibition suppresses cell growth, migration and invasion of salivary adenoid cystic carcinoma. Biol Res. 50:192017. View Article : Google Scholar : PubMed/NCBI

80 

Yamamoto T, Hirosue A, Nakamoto M, Yoshida R, Sakata J, Matsuoka Y, Kawahara K, Nagao Y, Nagata M, Takahashi N, et al: BRD4 promotes metastatic potential in oral squamous cell carcinoma through the epigenetic regulation of the MMP2 gene. Br J Cancer. 123:580–590. 2020. View Article : Google Scholar : PubMed/NCBI

81 

Griso AB, Acero-Riaguas L, Castelo B, Cebrian-Carretero JL and Sastre-Perona A: Mechanisms of cisplatin resistance in HPV negative head and neck squamous cell carcinomas. Cells. 11:5612022. View Article : Google Scholar : PubMed/NCBI

82 

Bonomi M, Patsias A, Posner M and Sikora A: The role of inflammation in head and neck cancer. Adv Exp Med Biol. 816:107–127. 2014. View Article : Google Scholar

83 

Gong Z, Liu G, Liu W, Zou H, Song R, Zhao H, Yuan Y, Gu J, Bian J, Zhu J, et al: The epigenetic regulator BRD4 is involved in cadmium-triggered inflammatory response in rat kidney. Ecotoxicol Environ Saf. 224:1126202021. View Article : Google Scholar : PubMed/NCBI

84 

Bao Y, Wu X, Chen J, Hu X, Zeng F, Cheng J, Jin H, Lin X and Chen LF: Brd4 modulates the innate immune response through Mnk2-eIF4E pathway-dependent translational control of IκBα. Proc Natl Acad Sci USA. 114:E3993–E4001. 2017. View Article : Google Scholar : PubMed/NCBI

85 

Xu Y and Vakoc CR: Brd4 is on the move during inflammation. Trends Cell Biol. 24:615–616. 2014. View Article : Google Scholar

86 

Jarausch J, Neuenroth L, Andag R, Leha A, Fischer A, Asif AR, Lenz C and Eidizadeh A: Influence of shear stress, inflammation and BRD4 inhibition on human endothelial cells: A holistic proteomic approach. Cells. 11:30862022. View Article : Google Scholar : PubMed/NCBI

87 

Chen L, Zhong X, Cao W, Mao M, Li W, Yang H, Li M, Shi M, Zhang Y, Deng Y, et al: JQ1 as a BRD4 inhibitor blocks inflammatory pyroptosis-related acute colon injury induced by LPS. Front Immunol. 12:6093192021. View Article : Google Scholar

88 

Nicodeme E, Jeffrey KL, Schaefer U, Beinke S, Dewell S, Chung CW, Chandwani R, Marazzi I, Wilson P, Coste H, et al: Suppression of inflammation by a synthetic histone mimic. Nature. 468:1119–1123. 2010. View Article : Google Scholar : PubMed/NCBI

89 

Khan YM, Kirkham P, Barnes PJ and Adcock IM: Brd4 is essential for IL-1β-induced inflammation in human airway epithelial cells. PLoS One. 9:e950512014. View Article : Google Scholar : PubMed/NCBI

90 

Jing SL, Afshari K and Guo ZC: Inflammatory response-related genes predict prognosis in patients with HNSCC. Immunol Lett. 259:46–60. 2023. View Article : Google Scholar

91 

Rassouli A, Saliba J, Castano R, Hier M and Zeitouni AG: Systemic inflammatory markers as independent prognosticators of head and neck squamous cell carcinoma. Head Neck. 37:103–110. 2015. View Article : Google Scholar : PubMed/NCBI

92 

Zhou S, Yuan H, Wang J, Hu X, Liu F, Zhang Y, Jiang B and Zhang W: Prognostic value of systemic inflammatory marker in patients with head and neck squamous cell carcinoma undergoing surgical resection. Future Oncol. 16:559–571. 2020. View Article : Google Scholar

93 

Charles KA, Harris BD, Haddad CR, Clarke SJ, Guminski A, Stevens M, Dodds T, Gill AJ, Back M, Veivers D, et al: Systemic inflammation is an independent predictive marker of clinical outcomes in mucosal squamous cell carcinoma of the head and neck in oropharyngeal and non-oropharyngeal patients. BMC Cancer. 16:1242016. View Article : Google Scholar : PubMed/NCBI

94 

Roman BR and Aragones A: Epidemiology and incidence of HPV-related cancers of the head and neck. J Surg Oncol. 124:920–922. 2021. View Article : Google Scholar

95 

Serrano B, Brotons M, Bosch FX and Bruni L: Epidemiology and burden of HPV-related disease. Best Pract Res Clin Obstet Gynaecol. 47:14–26. 2018. View Article : Google Scholar

96 

Betiol J, Villa LL and Sichero L: Impact of HPV infection on the development of head and neck cancer. Braz J Med Biol Res. 46:217–226. 2013. View Article : Google Scholar : PubMed/NCBI

97 

de Martel C, Georges D, Bray F, Ferlay J and Clifford GM: Global burden of cancer attributable to infections in 2018: A worldwide incidence analysis. Lancet Glob Health. 8:e180–e190. 2020. View Article : Google Scholar : PubMed/NCBI

98 

Kajitani N, Satsuka A, Kawate A and Sakai H: Productive lifecycle of human papillomaviruses that depends upon squamous epithelial differentiation. Front Microbiol. 3:1522012. View Article : Google Scholar

99 

Cricca M, Venturoli S, Leo E, Costa S, Musiani M and Zerbini M: Disruption of HPV 16 E1 and E2 genes in precancerous cervical lesions. J Virol Methods. 158:180–183. 2009. View Article : Google Scholar : PubMed/NCBI

100 

Pal A and Kundu R: Human Papillomavirus E6 and E7: The cervical cancer hallmarks and targets for therapy. Front Microbiol. 10:31162019. View Article : Google Scholar

101 

Yu L, Majerciak V and Zheng ZM: HPV16 and HPV18 genome structure, expression, and Post-Transcriptional regulation. Int J Mol Sci. 23:49432022. View Article : Google Scholar

102 

Helfer CM, Yan J and You J: The cellular bromodomain protein Brd4 has multiple functions in E2-mediated papillomavirus transcription activation. Viruses. 6:3228–3249. 2014. View Article : Google Scholar : PubMed/NCBI

103 

McBride AA, McPhillips MG and Oliveira JG: Brd4: Tethering, segregation and beyond. Trends Microbiol. 12:527–529. 2004. View Article : Google Scholar

104 

Jang MK, Shen K and McBride AA: Papillomavirus genomes associate with BRD4 to replicate at fragile sites in the host genome. PLoS Pathog. 10:e10041172014. View Article : Google Scholar : PubMed/NCBI

105 

McBride AA and Jang MK: Current understanding of the role of the Brd4 protein in the papillomavirus lifecycle. Viruses. 5:1374–1394. 2013. View Article : Google Scholar : PubMed/NCBI

106 

McKinney CC, Kim MJ, Chen D and McBride AA: Brd4 activates early viral transcription upon human papillomavirus 18 infection of primary keratinocytes. mBio. 7:e01644–16. 2016. View Article : Google Scholar : PubMed/NCBI

107 

Iftner T, Haedicke-Jarboui J, Wu SY and Chiang CM: Involvement of Brd4 in different steps of the papillomavirus life cycle. Virus Res. 231:76–82. 2017. View Article : Google Scholar : PubMed/NCBI

108 

Morse MA, Balogh KK, Brendle SA, Campbell CA, Chen MX, Furze RC, Harada IL, Holyer ID, Kumar U, Lee K, et al: BET bromodomain inhibitors show anti-papillomavirus activity in vitro and block CRPV wart growth in vivo. Antiviral Res. 154:158–165. 2018. View Article : Google Scholar : PubMed/NCBI

109 

Wang J, Li GL, Ming SL, Wang CF, Shi LJ, Su BQ, Wu HT, Zeng L, Han YQ, Liu ZH, et al: BRD4 inhibition exerts anti-viral activity through DNA damage-dependent innate immune responses. PLoS Pathog. 16:e10084292020. View Article : Google Scholar : PubMed/NCBI

110 

Chen J, Wang Z, Phuc T, Xu Z, Yang D, Chen Z, Lin Z, Kendrick S, Dai L, Li HY, et al: Oncolytic strategy using new bifunctional HDACs/BRD4 inhibitors against virus-associated lymphomas. PLoS Pathog. 19:e10110892023. View Article : Google Scholar : PubMed/NCBI

111 

Cohen EEW, Bell RB, Bifulco CB, Burtness B, Gillison ML, Harrington KJ, Le QT, Lee NY, Leidner R, Lewis RL, et al: The Society for Immunotherapy of Cancer consensus statement on immunotherapy for the treatment of squamous cell carcinoma of the head and neck (HNSCC). J Immunother Cancer. 7:1842019. View Article : Google Scholar : PubMed/NCBI

112 

Oosting SF and Haddad RI: Best practice in systemic therapy for head and neck squamous cell carcinoma. Front Oncol. 9:8152019. View Article : Google Scholar

113 

Rosenberg AJ and Vokes EE: Optimizing treatment De-Escalation in head and neck cancer: Current and future perspectives. Oncologist. 26:40–48. 2021. View Article : Google Scholar

114 

Swain M, Kannan S, Srinivasan S, Agarwal JP and Gupta T: Concurrent Cetuximab-based bioradiotherapy versus Cisplatin-based Chemoradiotherapy in the Definitive Management of Favourable Biology Human Papillomavirus-associated Oropharyngeal Squamous Cell Carcinoma: Systematic Review and Meta-analysis. Clin Oncol (R Coll Radiol). 34:786–795. 2022. View Article : Google Scholar : PubMed/NCBI

115 

Lv XX, Zheng XY, Yu JJ, Ma HR, Hua C and Gao RT: EGFR enhances the stemness and progression of oral cancer through inhibiting autophagic degradation of SOX2. Cancer Med. 9:1131–1140. 2020. View Article : Google Scholar : PubMed/NCBI

116 

Beck TN, Georgopoulos R, Shagisultanova EI, Sarcu D, Handorf EA, Dubyk C, Lango MN, Ridge JA, Astsaturov I, Serebriiskii IG, et al: EGFR and RB1 as dual biomarkers in HPV-Negative head and neck cancer. Mol Cancer Ther. 15:2486–2497. 2016. View Article : Google Scholar : PubMed/NCBI

117 

Alsahafi EN, Thavaraj S, Sarvestani N, Novoplansky O, Elkabets M, Ayaz B, Tavassoli M and Legends MF: EGFR overexpression increases radiotherapy response in HPV-positive head and neck cancer through inhibition of DNA damage repair and HPV E6 downregulation. Cancer Lett. 498:80–97. 2021. View Article : Google Scholar

118 

Rieckmann T and Kriegs M: The failure of cetuximab-based de-intensified regimes for HPV-positive OPSCC: A radiobiologists perspective. Clin Transl Radiat Oncol. 17:47–50. 2019.

119 

Krishnamurthy S, Ahmed I, Bhise R, Mohanti BK, Sharma A, Rieckmann T, Paterson C and Bonomo P: The dogma of Cetuximab and Radiotherapy in head and neck cancer-A dawn to dusk journey. Clin Transl Radiat Oncol. 34:75–81. 2022.

120 

Xu K, Chen D, Qian D, Zhang S, Zhang Y, Guo S, Ma Z and Wang S: AZD5153, a novel BRD4 inhibitor, suppresses human thyroid carcinoma cell growth in vitro and in vivo. Biochem Biophys Res Commun. 499:531–537. 2018. View Article : Google Scholar : PubMed/NCBI

121 

Cortiguera MG, Batlle-López A, Albajar M, Delgado MD and León J: MYC as therapeutic target in leukemia and lymphoma. Blood and Lymphatic Cancer: Targets and Therapy. 5:75–91. 2015.

122 

Wang C, Zhang Y, Zhou D, Cao G and Wu Y: miR-204 enhances p27 mRNA stability by targeting Brd4 in head and neck squamous cell carcinoma. Oncol Lett. 16:4179–4184. 2018.

123 

Zhang W, Ge H, Jiang Y, Huang R, Wu Y, Wang D, Guo S, Li S, Wang Y, Jiang H, et al: Combinational therapeutic targeting of BRD4 and CDK7 synergistically induces anticancer effects in head and neck squamous cell carcinoma. Cancer Lett. 469:510–523. 2020. View Article : Google Scholar

124 

Webber LP, Yujra VQ, Vargas PA, Martins MD, Squarize CH and Castilho RM: Interference with the bromodomain epigenome readers drives p21 expression and tumor senescence. Cancer Lett. 461:10–20. 2019. View Article : Google Scholar

125 

Dong J, Li J, Li Y, Ma Z, Yu Y and Wang CY: Transcriptional super-enhancers control cancer stemness and metastasis genes in squamous cell carcinoma. Nat Commun. 12:39742021. View Article : Google Scholar : PubMed/NCBI

126 

Jing C, Liu D, Lai Q, Li L, Zhou M, Ye B, Wu Y, Li H, Yue K, Wu Y, et al: JOSD1 promotes proliferation and chemoresistance of head and neck squamous cell carcinoma under the epigenetic regulation of BRD4. Cancer Cell Int. 21:3752021. View Article : Google Scholar

127 

Chen N, Golczer G, Ghose S, Lin B, Langenbucher A, Webb J, Bhanot H, Abt NB, Lin D, Varvares M, et al: YAP1 maintains active chromatin state in head and neck squamous cell carcinomas that promotes tumorigenesis through cooperation with BRD4. Cell Rep. 39:1109702022. View Article : Google Scholar : PubMed/NCBI

128 

Leonard B, Brand TM, O'Keefe RA, Lee ED, Zeng Y, Kemmer JD, Li H, Grandis JR and Bhola NE: BET Inhibition overcomes receptor tyrosine Kinase-Mediated cetuximab resistance in HNSCC. Cancer Res. 78:4331–4343. 2018. View Article : Google Scholar : PubMed/NCBI

129 

Araujo TG, Mota STS, Ferreira HSV, Ribeiro MA, Goulart LR and Vecchi L: Annexin A1 as a regulator of immune response in cancer. Cells. 10:22452021. View Article : Google Scholar : PubMed/NCBI

130 

Zhong L, Yang Z, Lei D, Li L, Song S, Cao D and Liu Y: Bromodomain 4 is a potent prognostic marker associated with immune cell infiltration in breast cancer. Basic Clin Pharmacol Toxicol. 128:169–182. 2021. View Article : Google Scholar

131 

Chen YR, Ouyang SS, Chen YL, Li P, Xu HW and Zhu SL: BRD4/8/9 are prognostic biomarkers and associated with immune infiltrates in hepatocellular carcinoma. Aging (Albany NY). 12:17541–17567. 2020. View Article : Google Scholar : PubMed/NCBI

132 

Lee M, Tayyari F, Pinnaduwage D, Bayani J, Bartlett JMS, Mulligan AM, Bull SB and Andrulis IL: Tumoral BRD4 expression in lymph node-negative breast cancer: association with T-bet+ tumor-infiltrating lymphocytes and disease-free survival. BMC Cancer. 18:7502018. View Article : Google Scholar : PubMed/NCBI

133 

Zhao L, Li P, Zhao L, Wang M, Tong D, Meng Z, Zhang Q, Li Q and Zhang F: Expression and clinical value of PD-L1 which is regulated by BRD4 in tongue squamous cell carcinoma. J Cell Biochem. 121:1855–1869. 2020. View Article : Google Scholar : PubMed/NCBI

134 

Jing X, Shao S, Zhang Y, Luo A, Zhao L, Zhang L, Gu S and Zhao X: BRD4 inhibition suppresses PD-L1 expression in triple-negative breast cancer. Exp Cell Res. 392:1120342020. View Article : Google Scholar : PubMed/NCBI

135 

Ye Y, Zhong W, Qian J, Zhang J, Xu T, Han R, Han J, Wang C, Song L, Zeng X, et al: Comprehensive analysis of the prognosis and immune infiltrates for the BET protein family reveals the significance of BRD4 in glioblastoma multiforme. Front Cell Dev Biol. 11:10424902023. View Article : Google Scholar

136 

Bhola NE, Njatcha C, Hu L, Lee ED, Shiah JV, Kim MO, Johnson DE and Grandis JR: PD-L1 is upregulated via BRD2 in head and neck squamous cell carcinoma models of acquired cetuximab resistance. Head Neck. 43:3364–3373. 2021. View Article : Google Scholar : PubMed/NCBI

137 

Zhang M, Wang G, Ma Z, Xiong G, Wang W, Huang Z, Wan Y, Xu X, Hoyle RG, Yi C, et al: BET inhibition triggers antitumor immunity by enhancing MHC class I expression in head and neck squamous cell carcinoma. Mol Ther. 30:3394–3413. 2022. View Article : Google Scholar : PubMed/NCBI

138 

Suarez-Alvarez B, Morgado-Pascual JL, Rayego-Mateos S, Rodriguez RM, Rodrigues-Diez R, Cannata-Ortiz P, Sanz AB, Egido J, Tharaux PL, Ortiz A, et al: Inhibition of bromodomain and extraterminal domain family proteins ameliorates experimental renal damage. J Am Soc Nephrol. 28:504–519. 2017. View Article : Google Scholar

139 

Xia L, Liu JY, Zheng ZZ, Chen YJ, Ding JC, Hu YH, Hu GS, Xia NS and Liu W: BRD4 inhibition boosts the therapeutic effects of epidermal growth factor receptor-targeted chimeric antigen receptor T cells in glioblastoma. Mol Ther. 29:3011–3026. 2021. View Article : Google Scholar : PubMed/NCBI

140 

Joshi S, Singh AR, Liu KX, Pham TV, Zulcic M, Skola D, Chun HB, Glass CK, Morales GA, Garlich JR, et al: SF2523: Dual PI3K/BRD4 inhibitor blocks tumor immunosuppression and promotes adaptive immune responses in cancer. Mol Cancer Ther. 18:1036–1044. 2019. View Article : Google Scholar : PubMed/NCBI

141 

Li X, Fu Y, Yang B, Guo E, Wu Y, Huang J, Zhang X, Xiao R, Li K, Wang B, et al: BRD4 Inhibition by AZD5153 promotes antitumor immunity via depolarizing M2 macrophages. Front Immunol. 11:892020. View Article : Google Scholar

142 

Fong CY, Gilan O, Lam EY, Rubin AF, Ftouni S, Tyler D, Stanley K, Sinha D, Yeh P, Morison J, et al: BET inhibitor resistance emerges from leukaemia stem cells. Nature. 525:538–542. 2015. View Article : Google Scholar : PubMed/NCBI

143 

Rathert P, Roth M, Neumann T, Muerdter F, Roe JS, Muhar M, Deswal S, Cerny-Reiterer S, Peter B, Jude J, et al: Transcriptional plasticity promotes primary and acquired resistance to BET inhibition. Nature. 525:543–547. 2015. View Article : Google Scholar : PubMed/NCBI

144 

Calder J, Nagelberg A, Luu J, Lu D and Lockwood WW: Resistance to BET inhibitors in lung adenocarcinoma is mediated by casein kinase phosphorylation of BRD4. Oncogenesis. 10:272021. View Article : Google Scholar : PubMed/NCBI

145 

Jin X, Yan Y, Wang D, Ding D, Ma T, Ye Z, Jimenez R, Wang L, Wu H and Huang H: DUB3 promotes BET inhibitor resistance and cancer progression by deubiquitinating BRD4. Mol Cell. 71:592–605. e5942018. View Article : Google Scholar

146 

Tai F, Gong K, Song K, He Y and Shi J: Enhanced JunD/RSK3 signalling due to loss of BRD4/FOXD3/miR-548d-3p axis determines BET inhibition resistance. Nat Commun. 11:2582020. View Article : Google Scholar : PubMed/NCBI

147 

Wang X, Wei X, Cao Y and Xing P: ZNF33A promotes tumor progression and BET inhibitor resistance in Triple-Negative breast cancer. Am J Pathol. 192:1458–1469. 2022. View Article : Google Scholar

148 

Shu S, Wu HJ, Ge JY, Zeid R, Harris IS, Jovanovic B, Murphy K, Wang B, Qiu X, Endress JE, et al: Synthetic lethal and resistance interactions with BET bromodomain inhibitors in Triple-Negative breast cancer. Mol Cell. 78:1096–1113.e8. 2020. View Article : Google Scholar

149 

Luan W, Pang Y, Li R, Wei X, Jiao X, Shi J, Yu J, Mao H and Liu P: Akt/mTOR-Mediated autophagy confers resistance to BET inhibitor JQ1 in ovarian cancer. Onco Targets Ther. 12:8063–8074. 2019. View Article : Google Scholar : PubMed/NCBI

150 

Andrikopoulou A, Liontos M, Koutsoukos K, Dimopoulos MA and Zagouri F: Clinical perspectives of BET inhibition in ovarian cancer. Cell Oncol (Dordr). 44:237–249. 2021. View Article : Google Scholar : PubMed/NCBI

151 

Wang B, Fan P, Zhao J and Wu H, Jin X and Wu H: FBP1 loss contributes to BET inhibitors resistance by undermining c-Myc expression in pancreatic ductal adenocarcinoma. J Exp Clin Cancer Res. 37:2242018. View Article : Google Scholar : PubMed/NCBI

152 

Zhang P, Wang D, Zhao Y, Ren S, Gao K, Ye Z, Wang S, Pan CW, Zhu Y, Yan Y, et al: Intrinsic BET inhibitor resistance in SPOP-mutated prostate cancer is mediated by BET protein stabilization and AKT-mTORC1 activation. Nat Med. 23:1055–1062. 2017. View Article : Google Scholar : PubMed/NCBI

153 

Piha-Paul SA, Sachdev JC, Barve M, LoRusso P, Szmulewitz R, Patel SP, Lara PN Jr, Chen X, Hu B, Freise KJ, et al: First-in-Human study of mivebresib (ABBV-075), an Oral Pan-Inhibitor of bromodomain and extra terminal proteins, in patients with Relapsed/Refractory solid tumors. Clin Cancer Res. 25:6309–6319. 2019. View Article : Google Scholar : PubMed/NCBI

154 

Aggarwal RR, Schweizer MT, Nanus DM, Pantuck AJ, Heath EI, Campeau E, Attwell S, Norek K, Snyder M, Bauman L, et al: A Phase Ib/IIa Study of the Pan-BET Inhibitor ZEN-3694 in combination with enzalutamide in patients with metastatic Castration-Resistant prostate cancer. Clin Cancer Res. 26:5338–5347. 2020. View Article : Google Scholar : PubMed/NCBI

155 

Faivre EJ, McDaniel KF, Albert DH, Mantena SR, Plotnik JP, Wilcox D, Zhang L, Bui MH, Sheppard GS, Wang L, et al: Selective inhibition of the BD2 bromodomain of BET proteins in prostate cancer. Nature. 578:306–310. 2020. View Article : Google Scholar : PubMed/NCBI

156 

Ferreira D, Moreira JN and Rodrigues LR: New advances in exosome-based targeted drug delivery systems. Crit Rev Oncol Hematol. 172:1036282022. View Article : Google Scholar

157 

Bhome R, Del Vecchio F, Lee GH, Bullock MD, Primrose JN, Sayan AE and Mirnezami AH: Exosomal microRNAs (exomiRs): Small molecules with a big role in cancer. Cancer Lett. 420:228–235. 2018. View Article : Google Scholar

158 

Alcitepe I, Salcin H, Karatekin I and Kaymaz BT: HDAC inhibitor Vorinostat and BET inhibitor Plx51107 epigenetic agents' combined treatments exert a therapeutic approach upon acute myeloid leukemia cell model. Med Oncol. 39:2572022. View Article : Google Scholar

159 

Liu S, Li F, Pan L, Yang Z, Shu Y, Lv W, Dong P and Gong W: BRD4 inhibitor and histone deacetylase inhibitor synergistically inhibit the proliferation of gallbladder cancer in vitro and in vivo. Cancer Sci. 110:2493–2506. 2019. View Article : Google Scholar

160 

Patel MR, Garcia-Manero G, Paquette R, Dinner S, Donnellan WB, Grunwald MR, Ribadeneira MD, Schroeder P, Brevard J, Wilson L, et al: Phase 1 Dose escalation and expansion study to determine safety, tolerability, pharmacokinetics, and pharmacodynamics of the BET inhibitor FT-1101 as a single agent in patients with relapsed or refractory hematologic malignancies. Blood. 134:3907. 2019. View Article : Google Scholar

161 

Piha-Paul SA, Hann CL, French CA, Cousin S, Brana I, Cassier PA, Moreno V, de Bono JS, Harward SD, Ferron-Brady G, et al: Phase 1 study of molibresib (GSK525762), a bromodomain and Extra-Terminal domain protein inhibitor, in NUT carcinoma and other solid tumors. JNCI Cancer Spectr. 4:pkz0932020. View Article : Google Scholar : PubMed/NCBI

162 

Dawson M, Stein EM, Huntly BJP, Karadimitris A, Kamdar M, Fernandez de Larrea C, Dickinson MJ, Yeh PS-H, Daver N, Chaidos A, et al: A Phase I study of GSK525762, a selective bromodomain (BRD) and extra terminal protein (BET) Inhibitor: Results from Part 1 of Phase I/II open label single agent study in patients with acute myeloid leukemia (AML). Blood. 130:13772017. View Article : Google Scholar : PubMed/NCBI

163 

Falchook G, Rosen S, LoRusso P, Watts J, Gupta S, Coombs CC, Talpaz M, Kurzrock R, Mita M, Cassaday R, et al: Development of 2 bromodomain and extraterminal inhibitors with distinct pharmacokinetic and pharmacodynamic profiles for the treatment of advanced malignancies. Clin Cancer Res. 26:1247–1257. 2020. View Article : Google Scholar : PubMed/NCBI

164 

Watts JM, Hunter AM, Iurlo A, Xicoy B, Palandri F, Reeves B, Vannucchi A, Bose P, Ayala Diaz R, Halpern AB, et al: Bromodomain and extra-terminal (BET) inhibitor INCB057643 (LIMBER-103) in patients (pts) with relapsed or refractory myelofibrosis (R/R MF) and other advanced myeloid neoplasms: A phase 1 study. HemaSphere. 7:e17929062023. View Article : Google Scholar

165 

Ameratunga M, Brana I, Bono P, Postel-Vinay S, Plummer R, Aspegren J, Korjamo T, Snapir A and de Bono JS: First-in-human Phase 1 open label study of the BET inhibitor ODM-207 in patients with selected solid tumours. Br J Cancer. 123:1730–1736. 2020. View Article : Google Scholar : PubMed/NCBI

166 

Dombret H, Preudhomme C, Berthon C, Raffoux E, Thomas X, Vey N, Gomez-Roca C, Ethell M, Yee K, Bourdel F, et al: A Phase 1 Study of the BET-Bromodomain inhibitor OTX015 in patients with advanced acute leukemia. Blood. 124:1172014. View Article : Google Scholar

167 

Doroshow DB, Eder JP and LoRusso PM: BET inhibitors: A novel epigenetic approach. Ann Oncol. 28:1776–1787. 2017. View Article : Google Scholar

168 

Lewin J, Soria JC, Stathis A, Delord JP, Peters S, Awada A, Aftimos PG, Bekradda M, Rezai K, Zeng Z, et al: Phase Ib Trial With Birabresib, a Small-Molecule inhibitor of bromodomain and extraterminal proteins, in patients with selected advanced solid tumors. J Clin Oncol. 36:3007–3014. 2018. View Article : Google Scholar

169 

Moreno V, Sepulveda JM, Vieito M, Hernandez-Guerrero T, Doger B, Saavedra O, Ferrero O, Sarmiento R, Arias M, De Alvaro J, et al: Phase I study of CC-90010, a reversible, oral BET inhibitor in patients with advanced solid tumors and relapsed/refractory non-Hodgkin's lymphoma. Ann Oncol. 31:780–788. 2020. View Article : Google Scholar

170 

Bhattacharya S, Piya S and Borthakur G: Bromodomain inhibitors: What does the future hold? Clin Adv Hematol Oncol. 16:504–515. 2018.

171 

Mascarenhas J, Kremyanskaya M, Hoffman R, Bose P, Talpaz M, Harrison CN, Gupta V, Leber B, Sirhan S, Kabir S, et al: MANIFEST, a Phase 2 Study of CPI-0610, a bromodomain and extraterminal domain inhibitor (BETi), as monotherapy or ‘Add-on’ to ruxolitinib, in patients with refractory or intolerant advanced myelofibrosis. Blood. 134:6702019. View Article : Google Scholar

172 

Senapati J, Fiskus WC, Daver N, Wilson NR, Ravandi F, Garcia-Manero G, Kadia T, DiNardo CD, Jabbour E, Burger J, et al: Phase I results of bromodomain and Extra-terminal inhibitor PLX51107 in combination with azacitidine in patients with Relapsed/Refractory myeloid malignancies. Clin Cancer Res. 29:4352–4360. 2023. View Article : Google Scholar : PubMed/NCBI

173 

Patnaik A, Carvajal RD, Komatsubara KM, Britten CD, Wesolowski R, Michelson G, Alcantar O, Zhang C, Powell B, Severson P, et al: Phase ib/2a study of PLX51107, a small molecule BET inhibitor, in subjects with advanced hematological malignancies and solid tumors. J Clin Oncol. 36:2550. 2018. View Article : Google Scholar

174 

Roboz GJ, Desai P, Lee S, Ritchie EK, Winer ES, DeMario M, Brennan B, Nuesch E, Chesne E, Brennan L, et al: A dose escalation study of RO6870810/TEN-10 in patients with acute myeloid leukemia and myelodysplastic syndrome. Leuk Lymphoma. 62:1740–1748. 2021. View Article : Google Scholar

175 

Shapiro GI, LoRusso P, Dowlati A, T Do K, Jacobson CA, Vaishampayan U, Weise A, Caimi PF, Eder JP, French CA, et al: A Phase 1 study of RO6870810, a novel bromodomain and extra-terminal protein inhibitor, in patients with NUT carcinoma, other solid tumours, or diffuse large B-cell lymphoma. Br J Cancer. 124:744–753. 2021. View Article : Google Scholar : PubMed/NCBI

176 

Wang JSZ, Vita SD, Karlix JL, Cook C, Littlewood GM, Hattersley MM, Moorthy G, Edlund H, Fabbri G, Sachsenmeier KF, et al: First-in-human study of AZD5153, a small molecule inhibitor of bromodomain protein 4 (BRD4), in patients (pts) with relapsed/refractory (RR) malignant solid tumor and lymphoma: Preliminary data. J Clin Oncol. 37:3085. 2019. View Article : Google Scholar

177 

Hilton J, Cristea M, Postel-Vinay S, Baldini C, Voskoboynik M, Edenfield W, Shapiro GI, Cheng ML, Vuky J, Corr B, et al: BMS-986158, a small molecule inhibitor of the bromodomain and extraterminal domain proteins, in patients with selected advanced solid tumors: Results from a Phase 1/2a trial. Cancers (Basel). 14:40792022. View Article : Google Scholar : PubMed/NCBI

178 

Brown JA, Bal J, Simeoni M, Williams P, Mander PK, Soden PE, Daga S, Fahy WA, Wong GK, Bloomer JC, et al: A randomized study of the safety and pharmacokinetics of GSK3358699, a mononuclear myeloid-targeted bromodomain and extra-terminal domain inhibitor. Br J Clin Pharmacol. 88:2140–2155. 2022. View Article : Google Scholar

179 

Li Y, Xiang J, Zhang J, Lin J, Wu Y and Wang X: Inhibition of Brd4 by JQ1 promotes functional recovery from spinal cord injury by activating autophagy. Front Cell Neurosci. 14:5555912020. View Article : Google Scholar

180 

Lee DU, Katavolos P, Palanisamy G, Katewa A, Sioson C, Corpuz J, Pang J, DeMent K, Choo E, Ghilardi N, et al: Nonselective inhibition of the epigenetic transcriptional regulator BET induces marked lymphoid and hematopoietic toxicity in mice. Toxicol Appl Pharmacol. 300:47–54. 2016. View Article : Google Scholar

181 

Bakshi S, McKee C, Walker K, Brown C and Chaudhry GR: Toxicity of JQ1 in neuronal derivatives of human umbilical cord mesenchymal stem cells. Oncotarget. 9:33853–33864. 2018. View Article : Google Scholar

182 

Leal AS, Williams CR, Royce DB, Pioli PA, Sporn MB and Liby KT: Bromodomain inhibitors, JQ1 and I-BET 762, as potential therapies for pancreatic cancer. Cancer Lett. 394:76–87. 2017. View Article : Google Scholar

183 

Piquereau J, Boet A, Pechoux C, Antigny F, Lambert M, Gressette M, Ranchoux B, Gambaryan N, Domergue V, Mumby S, et al: The BET bromodomain inhibitor I-BET-151 induces structural and functional alterations of the heart mitochondria in healthy male mice and rats. Int J Mol Sci. 20:15272019. View Article : Google Scholar

184 

Liu CS, Rioja I, Bakr A, Veldwijk MR, Sperk E, Herskind C, Weichenhan D, Prinjha RK, Plass C, Schmezer P, et al: Selective inhibitors of bromodomain BD1 and BD2 of BET proteins modulate radiation-induced profibrotic fibroblast responses. Int J Cancer. 151:275–286. 2022. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2024
Volume 51 Issue 6

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yongprayoon V, Wattanakul N, Khomate W, Apithanangsiri N, Kasitipradit T, Nantajit D and Tavassoli M: Targeting BRD4: Potential therapeutic strategy for head and neck squamous cell carcinoma (Review). Oncol Rep 51: 74, 2024
APA
Yongprayoon, V., Wattanakul, N., Khomate, W., Apithanangsiri, N., Kasitipradit, T., Nantajit, D., & Tavassoli, M. (2024). Targeting BRD4: Potential therapeutic strategy for head and neck squamous cell carcinoma (Review). Oncology Reports, 51, 74. https://doi.org/10.3892/or.2024.8733
MLA
Yongprayoon, V., Wattanakul, N., Khomate, W., Apithanangsiri, N., Kasitipradit, T., Nantajit, D., Tavassoli, M."Targeting BRD4: Potential therapeutic strategy for head and neck squamous cell carcinoma (Review)". Oncology Reports 51.6 (2024): 74.
Chicago
Yongprayoon, V., Wattanakul, N., Khomate, W., Apithanangsiri, N., Kasitipradit, T., Nantajit, D., Tavassoli, M."Targeting BRD4: Potential therapeutic strategy for head and neck squamous cell carcinoma (Review)". Oncology Reports 51, no. 6 (2024): 74. https://doi.org/10.3892/or.2024.8733