scholarly journals The BET Inhibitor JQ1 Augments the Antitumor Efficacy of Gemcitabine in Preclinical Models of Pancreatic Cancer

Cancers ◽  
2021 ◽  
Vol 13 (14) ◽  
pp. 3470
Author(s):  
Aubrey L. Miller ◽  
Patrick L. Garcia ◽  
Samuel C. Fehling ◽  
Tracy L. Gamblin ◽  
Rebecca B. Vance ◽  
...  

Gemcitabine is used to treat pancreatic cancer (PC), but is not curative. We sought to determine whether gemcitabine + a BET bromodomain inhibitor was superior to gemcitabine, and identify proteins that may contribute to the efficacy of this combination. This study was based on observations that cell cycle dysregulation and DNA damage augment the efficacy of gemcitabine. BET inhibitors arrest cells in G1 and allow increases in DNA damage, likely due to inhibition of expression of DNA repair proteins Ku80 and RAD51. BET inhibitors (JQ1 or I-BET762) + gemcitabine were synergistic in vitro, in Panc1, MiaPaCa2 and Su86 PC cell lines. JQ1 + gemcitabine was more effective in vivo than either drug alone in patient-derived xenograft models (P < 0.01). Increases in the apoptosis marker cleaved caspase 3 and DNA damage marker γH2AX paralleled antitumor efficacy. Notably, RNA-seq data showed that JQ1 + gemcitabine selectively inhibited HMGCS2 and APOC1 ~6-fold, compared to controls. These proteins contribute to cholesterol biosynthesis and lipid metabolism, and their overexpression supports tumor cell proliferation. IPA data indicated that JQ1 + gemcitabine selectively inhibited the LXR/RXR activation pathway, suggesting the hypothesis that this inhibition may contribute to the observed in vivo efficacy of JQ1 + gemcitabine.

2010 ◽  
Vol 999 (999) ◽  
pp. 1-11
Author(s):  
P. Ulivi ◽  
C. Arienti ◽  
W. Zoli ◽  
M. Scarsella ◽  
S. Carloni ◽  
...  

2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Anna M. Jermakowicz ◽  
Matthew J. Rybin ◽  
Robert K. Suter ◽  
Jann N. Sarkaria ◽  
Zane Zeier ◽  
...  

AbstractBromodomain and extraterminal domain (BET) proteins have emerged as therapeutic targets in multiple cancers, including the most common primary adult brain tumor glioblastoma (GBM). Although several BET inhibitors have entered clinical trials, few are brain penetrant. We have generated UM-002, a novel brain penetrant BET inhibitor that reduces GBM cell proliferation in vitro and in a human cerebral brain organoid model. Since UM-002 is more potent than other BET inhibitors, it could potentially be developed for GBM treatment. Furthermore, UM-002 treatment reduces the expression of cell-cycle related genes in vivo and reduces the expression of invasion related genes within the non-proliferative cells present in tumors as measured by single cell RNA-sequencing. These studies suggest that BET inhibition alters the transcriptional landscape of GBM tumors, which has implications for designing combination therapies. Importantly, they also provide an integrated dataset that combines in vitro and ex vivo studies with in vivo single-cell RNA-sequencing to characterize a novel BET inhibitor in GBM.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Wenyu Wang ◽  
Yen-An Tang ◽  
Qian Xiao ◽  
Wee Chyan Lee ◽  
Bing Cheng ◽  
...  

AbstractBRD4, a Bromodomain and Extraterminal (BET) protein family member, is a promising anti-cancer drug target. However, resistance to BET inhibitors targeting BRD4 is common in solid tumors. Here, we show that cancer-associated fibroblast (CAF)-activated stromal signaling, interleukin-6/8-JAK2, induces BRD4 phosphorylation at tyrosine 97/98 in colorectal cancer, resulting in BRD4 stabilization due to interaction with the deubiquitinase UCHL3. BRD4 phosphorylation at tyrosine 97/98 also displays increased binding to chromatin but reduced binding to BET inhibitors, resulting in resistance to BET inhibitors. We further show that BRD4 phosphorylation promotes interaction with STAT3 to induce chromatin remodeling through concurrent binding to enhancers and super-enhancers, supporting a tumor-promoting transcriptional program. Inhibition of IL6/IL8-JAK2 signaling abolishes BRD4 phosphorylation and sensitizes BET inhibitors in vitro and in vivo. Our study reveals a stromal mechanism for BRD4 activation and BET inhibitor resistance, which provides a rationale for developing strategies to treat CRC more effectively.


PLoS ONE ◽  
2014 ◽  
Vol 9 (2) ◽  
pp. e87868 ◽  
Author(s):  
Chongqing Lv ◽  
Hongru Kong ◽  
Guohua Dong ◽  
Lewei Liu ◽  
Kun Tong ◽  
...  

2021 ◽  
Vol 11 ◽  
Author(s):  
Yafei Jiang ◽  
Gangyang Wang ◽  
Haoran Mu ◽  
Xiaojun Ma ◽  
Zhuoying Wang ◽  
...  

Osteosarcoma is the most common primary malignant bone tumor, and there are few ideal clinically available drugs. The bromodomain and extraterminal domain (BET) protein is an emerging target for aggressive cancer, but therapies targeting the BET in osteosarcoma have been unsuccessful in clinical trials to date, and further exploration of specific BET inhibitors is of great significance. In our study, we demonstrated that NHWD-870, a potent BET inhibitor in a phase I clinical trial, significantly inhibited tumor proliferation and promoted cell apoptosis by reversing the oncogenic signature in osteosarcoma. More importantly, we identified NHWD-870 impeded binding of BRD4 to the promoter of GP130 leading to diminished activation of JAK/STAT3 signaling pathway. Furthermore, GP130 knockdown significantly sensitizes the chemosensitivity in vitro. In OS cell-derived xenografts, NHWD-870 effectively inhibited the growth of osteosarcoma. Beyond that, NHWD-870 effectively inhibited the differentiation and maturation of precursor osteoclasts in vitro and attenuated osteoclast-mediated bone loss in vivo. Finally, we confirmed the efficacy of synthetic lethal effects of NHWD-870 and cisplatin in antagonizing osteosarcoma in a preclinical PDX model. Taken together, these findings demonstrate that NHWD-870, as an effective BET inhibitor, may be a potential candidate for osteosarcoma intervention linked to its STAT3 signaling inhibitory activity. In addition, NHWD-870 appears to be a promising therapeutic strategy for bone-associated tumors, as it interferes with the vicious cycle of tumor progression and bone destruction.


2017 ◽  
Vol 35 (15_suppl) ◽  
pp. e14086-e14086 ◽  
Author(s):  
Kazuhiro Onuma ◽  
Yasuhiro Aga ◽  
Sayaka Ogi ◽  
Takashi Matsushita ◽  
Hidetoshi Sunamoto ◽  
...  

e14086 Background: Cyclin dependent kinase 7 (CDK7) modulates mRNA transcription and some oncogenes are reported to be sensitive to inhibition of transcription in certain cancer cells. CDK7 inhibitors have been considered as an intriguing approach to treat cancers that depend on transcriptional regulation of their oncogenes. We synthesized a novel highly selective CDK7 inhibitor, UD-017, and found that the compound showed antitumor potency in a variety of cancers in vitro and in vivo. We therefore explored underlying mechanisms especially focusing on an oncogenic driver, c-Myc. Methods: We examined CDK7 selectivity of UD-017 against the other CDKs and kinases. We evaluated an antiproliferative activity of UD-017 in over 200 multiple types of cancer cell lines including patients-derived cancer cells. We then investigated the correlation between c-Myc expression levels and an antiproliferative activity of UD-017 in cancer cells. Antitumor efficacy of UD-017 was assessed in multiple types of cancer xenograft models and patients-derived xenograft model. We determined whether an intratumoral c-Myc expression levels correlated with in vivo antitumor efficacy of UD-017 in xenograft models. Results: UD-017 inhibited CDK7 enzyme (IC50= 16 nM) highly selectively among the CDKs (over 300-fold) and almost mono-specifically in a panel of 313 kinases assay. In a cellular antiproliferative panel assay, UD-017 broadly inhibited the proliferation of a variety of cancer cells and c-Myc expression levels showed the good correlation with antiproliferative activity. UD-017 showed favorable PK profile and good oral absorbability and showed the potent antitumor activity in multiple types of cancer xenograft models in mice. In correlation with the PK, UD-017 reduced the intratumoral c-Myc mRNA levels time-dependently after dosing of UD-017 in the colorectal cancer xenograft model. Conclusions: We identified a highly selective and orally available CDK7 inhibitor that showed the broad in vitro and in vivo antitumor activity in a variety of cancers, modulating c-Myc as an oncogenic driver. These data support the rationale for further advancing towards clinical development.


2020 ◽  
Author(s):  
Yang Li ◽  
Zhiqiang Liu ◽  
Ying Sun ◽  
Hui Ding ◽  
Dianyun Ren ◽  
...  

Abstract Background Pancreatic cancer remains one of the deadliest cancers worldwide. The tumor microenvironment is closely related to the occurrence, growth, and metastasis of tumors. Collagen type XI alpha 1 chain (COL11A1), as a component of extracellular collagen, has been proven to be responsible for tumor development and drug resistance in various cancers. However, it’s role in pancreatic cancer remains unknown. Method The GEPIA (Gene Expression Profiling Interactive Analysis) web tool was used to clarify the differential expression of COL11A1 and clinical prognosis in pancreatic cancer. Functional experiments were performed to assess the effect of COL11A1 on the state of pancreatic cells in vitro. Mouse xenograft models and pulmonary metastasis models were established to investigate the influence of COL11A1 in vivo. Chromatin immunoprecipitation (ChIP) assays and dual-luciferase assays were applied to assess the relationship between muscle, intestine and stomach expression 1 (Mist1) and COL11A1.Results The upregulated expression of COL11A1 in pancreatic cancer led to a worse prognosis and overall survival for patients with pancreatic cancer. Knockdown of COL11A1 in pancreatic cancer cell lines inhibited their proliferation and invasion, while upregulating COL11A1 increased those abilities. The ChIP and dual-luciferase assays clarified Mist1 could bind to the promoter of COL11A1 as a transcription factor and repress its transcription. Meanwhile, we found that the N-terminal repressor region of Mist1 was capable of inhibiting COL11A1 expression.Conclusion We identified COL11A1 as a carcinogen in pancreatic cancer, and clarified a novel mechanism which Mist1 reverses the Epithelial-Mesenchymal Transition in pancreatic cancer by repressing COL11A1 expression.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 674-674 ◽  
Author(s):  
Anne-Louise Latif ◽  
John J Cole ◽  
Joana Monteiro Campos ◽  
William Clark ◽  
Lynn McGarry ◽  
...  

Abstract Background: There remains a critical requirement for novel therapies for Acute Myeloid Leukemia (AML). Bromodomain and extra-terminal domain (BET) inhibitors are emerging as exciting therapeutic agents for hematopoietic malignancies. Pharmacological inhibition of BET bromodomains targets malignant cells by preventing reading of acetylated lysine residues, thus disrupting chromatin-mediated signal transduction, which reduces transcription at oncogenic loci. Although a heterogeneous disease, most AML retains wild type p53. However, p53 is often rendered functionally deficient by over-expression of MDM2. Potentiating the p53 response though MDM2 antagonism is therefore potentially beneficial to most AML subtypes. We hypothesized that dual inhibition of MDM2 and BET would be synthetic lethal to p53 wild type AML. Methods: For in vitro experiments CPI203 (BET inhibitor, Constellation Pharmaceuticals) and nutlin-3 (MDM2 antagonist, Sigma) were assessed on p53 wild type cell lines (OCI-AML3, MOLM-13 and MV411) and p53 wild type primary murine AML. To assess the combination's dependency on wild type p53; p53 mutated cell lines (KG1a, KASUMI-1 and THP1) were tested. Cell viability was assessed using resazurin (Alamar blue dye) across numerous dose ratios on the OCI-AML3 cell line and analysed using the Envision Fluorescent Reader. Drug combination indices (CI) were evaluated using Calcusyn (version 2.0). Apoptosis was assessed using flow cytometry staining for Annexin V and propidium iodide (PI) on all p53 wild type and mutated cell lines. For in vivo experiments CPI0610 (clinical grade BET inhibitor, Constellation Pharmaceuticals) and RG7112 (MDM2 inhibitor, Roche) were tested as single agents, in combination and with relevant vehicle controls. RNA seq was performed on the GAIIX sequencer and gene ontology analysis was performed using DAVID/INGENUITY pathway analysis (IPA). Results: In the OCI-AML3 cell line, resazurin analysis demonstrated that combining CPI203 with nutlin-3 was potently synergistic in decreasing viable cells for a 1:12.5 (mean CI=0.07) and 1:25 ratio (mean CI=0.299), and synergistic for a 1:50(mean CI=0.44) and 1:100(mean CI= 0.66) ratios. There was no benefit in using the combination treatment on the p53 mutated cell lines. Apoptosis was enhanced at least 1.5 fold (median 1.7, range 1.5-2.65) by the drug combination versus the single agents, in the panel of p53 wild type cell lines tested. Analysis of whole genome RNA seq on OCI-AML3 treated cells, showed that genes up-regulated by the combination of CPI203 and nutlin-3, had a thirty-fold enrichment for p53 signalling (FDR (<0.05). Down-regulated genes were enriched for FOXM1-dependent cell cycle progression genes. To evaluate the combination in vivo, we used a Trib-2 driven primary AML where leukemogenesis is induced through inhibition of C/EBPα. Myeloblasts were transduced with GFP on the same retroviral construct asTrib-2 for disease tracking. Treatment was commenced in all mice (n=40), post confirmation of disease engraftment. Three mice from each treatment group were sacrificed after 48hrs and cells sorted for GFP to perform RNA seq in this in vivo setting. After 21 days of treatment all mice were sacrificed (n=27, one vehicle control succumbed to disease 15 days post engraftment). End of treatment results (primary read out was the GFP% which equates to the blast%) demonstrated superior in vivo efficacy of dual inhibition of MDM2 and BET in comparison with controls in eradicating AML, p<0.0001, (see figure). Importantly, normal haematopoiesis was spared - as evidenced by normal full blood counts and comparable myeloid, B-cell and T-cell populations with our C57bl6 wild type controls. RNA seq of the murine blasts revealed that many more genes significantly (FDR<0.05) changed expression in the combination treated mice than single agent treated mice. The p53 pathway was the most common up-stream regulator of genes changing expression post combination treatment, p<0.0001. The combination affected many more genes in the p53 pathway than RG7112 alone (120 genes versus 20 genes respectively), in line with our in vitro results. Conclusion: This combination of BET and MDM2 inhibition is effective and superior to single agent therapy on all p53 wild type AMLs tested, in vitro and in vivo. In both contexts this is associated with potentiating the p53 response and could be relevant to many patients with p53 wild type AML. Figure 1. Figure 1. Disclosures Latif: Novartis: Honoraria. Copland:Ariad: Honoraria, Membership on an entity's Board of Directors or advisory committees; BMS: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Pfizer: Honoraria, Membership on an entity's Board of Directors or advisory committees.


Sign in / Sign up

Export Citation Format

Share Document