Two novel histone deacetylase inhibitors NVP-LAQ824 and NVP-LBH589 are active against biliary tract cancer and potentiate the efficacy of gemcitabine

2006 ◽  
Vol 24 (18_suppl) ◽  
pp. 4149-4149 ◽  
Author(s):  
M. Wiedmann ◽  
T. Bluethner ◽  
M. Niederhagen ◽  
K. Schoppmeyer ◽  
J. Moessner ◽  
...  

4149 Background: Chromatin remodelling agents such as histone deacetylase inhibitors have been shown to modulate gene expression in tumor cells and inhibit tumor growth and angiogenesis. NVP-LAQ824 and NVP-LBH589 are two novel chemical entities belonging to a cinnamic hydroxamic acid class of compounds. Little is known about their efficacy for the treatment of biliary tract cancer. Methods: Cell-growth inhibition by NVP-LAQ824 and NVP-LBH589 was studied in vitro in 7 human biliary tract cancer cell lines by MTT assay. In addition, the anti-tumoral effect of NVP-LBH589 was studied in a chimeric mouse model. Anti-tumoral drug mechanism was assessed by immunoblotting for p21WAF-1, acH3Lys9 and acH4, cell cycle analysis, PARP assay, TUNEL assay, and immunhistochemistry for MIB-1. Results: In vitro treatment with both compounds significantly suppressed the growth of all cancer cell lines (mean IC50 (3d) 0.08 and 0.04 μM, respectively) and was associated with hyperacetylation of nucleosomal histones H3 and H4, increased expression of p21WAF-1, cell cycle arrest at G2/M-checkpoint, and induction of apoptosis (PARP cleavage). After 28 d, NVP-LBH589 reduced tumor mass by 66% (bile duct cancer) and 87% (gallbladder cancer) in vivo in comparison to placebo and potentiated the efficacy of gemcitabine. Further analysis of the tumor specimens revealed increased apoptosis (TUNEL) and reduced cell proliferation (MIB-1). Conclusions: Our findings suggest that NVP-LBH589 > NVP-LAQ824 are active against human biliary tract cancer in vitro. In addition, NVP-LBH589 demonstrated significant in vivo activity and potentiated the efficacy of gemcitabine. Therefore, further clinical evaluation of this new drug for the treatment of biliary tract cancer is recommended. No significant financial relationships to disclose.

2007 ◽  
Vol 25 (18_suppl) ◽  
pp. 4624-4624 ◽  
Author(s):  
M. Wiedmann ◽  
M. Haefner ◽  
M. Niederhagen ◽  
K. Schoppmeyer ◽  
C. Wittekind ◽  
...  

4624 Background: Pancreatic cancer is the fifth to sixth leading cause of cancer death in Europe and up to 90% of patients will present with locally advanced or metastatic disease. Unfortunately, little progress has been obtained from chemotherapy regimens in the past decade. Treatment with histone deacetylase inhibitors, like NVP-LAQ824 and NVP-LBH589, either alone or in combination with conventional chemotherapy may be a novel alternative. Methods: Cell-growth inhibition by NVP-LAQ824 and NVP-LBH589 was studied in vitro in 8 human pancreatic cancer cell lines by MTT assay. In addition, the anti-tumoral effect of NVP-LBH589 was studied in a chimeric mouse model. Anti-tumoral drug mechanism was assessed by immunoblotting for p21WAF-1, acH4, p42/p44, Phospho-p42/p44, AKT, Phospho- AKT, cell cycle analysis, TUNEL assay, and immunohistochemistry for MIB-1. Results: In vitro treatment with both compounds significantly suppressed the growth of all cancer cell lines (mean IC50 (6d) 0.06 and 0.03 μM, respectively) and was associated with hyperacetylation of nucleosomal histones H4, increased expression of p21WAF-1, and cell cycle arrest at G2/M-checkpoint. After 21 d, NVP-LBH589 alone reduced tumor mass in vivo by 70% and in combination with gemcitabine by 81% in comparison to placebo for cell line L3.6 pl. Further analysis of the tumor specimens revealed slightly increased apoptosis (TUNEL) and no significant reduction of cell proliferation (MIB-1). Protein levels of p42/p44, and AKT remained stable, whereas levels of Phospho-p42/p44 and Phospho-AKT increased. Conclusions: Our findings suggest that NVP-LBH589 > NVP-LAQ824 are active against human pancreatic cancer in vitro. In addition, NVP-LBH589 demonstrated significant in vivo activity and potentiated the efficacy of gemcitabine, although the precise mechanism of drug action is not yet completely understood. Therefore, further preclinical and clinical evaluation of this new drug for the treatment of pancreatic cancer is recommended. No significant financial relationships to disclose.


Cancers ◽  
2021 ◽  
Vol 13 (15) ◽  
pp. 3862
Author(s):  
Christian Mayr ◽  
Tobias Kiesslich ◽  
Sara Erber ◽  
Dino Bekric ◽  
Heidemarie Dobias ◽  
...  

Inhibition of histone deacetylases (HDACs) is a promising anti-cancer approach. For biliary tract cancer (BTC), only limited therapeutic options are currently available. Therefore, we performed a comprehensive investigation of HDAC expression and pharmacological HDAC inhibition into a panel of eight established BTC cell lines. The screening results indicate a heterogeneous expression of HDACs across the studied cell lines. We next tested the effect of six established HDAC inhibitors (HDACi) covering pan- and class-specific HDACis on cell viability of BTC cells and found that the effect (i) is dose- and cell-line-dependent, (ii) does not correlate with HDAC isoform expression, and (iii) is most pronounced for romidepsin (a class I HDACi), showing the highest reduction in cell viability with IC50 values in the low-nM range. Further analyses demonstrated that romidepsin induces apoptosis in BTC cells, reduces HDAC activity, and increases acetylation of histone 3 lysine 9 (H3K9Ac). Similar to BTC cell lines, HDAC 1/2 proteins were heterogeneously expressed in a cohort of resected BTC specimens (n = 78), and their expression increased with tumor grading. The survival of BTC patients with high HDAC-2-expressing tumors was significantly shorter. In conclusion, HDAC class I inhibition in BTC cells by romidepsin is highly effective in vitro and encourages further in vivo evaluation in BTC. In situ assessment of HDAC 2 expression in BTC specimens indicates its importance for oncogenesis and/or progression of BTC as well as for the prognosis of BTC patients.


2014 ◽  
Vol 2014 ◽  
pp. 1-7 ◽  
Author(s):  
Kahori Seto ◽  
Junichi Shoda ◽  
Tomohisa Horibe ◽  
Eiji Warabi ◽  
Masayuki Kohno ◽  
...  

It is known that the interleukin-4 receptorα(IL-4Rα) is highly expressed on the surface of various human solid tumors. We previously designed novel IL-4Rα-lytic hybrid peptide composed of binding peptide to IL-4Rαand cell-lytic peptide and reported that the designed IL-4Rα-lytic hybrid peptide exhibited cytotoxic and antitumor activity bothin vitroandin vivoagainst the human pancreatic cancer cells expressing IL-4Rα. Here, we evaluated the antitumor activity of the IL-4Rα-lytic hybrid peptide as a novel molecular targeted therapy for human biliary tract cancer (BTC). The IL-4Rα-lytic hybrid peptide showed cytotoxic activity in six BTC cell lines with a concentration that killed 50% of all cells (IC50) as low as 5 μM. We also showed that IL-4Rα-lytic hybrid peptide in combination with gemcitabine exhibited synergistic cytotoxic activityin vitro. In addition, intravenous administration of IL-4Rα-lytic hybrid peptide significantly inhibited tumor growth in a xenograft model of human BTCin vivo. Taken together, these results indicated that the IL-4Rα-lytic hybrid peptide is a potent agent that might provide a novel therapy for patients with BTC.


2020 ◽  
Vol 52 (3) ◽  
pp. 945-956 ◽  
Author(s):  
Ah-Rong Nam ◽  
Mei-Hua Jin ◽  
Ju-Hee Bang ◽  
Kyoung-Seok Oh ◽  
Hye-Rim Seo ◽  
...  

PurposeCurrently, the DNA damage response (DDR) pathway represents a key target for new cancer drug development. Advanced biliary tract cancer (BTC) has a poor prognosis because of the lack of efficacious treatment options. Although DNA repair pathway alterations have been reported in many patients with BTC, little is known regarding the effects of DDR-targeted agents against BTC.Materials and MethodsIn this study, nine BTC cell lines were exposed to the WEE1 inhibitor (AZD1775). In vitro, MTT assay, colony-forming assay, cell cycle analysis, phospho-histone H3 staining assay, Transwell migration assay, and western blot were performed. Then, to enhance the antitumor effect of AZD1775, the combination treatment of WEE1 inhibitor and ataxia telangiectasia mutated and Rad3 related (ATR) inhibitor (AZD6738) was conducted using MTT assay and comet assay. Finally, HuCCT-1 and SNU2670 xenograft models were established to confirm the anti-tumor effect of AZD1775 alone. Furthermore, the combination treatment was also evaluated in SNU2670 xenograft models.ResultsAZD1775 blocked the phosphorylation of CDC2 and CDC25C in all cell lines, but significantly increased apoptosis and S phase arrest in sensitive cells. However, increased p-ATR and phosphorylated ataxia telangiectasia mutated levels were observed in less sensitive cells. In addition, in vitro and in vivo data illustrated that AZD1775 combined with AZD6738 exerted more potent anti-tumor effects than either drug alone. Although WEE1 inhibition has promising anti-tumor effects in some BTC cells, the addition of ATR inhibitors could enhance its efficacy.ConclusionTaken together, this study supports further clinical development of DDR-targeted strategies as monotherapy or combination regimens for BTC.


Author(s):  
Y Moon ◽  
W Dahlberg ◽  
Y Yu ◽  
T Ohno ◽  
T Todoroki ◽  
...  

2013 ◽  
Vol 31 (4_suppl) ◽  
pp. 196-196
Author(s):  
Junyao Xu ◽  
Jennifer J. Knox ◽  
Emin Ibrahimov ◽  
Eric Xueyu Chen ◽  
Stefano Serra ◽  
...  

196 Background: MEK inhibition has clinical activity against biliary tract cancers, and might therefore be successfully combined with gemcitabine; one of the most active chemotherapy agents for these cancers. As gemcitabine is active in S-phase, and the ERK pathway has a major role driving cell cycle progression, concurrent use of a MEK inhibitor could potentially antagonize the effect of gemcitabine. We therefore tested the sequence dependence of the combination of gemcitabine and the MEK inhibitor AZD6244 in vitro and in vivo. Methods: Two biliary tract cancer cell lines and 4 xenografts established from patients were used in this study. In vitro, cell cycle effects of AZD6244 and their impact on gemcitabine sensitivity were detected by Flow cytometry, EdU uptake assay, MTS assay and Clonogenic survival assay. In vivo, tumor-bearing SCID mice were treated for 48hr with AZD6244 and then monitored for 48hr off treatment. Plasma and tumor drug levels were assessed by LC-MS. The time course for recovery of ERK signaling, cell cycle distribution and cell proliferation were measured by Immunofluorescence staining, Flow cytometry, EdU uptake assay and 18F-FLT PET imaging. Based on these results, two different treatment schedules combining AZD6244 with gemcitabine were tested in four different biliary tract cancer models. Results: DNA synthesis was suppressed during treatment with AZD6244, and re-entry into S-phase was delayed by 15hr in vitro and 48hr post-treatment in vivo. Concurrent treatment showed antagonistic effect (IC50=2.02±0.91uM) compared to gemcitabine alone (IC50=0.09±0.04uM) whereas when gemcitabine treatment was delayed for 24 hr after AZD6244 removal, enhanced cytotoxic effect was observed (IC50=0.051± 0.012uM). Consistantly, strong schedule dependence was seen in all four biliary cancer models tested: combined treatment with AZD6244 plus gemcitabine exerts enhanced antitumor effect when gemcitabine is given following a 48hr interruption in AZD6244 dosing, rather than concurrently. Conclusions: The combination of AZD6244 plus gemcitabine is highly schedule dependent, and predicted to be more effective in the clinic using sequential rather than simultaneous dosing protocols.


2011 ◽  
Vol 30 (6) ◽  
pp. 2148-2160 ◽  
Author(s):  
Hyun-Jin Nam ◽  
Hwang-Phill Kim ◽  
Young-Kwang Yoon ◽  
Sang-Hyun Song ◽  
Ah-Rum Min ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document