scholarly journals PO-1049: HNSCC Cell Lines with DNA Repair Defects show an Invasive Phenotype in vitro

2018 ◽  
Vol 127 ◽  
pp. S589
Author(s):  
P. Essers ◽  
E. Ploeg ◽  
M. Verheij ◽  
C. Vens
2019 ◽  
Vol 20 (6) ◽  
pp. 1306 ◽  
Author(s):  
Mario Scheurer ◽  
Roman Brands ◽  
Mohamed El-Mesery ◽  
Stefan Hartmann ◽  
Urs Müller-Richter ◽  
...  

Inflammation is a central aspect of tumour biology and can contribute significantly to both the origination and progression of tumours. The NFκB pathway is one of the most important signal transduction pathways in inflammation and is, therefore, an excellent target for cancer therapy. In this work, we examined the influence of four NFκB inhibitors—Cortisol, MLN4924, QNZ and TPCA1—on proliferation, inflammation and sensitisation to apoptosis mediated by the death ligand FasL in the HNSCC cell lines PCI1, PCI9, PCI13, PCI52 and SCC25 and in the human dermal keratinocyte cell line HaCaT. We found that the selection of the inhibitor is critical to ensure that cells do not respond by inducing counteracting activities in the context of cancer therapy, e.g., the extreme IL-8 induction mediated by MLN4924 or FasL resistance mediated by Cortisol. However, TPCA1 was qualified by this in vitro study as an excellent therapeutic mediator in HNSCC by four positive qualities: (1) proliferation was inhibited at low μM-range concentrations; (2) TNFα-induced IL-8 secretion was blocked; (3) HNSCC cells were sensitized to TNFα-induced cell death; and (4) FasL-mediated apoptosis was not disrupted.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 891-891
Author(s):  
Katia Beider ◽  
Valeria Voevoda ◽  
Hanna Bitner ◽  
Evgenia Rosenberg ◽  
Hila Magen ◽  
...  

Abstract Introduction: Multiple myeloma (MM) is a neoplastic disorder that is characterized by clonal proliferation of plasma cells in the bone marrow (BM). Despite the initial efficacious treatment, MM patients often become refractory to common anti-MM drugs, therefore novel therapies are in need. Pan-histone deacetylase (HDAC) inhibitor panobinostat exerts multiple cytotoxic actions in MM cells in vitro, and was approved for the treatment of relapsed/refractory MM in combination with bortezomib and dexamethasone. Although having promising anti-MM properties, panobinostat lacks therapeutic activity as monotherapy. The aim of the current study was to elucidate the mechanisms underlying MM resistance to panobinostat and to define strategies to overcome it. Results: Panobinostat at the low concentrations (IC50 5-30 nM) suppressed the viability in MM cell lines (n=7) and primary CD138+ cells from MM patients (n=8) in vitro. Sensitivity to panobinostat correlated with reduced expression of chemokine receptor CXCR4, while overexpression of CXCR4 or its ligand CXCL12 in RPMI8226 and CAG MM cell lines significantly (p<0.001) increased their resistance to panobinostat, pointing to the role of the CXCR4 axis in HDACi response. Notably, similar expression levels of class I HDACs (HDAC1-3) were detected in MM cells with either low or high CXCR4. Interaction with BM stromal cells that represent the source of CXCL12 also protected MM cells from panobinostat-induced apoptosis, further strengthening a role for CXCR4 downstream pathway. Decreased sensitivity to cytotoxic effect was concomitant with reduced histone (H3K9 and H4K8) acetylation in response to panobinostat treatment. In addition, resistance to HDACi was associated with the reversible G0/G1 cell growth arrest, whereas sensitivity was characterized by apoptotic cell death. Analysis of intra-cellular signaling mediators involved in CXCR4-mediated HDACi resistance revealed the pro-survival AKT/mTOR pathway to be regulated by both CXCR4 over-expression and interaction with BMSCs. Combining panobinostat with mTOR inhibitor everolimus abrogated the resistance and induced synergistic cell death of MM cell lines and primary MM cells, but not of normal mononuclear cells (CI<0.4). This effect was concurrent with the increase in DNA double strand breaks, histone H2AX phosphorylation, loss of Dψm, cytochrome c release, caspase 3 activation and PARP cleavage. The increase in DNA damage upon combinational treatment was not secondary to the apoptotic DNA fragmentation, as it occurred similarly when apoptosis onset was blocked by caspase inhibitor z-VAD-fmk. Kinetics studies also confirmed that panobinostat-induced DNA damage preceded apoptosis induction. Strikingly, combined panobinostat/everolimus treatment resulted in sustained DNA damage and irreversible suppression of MM cell proliferation accompanied by robust apoptosis, in contrast to the modest effects induced by single agent. Gene expression analysis revealed distinct genetic profiles of single versus combined exposures. Whereas panobinostat increased the expression of cell cycle inhibitors GADD45G and p21, co-treatment with everolimus abrogated the increase in p21 and synergistically downregulated DNA repair genes, including RAD21, Ku70, Ku80 and DNA-PKcs. Furthermore, combined treatment markedly decreased both mRNA and protein expression of anti-apoptotic factors survivin and BCL-XL, checkpoint regulator CHK1, and G2/M-specific factors PLK1, CDK1 and cyclin B1, therefore suppressing the DNA damage repair and inhibiting mitotic progression. Given the anti-apoptotic role of p21, the synergistic lethal effect of everolimus could be attributed to its ability to suppress p21 induction by panobinostat ensuing the shift in the DNA damage response toward apoptosis. Conclusions: Collectively, our findings indicate that CXCR4/CXCL12 activity promotes the resistance of MM cells to HDACi with panobinostat through mTOR activation. Inhibition of mTOR by everolimus synergizes with panobinostat by simultaneous suppression of p21, G2/M mitotic factors and DNA repair machinery, rendering MM cells incapable of repairing accumulated DNA damage and promoting their apoptosis. Our results unravel the mechanism responsible for strong synergistic anti-MM activity of dual HDAC and mTOR inhibition and provide the rationale for a novel therapeutic strategy to eradicate MM. Disclosures No relevant conflicts of interest to declare.


2017 ◽  
Vol 35 (15_suppl) ◽  
pp. e14105-e14105
Author(s):  
Janaki Parameswaran ◽  
Jong Woo Lee ◽  
Teresa Sandoval-Schaefer ◽  
Jaseok P. Koo ◽  
Barbara Burtness

e14105 Background: Aurora Kinase A (AURKA) is overexpressed in HNSCC, and correlates with poor prognosis. It has been identified as a potential therapeutic target, yet the response rate for the AURKA inhibitor MLN8237 is only 9% in treatment refractory HNSCC. We hypothesized that although AURKA inhibitors lead to defective spindle assembly, they may also reduce mitotic entry, undermining cytotoxic effect. We predicted that adding a WEE1 inhibitor to an AURKA inhibitor would mitigate this effect and enhance cell death Methods: Cell viability assays were performed on FaDu ( p53 mut.), Detroit562 ( p53 mut.), and UNC7 ( p53 WT) HPV negative HNSCC cell lines treated with AZD1775 (AZD), MLN8237 (MLN), and combination of AZD+MLN. Oncosphere formation assays were used to confirm findings of cell death, and western blot analysis and confocal microscopy were used to investigate mechanism of synergy. The above drugs were also given at varying doses via oral gavage to FaDu xenografted nude mice. Results: There was clear synergy of AZD and MLN in-vitro. Combination Indices were determined by the Chou-Talalay method: FaDu 0.4, Detroit562 0.5, and UNC7 0.6 (synergy = < 0.8). Oncopshere assays showed inability of AZD+MLN treated cells to re-differentiate. FaDu cells treated with MLN had increased p-CDK1 and reduced phospho-histone H3 (pHH3), suggesting reduced mitotic entry. AZD+MLN treated cells had reduced p-CDK1 and increased pHH3, similar to AZD treated cells; they also had spindle disarray with poor chromatin organization on confocal microscopy indicating mitotic catastrophe. In mice, the combination of AZD+MLN inhibited tumor growth, with no apparent toxicity. Mice treated with either drug alone had tumor volumes over 1000mm3 and were sacrificed at day 21; those treated with AZD 90mg/kg and MLN 30mg/kg had tumors volumes around 300mm3 on day 28. Conclusions: AZD and MLN synergistically enhance cell death in HNSCC cell lines and significantly inhibit tumor growth in mouse xenograft models. The ability of AZD to overcome intrinsic resistance to MLN may underlie mechanism of synergy. We recommend further investigation of AURKA and WEE1 inhibition in other cancers with high AURKA expression, and in patients with HNSCC.


2007 ◽  
Vol 2 (8) ◽  
pp. S382
Author(s):  
Kimio Yonesaka ◽  
Kreshnik Zejnullahu ◽  
Alison J. Homes ◽  
Bruce E. Johnson ◽  
Pasi A. Janne

2008 ◽  
Vol 139 (2_suppl) ◽  
pp. P92-P93
Author(s):  
James Russell Tysome ◽  
Ghassan Alusi ◽  
Nick Lemoine ◽  
Yaohe Wang

Problem Oncolytic viral therapy a promising new strategy for the treatment of cancer and an oncolytic adenovirus was first licensed for head and neck squamous cell carcinoma (HNSCC). However, the outcomes of clinical trials with viral monotherapy have been disappointing. Oncolytic vaccinia virus represents an attractive alternative as its replication is less dependent than adenovirus on the genetic make-up of tumor cells and it has been used safely as the smallpox vaccine in millions of patients. Methods The potency and replication of vaccinia virus and adenovirus were compared in a panel of HNSCC cell lines in vitro before assessing the tumor selectivity of systemically delivered vaccinia virus in vivo. In order to increase antitumor potency, a novel vaccinia virus expressing the angiogenesis inhibitory endostatin-angiostatin fusion protein was constructed. The expression and function of this protein was confirmed in vitro and antitumor efficacy assessed in vivo. Results Oncolytic vaccinia virus was more potent than adenovirus against all HNSCC cell lines and displayed high selectivity for cancer cells, sparing normal cells both in vitro and in animal tumour models in vivo. Vaccinia virus expressing the endostatin-angiostatin fusion protein inhibited new blood vessel formation as well as tumour growth by oncolysis. The protein was expressed in virus-infected HNSCC cells and demonstrated function by the inhibition of human umbilical vein epithelial cell proliferation and tube formation in vitro. Treatment of nude mice bearing FaDu HNSCC xenografts significantly prolonged survival when compared to the oncolytic adenovirus ONYX-015 used previously for HNSCC. Conclusion This novel vaccinia virus is a promising therapeutic agent for HNSCC, which improved survival in tumour bearing mice and requires further evaluation in vivo. Significance The combination of an oncolytic vaccinia virus that delivers tumor-specific angiogenesis inhibition may prove to be an effective treatment for patients with HNSCC, with the potential for systemic delivery to treat metastatic disease. Support Cancer Research UK, Royal College of Surgeons of England, Barts and the London Trustees.


2008 ◽  
Vol 139 (2_suppl) ◽  
pp. P89-P89
Author(s):  
Gregor Bran ◽  
Jens Stern-Straeter ◽  
Ulrich R. Goessler ◽  
Birgit Bran ◽  
Karl Hoermann ◽  
...  

Problem Angiogenesis is increased in head and neck squamous cell carcinoma (HNSCC), and correlates with tumor progression and metastasis. Vascular endothelial growth factor (VEGF) has been shown to be a key regulator of angiogenesis. Microvascular density is known to correlate with metastasis and aggressiveness. PDGF and PDGFR are expressed by many tumor types, including HNSCC. The PDGF/PDGFR-System is believed to play a pivotal role in tumorigenesis. The compound STI571 (Imatinib mesylate, Gleevec) inhibits PDGFR tyrosine kinases. In this study, we seek to determine whether targeting PDGFR signalling in HNSCC cell lines may affect expression and secretion of PDGF as well as VEGF in vitro. Methods Established human HNSCC cell lines were incubated with STI571 of different concentrations for 24h and 72h. PDGF and VEGF concentrations were determined by an ELISA technique, and gene expression was analyzed by RTPCR. Results In vitro treatment of HNSCC cell lines with STI571 resulted in a significant reduction of expression and secretion of PDGF and VEGF. We also observed a dose related effect for VEGF synthesis. Conclusion The results in this study suggest, that inhibition of PDGFR using STI571 might have an antitumor effect mediated in part by inhibition of VEGF-induced angiogenesis in HNSCC. Further studies need to be conducted to improve understanding of the molecular mechanisms. Significance Our study indicates that STI571 might play a role in establishing new therapeutic strategies against HNSCC.


2004 ◽  
Vol 131 (2) ◽  
pp. P176-P176
Author(s):  
Mark W El-Deiry ◽  
John H Lee ◽  
William C Spanos
Keyword(s):  

2021 ◽  
Vol 11 ◽  
Author(s):  
Sebastian Zahnreich ◽  
Senayit Gebrekidan ◽  
Gabriele Multhoff ◽  
Peter Vaupel ◽  
Heinz Schmidberger ◽  
...  

Abundance and signaling of the epidermal growth factor receptor (EGFR) and programmed cell death protein ligand 1 (PD-L1) in head and neck squamous cell carcinoma (HNSCC) are not only genetically determined but are also subject to the traits of the tumor microenvironment, which has hitherto not been clarified completely. We investigated the impact of hypoxia on the EGFR system and on PD-L1 in six HPV negative HNSCC cell lines in vitro and in FaDu xenografts in vivo. Protein levels of EGFR, AKT, pAKT, ERK1/2, pERK1/2, CA IX, cleaved PARP (apoptosis), LC3B (autophagy), and PD-L1 were quantified by western blot after oxygen deprivation or CoCl2, staurosporine, and erlotinib treatment. In FaDu xenograft tumors the expression of EGFR, CA IX andCD34 staining were analyzed. Reduced oxygen supply strongly downregulated EGFR protein levels and signaling in FaDu cells in vitro and in vivo, and a transient downregulation of EGFR signaling was found in three other HNSCC cell lines. PD-L1 was affected by oxygen deprivation in only one HNSCC cell line showing increased protein amounts. The results of this study indicate a significant impact of the traits of the tumor microenvironment on crucial molecular targets of cancer therapies with high clinical relevance for therapy resistance and response in HNSCC.


Sign in / Sign up

Export Citation Format

Share Document