Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Cancer Cell Int ; 24(1): 190, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38822309

ABSTRACT

BACKGROUND: Cancer-associated fibroblasts (CAFs) are the major cellular component of the tumor microenvironment and are known to affect tumor growth and response to various treatments. This study was undertaken to investigate the crosstalk between tumor-matched or unmatched CAFs and head and neck squamous cell carcinoma (HNSCC) cells regarding tumor growth and treatment response. METHODS: Three HNSCC cell lines (LK0412, LK0902 and LK0923), were cocultured in 2D or in 3D with their tumor-matched CAFs, site matched CAFs from other tumors or normal oral fibroblasts (NOFs). Cell proliferation was assessed as the amount of Ki67 positive cells/ spheroid area in formalin-fixed- paraffin-embedded 3D spheroids stained with Ki67 antibody. Viability after seven days of cisplatin treatment was measured with CellTiter-Glo 3D Viability Assay. The mRNA expression of CAF-associated markers (ACTA2, COL1A2, FAP, PDGFRα, PDGFRß, PDPN, POSTN and S100A4) in CAFs before and after coculture with tumor cells as well as mRNA expression of CAF-induced genes (MMP1, MMP9 and FMOD) in tumor cells separated from CAFs after co-culture was measured with RT-qPCR. The expression of selected protein biomarkers was validated with immunohistochemistry based on previous mRNA expression results. RESULTS: The proliferation of the LK0412 and LK0902 tumor spheroids varied significantly when cocultured with different CAFs and NOFs as shown by Ki-67 positive cells. RT‒qPCR analysis revealed different molecular profile of the analyzed HNSCC-derived CAFs concerning the expression of CAF-associated markers. The interaction between CAFs and HNSCC cells was more pronounced after coculture with unmatched CAFs as shown by changes in mRNA expression pattern of CAF-specific markers. Additionally, the unmatched CAFs significantly upregulated the mRNA expression of MMP1, MMP9 and FMOD in tumor cells compared to tumor-matched CAFs. CONCLUSION: Our results indicate that tumor-matched CAFs are unique for each tumor and affect the proliferation and the gene/protein expression of tumor cells in a distinct manner. The interaction between tumor unmatched CAFs and HNSCC cells in the tumor spheroids is associated with significant changes in the mRNA expression of CAF-specific markers and significant increases in FMOD and MMP9 in tumor cells compared to when cocultured with tumor-matched CAFs. Taken together, our results show how important the selection of CAFs is to get a reliable in vitro model that mimics the patients' tumor.

2.
Microvasc Res ; 153: 104666, 2024 May.
Article in English | MEDLINE | ID: mdl-38301938

ABSTRACT

OBJECTIVES: Laser Doppler Flowmetry (LDF) is a non-invasive technique for the assessment of tissue blood flow, but increased reproducibility would facilitate longitudinal studies. The aim of the study was to assess the interday reproducibility of Laser Doppler Flowmetry (LDF) at rest, at elevated local temperatures, and with the use of the vasodilator Methyl Nicotinate (MN) in six interconnected protocols for the measurement of the blood supply to the microvascular bed of the gingiva. METHODS: Ten healthy volunteers were included. Interweek LDF measurements with custom-made acrylic splints were performed. Six protocols were applied in separate regions of interest (ROI): 1; basal LDF, 2; LDF with thermoprobe 42 °C, 3; LDF with thermoprobe 45 °C, 4; LDF with thermoprobe 42 °C and MN, 5; LDF with thermoprobe 45 °C and MN and 6; LDF with MN. RESULTS: Intra-individual reproducibility was assessed by the within-subject coefficient of variation (wCV) and the intraclass correlation coefficient (ICC). Basal LDF measurements demonstrated high reproducibility with wCV 11.1 in 2 min and 10.3 in 5 min. ICC was 0.9 and 0.92. wCV after heat and MN was 4.9-10.3 and ICC 0.82-0.93. The topically applied MN yielded increased blood flow. CONCLUSION: This is the first study evaluating the reproducibility of basal LDF compared to single or multiple vasodilatory stimuli in gingiva. Multiple collector fibers probe and stabilizing acrylic splints are recommended. Vasodilatory stimulation showed a tendency toward higher reproducibility. Furthermore, MN yields vasodilation in gingiva.


Subject(s)
Gingiva , Skin , Humans , Microcirculation , Laser-Doppler Flowmetry/methods , Reproducibility of Results , Skin/blood supply , Regional Blood Flow
3.
ACS Appl Mater Interfaces ; 15(14): 17485-17494, 2023 Apr 12.
Article in English | MEDLINE | ID: mdl-36976817

ABSTRACT

Despite the enormous advancements in nanomedicine research, a limited number of nanoformulations are available on the market, and few have been translated to clinics. An easily scalable, sustainable, and cost-effective manufacturing strategy and long-term stability for storage are crucial for successful translation. Here, we report a system and method to instantly formulate NF achieved with a nanoscale polyelectrolyte coacervate-like system, consisting of anionic pseudopeptide poly(l-lysine isophthalamide) derivatives, polyethylenimine, and doxorubicin (Dox) via simple "mix-and-go" addition of precursor solutions in seconds. The coacervate-like nanosystem shows enhanced intracellular delivery of Dox to patient-derived multidrug-resistant (MDR) cells in 3D tumor spheroids. The results demonstrate the feasibility of an instant drug formulation using a coacervate-like nanosystem. We envisage that this technique can be widely utilized in the nanomedicine field to bypass the special requirement of large-scale production and elongated shelf life of nanomaterials.


Subject(s)
Nanoparticles , Nanostructures , Neoplasms , Humans , Feasibility Studies , Doxorubicin/pharmacology , Doxorubicin/chemistry , Neoplasms/pathology , Drug Carriers/chemistry , Nanoparticles/chemistry , Cell Line, Tumor , Drug Delivery Systems
4.
Oncol Rep ; 47(3)2022 Mar.
Article in English | MEDLINE | ID: mdl-35059742

ABSTRACT

Hypoxia has been linked with increased resistance to treatment in various solid tumors, including head and neck squamous cell carcinoma (HNSCC). The aim of the present study was to identify genes involved in hypoxia­mediated responses to radiotherapy in HNSCC. A total of three HNSCC cell lines with an epithelial phenotype were selected for this study and cultured under normoxic (21% O2) or hypoxic (1% O2) conditions. The sensitivity of the HNSCC cells to radiotherapy was assessed by a crystal violet assay. Western blotting (for protein expression), cDNA microarrays and reverse transcription­quantitative PCR (for gene expression) were also applied. Small interfering RNA silencing was used to knock down target genes. The results revealed that hypoxia negatively affected the response of HNSCC cells to radiotherapy. Of note, increased levels of N­cadherin, vimentin and fibronectin, as well as stem cell­associated transcription factors, were observed under hypoxia. The microarray analysis revealed a number of hypoxia­regulated genes that were involved in multiple biological functions. However, downregulation of hypoxia­regulated genes did not affect sensitivity to radiotherapy of the investigated cell lines. Taken together, the present findings indicated several important pathways and genes that were involved in hypoxia and radiotherapy resistance. It is hypothesized that panels of reported hypoxia­regulated genes may be useful for the prediction of radiotherapy responses in patients with HNSCC.


Subject(s)
Cell Hypoxia/genetics , Epithelial-Mesenchymal Transition/genetics , Neoplastic Stem Cells/pathology , Phenotype , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/radiotherapy , Down-Regulation , Gene Expression Regulation, Neoplastic , Humans
5.
Cancers (Basel) ; 13(13)2021 Jul 04.
Article in English | MEDLINE | ID: mdl-34283070

ABSTRACT

Cancer-associated fibroblasts (CAFs) are known to increase tumor growth and to stimulate invasion and metastasis. Increasing evidence suggests that CAFs mediate response to various treatments. HNSCC cell lines were co-cultured with their patient-matched CAFs in 2D and 3D in vitro models, and the tumor cell gene expression profiles were investigated by cDNA microarray and qRT-PCR. The mRNA expression of eight candidate genes was examined in tumor biopsies from 32 HNSCC patients and in five biopsies from normal oral tissue. Differences in overall survival (OS) were tested with Kaplan-Meier long-rank analysis. Thirteen protein coding genes were found to be differentially expressed in tumor cells co-cultured with CAFs in 2D and 81 in 3D when compared to tumor cells cultured without CAFs. Six of these genes were upregulated both in 2D and 3D (POSTN, GREM1, BGN, COL1A2, COL6A3, and COL1A1). Moreover, two genes upregulated in 3D, MMP9 and FMOD, were significantly associated with the OS. In conclusion, we demonstrated in vitro that CAF-derived signals alter the tumor cell expression of multiple genes, several of which are associated with differentiation, epithelial-to-mesenchymal transition (EMT) phenotype, and metastasis. Moreover, six of the most highly upregulated genes were found to be overexpressed in tumor tissue compared to normal tissue.

6.
Small ; 17(31): e2101440, 2021 08.
Article in English | MEDLINE | ID: mdl-34173333

ABSTRACT

Given the breadth of currently arising opportunities and concerns associated with nanoparticles for biomedical imaging, various types of nanoparticles have been widely exploited, especially for cellular/subcellular level probing. However, most currently reported nanoparticles either have inefficient delivery into cells or lack specificity for intracellular destinations. The absence of well-defined nanoplatforms remains a critical challenge hindering practical nano-based bio-imaging. Herein, the authors elaborate on a tailorable membrane-penetrating nanoplatform as a carrier with encapsulated actives and decorated surfaces to tackle the above-mentioned issues. The tunable contents in such a versatile nanoplatform offer huge flexibility to reach the expected properties and functions. Aggregation-induced emission luminogen (AIEgen) is applied to achieve sought-after photophysical properties, specific targeting moieties are installed to give high affinity towards different desired organelles, and critical grafting of cell-penetrating cyclic disulfides (CPCDs) to promote cellular uptake efficiency without sacrificing the specificity. Hereafter, to validate its practicability, the tailored nano products are successfully applied to track the dynamic correlation between mitochondria and lysosomes during autophagy. The authors believe that the strategy and described materials can facilitate the development of functional nanomaterials for various life science applications.


Subject(s)
Nanoparticles , Nanostructures , Lysosomes , Mitochondria , Organelles/metabolism
7.
Cancer Cell Int ; 20(1): 599, 2020 Dec 22.
Article in English | MEDLINE | ID: mdl-33353547

ABSTRACT

BACKGROUND: Head and neck squamous cell carcinoma (HNSCC) is a heterogeneous group of tumors for which the overall survival rate worldwide is around 60%. The tumor microenvironment, including cancer-associated fibroblasts (CAFs), is believed to affect the treatment response and migration of HNSCC. The aim of this study was to create a biologically relevant HNSCC in vitro model consisting of both tumor cells and CAFs cultured in 3D to establish predictive biomarkers for treatment response, as well as to investigate the impact of CAFs on phenotype, proliferation and treatment response in HNSCC cells. METHODS: Three different HNSCC patient-derived tumor cell lines were cultured with and without CAFs in a 3D model. Immunohistochemistry of the proliferation marker Ki67, epidermal growth factor receptor (EGFR) and fibronectin and a TUNEL-assay were performed to analyze the effect of CAFs on both tumor cell proliferation and response to cisplatin and cetuximab treatment in tumor spheroids (3D). mRNA expression of epithelial-mesenchymal transition (EMT) and cancer stem cells markers were analyzed using qRT-PCR. RESULTS: The results demonstrated increased cell proliferation within the tumor spheroids in the presence of CAFs, correlating with increased expression of EGFR. In spheroids with increased expression of EGFR, a potentiated response to cetuximab treatment was observed. Surprisingly, an increase in Ki67 expressing tumor cells were observed in spheroids treated with cisplatin for 3 days, correlating with increased expression of EGFR. Furthermore, tumor cells co-cultured with CAFs presented an increased EMT phenotype compared to tumor cells cultured alone in 3D. CONCLUSION: Taken together, our results reveal increased cell proliferation and elevated expression of EGFR in HNSCC tumor spheroids in the presence of CAFs. These results, together with the altered EMT phenotype, may influence the response to cetuximab or cisplatin treatment.

8.
Sci Rep ; 9(1): 20066, 2019 12 27.
Article in English | MEDLINE | ID: mdl-31882620

ABSTRACT

One of the hallmarks of cancers is their ability to develop resistance against therapeutic agents. Therefore, developing effective in vitro strategies to identify drug resistance remains of paramount importance for successful treatment. One of the ways cancer cells achieve drug resistance is through the expression of efflux pumps that actively pump drugs out of the cells. To date, several studies have investigated the potential of using 3-dimensional (3D) multicellular tumor spheroids (MCSs) to assess drug resistance; however, a unified system that uses MCSs to differentiate between multi drug resistance (MDR) and non-MDR cells does not yet exist. In the present report we describe MCSs obtained from post-diagnosed, pre-treated patient-derived (PTPD) cell lines from head and neck squamous cancer cells (HNSCC) that often develop resistance to therapy. We employed an integrated approach combining response to clinical drugs and screening cytotoxicity, monitoring real-time drug uptake, and assessing transporter activity using flow cytometry in the presence and absence of their respective specific inhibitors. The report shows a comparative response to MDR, drug efflux capability and reactive oxygen species (ROS) activity to assess the resistance profile of PTPD MCSs and two-dimensional (2D) monolayer cultures of the same set of cell lines. We show that MCSs provide a robust and reliable in vitro model to evaluate clinical relevance. Our proposed strategy can also be clinically applicable for profiling drug resistance in cancers with unknown resistance profiles, which consequently can indicate benefit from downstream therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/pathology , Humans , Spheroids, Cellular , Tumor Cells, Cultured
9.
Oncol Lett ; 18(5): 5155-5162, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31620196

ABSTRACT

The standard treatment for head and neck squamous cell carcinoma (HNSCC) is radiotherapy, often in combination with chemotherapy or surgery. However, a novel monoclonal antibody, cetuximab (Erbitux®), has also been approved for patient therapy. The aim of present study was to develop an in vitro method for the measurement of 18F-fluoro-2deoxy-D-glucose (FDG) to determine if cellular 18F-FDG uptake is associated with response to radiotherapy or cetuximab treatment. In the current study, HNSCC cell lines were treated with radiation or with cetuximab. Next, the uptake of 18F-FDG was measured using a gamma spectrometer (GS). Thereafter, uptake after radiation was measured first with GS and then compared with positron emission tomography (PET)/computed tomography (CT) imaging. Furthermore, the mRNA expression of glucose transporter 1 (GLUT1) was measured following cetuximab treatment via reverse transcription-quantitative PCR. A study protocol was developed to measure the cellular uptake of 18F-FDG via gamma-ray spectrometry and comparable results were obtained with those of clinical PET/CT. The results revealed a decrease in 18F-FDG after radiation and cetuximab treatment. The uptake of 18F-FDG following cetuximab treatment was significantly lower in the cetuximab-sensitive cell line UT-SCC-14 compared with the cetuximab-resistant cell lines UT-SCC-2 and UT-SCC-45. Furthermore, after treatment with cetuximab for 24 and 48 h, a significant increase in GLUT1 expression was detected in the sensitive cell line compared with the two resistant cell lines. In conclusion, a novel yet reliable method for the measurement of intracellular 18F-FDG via GS has been developed, and our results indicate that 18F-FDG uptake is associated with radiation and cetuximab response in HNSCC.

10.
Cancer Cell Int ; 19: 16, 2019.
Article in English | MEDLINE | ID: mdl-30651721

ABSTRACT

BACKGROUND: Head and Neck Squamous Cell Carcinoma (HNSCC) tumors are often resistant to therapies. Therefore searching for predictive markers and new targets for treatment in clinically relevant in vitro tumor models is essential. Five HNSCC-derived cell lines were used to assess the effect of 3D culturing compared to 2D monolayers in terms of cell proliferation, response to anti-cancer therapy as well as expression of EMT and CSC genes. METHODS: The viability and proliferation capacity of HNSCC cells as well as induction of apoptosis in tumor spheroids cells after treatment was assessed by MTT assay, crystal violet- and TUNEL assay respectively. Expression of EMT and CSC markers was analyzed on mRNA (RT-qPCR) and protein (Western blot) level. RESULTS: We showed that HNSCC cells from different tumors formed spheroids that differed in size and density in regard to EMT-associated protein expression and culturing time. In all spheroids, an up regulation of CDH1, NANOG and SOX2 was observed in comparison to 2D but changes in the expression of EGFR and EMT markers varied among the cell lines. Moreover, most HNSCC cells grown in 3D showed decreased sensitivity to cisplatin and cetuximab (anti-EGFR) treatment. CONCLUSIONS: Taken together, our study points at notable differences between these two cellular systems in terms of EMT-associated gene expression profile and drug response. As the 3D cell cultures imitate the in vivo behaviour of neoplastic cells within the tumor, our study suggest that 3D culture model is superior to 2D monolayers in the search for new therapeutic targets.

11.
Oncol Rep ; 38(4): 2062-2068, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28849066

ABSTRACT

The current treatment recommendation for T2-3N0M0 glottic squamous cell carcinoma (SCC) in the Nordic countries comprises of radiotherapy (RT) and chemoradiotherapy (CRT). Tumor radiosensitivity varies and another option is primary surgical treatment, which underlines the need for predictive markers in this patient population. The aim of the present study was to investigate the relation of the proteins WRAP53ß, survivin and p16INK4a to RT/CRT response and ultimate outcome of patients with T2-T3N0 glottic SCC. Protein expression was determined using immunohistochemistry on tumors from 149 patients consecutively treated with RT or CRT at Helsinki University Hospital, Karolinska University Hospital, and Linköping University Hospital during 1999-2010. Our results demonstrate a significantly better 5-year relapse-free survival, disease-free survival (DFS), disease-specific survival and overall survival of patients with T3N0 tumors treated with CRT compared with RT alone. Patients with tumors showing a cytoplasmic staining of WRAP53ß revealed significantly worse DFS compared with those with nuclear staining. For survivin, we observed a trend towards better 5-year DFS in patients with strong nuclear survivin expression compared with those with weak nuclear survivin expression (p=0.091). Eleven (7%) tumors showed p16 positivity, with predilection to younger patients, and this age group of patients with p16-positive SCC had a significantly better DFS compared with patients with p16-negative SCC. Taken together, our results highlight WRAP53ß as a potential biomarker for predicting RT/CRT response in T2-T3N0 glottic SCC. p16 may identify a small but distinct group of glottic SCC with favorable outcome. Furthermore, for T3N0 patients better outcome was observed following CRT compared to RT alone.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/therapy , Cisplatin/therapeutic use , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Glottis/pathology , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/therapy , Inhibitor of Apoptosis Proteins/biosynthesis , Laryngeal Neoplasms/metabolism , Laryngeal Neoplasms/therapy , Telomerase/biosynthesis , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/biosynthesis , Carcinoma, Squamous Cell/pathology , Chemoradiotherapy , Female , Glottis/drug effects , Glottis/radiation effects , Head and Neck Neoplasms/pathology , Humans , Immunohistochemistry , Laryngeal Neoplasms/pathology , Male , Middle Aged , Molecular Chaperones , Neoplasm Staging , Predictive Value of Tests , Squamous Cell Carcinoma of Head and Neck , Survivin
12.
Int J Mol Sci ; 18(5)2017 Apr 29.
Article in English | MEDLINE | ID: mdl-28468237

ABSTRACT

Despite advances in the head and neck squamous cell carcinoma (HNSCC) treatment modalities, drug resistance and cancer recurrence are often reported. Hypoxia signaling through hypoxia-inducible factor 1 (HIF-1) promotes angiogenesis and metastasis by inducing epithelial-mesenchymal-transition (EMT). The aim of this study was to evaluate the impact of hypoxia on response to therapy as well as EMT and expression of stem cell markers in HNSCC cells. Five HNSCC cell lines (UT-SCC-2, UT-SCC-14, LK0412, LK0827, and LK0923) were selected for this study. The treatment sensitivity for radiation, cisplatin, cetuximab, and dasatinib was assessed by crystal violet assay. Gene expression of EMT and cancer stem cell (CSC) markers as well as protein level of EGFR signaling molecules were analyzed by qPCR and western blotting, respectively. Unlike UT-SCC-14 and LK0827, the LK0412 cell line became significantly more sensitive to cetuximab in hypoxic conditions. This cetuximab sensitivity was efficiently reversed after suppression of HIF-1α with siRNA. Additionally, hypoxia-induced EMT and expression of stem cell markers in HNSCC cells was partially revoked by treatment with cetuximab or knockdown of HIF-1α. In summary, our study shows that hypoxia might have a positive influence on the anti-EGFR therapy effectiveness in HNSCC. However, due to heterogeneity of HNSCC lesions, targeting HIF-1α may not be sufficient to mediate such a response. Further studies identifying a trait of hypoxia-specific response to cetuximab in HNSCC are advisable.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Carcinoma, Squamous Cell/complications , Carcinoma, Squamous Cell/drug therapy , Cetuximab/pharmacology , Dasatinib/pharmacology , ErbB Receptors/antagonists & inhibitors , Head and Neck Neoplasms/complications , Head and Neck Neoplasms/drug therapy , Hypoxia/complications , Protein Kinase Inhibitors/pharmacology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cisplatin/pharmacology , Epithelial-Mesenchymal Transition/drug effects , ErbB Receptors/metabolism , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Hypoxia/metabolism , Hypoxia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Signal Transduction/drug effects , Squamous Cell Carcinoma of Head and Neck
13.
J Oral Pathol Med ; 46(9): 717-724, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28036101

ABSTRACT

BACKGROUND: The aims of this study were to validate in vitro drug sensitivity testing of head and neck squamous cell carcinoma (HNSCC) cell lines in an in vivo xenograft model and to identify treatment-induced changes in the epidermal growth factor receptor (EGFR) signaling pathway that could be used as markers for cetuximab treatment response. MATERIALS AND METHODS: The in vitro and in vivo cetuximab sensitivity of two HNSCC cell lines, UT-SCC-14 and UT-SCC-45, was assessed using a crystal violet assay and xenografts in nude mice, respectively. The expression of EGFR, phosphorylated EGFR (pEGFR), phosphorylated Src (pSrc), and Ki-67 was investigated by immunohistochemistry. To verify these results, the in vitro expression of EGFR and pEGFR was analyzed with ELISA in a panel of 10 HNSCC cell lines. RESULTS: A close correlation was found between in vitro and in vivo cetuximab sensitivity data in the two investigated HNSCC cell lines. In treatment sensitive UT-SCC-14 xenografts, there was a decrease in EGFR, pEGFR, and pSrc upon cetuximab treatment. Interestingly, in insensitive UT-SCC-45 xenografts, an increased expression of these three proteins was found. The change in EGFR and pEGFR expression in vivo was confirmed in cetuximab-sensitive and cetuximab-insensitive HNSCC cell lines using ELISA. CONCLUSION: High sensitivity to cetuximab was strongly associated with a treatment-induced reduction in pEGFR both in vivo and in vitro in a panel of HNSCC cell lines, suggesting that EGFR and pEGFR dynamics could be used as a predictive biomarker for cetuximab treatment response.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Cetuximab/pharmacology , Cetuximab/therapeutic use , ErbB Receptors/drug effects , Head and Neck Neoplasms/drug therapy , Oncogene Protein pp60(v-src)/drug effects , Animals , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Drug Screening Assays, Antitumor , ErbB Receptors/biosynthesis , Female , Head and Neck Neoplasms/metabolism , Heterografts , Humans , Mice , Mice, Inbred BALB C , Oncogene Protein pp60(v-src)/biosynthesis , Phosphorylation , Squamous Cell Carcinoma of Head and Neck
14.
J Oral Pathol Med ; 45(9): 640-646, 2016 Oct.
Article in English | MEDLINE | ID: mdl-26757432

ABSTRACT

BACKGROUND: Head and neck squamous cell carcinoma (HNSCC) tumors are often therapy resistant and may originate from cancer stem cells or tumor cells with an epithelial-to-mesenchymal transition (EMT) phenotype. The aim of this study was to characterize HNSCC cell lines with regard to EMT profile and to investigate the influence of EMT on the response to treatment. METHODS: mRNA expression of the EMT-associated genes CDH1 (E-cadherin), CDH2 (N-cadherin), FOXC2, TWIST1, VIM (vimentin), and FN1 (fibronectin) was determined using quantitative real-time PCR. Cell morphology and migration were investigated by phase-contrast microscopy and Boyden chamber assay, respectively. The cell surface expression of CD44 and epidermal growth factor receptor (EGFR) was examined by flow cytometry. The response to radiotherapy, cetuximab, and dasatinib was assessed by crystal violet staining. RESULTS: A total of 25 cell lines investigated differed greatly with regard to EMT phenotype. Cell lines with an EMT expression profile showed a mesenchymal morphology and a high migratory capacity. In addition, they exhibited a high cell surface expression of CD44 and a low expression of EGFR, a pattern previously associated with stemness. When the EMT inducer transforming growth factor-ß (TGF-ß) was added to non-EMT cells, changes in treatment responses were observed. Moreover, the expression of TWIST1 was found to correlate with radioresistance. CONCLUSIONS: The data presented in this report suggest that EMT is associated with a CD44high /EGFRlow phenotype and possibly negative impact on radiotherapy response in HNSCC cell lines.


Subject(s)
Epithelial-Mesenchymal Transition , ErbB Receptors/metabolism , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/therapy , Hyaluronan Receptors/metabolism , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Movement , Humans , Nuclear Proteins/metabolism , Phenotype , Prognosis , RNA, Messenger/metabolism , Radiotherapy , Twist-Related Protein 1/metabolism
15.
J Oral Pathol Med ; 45(1): 9-16, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25677871

ABSTRACT

BACKGROUND: Head and neck squamous cell carcinoma is frequently associated with aberrant epidermal growth factor receptor (EGFR) signaling, which contributes to tumor growth. Here, the functional importance of EGFR ligands in relation to proliferation and sensitivity to the EGFR-targeted therapy cetuximab was investigated in three tongue cancer cell lines. METHODS: The influence of epidermal growth factor (EGF), amphiregulin (AR), and epiregulin (EPR) on tumor cell proliferation and cetuximab response was evaluated by the addition of recombinant human (rh) proteins or by siRNA-mediated downregulation of the endogenous ligand production. The expression, activation and cellular distribution of EGFR were assessed by Western blot analysis and immunocytochemistry. RESULTS: EGF downregulation suppressed the proliferation of all investigated tumor cell lines, whereas the response to an increased level of EGF differed between EGFR-overexpressing and EGFR-non-overexpressing cell lines. Furthermore, tumor cells consistently displayed increased cetuximab resistance upon the addition of rhEGF, whereas EGF silencing was associated with an improved cetuximab response. The data regarding AR and EPR were inconclusive. CONCLUSIONS: Our data suggest that the amount of EGF is a determinant of the tumor cell proliferation rate and the response to cetuximab treatment in tongue cancer. Thus, EGF is a potential predictive biomarker of poor cetuximab response and a possible treatment target.


Subject(s)
Biomarkers, Tumor/biosynthesis , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Cetuximab/pharmacology , Epidermal Growth Factor/biosynthesis , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Tongue Neoplasms/drug therapy , Tongue Neoplasms/metabolism , Amphiregulin/deficiency , Amphiregulin/genetics , Amphiregulin/pharmacology , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/analysis , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation , Drug Resistance, Neoplasm , Epidermal Growth Factor/analysis , Epiregulin/deficiency , Epiregulin/genetics , Epiregulin/pharmacology , ErbB Receptors/biosynthesis , ErbB Receptors/metabolism , Head and Neck Neoplasms/pathology , Humans , Immunohistochemistry , Ligands , Molecular Targeted Therapy , Squamous Cell Carcinoma of Head and Neck , Tongue Neoplasms/pathology
16.
Oncotarget ; 6(12): 10134-45, 2015 Apr 30.
Article in English | MEDLINE | ID: mdl-25912307

ABSTRACT

Salinomycin has been used as treatment for malignant tumors in a small number of humans, causing far less side effects than standard chemotherapy. Several studies show that Salinomycin targets cancer-initiating cells (cancer stem cells, or CSC) resistant to conventional therapies. Numerous studies show that Salinomycin not only reduces tumor volume, but also decreases tumor recurrence when used as an adjuvant to standard treatments. In this study we show that starvation triggered different stress responses in cancer cells and primary normal cells, which further improved the preferential targeting of cancer cells by Salinomycin. Our in vitro studies further demonstrate that the combined use of 2-Fluoro 2-deoxy D-glucose, or 2-deoxy D-glucose with Salinomycin is lethal in cancer cells while the use of Oxamate does not improve cell death-inducing properties of Salinomycin. Furthermore, we show that treatment of cancer cells with Salinomycin under starvation conditions not only increases the apoptotic caspase activity, but also diminishes the protective autophagy normally triggered by the treatment with Salinomycin alone. Thus, this study underlines the potential use of Salinomycin as a cancer treatment, possibly in combination with short-term starvation or starvation-mimicking pharmacologic intervention.


Subject(s)
Cell Hypoxia/physiology , Glucose/administration & dosage , Neoplasms/drug therapy , Neoplastic Stem Cells/metabolism , Pyrans/antagonists & inhibitors , Pyrans/pharmacology , Autophagy/drug effects , Cell Death/drug effects , Cell Line, Tumor , Drug Screening Assays, Antitumor , Glucose/metabolism , Humans , Neoplasms/metabolism , Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology
17.
Oral Oncol ; 51(1): 24-30, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25456005

ABSTRACT

OBJECTIVES: Today there are no reliable predictive markers for radiotherapy response in head and neck squamous cell carcinoma (HNSCC), leading to both under- and over-treatment of patients, personal suffering, and negative socioeconomic effects. Inherited mutation in WRAP53ß (WD40 encoding RNA Antisense to p53), a protein involved in intracellular trafficking, dramatically increases the risk of developing HNSCC. The purpose of this study was to investigate whether WRAP53ß can predict response to radiotherapy in patients with HNSCC. MATERIALS AND METHODS: Tumor biopsies from patients with HNSCC classified as responders or non-responders to radiotherapy were examined for the expression of the WRAP53ß protein and single nucleotide polymorphisms in the corresponding gene employing immunohistochemistry and allelic discrimination, respectively. In addition, the effect of RNAi-mediated downregulation of WRAP53ß on the intrinsic radiosensitivity of two HNSCC cell lines was assed using crystal violet and clonogenic assays. RESULTS: Nuclear expression of WRAP53ß was significantly associated with better response to radiotherapy and improved patient survival. Downregulation of WRAP53ß with siRNA in vitro enhanced cellular resistance to radiation. CONCLUSIONS: Our findings suggest that nuclear expression of WRAP53ß promotes tumor cell death in response to radiotherapy and is a promising predictor of radiotherapy response in patients with HNSCC.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Cell Nucleus/metabolism , Head and Neck Neoplasms/metabolism , Telomerase/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Biopsy , Carcinoma, Squamous Cell/physiopathology , Carcinoma, Squamous Cell/radiotherapy , Female , Head and Neck Neoplasms/physiopathology , Head and Neck Neoplasms/radiotherapy , Humans , Male , Middle Aged , Molecular Chaperones , Survival Analysis
18.
Oral Oncol ; 50(9): 832-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24993889

ABSTRACT

OBJECTIVES: Targeted therapy against the epidermal growth factor receptor (EGFR) only variably represents a therapeutic advance in head and neck squamous cell carcinoma (HNSCC). This study addresses the need of biomarkers of treatment response to the EGFR-targeting antibody cetuximab (Erbitux®). MATERIALS AND METHODS: The intrinsic cetuximab sensitivity of HNSCC cell lines was assessed by a crystal violet assay. Gene copy number analysis of five resistant and five sensitive cell lines was performed using the Affymetrix SNP 6.0 platform. Quantitative real-time PCR was used for verification of selected copy number alterations and assessment of mRNA expression. The functional importance of the findings on the gene and mRNA level was investigated employing siRNA technology. The data was statistically evaluated using Mann-Whitney U-test and Spearman's correlation test. RESULTS: Analysis of the intrinsic cetuximab sensitivity of 32 HNSCC cell lines characterized five and nine lines as cetuximab sensitive or resistant, respectively. Gene copy number analysis of five resistant versus five sensitive cell lines identified 39 amplified protein-coding genes, including YAP1, in the genomic regions 11q22.1 or 5p13-15. Assessment using qPCR verified that YAP1 amplification associated with cetuximab resistance. Amplification of YAP1 correlated to higher mRNA levels, and RNA knockdown resulted in increased cetuximab sensitivity. Assessment of several independent clinical data sets in the public domain confirmed YAP1 amplifications in multiple tumor types including HNSCC, along with highly differential expression in a subset of HNSCC patients. CONCLUSION: Taken together, we provide evidence that YAP1 could represent a novel biomarker gene of cetuximab resistance in HNSCC cell lines.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Antibodies, Monoclonal, Humanized/therapeutic use , Biomarkers/metabolism , Carcinoma, Squamous Cell/metabolism , Head and Neck Neoplasms/metabolism , Phosphoproteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Base Sequence , Carcinoma, Squamous Cell/drug therapy , Cell Line, Tumor , Cetuximab , DNA Primers , Drug Resistance, Neoplasm , Gene Silencing , Head and Neck Neoplasms/drug therapy , Humans , Phosphoproteins/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Transcription Factors , YAP-Signaling Proteins
19.
Int J Cancer ; 133(8): 1994-2003, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23564498

ABSTRACT

Head and neck squamous cell carcinoma (HNSCC) is a malignancy that is associated with severe mortality despite advances in therapy. Today's standard treatment most commonly includes radiotherapy, often combined with chemotherapy or surgery. There are so far no established biomarkers to predict response to radiation, and thus the aim of this study was to investigate a series of markers that could potentially identify HNSCC patients who would benefit from radiotherapy. The selected markers, both proteins (epidermal growth factor receptor, survivin and p53), and single nucleotide polymorphisms (SNPs) in the genes of XRCC3, XRCC1, XPC, XPD, MDM2, p53 and FGFR4 were correlated to the response to radiotherapy and overall survival. Investigations were performed on pretreatment tumor biopsies from patients classified as responders or nonresponders to radiotherapy. Protein expression was examined using immunohistochemistry and the genotyping of specific SNPs was analyzed using PCR-RFLP or pyrosequencing. We found that survivin expression was significantly stronger in the responder group (p = 0.003) and that patients with a strong survivin expression had a significantly better overall survival (p < 0.001). Moreover, downregulation of survivin by siRNA in two HNSCC cell lines significantly decreased their sensitivity to radiation. Among the SNPs analyzed, patients with the XPD Lys751Gln SNP had a significantly shorter overall survival (p = 0.048), and patients with the FGFR4 Gly388Arg SNP had a significantly longer overall survival (p = 0.010). In conclusion, our results suggest that survivin plays an important role in the response to radiotherapy and may be a useful marker for predicting radiotherapy response in patients with HNSCC.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/radiotherapy , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/radiotherapy , Inhibitor of Apoptosis Proteins/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/mortality , Cell Line, Tumor , Down-Regulation , Female , Head and Neck Neoplasms/mortality , Humans , Inhibitor of Apoptosis Proteins/biosynthesis , Inhibitor of Apoptosis Proteins/genetics , Male , Middle Aged , Polymorphism, Single Nucleotide , RNA Interference , RNA, Small Interfering , Receptor, Fibroblast Growth Factor, Type 4/genetics , Survivin , Treatment Outcome , Xeroderma Pigmentosum Group D Protein/genetics
20.
J Cell Mol Med ; 17(1): 12-29, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23301705

ABSTRACT

The rapid accumulation of knowledge on apoptosis regulation in the 1990s was followed by the development of several experimental anticancer- and anti-ischaemia (stroke or myocardial infarction) drugs. Activation of apoptotic pathways or the removal of cellular apoptotic inhibitors has been suggested to aid cancer therapy and the inhibition of apoptosis was thought to limit ischaemia-induced damage. However, initial clinical studies on apoptosis-modulating drugs led to unexpected results in different clinical conditions and this may have been due to co-effects on non-apoptotic interconnected cell death mechanisms and the 'yin-yang' role of autophagy in survival versus cell death. In this review, we extend the analysis of cell death beyond apoptosis. Upon introduction of molecular pathways governing autophagy and necrosis (also called necroptosis or programmed necrosis), we focus on the interconnected character of cell death signals and on the shared cell death processes involving mitochondria (e.g. mitophagy and mitoptosis) and molecular signals playing prominent roles in multiple pathways (e.g. Bcl2-family members and p53). We also briefly highlight stress-induced cell senescence that plays a role not only in organismal ageing but also offers the development of novel anticancer strategies. Finally, we briefly illustrate the interconnected character of cell death forms in clinical settings while discussing irradiation-induced mitotic catastrophe. The signalling pathways are discussed in their relation to cancer biology and treatment approaches.


Subject(s)
Apoptosis/genetics , Autophagy/genetics , Necrosis/genetics , Neoplasms/pathology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Caspases/genetics , Caspases/metabolism , Gene Expression Regulation, Neoplastic , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Molecular Targeted Therapy , Necrosis/drug therapy , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Death Domain/genetics , Receptors, Death Domain/metabolism , Signal Transduction , Stress, Physiological
SELECTION OF CITATIONS
SEARCH DETAIL
...