Abstract
Laryngeal cancer (LC) is one of the common head and neck neoplasms and is characterized by resistance to conventional therapy and poor prognosis. This may result from the presence of cancer stem cells (CSCs), which form a small population in tumors with metastatic potential, high invasive capacity, self-renewal, and differentiation. This study aimed to evaluate the effectiveness of 5-fluorouracil and cisplatin individually, as well as the combination of cetuximab and paclitaxel in a CSC subpopulation separated with biomarkers related to tumoral growth (CD44, CD117, and CD133). In addition, expression of TrkB, KRAS, HIF-1α, and VEGF-A genes and proteins related to cell proliferation were evaluated in this subpopulation. The CD44, CD133, and CD117 biomarkers were used to analyze the identification and separation of both subpopulations using FACSAria Fusion. Subpopulations positive for CD44, CD133, and CD117 or lacking these biomarkers were classified as laryngeal cancer stem cells (LCSCs) or laryngeal cancer non-stem cells (non-LCSCs), respectively. Matrigel invasion and colony forming assays were performed to confirm CSC presence. Subpopulations were cultured and exposed to 5-fluorouracil, cisplatin, and cetuximab/paclitaxel drugs for 24 h. Cell proliferation was determined using MTS assay. KRAS and TrkB gene expression levels were evaluated using quantitative real time PCR with TaqMan® Assay in both subpopulations. The non-LCSC subpopulation was considered as the control for relative expression. We found that the LCSC subpopulation demonstrated more resistance to cetuximab and paclitaxel combination chemotherapy when compared with the non-LCSC subpopulation of the cell line. These LCSC subpopulations presented up-regulated expression of KRAS, HIF-1α, and VEGF-A genes and proteins and no TrkB gene expression, but TrkB protein expression was up-regulated in the LC cell line when compared to the non-CSC subpopulation. “In conclusion, the combination of CD44, CD133, and CD117 biomarkers has stem cell properties. Moreover, LCSCs, are capable of resisting treatment and present high KRAS, HIF-1α, and VEGF-A gene expression”.
Keywords: Cancer stem cells, chemotherapy, head and neck neoplasms, gene expression, cell line
Introduction
Laryngeal cancer (LC) is one of the most common head and neck neoplasms, representing 2% of all malignant neoplasms [1]. Estimates show that by 2020, 9,491 new cases and 5,202 deaths may occur owing to this disease [2]. Chemotherapy with docetaxel, bleomycin, hydroxyurea, pembrolizaumab, nivolumab, methotrexate, cetuximab [3], and paclitaxel [4] drugs can be used for treating LC. Despite advances in drug therapy, individuals with LC show low survival due to the locoregional recurrence and metastasis onset [5].
A small group of cells known as cancer stem cells (CSCs) may be responsible for tumor maintenance and dissemination. These cells possess self-renewal and differentiation potential and also play an important role in tumor initiation and progression [6]. These features can be associated with poor prognosis [7] and provide tumoral resistance, leading to ineffective drug treatment [8-10]. CSCs can be identified by cell surface biomarkers such as CD44, CD117, and CD133 related to tumoral growth [6,11-14].
Literature also show that genes related to the cell proliferation pathway may be associated with increase of tumoral progression and poor prognosis; for example, tropomyosin-related kinase B (TrkB), rat sarcoma (RAS), epidermal growth factor receptor (EGFR), Hypoxia-Inducible Factor 1 alpha (HIF1-α) and vascular endothelial growth factor (VEGF) genes are overexpressed in different tumor types [15-22]. Both EGFR and TrkB are cell surface receptors that are activated by binding to epidermal growth factor (EGF) and brain-derived neurotrophic factor (BDNF), respectively. These tyrosine kinase receptors are responsible for activating some downstream intracellular signals, such as the Ras-Raf-MEK-ERK pathway [16,23].
The RAS oncogene family has three isoforms: Harvey (HRAS), neuroblastoma (NRAS), and Kirsten (KRAS) [17]. They encode small GTPase proteins, which have essential roles in cell proliferation, growth, survival, migration, and epithelial-mesenchymal transition (EMT), as well as important roles in tumor relapse and chemotherapeutic resistance [19,20]. Alterations in KRAS are associated with benefits from anti-EGFR antibody therapy, consequently improving progression-free survival and overall survival [17]. Nevertheless, mutated KRAS can regulate the GDP-GTP process and activate Ras-Raf-MEK-ERK downstream effectors independent of EGFR and TrkB receptor activation, leading to chemotherapy resistance [17,24].
Depending on the alterations in the KRAS gene, the overexpression of this gene may occur with different stimuli that activate signaling pathways with distinct impacts on the production of basal genes [25]; for example, HIF-1α, a nuclear transcription factor important in the hypoxia response, leads to activation of VEGF-A [21], which is responsible for angiogenesis as well as preservation of blood vessels for tumors [25,26].
This study aimed to evaluate the effectiveness of 5-fluorouracil and cisplatin individually as well as the combination of cetuximab and paclitaxel in a CSC subpopulation separated with biomarkers related to tumoral growth, CD44, CD117, and CD133. In addition, TrkB, KRAS, HIF-1α and VEGF-A gene and protein expressions related with cell proliferation were evaluated in this subpopulation.
Materials and methods
Sample
Hep2 cell line, originally established from laryngeal squamous cell carcinoma and described with HeLa cell contamination (American Type Culture Collection, ATCC, Rockville, MD, USA), was utilized in the present study. Hep2 authentication was performed using the AmpFLSTR Identifier PCR Amplification kit (Life Technologies, Carlsbad, CA, USA) at the Special Techniques Laboratory, Hospital Israelita Albert Einstein (LATE-HIAE), São Paulo, and our cell line showed 100% identify compared to the ATCC database. Cells were cultured in Dulbecco’s Modified Eagle Medium (DMEM; Sigma-Aldrich, St. Louis, MO, USA) supplemented with 10% fetal bovine serum (FBS; Gibco™, Carlsbad, CA, USA), 1% L-glutamine (Gibco™), and 1% penicillin/streptomycin/amphotericin B (Gibco™) in a humidified 5% CO2 atmosphere.
Cell sorting
Two Hep2 cell subpopulations were identified using the combination of three antibodies: CD44, phycoerythrin (PE; BD Biosciences, San Jose, CA, USA); CD117, fluorescein isothiocyanate (FITC; BD Biosciences); and CD133, allophyllocyanine (APC; Miltenyi Biotec, Bergisch Gladbach, Germany), and sorted by fluorescence-activated cell sorting (FACS) using FACSAria Fusion equipment (BD Biosciences) and FACSDiva Software Version 6.1.3 for analysis. Positively labeled cells (CD44+/CD117+/CD133+) were classified as laryngeal cancer stem cells (LCSCs), and negatively labeled cells (CD44-/CD117-/CD133-) were considered laryngeal cancer non-stem cells (non-LCSCs). Both cell subpopulations were cultured in DMEM to obtain enough cells for subsequent analysis.
Invasion assay
Quantitative analysis of invasive potential was performed using Matrigel invasion chambers with 8 µm PET membranes in 24-well plates (Corning® BioCoat™, Corning Inc., Corning, NY, USA). Cells were seeded in the upper compartment of the transwell chamber at a density of 2×104 cells per insert in 100 µL serum-free DMEM. Well bottoms were filled with 750 μL DMEM supplemented with 10% FBS, which acts as a chemoattractant. Cells were then incubated for 24 h at 37°C. Cells that invaded the lower membrane surface were fixed with 4% paraformaldehyde for 20 min and stained with 5% Giemsa for 10 min. Four fields were photographed from each insert at 100× magnification using an Olympus BX53 Microscope (Olympus Life Science, Waltham, MA, USA), and the cells were counted.
Sphere-forming assay
Clonogenicity characteristics were evaluated by observing the capacity of cells to generate tumor spheres. LCSC and non-LCSC cells were cultured in low-adherence 6-well plates (Ultra-low Attachment Plates, Corning) in triplicates. Then, 1×104 cells/well were cultured in DMEM without FBS and supplemented with 10 ng/mL EGF, 10 ng/mL fibroblast growth factor, and 1% antibiotic/antimycotic solution. Cells were incubated at 37°C in a humidified atmosphere of 5% CO2 for 5 days (120 h). The former colonies were counted and photographed.
Treatments and MTS assay
Cell viability was determined colorimetrically by MTS assay using the Cell Titer 96 Aqueous One Solution Cell Proliferation Assay (Promega, Madison, WI, USA), following the manufacturer instructions. A total of 5×103 cells were seeded into 96-well plates and treated with 0.37 mg/mL 5-fluorouracil, 2.0 mg/mL cisplatin, or 0.06 mg/mL cetuximab combined with 0.05 mg/mL paclitaxel. After 24 h of treatment, cell viability was determined by absorbance analysis on an ELISA plate reader (Multiskan FC; Thermo Fisher Scientific - Uniscience, São Paulo, Brazil) at 490 nm.
Gene expression
RNA was extracted from 1×106 cells by cell lysis with 750 μL Trizol® (Invitrogen, Carlsbad, CA, USA), according to the manufacturer’s instructions. The RNA concentration was estimated using the Qubit™ RNA HS Assay Kit with the Qubit® 2.0 Fluorometer (Life Technologies). Total RNA (1 μg) was reverse transcribed into cDNA using the High-Capacity cDNA Reverse Transcription Kit (Applied Biosystems™, Foster City, CA, USA). For real-time PCR, TaqMan™ (Applied Biosystems™) probes for the TrkB (HS00178811_m1), KRAS (HS00364284_g1), HIF-1α (HS00153153_m1), and VEGFA (HS00900055_m1) genes were used in custom microplates using the TaqMan™ Universal Master Mix (Thermo Fisher Scientific, Waltham, MA, USA) and the CFX96 Touch™ Deep Well Real-Time PCR Detection System (BioRad, Hercules, CA, USA). The comparative expression level of each condition was calculated as 2-ΔΔCt (ΔΔCt1 method). The Ct values of the samples and controls were normalized by the amount of β-actin and GAPDH.
Protein expression
Proteins were extracted using Trizol® (Invitrogen,) and the concentration was estimated using Pierce™ BCA Protein Assay Kit (Thermo Fisher Scientific, Rockford, USA), according to the manufacturer’s instructions. The expression levels of KRAS, TrkB, HIF-1α VEGF-A, and β-actin were also measured by western blot analysis.
Western blotting
Equal amounts of proteins were loaded on 12% SDS-PAGE gels and subsequent electrophoretic transfer was performed on iBlotR Gel Transfer Stacks PVDF, Regular (Invitrogen by Thermo Fisher Scientific). Blocking was done for 1 h in 3% BSA in 0.5% Tris buffered saline (TBS)-T; primary antibody was in 3% BSA in 0.5% TBS-T or PBS and incubated at 4°C overnight. Then, HRP-conjugated secondary antibodies in 3% BSA in 0.5% TBS-T were incubated at room temperature for 1 h. Enhanced chemiluminescence reagent (Invitrogen by Thermo Fisher Scientific) was used to detect immuno-reactive secondary antibodies still bound to the membrane.
These data were quantified to evaluate band intensity of mean grey values by densitometric analysis using ImageJ v4.0 software, and the relative expression levels of the samples and controls were normalized by the internal standard β-actin [27,28].
Enzyme-linked immunosorbent assay (ELISA)
The ELISA sandwich assay was utilized because the western blot of VEGF-A protein did not present good results. Thereby, specific Quantikine™ ELISA kits used were human VEGF-A (R&D Systems) according to protocol manufacturer’s instructions. The plate was read at 450 nm. Data capture for the colorimetric ELISA assays was performed with an ELISA plate reader (Multiskan FC).
Statistical analysis
Results were expressed independently as the mean ± standard deviation. Functional assays (sphere-forming and invasion assay) were compared by one-way variance analysis (ANOVA) with the Bonferroni correction, treatment and protein expressions were compared by two-way variance analysis (ANOVA) with the Bonferroni correction and gene were compared by t-test analysis. Analyses were performed using the GraphPad PRISM 8 software. Significance was set at P<0.05.
Results
CD44+/CD133+/CD117+ subpopulation has cancer stem cell properties
Cells from a Hep2 cell line were sorted using the set of CD44, CD133, and CD117 biomarkers. LCSCs were representative in 0.8% of cells, whereas non-LCSCs were representative in 4.8% of cells (Figure 1). The invasive potential of the LCSC and non-LCSC subpopulations was evaluated in vitro. Figure 2 shows increased invasive capacity of the LCSCs when compared with the non-LCSC subpopulation after 24 h; LCSCs have a significantly higher invasive potential than non-LCSCs (P=0.0022).
Figure 1.
Cell sorting graphics with CD44, CD117, and CD133 in FACSAria Fusion using FACSDiva Software. Cells in quadrants above 103 (P2, P3, and P4) were considered positive for the markers, and cells in quadrants below 103 (P5, P6, and P7) were considered negative for the markers. The positive cells for FITC-CD117 (P2) were selected from these cells, then those that were positive for the marker PE-CD44 (P3) were selected, and then those positive for the APC-CD133 (P4) were selected. This formed the triple cell positive group for the three tumor stem cell biomarkers. For triple cell negative group, we selected negative cells for FITC-CD117 (P5) fom the quadrants below 103, then selected those that were negative for PE-CD44 (P6), then those that were negative for APC-CD133 (P7).
Figure 2.
Cell invasion assay of LCSC and non-LCSC subpopulations of the Hep2 cell line. Cells were seeded in matrigel inserts and cultured for 24 h. A. LCSC subpopulation; B. non-LCSC subpopulation. Grayscale pictures at 24 h were observed under an optical microscope (×100). The arrows point to the cells that invaded through the matrigel insert. C. Graphic showing the comparative between LCSCs and non-LCSCs invasion. Analysis were performed in triplicate and *P<0.05 versus non-LCSCs. Statistically significant difference was determined using one-way ANOVA with Bonferroni corrections.
The colony-forming assay was conducted for the LCSC and non-LCSC subpopulations of the Hep2 cell line (Figure 3). Clone formation was quantified, and LCSCs presented more colonies than non-LCSCs (p=0.0117).
Figure 3.
Colony-forming LCSC and non-LCSC subpopulations of the Hep2 cell line. The cells were seeded in ultra-low attachment surface 6-well plates and cultured for five days (120 h). (A) Non-LCSC and (C) LCSC subpopulations at 0 h; (B) Non-LCSC subpopulation after five days; and (D) colonies formed in the Hep2 LCSC subpopulation after five days. Grayscale pictures at 120 h were observed under a phase contrast microscopy (×100). (E) Graphic showing the comparative between LCSCs and non-LCSCs invasion. Analysis were performed in triplicate and *P<0.05 versus non-LCSCs. Statistically significant difference was determined using one-way ANOVA with Bonferroni corrections.
LCSCs are treatment-resistant
The results showed no statistical differences between LCSCs and non-LCSCs when treated with 5-fluorouracil, but statistically significant differences were found with cisplatin (P=0.0024) as well as cetuximab combined with paclitaxel (P=0.0069) treatments (Figure 4A). LCSCs had higher viability than non-LCSCs. Furthermore, cetuximab and paclitaxel combination treatment had a greater influence on subpopulation elimination than 5-fluorouracil and cisplatin treatments (Figure 4B and 4C).
Figure 4.
Cell viability after 24 h in Hep2. (A) LCSC and non-LCSC subpopulations treated with 5-fluorouracil, cisplatin, and the combination of cetuximab and paclitaxel. Comparison of responses to 5-fluorouracil, cisplatin, and the combination of cetuximab and paclitaxel in Hep2 LCSC and non-LCSC subpopulations *P≤0.05 versus non-LCSCs. Data and p-values are shown for the comparison between treatments with others in (B) LCSCs. ***P≤0.0001 comparison one treatment with others. Data and p-values are shown for the comparison between treatments with others in (C) non-LCSCs *p≤0.05; ** P≤0.001; ***P≤0.0001 comparison one treatment with others. Statistically significant difference was determined using one-way ANOVA with Bonferroni corrections.
High KRAS, HIF-1α and VEGF-A gene expression in LCSC subpopulation
The KRAS, HIF-1α and VEGF-A genes presented up-regulation in LCSCs compared with non-LCSCs (Figure 5). The TrkB gene showed delayed expression in both subpopulations of the Hep2 cell line.
Figure 5.
Graph showing the relative values of differential expression of the KRAS, TRKB, HIF1α and VEGFA genes, comparing LCSC with control. Statistically significant was determined using one sample t-test analysis compared with a hypothetical mean (1).
High KRAS, TrkB, HIF-1α and VEGF-A protein expression in LCSC subpopulation
From Western blot assay, KRAS, TrkB, HIF-1α and VEGF-A protein expression was up-regulated in LCSCs compared with non-LCSCs (Figure 6A, 6B). Moreover, ELISA assay showed that LCSCs had higher VEGF-A protein expression than non-LCSCs (Figure 6C).
Figure 6.
Protein expression data. (A) Subjected to western blot analysis of TrkB, HIF-1α, KRAS, VEGF-A and β-actin expression (B) Histogram showing quantitative fold change in protein expression normalized to β-actin expression by Image J analysis. (C) ELISA assay graph showing VEGF-A protein expression levels in both cell subpopulations in triplicate. *P<0.05 versus non-LCSCs using 2 way ANOVA with Bonferroni corretions.
Discussion
In our previous study, we showed that CD44+/CD133+/CD117+ cells, classified as LCSCs and obtained from a Hep2 cell line, presented 81% more migration capacity than CD44-/CD13-/CD117- cells, designated as non-LCSCs [29]. In the current study, we found that CD44+/CD133+/CD117+ has cancer stem cell properties, similar to our previous study. Furthermore, we confirmed CSC presence in subpopulation using the invasion and colony-forming assays. The results of these assays demonstrated an increased tumorigenic potential in the LCSC subpopulation of the Hep2 cell line.
Regarding treatment, we found that 5-fluorouracil was ineffective at eliminating either subpopulation. The LCSC subpopulation demonstrated greater resistance to cisplatin and the combination of cetuximab and paclitaxel compared with the non-LCSC subpopulation of the Hep2 cell line. Moreover, the cetuximab and paclitaxel combination treatment was most effective in both subpopulations compared to the other treatments, especially in the non-LCSC subpopulation. Previously, our research group demonstrated that individual cetuximab and paclitaxel treatments showed no statistical differences between LCSCs and non-LCSCs from the Hep2 cell line [29]. These drugs were chosen because they are the most commonly used to treat head and neck cancer (HNC) patients. Cisplatin reacts with DNA to produce crosslinks, and 5-fluorouracil is an antineoplastic antimetabolite; both drugs impair DNA replication and transcription [30,31]. Cetuximab is a monoclonal antibody that functions by blocking EGF from binding to EGFR [18], thereby interrupting the cascade that activates KRAS [15]. Paclitaxel is a chemotherapeutic that inhibits mitotic spindle fiber dynamics [32].
Our results align with those of other studies performed in CSCs from head and neck cancers, which showed resistance to 5-fluorouracil, cisplatin, and cetuximab when used individually [33-36]. Grau et al. [33] observed cisplatin and cetuximab resistance in CSCs from head and neck carcinoma squamous cell (HNCSC) lines that had high expression of the CD44 biomarker. It has also been shown that CSCs from HNCSC cell lines, which used Aldehyde dehydrogenases (ALDH) as a biomarker, were resistant to 5-fluorouracil, cisplatin, and etoposide [35]. Other studies in HNCSC cell lines, which were conducted with FACS to isolate CSCs using both CD44 and ALDH biomarkers, also showed resistance to docetaxel, cetuximab, and PI3K inhibitor (ZSTK474 and PX-866) in these subpopulations, in addition to radiation, photon irradiation (2 Gy/min), and carbon ion irradiation (75MeV/n) resistance [34,36]. In contrast, CSCs from HNCSC cell lines sorted with CD44high/EGFRlow presented sensitivity to cisplatin, cetuximab, gefitinib, and radiation compared to CD44high/EGFRhigh cells [37].
The combined therapy with cetuximab and paclitaxel has been evaluated in head neck cancer clinical studies, with a better response found in oral cancer patients [38]. Furthermore, improved progression-free survival and overall survival have been observed in patients with head and neck cancer [39] mainly after failure of platinum therapy [40].
To our knowledge, to date, there are no studies on combination therapy in LCSCs. Herein, we hypothesized that the combined action of cetuximab and paclitaxel drugs may contribute to eliminating LCSCs, consequently reducing tumor aggressiveness and recurrence. Cetuximab does not have apoptosis-inducing activity; similar to our study, other researchers also observed that cetuximab might be acting as an enhancer of the paclitaxel possibly by induced apoptosis [41]. However, the precise action mechanism of cetuximab and paclitaxel combined treatment responsible for the antitumor effects is still not clear [41].
Similar to our findings, other studies using in vitro and in vivo models have demonstrated that drug combinations, related to the EGFR inhibition pathway combined with other treatments such as tyrosine kinase inhibition, immunotherapy or radiation, have higher therapeutic effectiveness in cancer stem cells of the head and neck cancer [35,42]. Studies using in vivo models have shown that the tyrosine kinase receptors crosstalk with each other and the ligands are able to bind with other receptors to activate the signaling pathways [35,42] that can activate the KRAS gene, resulting in tumor relapse and chemotherapeutic resistance [19,20]. Further studies of combination therapy related to surface biomarkers are required to better understand the therapy response in LCSCs in in vivo models to improve clinical outcomes.
This is the first study to evaluate TrkB and KRAS gene expression in CSC and non-CSC subpopulations of head and neck cancer. Considering the role of these two genes in cell proliferation, we expected that both genes would be overexpressed in the Hep2 cell line, especially in the CSC subpopulation; however, the TrkB gene was not expressed. Recently, TrkB and BDNF were found to be expressed in 30-50% of human HNCSCs [43-45]. One limitation in our study is that only one cell line was assessed; hence, results may not be representative. Therefore, studies with a larger sample size are needed, since TrkB activation has been associated with cell migration, invasion, EMT, cisplatin resistance, and poor prognosis in vivo [43,44,46-48]. Indeed, some studies in head and neck cancer have shown that TrkB inhibition can suppress tumor growth, cell proliferation, and migration, as well as sensitize cells to cisplatin [43,49-52]. In the present study, KRAS gene and protein presented high expression, which may be explained by EGFR-mediated signaling responsible for phosphorylating and activating KRAS, as shown in Figure 7 (adapted from [24,53]). In our previous study, we observed EGFR gene overexpression in LCSCs from the Hep2 cell line [29]; therefore, we suggested that this CSC subpopulation may contribute via EGFR-signaling to promote tumor cell growth, chemotherapy resistance, invasion, and migration, resulting in head and neck cancer progression.
Figure 7.
Summarized molecular mechanisms of the signaling pathway involving the EGFR, TrkB, KRAS HIF-1α and VEGF-A genes; adapted from [24,53]. 1) Phosphorylation resulting from BDNF/TrkB binding can also activate KRAS; however, TrkB gene expression was not found in either subpopulation. This suggests that only EGFR is activating the KRAS gene. 2) Phosphorylation resulting from EGFR/EGF binding activates KRAS, which leads to cell proliferation. The results of our present and previous studies showed high KRAS and EGFR expression in the CSC subpopulation [29]. 3) Only cetuximab [29] binds with EGFR, which blocks EGFR/EGF binding; we suggest that this isolated treatment does not inhibit KRAS inactivation. 4) KRAS gene can active HIF1α and consequently VEGF-A. In this study, these genes were highly expressed in the CSC subpopulation. 5) VEGF-A/VEGFR binding can activate KRAS by the PCLy pathway leading to vascular proliferation. Created with BioRender.com.
In the present study, we showed that the KRAS gene leads to activation of the HIF-1α and VEGF-A genes; all genes and proteins were up-regulated in the LCSC subpopulation. These high expressions are related to CSC features, such as more migration, invasion, colony forming, chemotherapy resistance, and angiogenesis, which lead to metastasis and poor prognosis. The molecular mechanism for explaining this relationship is still unclear, but one limitation of our study is that we evaluated only gene and protein expression and not mutations in the KRAS gene. However, it has been found that different KRAS alterations can be activated to signaling pathways with distinct impacts [25]. The ASP13 mutation in the KRAS gene leads to increased expression of the VEGF-A gene even in the absence or low expression of the HIF-1α gene [25,54]. The underlying molecular mechanisms responsible for the differential overexpression of VEGF-A may be mediated by a distinct activation of the Raf-ERKs pathway and AP2/Sp1 elements in the proximal VEGF-A promoter [25] mainly induced by EGF [55]. Additionally, the CYS12 mutation in the KRAS gene promotes HIF-1α-dependent induction of glycolytic enzymes, supporting the role of HIF-1α in changing tumor metabolism [25,56,57].
The KRAS oncogene has been reported to increase VEGF-A expression in different tumor types [58]. Moreover, mutations in the KRAS gene have been associated with PI3k-dependent up-regulation of VEGF-A in colon tumors [59]. Another study did not observe any association between KRAS mutation status and individual expression of VEGF-A, but showed that up-regulation of VEGF-A can be associated with different types mutation in the EGFR gene [60]. Subsequently, a study also evaluated tumor stem cells in glioma carcinoma and found elevated levels of VEGF-A gene and protein expression under normal and hypoxia conditions compared to the non-tumor stem cell population [61]. We suggest that the up-regulation of VEGF-A in cancer stem cells may be associated with signaling of the KRAS, which may be associated with HIF-1α-dependent KRAS downstream signaling by different types of EGFR mutations in head and neck cancer.
Our results, although limited, suggest for the first time that the combined action of cetuximab and paclitaxel drugs may be more efficient at eliminating CSC subpopulations classified by CD44, CD117, and CD133 biomarkers of a laryngeal cancer cell line than isolated therapies. We provide evidence that higher KRAS expression in LCSCs could contribute to aggressive tumor behavior and poor prognosis in LC. Thus, understanding of the molecular mechanisms that control CSCs proliferation may contribute to better strategies for treating head and neck cancer. Future clinical studies with patients with laryngeal cancer undergoing treatment with cetuximab and paclitaxel are important for further understanding our current findings. In addition, evaluating the expression and mutations of the KRAS gene in these patients can assist in developing specific protocols to stop tumor aggression and improve the prognosis.
Acknowledgements
This study was financed in part by the Coordenação de Aperfeiçoamento de Pessoal de Nível Superior - Brazil (CAPES) - Finance Code 001, The Brazilian National Council for Scientific and Technological Development (CNPq), grant #310582/2014-8, and grant #2016/20087-1, Nº #2015/04403-8 #2014/15009-6, São Paulo Research Foundation (FAPESP). Support was also provided by FAMERP/FUNFARME. The authors thank Carlos Henrique Viesi do Nascimento, Lennon Pereira Caires, Maria Antonia dos Santos Bezerra for technical support, and to Prof. Adília Maria Pires Sciarra (PhD) for support with the English language.
Disclosure of conflict of interest
None.
Abbreviations
- BDNF
Brain-derived neurotrophic factor
- CSCs
Cancer stem cells
- DMEM
Dulbecco’s modified Eagle medium
- EGF
Epidermal growth factor
- EGFR
Epidermal growth factor receptor
- EMT
Epithelial-mesenchymal transition
- FBS
Fetal bovine serum
- HRAS
Harvey rat sarcoma
- KRAS
Kirsten rat sarcoma
- LC
Laryngeal cancer
- LCSCs
Laryngeal cancer stem cells
- MTS
3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium
- non-CSCs
Cancer non-stem cells
- non-LCSCs
Laryngeal cancer non-stem cells
- NRAS
Neuroblastoma rat sarcoma
- RAS
Rat sarcoma
- SCF
Stem cell factor
- TrkB
Tropomyosin-related kinase B
- HIF-1α
Hypoxia-Inducible Factor 1 alpha
- VEGF-A
Vascular endothelial growth factor
References
- 1.Instituto Nacional de Câncer José Alencar Gomes da Silva. Coordenação de Prevenção e Vigilância. Estimativa 2018: incidência de câncer no Brasil/Instituto Nacional de Câncer José Alencar Gomes da Silva. 2019. Updated 2019; cited 2019 09/02. Available from: https://www.inca.gov.br/tipos-de-cancer.
- 2.World Health Organization. Cancer Tomorrow. 2018. Cited 2019 01/22. Available from: http://gco.iarc.fr/tomorrow/graphicbar?type=1&population=900&mode=population&sex=0&cancer=39&age_group=value&apc_male=0&apc_female=0.
- 3.Parikh P, Agarwal JP, Chaturvedi P, Vaidya A, Rathod S, Noronha V, Joshi A, Jamshed A, Bhattacharya GS, Gupta S, Desai C, Advani SH, Pai P, Laskar S, Ramesh A, Mohapatra PN, Vaid AK, Deshpande M, Ranade AA, Vora A, Baral R, Hussain MA, Rajan B, Dcruz AK, Prabhash K. Guidelines for treatment of recurrent or metastatic head and neck cancer. Indian J Cancer. 2014;51:6. doi: 10.4103/0019-509X.137896. [DOI] [PubMed] [Google Scholar]
- 4.Sociedade Brasileira de Oncologia Clínica. Cabeça e pescoço metastático e recorrente. 2017. Cited 2019 01/18. Available from: https://www.sboc.org.br/images/diretrizes/diretrizes_pdfs/Cabe%C3%A7a_e_pescoco_meta_vf_2017.pdf.
- 5.Shrivastava S, Steele R, Sowadski M, Crawford SE, Varvares M, Ray RB. Identification of molecular signature of head and neck cancer stem-like cells. Sci Rep. 2015;5:7819. doi: 10.1038/srep07819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Silva Galbiatti-Dias AL, Pavarino EC, Kawasaki-Oyama RS, Maniglia JV, Maniglia EJ, Goloni Bertollo EM. Cancer stem cells in head and neck cancer: a mini review. Cell Mol Biol (Noisy-le-grand) 2015;61:39–43. [PubMed] [Google Scholar]
- 7.Papaccio F, Paino F, Regad T, Papaccio G, Desiderio V, Tirino V. Concise Review: cancer cells, cancer stem cells, and mesenchymal stem cells: influence in cancer development. Stem Cells Transl Med. 2017;6:2115–2125. doi: 10.1002/sctm.17-0138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Colvin H, Mori M. Getting to the heart of the matter in cancer: novel approaches to targeting cancer stem cells. Proc Jpn Acad Ser B Phys Biol Sci. 2017;93:146–154. doi: 10.2183/pjab.93.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Tirino V, Desiderio V, Paino F, De Rosa A, Papaccio F, La Noce M, Laino L, De Francesco F, Papaccio G. Cancer stem cells in solid tumors: an overview and new approaches for their isolation and characterization. FASEB J. 2013;27:13–24. doi: 10.1096/fj.12-218222. [DOI] [PubMed] [Google Scholar]
- 10.Wiechec E, Hansson KT, Alexandersson L, Jonsson JI, Roberg K. Hypoxia mediates differential response to anti-EGFR therapy in HNSCC cells. Int J Mol Sci. 2017;18:943. doi: 10.3390/ijms18050943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Curtarelli RB, Gonçalves JM, Dos Santos LGP, Savi MG, Nör JE, Mezzomo LAM, Rodríguez Cordeiro MM. Expression of cancer stem cell biomarkers in human head and neck carcinomas: a systematic review. Stem Cell Rev Rep. 2018;14:769–784. doi: 10.1007/s12015-018-9839-4. [DOI] [PubMed] [Google Scholar]
- 12.Miettinen M, Lasota J. KIT (CD117): a review on expression in normal and neoplastic tissues, and mutations and their clinicopathologic correlation. Appl Immunohistochem Mol Morphol. 2005;13:205–220. doi: 10.1097/01.pai.0000173054.83414.22. [DOI] [PubMed] [Google Scholar]
- 13.Baillie R, Tan ST, Itinteang T. Cancer stem cells in oral cavity squamous cell carcinoma: a review. Front Oncol. 2017;7:112. doi: 10.3389/fonc.2017.00112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Wang L, Zuo X, Xie K, Wei D. The role of CD44 and cancer stem cells. Methods in Mollecular Biology. 2018;1692:12. doi: 10.1007/978-1-4939-7401-6_3. [DOI] [PubMed] [Google Scholar]
- 15.Kupferman ME, Jiffar T, El-Naggar A, Yilmaz T, Zhou G, Xie T, Feng L, Wang J, Holsinger FC, Yu D, Myers JN. TrkB induces EMT and has a key role in invasion of head and neck squamous cell carcinoma. Oncogene. 2010;29:2047–2059. doi: 10.1038/onc.2009.486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Amatu A, Sartore-Bianchi A, Siena S. NTRK gene fusions as novel targets of cancer therapy across multiple tumour types. ESMO Open. 2016;1:e000023. doi: 10.1136/esmoopen-2015-000023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Bahrami A, Hassanian SM, ShahidSales S, Farjami Z, Hasanzadeh M, Anvari K, Aledavood A, Maftouh M, Ferns GA, Khazaei M, Avan A. Targeting RAS signaling pathway as a potential therapeutic target in the treatment of colorectal cancer. J Cell Physiol. 2018;233:2058–2066. doi: 10.1002/jcp.25890. [DOI] [PubMed] [Google Scholar]
- 18.Fujiwara T, Eguchi T, Sogawa C, Ono K, Murakami J, Ibaragi S, Asaumi JI, Okamoto K, Calderwood SK, Kozaki KI. Anti-EGFR antibody cetuximab is secreted by oral squamous cell carcinoma and alters EGF-driven mesenchymal transition. Biochem Biophys Res Commun. 2018;503:1267–1272. doi: 10.1016/j.bbrc.2018.07.035. [DOI] [PubMed] [Google Scholar]
- 19.Manchado E, Weissmueller S, Morris JP 4th, Chen CC, Wullenkord R, Lujambio A, de Stanchina E, Poirier JT, Gainor JF, Corcoran RB, Engelman JA, Rudin CM, Rosen N, Lowe SW. A combinatorial strategy for treating KRAS-mutant lung cancer. Nature. 2016;534:647–651. doi: 10.1038/nature18600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Samatar AA, Poulikakos PI. Targeting RAS-ERK signalling in cancer: promises and challenges. Nat Rev Drug Discov. 2014;13:928–942. doi: 10.1038/nrd4281. [DOI] [PubMed] [Google Scholar]
- 21.Zang J, Li C, Zhao LN, Shi M, Zhou YC, Wang JH, Li X. Prognostic value of vascular endothelial growth factor in patients with head and neck cancer: a meta-analysis. Head Neck. 2013;35:1507–1514. doi: 10.1002/hed.23156. [DOI] [PubMed] [Google Scholar]
- 22.Gong L, Zhang W, Zhou J, Lu J, Xiong H, Shi X, Chen J. Prognostic value of HIFs expression in head and neck cancer: a systematic review. PLoS One. 2013;8:e75094. doi: 10.1371/journal.pone.0075094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Meng L, Liu B, Ji R, Jiang X, Yan X, Xin Y. Targeting the BDNF/TrkB pathway for the treatment of tumors. Oncol Lett. 2019;17:2031–2039. doi: 10.3892/ol.2018.9854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Fernandes GMM, Castanhole-Nunes MMU, Rodrigues-Fleming GH, Netinho JG, Pavarino EC, Goloni-Bertollo EM. A summary of the main biomarkers for diagnosis and prognosis of sporadic colorectal cancer: a review. Adv Res Gastroentero Hepatol. 2019;11:7. [Google Scholar]
- 25.Figueras A, Arbos MA, Quiles MT, Viñals F, Germà JR, Capellà G. The impact of KRAS mutations on VEGF-A production and tumour vascular network. BMC Cancer. 2013;13:125. doi: 10.1186/1471-2407-13-125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Huang J, Lu Z, Xiao Y, He B, Pan C, Zhou X, Xu N, Liu X. Inhibition of Siah2 ubiquitin ligase by vitamin K3 attenuates chronic myeloid leukemia chemo-resistance in hypoxic microenvironment. Med Sci Monit. 2018;24:727–735. doi: 10.12659/MSM.908553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Schneider CA, Rasband WS, Eliceiri KW. NIH Image to ImageJ: 25 years of image analysis. Nat Methods. 2012;9:671–675. doi: 10.1038/nmeth.2089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Hossein D. Quantifications of Western Blots with ImageJ. 2017. Available from: http://www.yorku.ca/yisheng/Internal/Protocols/ImageJ.pdf.
- 29.Silva Galbiatti-Dias AL, Fernandes GMM, Castanhole-Nunes MMU, Hidalgo LF, Nascimento Filho CHV, Kawasaki-Oyama RS, Ferreira LAM, Biselli-Chicote PM, Pavarino ÉC, Goloni-Bertollo EM. Relationship between CD44 (high)/CD133 (high)/CD117 (high) cancer stem cells phenotype and Cetuximab and Paclitaxel treatment response in head and neck cancer cell lines. Am J Cancer Res. 2018;8:1633–1641. [PMC free article] [PubMed] [Google Scholar]
- 30.Jordan VC. A retrospective: on clinical studies with 5-fluorouracil. Cancer Res. 2016;76:767–768. doi: 10.1158/0008-5472.CAN-16-0150. [DOI] [PubMed] [Google Scholar]
- 31.Aoki K, Ogawa T, Ito Y, Nakashima S. Cisplatin activates survival signals in UM-SCC-23 squamous cell carcinoma and these signal pathways are amplified in cisplatin-resistant squamous cell carcinoma. Oncol Rep. 2004;11:375–379. [PubMed] [Google Scholar]
- 32.Weaver BA. How taxol/paclitaxel kills cancer cells. Mol Biol Cell. 2014;25:2677–2681. doi: 10.1091/mbc.E14-04-0916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Grau JJ, Mesia R, de la Iglesia-Vicente M, Williams ES, Taberna M, Caballero M, Larque AB, de la Oliva J, Cordon-Cardo C, Domingo-Domenech J. Enrichment of cells with cancer stem cell-like markers in relapses of chemoresistant patients with locally advanced head and neck squamous cell carcinoma. Oncology. 2016;90:267–272. doi: 10.1159/000445118. [DOI] [PubMed] [Google Scholar]
- 34.Keysar SB, Le PN, Miller B, Jackson BC, Eagles JR, Nieto C, Kim J, Tang B, Glogowska MJ, Morton JJ, Padilla-Just N, Gomez K, Warnock E, Reisinger J, Arcaroli JJ, Messersmith WA, Wakefield LM, Gao D, Tan AC, Serracino H, Vasiliou V, Roop DR, Wang XJ, Jimeno A. Regulation of head and neck squamous cancer stem cells by PI3K and SOX2. J Natl Cancer Inst. 2017;109:djw189. doi: 10.1093/jnci/djw189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Leong HS, Chong FT, Sew PH, Lau DP, Wong BH, Teh BT, Tan DS, Iyer NG. Targeting cancer stem cell plasticity through modulation of epidermal growth factor and insulin-like growth factor receptor signaling in head and neck squamous cell cancer. Stem Cells Transl Med. 2014;3:1055–1065. doi: 10.5966/sctm.2013-0214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Moncharmont C, Guy JB, Wozny AS, Gilormini M, Battiston-Montagne P, Ardail D, Beuve M, Alphonse G, Simoens X, Rancoule C, Rodriguez-Lafrasse C, Magne N. Carbon ion irradiation withstands cancer stem cells’ migration/invasion process in Head and Neck Squamous Cell Carcinoma (HNSCC) Oncotarget. 2016;7:47738–47749. doi: 10.18632/oncotarget.10281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.La Fleur L, Johansson AC, Roberg K. A CD44high/EGFRlow subpopulation within head and neck cancer cell lines shows an epithelial-mesenchymal transition phenotype and resistance to treatment. PLoS One. 2012;7:e44071. doi: 10.1371/journal.pone.0044071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Bernad IP, Trufero JM, Urquizu LC, Pazo Cid RA, de Miguel AC, Agustin MJ, Lanzuela M, Antón A. Activity of weekly paclitaxel-cetuximab chemotherapy in unselected patients with recurrent/metastatic head and neck squamous cell carcinoma: prognostic factors. Clin Transl Oncol. 2017;19:769–776. doi: 10.1007/s12094-016-1604-z. [DOI] [PubMed] [Google Scholar]
- 39.Nakano K, Marshall S, Taira S, Sato Y, Tomomatsu J, Sasaki T, Shimbashi W, Fukushima H, Yonekawa H, Mitani H, Kawabata K, Takahashi S. A comparison of weekly paclitaxel and cetuximab with the EXTREME regimen in the treatment of recurrent/metastatic squamous cell head and neck carcinoma. Oral Oncol. 2017;73:21–26. doi: 10.1016/j.oraloncology.2017.07.022. [DOI] [PubMed] [Google Scholar]
- 40.Patil VM, Noronha V, Joshi A, Agarwala V, Muddu V, Ramaswamy A, Chandrasekharan A, Dhumal S, Juvekar S, Arya A, Bhattacharjee A, Prabhash K. Comparison of paclitaxel-cetuximab chemotherapy versus metronomic chemotherapy consisting of methotrexate and celecoxib as palliative chemotherapy in head and neck cancers. Indian J Cancer. 2017;54:20–24. doi: 10.4103/ijc.IJC_160_17. [DOI] [PubMed] [Google Scholar]
- 41.Harada K, Ferdous T, Kobayashi H, Ueyama Y. Paclitaxel in combination with cetuximab exerts antitumor effect by suppressing NF-κB activity in human oral squamous cell carcinoma cell lines. Int J Oncol. 2014;45:2439–2445. doi: 10.3892/ijo.2014.2655. [DOI] [PubMed] [Google Scholar]
- 42.Macha MA, Rachagani S, Qazi AK, Jahan R, Gupta S, Patel A, Seshacharyulu P, Lin C, Li S, Wang S, Verma V, Kishida S, Kishida M, Nakamura N, Kibe T, Lydiatt WM, Smith RB, Ganti AK, Jones DT, Batra SK, Jain M. Afatinib radiosensitizes head and neck squamous cell carcinoma cells by targeting cancer stem cells. Oncotarget. 2017;8:20961–20973. doi: 10.18632/oncotarget.15468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.de Moraes JK, Wagner VP, Fonseca FP, Vargas PA, de Farias CB, Roesler R, Martins MD. Uncovering the role of brain-derived neurotrophic factor/tyrosine kinase receptor B signaling in head and neck malignancies. J Oral Pathol Med. 2018;47:221–227. doi: 10.1111/jop.12611. [DOI] [PubMed] [Google Scholar]
- 44.Gotz R, Sendtner M. Cooperation of tyrosine kinase receptor TrkB and epidermal growth factor receptor signaling enhances migration and dispersal of lung tumor cells. PLoS One. 2014;9:e100944. doi: 10.1371/journal.pone.0100944. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Li L, Zhu L. Expression and clinical significance of TrkB in sinonasal squamous cell carcinoma: a pilot study. Int J Oral Maxillofac Surg. 2017;46:144–150. doi: 10.1016/j.ijom.2016.09.027. [DOI] [PubMed] [Google Scholar]
- 46.Yilmaz T, Jiffar T, de la Garza G, Lin H, Milas Z, Takahashi Y, Hanna E, MacIntyre T, Brown JL, Myers JN, Kupferman ME. Theraputic targeting of Trk supresses tumor proliferation and enhances cisplatin activity in HNSCC. Cancer Biol Ther. 2010;10:644–653. doi: 10.4161/cbt.10.6.12782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Zhang L, Hu Y, Sun CY, Li J, Guo T, Huang J, Chu ZB. Lentiviral shRNA silencing of BDNF inhibits in vivo multiple myeloma growth and angiogenesis via down-regulated stroma-derived VEGF expression in the bone marrow milieu. Cancer Sci. 2010;101:1117–1124. doi: 10.1111/j.1349-7006.2010.01515.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Sasahira T, Ueda N, Yamamoto K, Bhawal UK, Kurihara M, Kirita T, Kuniyasu H. Trks are novel oncogenes involved in the induction of neovascularization, tumor progression, and nodal metastasis in oral squamous cell carcinoma. Clin Exp Metastasis. 2013;30:165–176. doi: 10.1007/s10585-012-9525-x. [DOI] [PubMed] [Google Scholar]
- 49.Jang JW, Song Y, Kim SH, Kim J, Seo HR. Potential mechanisms of CD133 in cancer stem cells. Life Sci. 2017;184:25–29. doi: 10.1016/j.lfs.2017.07.008. [DOI] [PubMed] [Google Scholar]
- 50.Lee J, Jiffar T, Kupferman ME. A novel role for BDNF-TrkB in the regulation of chemotherapy resistance in head and neck squamous cell carcinoma. PLoS One. 2012;7:e30246. doi: 10.1371/journal.pone.0030246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Tang YL, Fan YL, Jiang J, Li KD, Zheng M, Chen W, Ma XR, Geng N, Chen QM, Chen Y, Liang XH. C-kit induces epithelial-mesenchymal transition and contributes to salivary adenoid cystic cancer progression. Oncotarget. 2014;5:1491–1501. doi: 10.18632/oncotarget.1606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Wang SJ, Bourguignon LY. Hyaluronan and the interaction between CD44 and epidermal growth factor receptor in oncogenic signaling and chemotherapy resistance in head and neck cancer. Arch Otolaryngol Head Neck Surg. 2006;132:771–778. doi: 10.1001/archotol.132.7.771. [DOI] [PubMed] [Google Scholar]
- 53.New approach for understanding genome variation in KEGG. Internet. 2019. Available from: https://www.genome.jp/kegg/kegg1.html Available from: https://www.genome.jp/kegg-bin/show_pathway?map04010 Available from: https://www.genome.jp/kegg-bin/show_pathway?map04066 Available from: and https://www.genome.jp/kegg-bin/show_pathway?map04370. [DOI] [PMC free article] [PubMed]
- 54.Carmeliet P, Jain RK. Molecular mechanisms and clinical applications of angiogenesis. Nature. 2011;473:298–307. doi: 10.1038/nature10144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Pagès G, Pouysségur J. Transcriptional regulation of the vascular endothelial growth factor gene-a concert of activating factors. Cardiovasc Res. 2005;65:564–573. doi: 10.1016/j.cardiores.2004.09.032. [DOI] [PubMed] [Google Scholar]
- 56.Vizan P, Boros LG, Figueras A, Capella G, Mangues R, Bassilian S, Lim S, Lee WN, Cascante M. K-ras codon-specific mutations produce distinctive metabolic phenotypes in NIH3T3 mice [corrected] fibroblasts. Cancer Res. 2005;65:5512–5515. doi: 10.1158/0008-5472.CAN-05-0074. [DOI] [PubMed] [Google Scholar]
- 57.Zhou W, Choi M, Margineantu D, Margaretha L, Hesson J, Cavanaugh C, Blau CA, Horwitz MS, Hockenbery D, Ware C, Ruohola-Baker H. HIF1α induced switch from bivalent to exclusively glycolytic metabolism during ESC-to-EpiSC/hESC transition. EMBO J. 2012;31:2103–2116. doi: 10.1038/emboj.2012.71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Krajnović M, Marković B, Knežević-Ušaj S, Nikolić I, Stanojević M, Nikolić V, Šiljić M, Jovanović Ćupić S, Dimitrijević B. Locally advanced rectal cancers with simultaneous occurrence of KRAS mutation and high VEGF expression show invasive characteristics. Pathol Res Pract. 2016;212:598–603. doi: 10.1016/j.prp.2016.02.018. [DOI] [PubMed] [Google Scholar]
- 59.Kang YH, Kim KS, Yu YK, Lim SC, Kim YC, Park KO. The relationship between microvessel count and the expression of vascular endothelial growth factor, p53, and K-ras in non-small cell lung cancer. J Korean Med Sci. 2001;16:417–423. doi: 10.3346/jkms.2001.16.4.417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Yuan XH, Yang J, Wang XY, Zhang XL, Qin TT, Li K. Association between EGFR/KRAS mutation and expression of VEGFA, VEGFR and VEGFR2 in lung adenocarcinoma. Oncol Lett. 2018;16:2105–2112. doi: 10.3892/ol.2018.8901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Bao S, Wu Q, Sathornsumetee S, Hao Y, Li Z, Hjelmeland AB, Shi Q, McLendon RE, Bigner DD, Rich JN. Stem cell-like glioma cells promote tumor angiogenesis through vascular endothelial growth factor. Cancer Res. 2006;66:7843–7848. doi: 10.1158/0008-5472.CAN-06-1010. [DOI] [PubMed] [Google Scholar]