Transcutaneous carbon dioxide enhances the antitumor effect of radiotherapy on oral squamous cell carcinoma

  • Authors:
    • Eiji Iwata
    • Takumi Hasegawa
    • Takeshi Ueha
    • Daisuke Takeda
    • Izumi Saito
    • Teruya Kawamoto
    • Toshihiro Akisue
    • Yoshitada Sakai
    • Ryohei Sasaki
    • Ryosuke Kuroda
    • Takahide Komori
  • View Affiliations

  • Published online on: May 16, 2018     https://doi.org/10.3892/or.2018.6444
  • Pages: 434-442
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Radiotherapy (RT) is one of the main treatment modalities for oral squamous cell carcinoma (OSCC), however, radioresistance is a major impediment to its clinical success and poses as a concern that needs to be addressed. Tumor hypoxia is known to be significantly associated with radioresistance in various malignancies, hence, resolving the hypoxic state of a tumor may improve the antitumor effect of RT on OSCC. We have previously revealed that transcutaneous CO2 induced mitochondrial apoptosis and suppressed tumor growth in OSCC by resolving hypoxia. Considering the previous study, we hypothesized that transcutaneous CO2 may enhance the antitumor effect of RT on OSCC by improving intratumoral hypoxia, thereby overcoming radioresistance. In the present study, the combination of transcutaneous CO2 and RT significantly inhibited tumor growth compared with other treatments. This combination therapy also led to decreased expression of HIF-1α in parallel with increased expression of the cleaved forms of caspase-3-8-9 and PARP, which play essential roles in mitochondrial apoptosis. Additionally, the combination therapy increased the expression of ROS modulator 1 and subsequent mitochondrial ROS production, compared to RT alone. These results indicated that transcutaneous CO2 could potentially improve the antitumor effect of RT by decreasing the intratumoral hypoxia and increasing the mitochondrial apoptosis. Our findings indicated that CO2 therapy may be a novel adjuvant therapy in combination with RT for OSCC.

Introduction

At present, the main treatments for oral squamous cell carcinoma (OSCC) are surgery, chemotherapy and radiotherapy (RT). RT is a particularly effective strategy for OSCC, since it is employed as both a primary modality and an adjuvant treatment following surgery. Despite modern radiation techniques, many studies have highlighted several complications associated with RT, including bone marrow suppression (13). Disregarding the associated complications, the therapeutic success of RT is also challenged by the development of tumor-cell radioresistance, which can be attributed to cell cycle, differentiation and hypoxia (46). Among these, tumor hypoxia is known to exhibit a significantly strong association with radioresistance (7,8). Therefore, resolving tumor hypoxia could be a key strategy in overcoming radioresistance and improving the antitumor effect of RT.

The importance of improving tumor oxygenation for potential clinical use has already been recognized and some strategies, including the use of radio-sensitizers and hypoxic cytotoxins, have been developed to overcome hypoxia-mediated tumor radioresistance (9). However, these strategies often lead to the development of more severe side-effects (10,11).

Tumor hypoxia is a characteristic feature of malignant tumors, including OSCC (12). In hypoxic conditions, hypoxia inducible factor-1α (HIF-1α), an oxygen-dependent α subunit of HIF, activates the transcription of specific metastatic genes, thus playing an important role in the growth and survival of malignant tumors (13,14). In addition, a recent study has revealed that HIF-1α plays an important role in hypoxia-related tumor radioresistance (15).

We previously revealed that transcutaneous CO2 application caused absorption of CO2 and an increase in the O2 pressure in treated tissues, potentially causing an ‘artificial Bohr effect’ (16). We have also reported that CO2 therapy induced mitochondrial apoptosis and suppressed tumor growth in OSCC by resolving hypoxia (17). Furthermore, we have revealed that transcutaneous CO2 decreased the expression of HIF-1α in OSCC. Considering our previous study, we hypothesized that transcutaneous CO2 may enhance the antitumor effect of RT on OSCC by improving intratumoral hypoxia, thereby overcoming radioresistance.

In the present study, we investigated the antitumor effects of a combination treatment of transcutaneous CO2 with RT by using human OSCC xenograft models in vivo.

Materials and methods

Cell culture

A human OSCC cell line, HSC-3, was used in our study (Health Science Research Resources Bank, Osaka, Japan). The HSC-3 cell line was established from a metastatic deposit of poorly differentiated OSCC of the tongue in a mid-internal jugular lymph node from a 64-year-old man (18). The HSC-3 cells were routinely cultured in Eagle's minimum essential medium (EMEM; Sigma-Aldrich, St. Louis, MO, USA) supplemented with 10% fetal bovine serum (FBS; Sigma-Aldrich) and 1,000 U/ml penicillin/streptomycin solution (Sigma-Aldrich) in an incubator with 5% CO2 at 37°C. Trypsin (0.25%) and ethylenediaminetetraacetic acid (EDTA, 0.02%) (Sigma-Aldrich) solution were used to isolate the cells for subculture, as previously described (17,19).

X-ray irradiation

X-ray irradiation was performed at a dose rate of 0.75–0.78 Gy/min using a 150 kV X-ray generator unit operating at 5 mA with an external 0.1 mm aluminum filter (MBR-1505122; Hitachi Medical Co., Tokyo, Japan).

Colony formation assay

Colony formation assays for the X-ray irradiated HSC-3 cells were performed to evaluate the response of these cells to X-ray irradiation. HSC-3 cells in T-25 flasks (Corning Japan, Tokyo, Japan) were treated by X-ray irradiation at three different doses (0, 2 or 8 Gy). Immediately after X-ray irradiation, the cells were trypsinized, seeded at a density of 1,000 cells/well in 6-well plates, and incubated in a humidified atmosphere of 5% CO2 at 37°C. After 2 weeks of incubation, the cells were stained by Giemsa (Muto Pure Chemicals Co., Tokyo, Japan) and the number of colonies was counted.

Animal models

Twenty-four male athymic BALB/c nude mice, 7-week-old, were obtained from CLEA Japan (Tokyo, Japan). The animals were maintained under pathogen-free conditions, in accordance with the institutional guidelines. All animal experiments were performed in accordance with the Guidelines for Animal Experimentation of Kobe University Animal Experimentation Regulations (permission no. P120602) and were approved by the Institutional Animal Care and Use Committee. HSC-3 cells at doses of 4.0×106 cells in 500 µl phosphate buffered saline (PBS) were implanted into the back of 24 mice to create the human OSCC xenograft models.

Transcutaneous CO2 therapy

Transcutaneous CO2 therapy was performed as previously described (17,19). Briefly, the area of skin around the implanted tumor was covered with CO2 hydrogel. This area was then sealed with a polyethylene bag, and 100% CO2 gas was administered into the bag. Treatment was performed for 20 min each, twice a week for 2 weeks. Animals in the control group were treated in a similar manner by replacing CO2 with room air.

In vivo studies

The mice implanted with HSC-3 cells were randomly divided into four groups: control group (n=6), RT group (n=6), CO2 group (n=6) or the combination group (n=6). RT was performed at a dose of 5.0 Gy each and in the RT group, the mice were treated by RT alone twice a week for 2 weeks (20 Gy in total). Mice in the CO2 group were treated by transcutaneous CO2 therapy alone. In the combination group, the mice were treated by CO2 therapy, immediately followed by RT. Tumor volume and body weight were monitored and calculated as previously described (17,19).

Treated tumors were removed 24 h after the final treatment and the total RNA and cell lysates were immediately extracted from the half of each tumor. The other half of each tumor was formalin-fixed and paraffin-embedded for staining. Serial 10-μm thick transverse sections were prepared from each block. Bone marrow suppression was investigated by the collection of 10 µl blood samples by inserting a heparinized capillary tube just below the eye ball. The obtained samples were diluted with Turk's solution (190 µl) and the leukocytes (white blood cells) in the samples were counted using a heamocytometer by a clinical laboratory technician.

Fluorescence activated cell scanning (FACS) assay

Flow cytometry was performed using a FACS Calibur™ flow cytometer (BD Pharmingen; BD Biosciences, Franklin Lakes, NJ, USA) as previously described (17). The apoptotic activity in treated tumors was evaluated by DNA fragmentation using the Apo-Direct kit according to the manufacturer's protocol (BD Pharmingen; BD Biosciences). In addition, ROS production was evaluated using anti-human ROS modulator 1 (ROMO-1) antibody (1:1,000; cat. no. ab121379; Abcam, Cambridge, UK) as an indicator. The data were analyzed using BD CellQuest software (BD Biosciences), and apoptotic or ROMO-1 expression cells were quantified as a percentage of the total number of cells.

Immunohistochemical analysis

The formalin-fixed and paraffin-embedded tumor sections were pretreated with citrate buffer for 40 min at 95°C, quenched with 0.05% H2O2 and incubated overnight at 4°C with the following primary antibodies in Can Get Signal immunostain solution A (Toyobo, Osaka, Japan): rabbit anti-human HIF-1α antibody (1:1,000; cat. no. ab82832; Abcam) and anti-human ROS modulator 1 (ROMO-1) antibody (1:1,000; cat. no. ab121379; Abcam). Following the treatment, the sections were incubated with horseradish peroxidase (HRP)-conjugated goat anti-rabbit IgG polyclonal antibody (1:1,000; cat. no. 6721; Nichirei Bioscience, Tokyo, Japan) for 30 min at room temperature. Signals were developed as a brown reaction product using peroxidase substrate 3′,3′-diaminobenzidine (Nichirei Bioscience). The sections were counterstained with hematoxylin and examined with a BZ-8000 confocal microscope (Keyence, Osaka, Japan). Immunohistochemical-positive staining was semi-quantified by densitometric analysis using the ImageJ software, version 1.47 (National Institutes of Health, Bethesda, MD, USA) (http://rsbweb.nih.gov/ij/download.html). Values were normalized against the control group and were presented as ratios.

Immunofluorescence staining

To assess the apoptotic activity in treated tumors, we performed the immunofluorescence staining by using the Apo-Direct kit following the manufacturer's protocol (BD Biosciences). The nucleus was stained with DAPI. The images were captured using a BZ-8000 confocal microscope (Keyence).

Immunoblot analysis

The cell lysates were prepared from treated tumors by using a whole cell lysis buffer supplemented with Halt protease and phosphatase inhibitor cocktail (Mammalian Protein Extraction Reagent; Thermo Fisher Scientific, Rockford, IL, USA). The protein samples were processed using standard western immunoblot procedures. The membranes were incubated overnight at 4°C with the following primary antibodies in Can Get Signal Immunoreaction Enhancer Solution 1 (Toyobo): anti-human caspase-8 antibody (1:1,000; cat. no. 9496; Cell Signaling Technology, Danvers, MA, USA), anti-human caspase-9 antibody (1:1,000; cat. no. 7237; Cell Signaling Technology), anti-human caspase-3 antibody (1:1,000; cat. no. 9664; Cell Signaling Technology), anti-human PARP antibody (1:1,000; cat. no. 5625; Cell Signaling Technology) and anti-human α-tubulin antibody (1:2,000; cat. no. 6074; Sigma-Aldrich). After washing, the membranes were incubated with the appropriate secondary antibody conjugated to horseradish peroxidase (GE Healthcare Bio-Sciences, Piscataway, NJ, USA) in Can Get Signal Immunoreaction Enhancer Solution 2 (Toyobo) and exposed with ECL Prime Plus Western Blotting Detection System Reagent (GE Healthcare Bio-Sciences, Pittsburgh, PA, USA). The signals were detected using a Chemilumino analyzer LAS-3000 mini (Fujifilm, Tokyo, Japan). Positive bands in the immunoblots were semi-quantified by densitometric analysis using ImageJ software version 1.47. The values were normalized against α-tubulin and were presented as ratios.

Statistical analysis

StatView J-4.5 software (Hulinks, Inc., Tokyo, Japan) was used for statistical analysis performed on all data for four groups by the Scheffe's test. Data was presented as the mean value ± standard error. P-value <0.05 was considered to indicate a statistically significant difference.

Results

RT induces ROS-related apoptosis in human HSC-3 cells in vitro

Colony formation assays for HSC-3 cells irradiated by various doses of X-rays were performed to evaluate the response of these cells to RT. The number of HSC-3 cell colonies decreased dose-dependently after RT (Fig. 1A). The half maximal inhibitory concentration (IC50) in colony formation was achieved at a dose of 5.0 Gy (Fig. 1B).

Subsequently, the reliability of ROS-related apoptosis in X-ray irradiated HSC-3 cells in vitro was studied. There was a correlation between the apoptotic activity and the expression of ROMO-1 in the cells irradiated with 5.0 Gy X-rays (Fig. 2).

Transcutaneous CO2 enhances the effect of RT on OSCC tumor growth with no observable side-effects in vivo

The effect of the transcutaneous CO2 therapy on RT was investigated in vivo. Tumor volume in the combination group significantly decreased compared with that in the control group (P<0.01), in the RT group (P<0.05) and in the CO2 group (P<0.05). At the end of the experiment, tumor volume in the combination group was reduced to 23, 38 and 37% of that in the control, RT, and CO2 groups, respectively (Fig. 3). In addition, significant decrease in tumor volume was observed in the CO2 and the RT groups compared with the control group (P<0.05). No significant difference in body weight was observed among the four groups (Fig. 4A), however the white blood cell number in the RT group was significantly decreased compared with that in the control and the CO2 group (Fig. 4B).

Transcutaneous CO2 with RT suppresses tumor growth by inducing apoptosis and ROS production in HSC-3 cells in vivo

Subsequently, we examined the in vivo effects of transcutaneous CO2 with RT on the expression of HIF-1α, the apoptotic activity and the production of ROS in HSC-3 cells. Immunohistochemical positive staining for HIF-1α was hardly detectable in the CO2 and the combination group compared with the other two groups (Fig. 5A). In contrast, positive staining for ROMO-1 (indicative of ROS production) was significantly higher in the combination group compared with that in the other three groups (Fig. 5B). Furthermore, immunofluorescence staining revealed that apoptotic activity was increased in both the CO2 and the combination groups (Fig. 6A). Quantitative analysis of the immunofluorescence-positive staining revealed that apoptotic cells were significantly increased. Relative positive staining compared to the control was increased 19.0-, 46.0-, and 66.0-fold in the CO2, RT and combination groups, respectively (Fig. 6B).

Immunoblot analysis revealed an increased protein expression of the cleaved forms of caspase-3 and PARP in the combination group compared with the other groups (Fig. 7A)

The protein expression of cleaved caspase-8 was increased in the RT and the combination group, compared with that in the other two groups, whereas increased protein expression of cleaved caspase-9 was observed in the CO2 and the combination group (Fig. 7A). Positive bands in the immunoblots were semi-quantified by densitometric analysis based on the concentrations of α-tubulin: caspase-3 (0.22, 0.62, 0.71, 0.89), caspase-8 (0.39, 0.41, 0.83, 0.93), caspase-9 (0.41, 0.79, 0.41, 1.00) and PARP (0.22, 0.53, 0.52, 0.93) in the control, CO2, RT and combination group, respectively (Fig. 7B).

Discussion

RT is one of the main treatments for OSCC. However, radioresistance is a recognized obstacle responsible for poor clinical success and patient prognosis. Radioresistance of tumors is caused by cell cycle, differentiation and hypoxia of which, tumor hypoxia is known to demonstrate a strong correlation with radioresistance (48). Hypoxia, characterized by an inadequate supply of oxygen to the tissues, disturbs the radiolysis of H2O and reduces the production of reactive and cytotoxic species, and that radiation-induced DNA damage is fixed under normoxic conditions (20,21).

Recent studies have revealed that HIF-1α plays a major role in hypoxia-related tumor radioresistance (15). Under hypoxic conditions, HIF-1α in tumor cells is activated; this activation promotes tumor growth. In contrast, normoxic conditions induce the proteolysis of HIF-1α and the loss of HIF-1α activity, which results in a dramatic decrease in tumor growth, angiogenesis and cellular energy metabolism (22).

RT results in tumor reoxygenation, upregulated production of ROS and activation and stabilization of HIF-1α in solid tumors, including OSCC (23). The cytokines induced by HIF-1α activate anti-apoptotic signals to tumor vessels, thereby imparting radioresistance to tumor cells (24). Therefore, it stands to reason that blocking either HIF-1α or these cytokines could considerably increase the radiosensitivity of tumor vasculature, thus causing a decreased overall tumor radioresistance (2528). Hence, resolving hypoxia via blocking the activation of HIF-1α may prevent the development of radioresistance and improve the prognosis of OSCC patients.

In support of this, several studies have revealed the correlation between the survival rate and the value of hypoxia in irradiated solid tumors, including OSCC (2933). Several strategies to address hypoxia have been explored, including the use of radio-sensitizers and hypoxic cytotoxins (9). However, when used in combination, these strategies resulted in a significant increase in the rate of both severe radiation tissue injury and the chance of seizures during therapy, presumably due to oxygen (O2) toxicity (10,11). Strategies for improving hypoxia employ 100% O2 or carbogen (95% O2 + 5% CO2), involve direct inhalation and do not inflict any toxicity (34). Breathing carbogen in combination with nicotinamide administration was used to improve the tumor response to accelerated RT in head-and-neck tumors (35).

We have previously reported that a transcutaneous CO2 system could combat hypoxia in treated tissues, potentially resulting in an ‘artificial Bohr effect’ (16). We have also demonstrated that transcutaneous CO2 induced mitochondrial apoptosis and suppressed tumor growth in OSCC by resolving hypoxia (17) and decreasing the expression of HIF-1α in OSCC. In light of these findings, we hypothesized that transcutaneous CO2 could improve hypoxic conditions and enhance the antitumor effect of RT on OSCC. CO2 was used instead of O2 because O2 has an inferior ability to dissolve in tissues and has no apoptotic action. Antitumor effects did not arise when O2 was directly used. Compared to carbogen, CO2 therapy is localized because the tumor is covered directly with CO2 hydrogel and the administered 100% CO2 gas.

In the present study, the combination therapy of transcutaneous CO2 with RT decreased the expression of HIF-1α and significantly suppressed in vivo OSCC tumor growth compared with RT alone. The results indicated that addressing hypoxia by transcutaneous CO2 could mitigate intratumoral radioresistance.

Apoptosis plays an important role in determining the cellular fate (36,37) and is mediated by two distinct cell death pathways: the intrinsic and the extrinsic pathway. The intrinsic cell death pathway is initiated by mitochondrial outer membrane damage, followed by the release of cytochrome c from the mitochondria into the cytoplasm, activation of an initiator caspase, caspase-9, which activates the downstream protein, caspase-3. The extrinsic cell death pathway is initiated when a ligand binds to its receptor causing the activation of an initiator caspase, caspase-8, which then activates the downstream protein caspase-3. In both pathways, caspase-3 is responsible for the cleavage of poly (ADP-ribose) polymerase (PARP) during cell death (3840). In the present study, the expression of cleaved caspase-8 was observed in both the RT and the combination groups, whereas cleaved caspase-9 was observed in both the CO2 and the combination groups. These results indicated that the effect of RT on OSCC results from the activation of the death receptor-mediated pathway, whereas the effect of transcutaneous CO2 occurs via the mitochondrial apoptotic pathway. Furthermore, the combination therapy of transcutaneous CO2 and RT demonstrated a strong antitumor effect mediated by both the death receptor and the mitochondrial apoptotic pathways.

In the present study, CO2 did not induce significant oxidative stress but did induce obvious apoptosis. We previously reported that transcutaneous CO2 inhibited SCC tumor growth in vivo by increasing the number of mitochondria and activating mitochondrial apoptosis by reducing intratumoral hypoxia (17). Mitochondria play important roles in cellular energy metabolism and apoptosis. Mitochondria proliferate independently from cancer cells, and the rates of cancer cell division and proliferation are likely faster than those of mitochondria under hypoxic conditions (41). Therefore, while cancer cells exhibit abnormal proliferation, the number of mitochondria decreases in cancer cells under hypoxic conditions, and mitochondrial dysfunction (the Warburg effect) prevents apoptosis in tumor tissue (42).

The antitumor effect of RT is also impacted by the generation of ROS (43), through a relationship that still requires further clarification. Radiation generates ROS when it passes through biological systems. ROS are very reactive, attack the critical cellular macromolecules such as DNA, and lead to cell damage and death (44). ROS have an important function in cell survival and cell death triggered by tumor necrosis factor-α (TNF-α) signaling, and the main source of these ROS is the mitochondria (45,46). Specifically, ROMO-1 is a mitochondrial membrane protein responsible for TNF-α-induced ROS production (47). ROMO-1 expression is upregulated in most cancer cells and in senescent cells, and it is induced by external stimuli (48). TNF-α treatment triggers the interaction between TNF complex II and the C-terminus of ROMO-1 exposed to the outside of the mitochondrial outer membrane (49). Simultaneously, ROMO-1 reduces the mitochondrial membrane potential, resulting in ROS generation and apoptosis. Thus, the ROS level is significantly increased by ROMO-1 overexpression. In the present study, we observed a correlation between apoptotic activity and ROMO-1 expression in HSC-3 cells after X-ray irradiation in vitro, indicating that RT could induce ROS-related apoptosis in human HSC-3 cells. Additionally, the in vivo overexpression of ROMO-1 was higher in the combination group than that in the RT group. In summary, transcutaneous CO2 may not interfere with ROS-related apoptosis.

In the present study, RT was performed in vivo at the in vitro IC50 dose of 5.0 Gy. Various studies have revealed that mice can be irradiated with different doses (5052). In addition, Fujii et al reported that a single dose of radiation at 5.0 Gy rapidly increased pO2 in SCC compared to other doses (53). Thus, we hypothesized that a combination therapy with CO2 and 5.0 Gy (IC50) would be effective in the present study. However, it may be more appropriate to use a dose other than 5.0 Gy.

In conclusion, this is the first study to demonstrate that transcutaneous CO2 enhanced the antitumor effect of RT on OSCC by improving intratumoral hypoxia, whilst eliciting no observable side-effects. Although further studies are required, transcutaneous CO2 may be a novel adjuvant therapy in combination with RT for OSCC.

Acknowledgements

We would like to thank Ms. Minako Nagata, Ms. Maya Yasuda and Ms. Kyoko Tanaka for their expert technical assistance.

Funding

No funding was received.

Availability of data and materials

The datasets used during the present study are available from the corresponding author upon reasonable request.

Authors' contributions

EI, TH, TU, DT, IS and TKa conceived and designed the study. EI and TU performed the experiments. EI, TH and DT wrote the paper. TU, TKa and RK reviewed and edited the manuscript. TA, YS, RS, RK and TKo supervised all aspects of this study. All authors read and approved the manuscript and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

All animal experiments were performed in accordance with the Guidelines for Animal Experimentation of Kobe University Animal Experimentation Regulations (permission no. P120602) and were approved by the Institutional Animal Care and Use Committee.

Consent for publication

Not applicable.

Competing interests

The authors state that they have no competing interests.

References

1 

Harrison LB, Zelefsky MJ, Pfister DG, Carper E, Raben A, Kraus DH, Strong EW, Rao A, Thaler H, Polyak T, et al: Detailed quality of life assessment in patients treated with primary radiotherapy for squamous cell cancer of the base of the tongue. Head Neck. 19:169–175. 1997. View Article : Google Scholar : PubMed/NCBI

2 

Cooper JS, Fu K, Marks J and Silverman S: Late effects of radiation therapy in the head and neck region. Int J Radiat Oncol Biol Phys. 31:1141–1164. 1995. View Article : Google Scholar : PubMed/NCBI

3 

Verastegui EL, Morales RB, Barrera-Franco JL, Poitevin AC and Hadden J: Long-term immune dysfunction after radiotherapy to the head and neck area. Int Immunopharmacol. 3:1093–1104. 2003. View Article : Google Scholar : PubMed/NCBI

4 

Hockel M, Schlenger K, Aral B, Mitze M, Schaffer U and Vaupel P: Association between tumor hypoxia and malignant progression in advanced cancer of the uterine cervix. Cancer Res. 56:4509–4515. 1996.PubMed/NCBI

5 

Chen XY, Wang Z, Li B, Zhang YJ and Li YY: Pim-3 contributes to radioresistance through regulation of the cell cycle and DNA damage repair in pancreatic cancer cells. Biochem Biophys Res Commun. 473:296–302. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Wang S, Wang Z, Yang YU, Shi MO and Sun Z: Overexpression of Ku80 correlates with aggressive clinicopathological features and adverse prognosis in esophageal squamous cell carcinoma. Oncol Lett. 10:2705–2712. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Koukourakis MI, Giatromanolaki A, Danielidis V and Sivridis E: Hypoxia inducible factor (HIf1alpha and HIF2alpha) and carbonic anhydrase 9 (CA9) expression and response of head-neck cancer to hypofractionated and accelerated radiotherapy. Int J Radiat Biol. 84:47–52. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Harrison LB, Chadha M, Hill RJ, Hu K and Shasha D: Impact of tumor hypoxia and anemia on radiation therapy outcomes. Oncologist. 7:492–508. 2002. View Article : Google Scholar : PubMed/NCBI

9 

Moeller BJ, Richardson RA and Dewhirst MW: Hypoxia and radiotherapy: Opportunities for improved outcomes in cancer treatment. Cancer Metastasis Rev. 26:241–248. 2007. View Article : Google Scholar : PubMed/NCBI

10 

Bennett M, Feldmeier J, Smee R and Milross C: Hyperbaric oxygenation for tumour sensitisation to radiotherapy. Cochrane Database Syst Rev. 19:CD0050072005.

11 

Henke M, Laszig R, Rübe C, Schäfer U, Haase KD, Schilcher B, Mose S, Beer KT, Burger U, Dougherty C, et al: Erythropoietin to treat head and neck cancer patients with anaemia undergoing radiotherapy: Randomised, double-blind, placebo-controlled trial. Lancet. 362:1255–1260. 2003. View Article : Google Scholar : PubMed/NCBI

12 

Teppo S, Sundquist E, Vered M, Holappa H, Parkkisenniemi J, Rinaldi T, Lehenkari P, Grenman R, Dayan D, Risteli J, et al: The hypoxic tumor microenvironment regulates invasion of aggressive oral carcinoma cells. Exp Cell Res. 319:376–389. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Maxwell PH, Dachs GU, Gleadle JM, Nicholls LG, Harris AL, Stratford IJ, Hankinson O, Pugh CW and Ratcliffe PJ: Hypoxia-inducible factor-1 modulates gene expression in solid tumors and influences both angiogenesis and tumor growth. Proc Natl Acad Sci USA. 94:8104–8109. 1997. View Article : Google Scholar : PubMed/NCBI

14 

Jing SW, Wang YD, Chen LQ, Sang MX, Zheng MM, Sun GG, Liu Q, Cheng YJ and Yang CR: Hypoxia suppresses E-cadherin and enhances matrix metalloproteinase-2 expression favoring esophageal carcinoma migration and invasion via hypoxia inducible factor-1 alpha activation. Dis Esophagus. 26:75–83. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Moeller BJ and Dewhirst MW: HIF-1 and tumour radiosensitivity. Br J Cancer. 95:1–5. 2006. View Article : Google Scholar : PubMed/NCBI

16 

Sakai Y, Miwa M, Oe K, Ueha T, Koh A, Niikura T, Iwakura T, Lee SY, Tanaka M and Kurosaka M: A novel system for transcutaneous application of carbon dioxide causing an ‘artificial Bohr effect’ in the human body. PLoS One. 6:e241372011. View Article : Google Scholar : PubMed/NCBI

17 

Takeda D, Hasegawa T, Ueha T, Imai Y, Sakakibara A, Minoda M, Kawamoto T, Minamikawa T, Shibuya Y, Akisue T, et al: Transcutaneous carbon dioxide induces mitochondrial apoptosis and suppresses metastasis of oral squamous cell carcinoma in vivo. PLoS One. 9:e1005302014. View Article : Google Scholar : PubMed/NCBI

18 

Matsui T, Ota T, Ueda Y, Tanino M and Odashima S: Isolation of a highly metastatic cell line to lymph node in human oral squamous cell carcinoma by orthotopic implantation in nude mice. Oral Oncol. 34:253–256. 1998. View Article : Google Scholar : PubMed/NCBI

19 

Iwata E, Hasegawa T, Takeda D, Ueha T, Kawamoto T, Akisue T, Sakai Y and Komori T: Transcutaneous carbon dioxide suppresses epithelial-mesenchymal transition in oral squamous cell carcinoma. Int J Oncol. 48:1493–1498. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Thomlinson RH and Gray LH: The histological structure of some human lung cancers and the possible implications for radiotherapy. Br J Cancer. 9:539–549. 1955. View Article : Google Scholar : PubMed/NCBI

21 

Brown JM and Wilson WR: Exploiting tumour hypoxia in cancer treatment. Nat Rev Cancer. 4:437–447. 2004. View Article : Google Scholar : PubMed/NCBI

22 

Ryan HE, Poloni M, McNulty W, Elson D, Gassmann M, Arbeit JM and Johnson RS: Hypoxia-inducible factor-1α is a positive factor in solid tumor growth. Cancer Res. 60:4010–4015. 2000.PubMed/NCBI

23 

Harada H, Itasaka S, Zhu Y, Zeng L, Xie X, Morinibu A, Shinomiya K and Hiraoka M: Treatment regimen determines whether an HIF-1 inhibitor enhances or inhibits the effect of radiation therapy. Br J Cancer. 100:747–757. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Geng L, Donnelly E, McMahon G, Lin PC, Sierra-Rivera E, Oshinka H and Hallahan DE: Inhibition of vascular endothelial growth factor receptor signaling leads to reversal of tumor resistance to radiotherapy. Cancer Res. 61:2413–2419. 2001.PubMed/NCBI

25 

Hess C, Vuong V, Hegyi I, Riesterer O, Wood J, Fabbro D, Glanzmann C, Bodis S and Pruschy M: Effect of VEGF receptor inhibitor PTK787/ZK222584 [correction of ZK222548] combined with ionizing radiation on endothelial cells and tumour growth. Br J Cancer. 85:2010–2016. 2001. View Article : Google Scholar : PubMed/NCBI

26 

Kozin SV, Boucher Y, Hicklin DJ, Bohlen P, Jain RK and Suit HD: Vascular endothelial growth factor receptor-2-blocking antibody potentiates radiation-induced long-term control of human tumor xenografts. Cancer Res. 61:39–44. 2001.PubMed/NCBI

27 

Lund EL, Bastholm L and Kristjansen PE: Therapeutic synergy of TNP-470 and ionizing radiation: Effects on tumor growth, vessel morphology, and angiogenesis in human glioblastoma multiforme xenografts. Clin Cancer Res. 6:971–978. 2000.PubMed/NCBI

28 

Ning S, Laird D, Cherrington JM and Knox SJ: The antiangiogenic agents SU5416 and SU6668 increase the antitumor effects of fractionated irradiation. Radiat Res. 157:45–51. 2002. View Article : Google Scholar : PubMed/NCBI

29 

Dunst J, Stadler P, Becker A, Lautenschläger C, Pelz T, Hänsgen G, Molls M and Kuhnt T: Tumor volume and tumor hypoxia in head and neck cancers. The amount of the hypoxic volume is important. Strahlenther Onkol. 179:521–526. 2003. View Article : Google Scholar : PubMed/NCBI

30 

Nordsmark M and Overgaard J: A confirmatory prognostic study on oxygenation status and loco-regional control in advanced head and neck squamous cell carcinoma treated by radiation therapy. Radiother Oncol. 57:39–43. 2000. View Article : Google Scholar : PubMed/NCBI

31 

Stadler P, Becker A, Feldmann HJ, Hänsgen G, Dunst J, Würschmidt F and Molls M: Influence of the hypoxic subvolume on the survival of patients with head and neck cancer. Int J Radiat Oncol Biol Phys. 44:749–754. 1999. View Article : Google Scholar : PubMed/NCBI

32 

Nordsmark M, Overgaard M and Overgaard J: Pretreatment oxygenation predicts radiation response in advanced squamous cell carcinoma of the head and neck. Radiother Oncol. 41:31–39. 1996. View Article : Google Scholar : PubMed/NCBI

33 

Nordsmark M, Bentzen SM, Rudat V, Brizel D, Lartigau E, Stadler P, Becker A, Adam M, Molls M, Dunst J, et al: Prognostic value of tumor oxygenation in 397 head and neck tumors after primary radiation therapy. An international multi-center study. Radiother Oncol. 77:18–24. 2005. View Article : Google Scholar : PubMed/NCBI

34 

Overgaard J and Horsman MR: Modification of hypoxia-induced radioresistance in tumors by the use of oxygen and sensitizers. Semin Radiat Oncol. 6:10–21. 1996. View Article : Google Scholar : PubMed/NCBI

35 

Saunders MI, Hoskin PJ, Pigott K, Powell ME, Goodchild K, Dische S, Denekamp J, Stratford MR, Dennis MF and Rojas AM: Accelerated radiotherapy, carbogen and nicotinamide (ARCON) in locally advanced head and neck cancer: A feasibility study. Radiother Oncol. 45:159–166. 1997. View Article : Google Scholar : PubMed/NCBI

36 

Fan TJ, Han LH, Cong RS and Liang J: Caspase family proteases and apoptosis. Acta Biochim Biophys Sin (Shanghai). 37:719–727. 2005. View Article : Google Scholar : PubMed/NCBI

37 

Bosch M, Poulter NS, Vatovec S and Franklin-Tong VE: Initiation of programmed cell death in self-incompatibility: Role for cytoskeleton modifications and several caspase-like activities. Mol Plant. 1:879–887. 2008. View Article : Google Scholar : PubMed/NCBI

38 

Kuwana T and Newmeyer DD: Bcl-2-family proteins and the role of mitochondria in apoptosis. Curr Opin Cell Biol. 15:691–699. 2003. View Article : Google Scholar : PubMed/NCBI

39 

Sharpe JC, Arnoult D and Youle RJ: Control of mitochondrial permeability by Bcl-2 family members. Biochim Biophys Acta. 1644:107–113. 2004. View Article : Google Scholar : PubMed/NCBI

40 

Festjens N, van Gurp M, van Loo G, Saelens X and Vandenabeele P: Bcl-2 family members as sentinels of cellular integrity and role of mitochondrial intermembrane space proteins in apoptotic cell death. Acta Haematol. 111:7–27. 2004. View Article : Google Scholar : PubMed/NCBI

41 

Sagan L: On the origin of mitosing cells. J Theor Biol. 14:255–274. 1967. View Article : Google Scholar : PubMed/NCBI

42 

Graeber TG, Osmanian C, Jacks T, Housman DE, Koch CJ, Lowe SW and Giaccia AJ: Hypoxia-mediated selection of cells with diminished apoptotic potential in solid tumours. Nature. 379:88–91. 1996. View Article : Google Scholar : PubMed/NCBI

43 

Prasad S, Gupta SC and Tyagi AK: Reactive oxygen species (ROS) and cancer: Role of antioxidative nutraceuticals. Cancer Lett. 387:95–105. 2017. View Article : Google Scholar : PubMed/NCBI

44 

Jing L, He MT, Chang Y, Mehta SL, He QP, Zhang JZ and Li PA: Coenzyme Q10 protects astrocytes from ROS-induced damage through inhibition of mitochondria-mediated cell death pathway. Int J Biol Sci. 11:59–66. 2015. View Article : Google Scholar : PubMed/NCBI

45 

Lo YY and Cruz TF: Involvement of reactive oxygen species in cytokine and growth factor induction of c-fos expression in chondrocytes. J Biol Chem. 270:11727–11730. 1995. View Article : Google Scholar : PubMed/NCBI

46 

Locksley RM, Killeen N and Lenardo MJ: The TNF and TNF receptor superfamilies: Integrating mammalian biology. Cell. 104:487–501. 2001. View Article : Google Scholar : PubMed/NCBI

47 

Chung YM, Lee SB, Kim HJ, Park SH, Kim JJ, Chung JS and Yoo YD: Replicative senescence induced by Romo1-derived reactive oxygen species. J Biol Chem. 283:33763–33771. 2008. View Article : Google Scholar : PubMed/NCBI

48 

Kim JJ, Lee SB, Park JK and Yoo YD: TNF-alpha-induced ROS production triggering apoptosis is directly linked to Romo1 and Bcl-X(L). Cell Death Differ. 17:1420–1434. 2010. View Article : Google Scholar : PubMed/NCBI

49 

Lee SB, Kim JJ, Kim TW, Kim BS, Lee MS and Yoo YD: Serum deprivation-induced reactive oxygen species production is mediated by Romo1. Apoptosis. 15:204–218. 2010. View Article : Google Scholar : PubMed/NCBI

50 

Zheng C, Cotrim AP, Rowzee A, Swaim W, Sowers A, Mitchell JB and Baum BJ: Prevention of radiation-induced salivary hypofunction following hKGF gene delivery to murine submandibular glands. Clin Cancer Res. 17:2842–2851. 2011. View Article : Google Scholar : PubMed/NCBI

51 

Harada K, Ferdous T and Yoshida H: Investigation of optimal schedule of concurrent radiotherapy with S-1 for oral squamous cell carcinoma. Oncol Rep. 18:1077–1083. 2007.PubMed/NCBI

52 

Chiang IT, Liu YC, Hsu FT, Chien YC, Kao CH, Lin WJ, Chung JG and Hwang JJ: Curcumin synergistically enhances the radiosensitivity of human oral squamous cell carcinoma via suppression of radiation-induced NF-κB activity. Oncol Rep. 31:1729–1737. 2014. View Article : Google Scholar : PubMed/NCBI

53 

Fujii H, Sakata K, Katsumata Y, Sato R, Kinouchi M, Someya M, Masunaga S, Hareyama M, Swartz HM and Hirata H: Tissue oxygenation in a murine SCC VII tumor after X-ray irradiation as determined by EPR spectroscopy. Radiother Oncol. 86:354–360. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2018
Volume 40 Issue 1

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Iwata E, Hasegawa T, Ueha T, Takeda D, Saito I, Kawamoto T, Akisue T, Sakai Y, Sasaki R, Kuroda R, Kuroda R, et al: Transcutaneous carbon dioxide enhances the antitumor effect of radiotherapy on oral squamous cell carcinoma. Oncol Rep 40: 434-442, 2018
APA
Iwata, E., Hasegawa, T., Ueha, T., Takeda, D., Saito, I., Kawamoto, T. ... Komori, T. (2018). Transcutaneous carbon dioxide enhances the antitumor effect of radiotherapy on oral squamous cell carcinoma. Oncology Reports, 40, 434-442. https://doi.org/10.3892/or.2018.6444
MLA
Iwata, E., Hasegawa, T., Ueha, T., Takeda, D., Saito, I., Kawamoto, T., Akisue, T., Sakai, Y., Sasaki, R., Kuroda, R., Komori, T."Transcutaneous carbon dioxide enhances the antitumor effect of radiotherapy on oral squamous cell carcinoma". Oncology Reports 40.1 (2018): 434-442.
Chicago
Iwata, E., Hasegawa, T., Ueha, T., Takeda, D., Saito, I., Kawamoto, T., Akisue, T., Sakai, Y., Sasaki, R., Kuroda, R., Komori, T."Transcutaneous carbon dioxide enhances the antitumor effect of radiotherapy on oral squamous cell carcinoma". Oncology Reports 40, no. 1 (2018): 434-442. https://doi.org/10.3892/or.2018.6444