1
|
Shi J, Li J, Li Z, Li Y, Xu L, Zhang Y. Prediction of pathological response grading for esophageal squamous carcinoma after neoadjuvant chemoradiotherapy based on MRI imaging using PDX. Front Oncol 2023; 13:1160815. [PMID: 37377911 PMCID: PMC10292012 DOI: 10.3389/fonc.2023.1160815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Introduction To confirm the efficacy of magnetic resonance-diffusion weighted imaging (MR-DWI) in esophageal squamous cell carcinoma (ESCC) early pathological response prediction and assessment to neoadjuvant chemoradiotherapy (nCRT) using patient-derived xenografts (PDXs). Methods PDX-bearing mice were randomly divided into two groups: the experimental group receiving cisplatin combined with radiotherapy, whereas the control group receiving normal saline. MRI scans were performed in treatment groups in the before, middle, and end of treatment. The correlations between tumor volumes, ADC values and tumor pathological response at different time nodes were explored. Then, expression of proliferation marker and apoptotic marker were detected using immunohistochemistry, and apoptosis rate was detected by TUNEL assay to further verify the results observed in the PDX models. Results The ADC values of the experimental group were significantly higher than the control group in the both middle and end stage of treatment (all P< 0.001), however, significant difference was only observed in tumor volume at the end stage of treatment (P< 0.001). Furthermore, the △ADCmid-pre in our study may able to identify tumors with or without pCR to nCRT at an early stage, due to these changes were prior to the changes of tumor volume after treatment. Finally, TUNEL results also showed that the apoptosis rate of the experiment groups increased the most in the middle stage of treatment, especially the groups with pCR, but the highest apoptosis rate occurred in the end of the treatment. Further, the two PDX models with pCR exhibited the highest levels of apoptotic marker (Bax), and lowest levels of proliferation marker (PCNA and Ki-67) in the both middle and end stage of the treatment. Conclusions ADC values could be used to determine the tumor's response to nCRT, especially in the middle stages of treatment and before the tumor tissue morphology changes, and further, the ADC values were consistent with the potential biomarkers reflecting histopathological changes. Therefore, we suggest that radiation oncologists could refer to the ADC values in the middle stages of treatment when predicting the tumor histopathological response to n CRT in patients with ESCC.
Collapse
Affiliation(s)
- Jingzhen Shi
- Department of Oncology, The Second Hospital of Tianjin Medical University, Tianjin, China
- School of Medicine, Shandong University, Jinan, China
| | - Jianbin Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Zhenxiang Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yankang Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Liang Xu
- Department of Medical Imaging, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yingjie Zhang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
2
|
Zhang AD, Su XH, Wang YF, Shi GF, Han C, Zhang N. Predicting the effects of radiotherapy based on diffusion kurtosis imaging in a xenograft mouse model of esophageal carcinoma. Exp Ther Med 2021; 21:327. [PMID: 33732300 PMCID: PMC7903468 DOI: 10.3892/etm.2021.9758] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 11/20/2020] [Indexed: 02/06/2023] Open
Abstract
The aim of the present study was to assess the predictive value of diffusion kurtosis imaging (DKI) on the effects of radiotherapy in a xenograft model of esophageal cancer. A total of 40 tumor-bearing mice, established by injection of Eca-109 cells in nude mice, were used. The experimental group (n=24) received a single dose of 15 Gy (6 MV by X-ray), and the control group (n=16) did not receive any treatment. Tumor volume, apparent diffusion coefficient (ADC), mean kurtosis (MK) and mean diffusivity (MD) of the two groups were compared, and the expression of aquaporin (AQP) 3 and necrosis ratio at matched time points in xenografts were also observed. There was a significant difference between the two groups from the 7th day of radiotherapy onwards; the xenograft volume of the experimental group was significantly smaller compared with the control group (P<0.05). On the 3rd day, the ADC and MD of the experimental group was significantly higher compared with the control group, and MK was significantly lower compared with the control group (P<0.05). On the 3rd day, AQP3 expression in the experimental group was lower compared with the control group, and the proportion of necrotic cells was higher compared with the control group (P<0.05). Single large fraction dose radiotherapy inhibited the growth of a xenografted esophageal tumor. Changes in ADC, MK and MD were observed prior to morphological changes in the tumor. The change in AQP3 expression and necrosis ratio was in also agreement with the DKI parameters assessed. DKI may thus provide early predictive ability on the effect of radiotherapy in esophageal carcinoma.
Collapse
Affiliation(s)
- An-Du Zhang
- Department of Radiotherapy, Hebei Medical University Fourth Affiliated Hospital/Hebei Provincial Tumor Hospital, Shijiazhuang, Hebei 050011, P.R. China
| | - Xiao-Hua Su
- Department of Oncology, Hebei General Hospital, Shijiazhuang, Hebei 050011, P.R. China
| | - Yan-Fei Wang
- Department of CT and MRI, Hebei Medical University Fourth Affiliated Hospital/Hebei Provincial Tumor Hospital, Shijiazhuang, Hebei 050011, P.R. China
| | - Gao-Feng Shi
- Department of CT and MRI, Hebei Medical University Fourth Affiliated Hospital/Hebei Provincial Tumor Hospital, Shijiazhuang, Hebei 050011, P.R. China
| | - Chun Han
- Department of Radiotherapy, Hebei Medical University Fourth Affiliated Hospital/Hebei Provincial Tumor Hospital, Shijiazhuang, Hebei 050011, P.R. China
| | - Nan Zhang
- Department of Radiotherapy, Hebei Medical University Fourth Affiliated Hospital/Hebei Provincial Tumor Hospital, Shijiazhuang, Hebei 050011, P.R. China
| |
Collapse
|
3
|
Mori H, Saito YU, Iwahashi S, Ikemoto T, Imura S, Morine Y, Shimada M. Impact of Bevacizumab on Liver Damage After Massive Hepatectomy in Rats. In Vivo 2019; 33:1469-1476. [PMID: 31471394 DOI: 10.21873/invivo.11626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/08/2019] [Accepted: 07/12/2019] [Indexed: 11/10/2022]
Abstract
BACKGROUND The aim of this study was to evaluate the impact of pretreatment with bevacizumab on liver damage in a rat model of massive hepatectomy (Hx) model, as a surrogate model of massive Hx for liver metastasis from colorectal cancer. MATERIALS AND METHODS Male Wister rats (n=24) were separated into the following two groups: 90% Hx and 90% Hx plus bevacizumab group. Bevacizumab (5 mg/kg) was injected intraperitoneally 7 days before Hx. Samples of blood and remnant liver tissue were obtained 24 hours after hepatectomy and the following parameters were evaluated: Biochemical analysis; liver regeneration rate; survival rate; and real-time polymerase chain reaction for interleukin-1 beta (Il1b), tumor necrosis factor alpha (Tnfa), matrix metalloproteinase (Mmp) 2 and Mmp9 mRNA. In addition, samples of whole liver tissue were obtained immediately before Hx and real-time polymerase chain reaction was performed for X-box binding protein 1 (Xbp1), activating transcription factor 6 (Atf6), C/EBP homologous protein (Chop), glucose-regulated protein 78 (Grp78) and heat-shock protein 70 (Hsp70), as markers of endoplasmic reticulum stress response. RESULTS The levels of transaminases 24 hours after Hx were significantly reduced in the group pretreated with bevacizumab compared to that not pretreated (p<0.05). The liver regeneration rate at 24 hours after Hx was significantly increased in the group pretreated with bevacizumab compared with the group which underwent Hx alone (p<0.05). The survival rate for the group pretreated with bevacizumab tended to be higher than that of the Hx-only group, 72 hours after Hx (p=0.09). The expressions of Il1b, Mmp2 and Mmp9 mRNA 24 hours after Hx in the group pretreated with bevacizumab tended to be lower than that of rats which underwent Hx alone (p=0.11, 0.09 and 0.15, respectively). The expression of Xbp1, Chop, Grp78 and Hsp70 mRNA immediately before Hx in the group pretreated with bevacizumab were significantly higher than the 90% Hx group (p<0.05). CONCLUSION Bevacizumab pretreatment had protective effects on liver injury after massive hepatectomy in rats, apparently via the induction of the endoplasmic reticulum stress response, i.e. the so-called unfolded protein response.
Collapse
Affiliation(s)
- Hiroki Mori
- Department of Surgery, The University of Tokushima, Tokushima, Japan
| | - Y U Saito
- Department of Surgery, The University of Tokushima, Tokushima, Japan
| | - Shuichi Iwahashi
- Department of Surgery, The University of Tokushima, Tokushima, Japan
| | - Tetsuya Ikemoto
- Department of Surgery, The University of Tokushima, Tokushima, Japan
| | - Satoru Imura
- Department of Surgery, The University of Tokushima, Tokushima, Japan
| | - Yuji Morine
- Department of Surgery, The University of Tokushima, Tokushima, Japan
| | - Mitsuo Shimada
- Department of Surgery, The University of Tokushima, Tokushima, Japan
| |
Collapse
|
4
|
Zheng X, Chen Y, Xiao Y, Zheng D, Chen W. Early diagnosis of radio-insensitive human nasopharyngeal carcinoma xenograft models by diffusion kurtosis imaging. Magn Reson Imaging 2018; 55:128-132. [PMID: 30098385 DOI: 10.1016/j.mri.2018.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 08/05/2018] [Accepted: 08/07/2018] [Indexed: 01/17/2023]
Abstract
OBJECTIVE To investigate the feasibility of DKI in early detection of radio-insensitive nasopharyngeal carcinoma (NPC) xenografts in nude mice. MATERIALS AND METHODS Seventy-two nude mice were implanted with CNE-1 (low radio-sensitive) and CNE-2 (high radio-sensitive) NPC cell lines, and their respective xenografts were obtained. Then, the NPC-bearing nude mice were exposed to different doses of fraction irradiation, which are divided into non-irradiated group (G0), 10Gy group (G1), 20Gy group (G2), 30Gy group (G3), 3rd (G4) and 5th (G5) days after the entire dose (30y) of irradiation. Subsequently, DKI was performed on each group. Tumor volumes, shrink rates, D and K parameters were measured by two experienced radiologists. Student's t-test and receiver operating characteristic (ROC) curve analysis were conducted in this study. RESULTS The differences of volume shrinkage rate between CNE-1 and -2 were observed in G2 (P = 0.032), with the shrink rates of 5.954% and 27.716%, respectively. The D values were reduced at G1 (DG1, P = 0.001) and then increased gradually after irradiation. The K values were increased at G1 (KG1, P = 0.001) and then declined sharply in CNE-2 (P < 0.01), but not in CNE-1 xenografts (P > 0.05). The respective AUC values for DG1 and KG1 were 0.875 and 0.917, with 66.7% and 83.3% sensitivity and 100% specificity, at the cutoff values of 1.27 × 10-3 mm2/s for parameter D and 0.88 for parameter K. CONCLUSION DKI can be used for early detection of radio-insensitive NPC xenografts prior to morphological change, where DG1 and KG1 may be the most valuable indicators.
Collapse
Affiliation(s)
- Xiang Zheng
- Department of Radiologic Diagnosis, Fujian Medical University Cancer Hospital, Fuzhou, Fujian, China
| | - Yunbin Chen
- Department of Radiologic Diagnosis, Fujian Medical University Cancer Hospital, Fuzhou, Fujian, China; Department of Radiologic Diagnosis, Fujian Cancer Hospital, Fuzhou, Fujian, China.
| | - Youping Xiao
- Department of Radiologic Diagnosis, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Dechun Zheng
- Department of Radiologic Diagnosis, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | | |
Collapse
|
5
|
Sun JS, Yang XH. Expression of DNA-dependent protein kinase catalytic subunit in laryngeal squamous cell carcinoma and its importance. Exp Ther Med 2018; 15:3295-3301. [PMID: 29545847 PMCID: PMC5840916 DOI: 10.3892/etm.2018.5826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 06/08/2017] [Indexed: 11/05/2022] Open
Abstract
The aim of the present study was to explore the expression and distribution of DNA-dependent protein kinase catalytic subunit (DNA-PKcs) in tumor tissues and adjacent normal mucosa tissues of patients with laryngeal squamous cell carcinoma (LSCC), and further analyze the association between the expression and the clinicopathological parameters of patients with LSCC. Clinical data of tumor tissues and corresponding adjacent normal mucosa tissues of pathologically diagnosed LSCC in 96 cases were collected in the present study. Of these specimens, the mRNA and protein expression levels of DNA-PKcs in LSCC tissues and the adjacent normal mucosa tissues were analyzed via reverse transcription-quantitative polymerase chain reaction and western blot analysis. Immunohistochemistry was used to detect expression and distribution of DNA-PKcs protein in LSCC tissues and corresponding adjacent normal mucosa tissues. The association between DNA-PKcs expression and the specific clinicopathologic features was evaluated by the χ2 test. Kaplan-Meier and Cox proportional hazards regression models were used to analyze the data. It was revealed that the expression of DNA-PKcs mRNA and protein was significantly higher in LSCC tissues than the adjacent normal mucosa tissues (P<0.05). DNA-PKcs was expressed predominantly in the nucleus. DNA-PKcs expression showed significant correlation with the differentiation degree of LSCC (P<0.05), and changes of DNA-PKcs expression gradually increased with the decrease of the differentiation degree. However, DNA-PKcs expression was not significantly associated with sex, age, lymph node metastasis or TMN stage (P>0.05). Patients with LSCC exhibited higher DNA-PKcs expression had markedly shorter survival than those with lower DNA-PKcs expression. In conclusion, the present results suggested that the expression levels of DNA-PKcs were significantly increased in LSCC tumor tissues than in adjacent normal mucosa. DNA-PKcs expression was correlated with differentiation of LSCC, and may become a novel prognostic marker for patients with LSCC.
Collapse
Affiliation(s)
- Jian-Song Sun
- Department of Otolaryngology-Head and Neck Surgery, The People's Hospital of Guizhou Province, Guiyang, Guizhou 550002, P.R. China
| | - Xiu-Hai Yang
- Department of Otolaryngology-Head and Neck Surgery, The People's Hospital of Guizhou Province, Guiyang, Guizhou 550002, P.R. China
| |
Collapse
|
6
|
Silva APS, Coelho PV, Anazetti M, Simioni PU. Targeted therapies for the treatment of non-small-cell lung cancer: Monoclonal antibodies and biological inhibitors. Hum Vaccin Immunother 2016; 13:843-853. [PMID: 27831000 PMCID: PMC5404364 DOI: 10.1080/21645515.2016.1249551] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The usual treatments for patients with non-small-cell lung cancer (NSCLC), such as advanced lung adenocarcinoma, are unspecific and aggressive, and include lung resection, radiotherapy and chemotherapy. Recently, treatment with monoclonal antibodies and biological inhibitors has emerged as an effective alternative, generating effective results with few side effects. In recent years, several clinical trials using monoclonal antibodies presented potential benefits to NSCLC, and 4 of them are already approved for the treatment of NSCLC, such as cetuximab, bevacizumab, nivolumab and pembrolizumab. Also, biological inhibitors are attractive tolls for biological applications. Among the approved inhibitors are crizotinib, erlotinib, afatinib and gefitinib, and side effects are usually mild to intense. Nevertheless, biological molecule treatments are under development, and several new monoclonal antibodies and biological inhibitors are in trial to treat NSCLC. Also under trial study are as follows: anti-epidermal growth factor receptor (EGFR) antibodies (nimotuzumab and ficlatuzumab), anti-IGF 1 receptor (IGF-1R) monoclonal antibody (figitumumab), anti-NR-LU-10 monoclonal antibody (nofetumomab) as well as antibodies directly affecting the cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) molecule (ipilimumab and tremelimumab), to receptor activator of nuclear factor-kappa B ligand (RANKL) (denosumab) or to polymerase enzyme (veliparib and olaparib). Among new inhibitors under investigation are poly-ADP ribose polymerase (PARP) inhibitors (veliparib and olaparib) and phosphatidylinositol 3-kinase (PI3K) inhibitor (buparlisib). However, the success of immunotherapies still requires extensive research and additional controlled trials to evaluate the long-term benefits and side effects.
Collapse
Affiliation(s)
- Ana P S Silva
- a Department of Biomedical Science , Faculty of Americana , Americana , SP , Brazil
| | - Priscila V Coelho
- a Department of Biomedical Science , Faculty of Americana , Americana , SP , Brazil
| | - Maristella Anazetti
- a Department of Biomedical Science , Faculty of Americana , Americana , SP , Brazil.,b Department of Health Science , Faculty DeVry Metrocamp , Campinas , SP , Brazil
| | - Patricia U Simioni
- a Department of Biomedical Science , Faculty of Americana , Americana , SP , Brazil.,c Department of Genetics , Evolution and Bioagents, Institute of Biology, University of Campinas (UNICAMP) , Campinas , SP , Brazil.,d Department of Biochemistry and Microbiology , Institute of Biosciences, Universidade Estadual Paulista, UNESP , Rio Claro , SP , Brazil
| |
Collapse
|
7
|
Becker S, Bohn P, Bouyeure-Petit AC, Modzelewski R, Gensanne D, Picquenot JM, Dubray B, Vera P. Bevacizumab enhances efficiency of radiotherapy in a lung adenocarcinoma rodent model: Role of αvβ3 imaging in determining optimal window. Nucl Med Biol 2015; 42:923-30. [DOI: 10.1016/j.nucmedbio.2015.08.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 07/30/2015] [Accepted: 08/10/2015] [Indexed: 11/25/2022]
|
8
|
Zhang CC, Yan Z, Giddabasappa A, Lappin PB, Painter CL, Zhang Q, Li G, Goodman J, Simmons B, Pascual B, Lee J, Levkoff T, Nichols T, Xie Z. Comparison of dynamic contrast-enhanced MR, ultrasound and optical imaging modalities to evaluate the antiangiogenic effect of PF-03084014 and sunitinib. Cancer Med 2014; 3:462-71. [PMID: 24573979 PMCID: PMC4101737 DOI: 10.1002/cam4.215] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 01/21/2014] [Accepted: 01/22/2014] [Indexed: 02/06/2023] Open
Abstract
Noninvasive imaging has been widely applied for monitoring antiangiogenesis therapy in cancer drug discovery. In this report, we used different imaging modalities including high-frequency ultrasound (HFUS), dynamic contrast enhanced-MR (DCE-MR), and fluorescence molecular tomography (FMT) imaging systems to monitor the changes in the tumor vascular properties after treatment with γ-secretase inhibitor PF-03084014. Sunitinib was tested in parallel for comparison. In the MDA-MB-231Luc model, we demonstrated that antiangiogenesis was one of the contributing mechanisms for the therapeutic effect of PF-03084014. By immunohistochemistry and FITC-lectin perfusion assays, we showed that the vascular defects upon treatment with PF-03084014 were associated with Notch pathway modulation, evidenced by a decrease in the HES1 protein and by the changes in VEGFR2 and HIF1α levels, which indicates down-stream effects. Using a 3D power Doppler scanning method, ultrasound imaging showed that the% vascularity in the MDA-MB-231Luc tumor decreased significantly at 4 and 7 days after the treatment with PF-03084014. A decrease in the tumor vessel function was also observed through contrast-enhanced ultrasound imaging with microbubble injection. These findings were consistent with the PF-03084014-induced functional vessel changes measured by suppressing the K(trans) values using DCE-MRI. In contrast, the FMT imaging with the AngioSence 680EX failed to detect any treatment-associated tumor vascular changes. Sunitinib demonstrated an outcome similar to PF-03084014 in the tested imaging modalities. In summary, ultrasound and DCE-MR imaging successfully provided longitudinal measurement of the phenotypic and functional changes in tumor vasculature after treatment with PF-03084014 and sunitinib.
Collapse
Affiliation(s)
- Cathy C Zhang
- Oncology Research Unit, Pfizer Global Research and Development, La Jolla, California
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Chen W, Ye F, Cui M, Sikora AG, Wang X, Wang P, Cui X, Guo X, Zhu W, Zhang DY. Protein marker profiling in different T classification in laryngeal squamous cell carcinoma. Head Neck 2014; 37:357-65. [PMID: 24753215 DOI: 10.1002/hed.23607] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 10/22/2013] [Accepted: 01/08/2014] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND The identification of specific biomarkers related to laryngeal squamous cell carcinoma (SCC) will be helpful in early detection and determination of reasonable treatment options, which are crucial for the prognosis of patients with laryngeal SCC. The purpose of this study was to profile the protein markers that can be used for diagnosis and prognosis of laryngeal SCC and to observe laryngeal SCC progression with distinct stages of malignant transformation. METHODS Two hundred twenty-five proteins were tested in 84 pairs of tumors and adjacent nontumor mucosa samples using protein pathway arrays (PPAs). Ingenuity pathway analysis (IPA) enrichment analysis was performed and protein expression profiles in different T classification were mapped by grid analysis of time-series expression (GATE). RESULTS Among 16 proteins differently expressed between tumors and normal tissues, we selected 9 proteins (TTF-1, CDK2, Eg5, proliferating cell nuclear antigen [PCNA], Bcl-xL, 14-3-3β, p27, SRC-1, cytokeratin 18) as markers for classification. From the IPA analysis, we observed a more malignant transformation from T3 to T4 at the protein level and described the changing patterns of the proteins' expression in this progression. JAK2, keratin 10, and IL-3Rα were identified as markers for prognosis. The risk model based on histological grade, T classification, N classification, JAK2, and IL-3Rα can predict the prognosis with 85.5% accuracy. CONCLUSION This study indicated that dysregulated signaling proteins can be selected as useful biomarkers for tumor classification and predicting the outcome in patients with laryngeal SCC. The changing patterns of the proteins' expression in different stages were related to the more malignant transformation and further studies will focus on the role of these proteins in laryngeal SCC progression.
Collapse
Affiliation(s)
- Weilun Chen
- Department of Otolaryngology Head and Neck Surgery, First Hospital of Jilin University, Changchun, Jilin, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
|
11
|
Microenvironment and radiation therapy. BIOMED RESEARCH INTERNATIONAL 2012; 2013:685308. [PMID: 23509762 PMCID: PMC3591225 DOI: 10.1155/2013/685308] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 07/13/2012] [Indexed: 12/19/2022]
Abstract
Dependency on tumor oxygenation is one of the major features of radiation therapy and this has led many radiation biologists and oncologists to focus on tumor hypoxia. The first approach to overcome tumor hypoxia was to improve tumor oxygenation by increasing oxygen delivery and a subsequent approach was the use of radiosensitizers in combination with radiation therapy. Clinical use of some of these approaches was promising, but they are not widely used due to several limitations. Hypoxia-inducible factor 1 (HIF-1) is a transcription factor that is activated by hypoxia and induces the expression of various genes related to the adaptation of cellular metabolism to hypoxia, invasion and metastasis of cancer cells and angiogenesis, and so forth. HIF-1 is a potent target to enhance the therapeutic effects of radiation therapy. Another approach is antiangiogenic therapy. The combination with radiation therapy is promising, but several factors including surrogate markers, timing and duration, and so forth have to be optimized before introducing it into clinics. In this review, we examined how the tumor microenvironment influences the effects of radiation and how we can enhance the antitumor effects of radiation therapy by modifying the tumor microenvironment.
Collapse
|
12
|
Abstract
Selective inhibition of vascular endothelial growth factor (VEGF) increases the efficacy of chemotherapy and has beneficial effects on multiple advanced cancers, but response is often limited and the disease eventually progresses. Changes in the tumour microenvironment--hypoxia among them--that result from vascular pruning, suppressed angiogenesis and other consequences of VEGF inhibition can promote escape and tumour progression. New therapeutic approaches that target pathways that are involved in the escape mechanisms add the benefits of blocking tumour progression to those of slowing tumour growth by inhibiting angiogenesis.
Collapse
Affiliation(s)
- Barbara Sennino
- The UCSF Helen Diller Family Comprehensive Cancer Center, Cardiovascular Research Institute and Department of Anatomy, University of California, San Francisco, San Francisco, California 94143-0452, USA
| | | |
Collapse
|
13
|
Titz B, Kozak KR, Jeraj R. Computational modelling of anti-angiogenic therapies based on multiparametric molecular imaging data. Phys Med Biol 2012; 57:6079-101. [PMID: 22972469 DOI: 10.1088/0031-9155/57/19/6079] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Computational tumour models have emerged as powerful tools for the optimization of cancer therapies; ideally, these models should incorporate patient-specific imaging data indicative of therapeutic response. The purpose of this study was to develop a tumour modelling framework in order to simulate the therapeutic effects of anti-angiogenic agents based upon clinical molecular imaging data. The model was applied to positron emission tomography (PET) data of cellular proliferation and hypoxia from a phase I clinical trial of bevacizumab, an antibody that neutralizes the vascular endothelial growth factor (VEGF). When using pre-therapy PET data in combination with literature-based dose response parameters, simulated follow-up hypoxia data yielded good qualitative agreement with imaged hypoxia levels. Improving the quantitative agreement with follow-up hypoxia and proliferation PET data required tuning of the maximum vascular growth fraction (VGF(max)) and the tumour cell cycle time to patient-specific values. VGF(max) was found to be the most sensitive model parameter (CV = 22%). Assuming availability of patient-specific, intratumoural VEGF levels, we show how bevacizumab dose levels can potentially be 'tailored' to improve levels of tumour hypoxia while maintaining proliferative response, both of which are critically important in the context of combination therapy. Our results suggest that, upon further validation, the application of image-driven computational models may afford opportunities to optimize dosing regimens and combination therapies in a patient-specific manner.
Collapse
Affiliation(s)
- Benjamin Titz
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| | | | | |
Collapse
|
14
|
Saha D, Dunn H, Zhou H, Harada H, Hiraoka M, Mason RP, Zhao D. In vivo bioluminescence imaging of tumor hypoxia dynamics of breast cancer brain metastasis in a mouse model. J Vis Exp 2011:3175. [PMID: 21989061 DOI: 10.3791/3175] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
It is well recognized that tumor hypoxia plays an important role in promoting malignant progression and affecting therapeutic response negatively. There is little knowledge about in situ, in vivo, tumor hypoxia during intracranial development of malignant brain tumors because of lack of efficient means to monitor it in these deep-seated orthotopic tumors. Bioluminescence imaging (BLI), based on the detection of light emitted by living cells expressing a luciferase gene, has been rapidly adopted for cancer research, in particular, to evaluate tumor growth or tumor size changes in response to treatment in preclinical animal studies. Moreover, by expressing a reporter gene under the control of a promoter sequence, the specific gene expression can be monitored non-invasively by BLI. Under hypoxic stress, signaling responses are mediated mainly via the hypoxia inducible factor-1α (HIF-1α) to drive transcription of various genes. Therefore, we have used a HIF-1α reporter construct, 5HRE-ODD-luc, stably transfected into human breast cancer MDA-MB231 cells (MDA-MB231/5HRE-ODD-luc). In vitro HIF-1α bioluminescence assay is performed by incubating the transfected cells in a hypoxic chamber (0.1% O₂) for 24 hr before BLI, while the cells in normoxia (21% O₂) serve as a control. Significantly higher photon flux observed for the cells under hypoxia suggests an increased HIF-1α binding to its promoter (HRE elements), as compared to those in normoxia. Cells are injected directly into the mouse brain to establish a breast cancer brain metastasis model. In vivo bioluminescence imaging of tumor hypoxia dynamics is initiated 2 wks after implantation and repeated once a week. BLI reveals increasing light signals from the brain as the tumor progresses, indicating increased intracranial tumor hypoxia. Histological and immunohistochemical studies are used to confirm the in vivo imaging results. Here, we will introduce approaches of in vitro HIF-1α bioluminescence assay, surgical establishment of a breast cancer brain metastasis in a nude mouse and application of in vivo bioluminescence imaging to monitor intracranial tumor hypoxia.
Collapse
Affiliation(s)
- Debabrata Saha
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, USA
| | | | | | | | | | | | | |
Collapse
|
15
|
Yoon MS, Nam TK, Lee JS, Cho SH, Song JY, Ahn SJ, Chung IJ, Jeong JU, Chung WK, Nah BS. VEGF as a predictor for response to definitive chemoradiotherapy and COX-2 as a prognosticator for survival in esophageal squamous cell carcinoma. J Korean Med Sci 2011; 26:513-20. [PMID: 21468258 PMCID: PMC3069570 DOI: 10.3346/jkms.2011.26.4.513] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Accepted: 02/17/2011] [Indexed: 12/14/2022] Open
Abstract
We investigated the patterns of pretreatment expression of the epidermal growth factor receptor (EGFR), vascular endothelial growth factor (VEGF), and cyclooxygenase-2 (COX-2) by immunohistochemical staining and determined their correlation with treatment response and survival in 44 patients with esophageal squamous cell carcinoma (ESCC) treated with definitive concurrent chemoradiotherapy (CCRT). The definitive CCRT consisted of a median dose of 54 Gy (range: 40.0-68.4 Gy) and two cycles of concurrent administration of mostly 5-fluorouracil + cisplatinum. High expression of EGFR, VEGF, and COX-2 was found in 79.5%, 31.8%, and 38.6%, respectively. The Cox regression analysis for overall survival (OS) showed that both the treatment response and COX-2 expression were significant. The 3-yr OS rates of patients that achieved a complete response and those that did not were 46.7% and 5.3%, respectively (P = 0.006). The logistic regression analysis for treatment response with various parameters showed that only a high expression of VEGF was significantly associated with a complete response. Unlike other well-known studies, higher expression of VEGF was significantly correlated with a complete response to CCRT in this study. However, higher expression of COX-2 was significantly associated with shorter survival. These results suggest that VEGF might be a predictive factor for treatment response and COX-2 a prognostic factor for OS in patients with ESCC after definitive CCRT.
Collapse
Affiliation(s)
- Mee Sun Yoon
- Department of Radiation Oncology, Chonnam National University Medical School, Gwangju, Korea
| | - Taek-Keun Nam
- Department of Radiation Oncology, Chonnam National University Medical School, Gwangju, Korea
| | - Ji-Shin Lee
- Department of Pathology, Chonnam National University Medical School, Gwangju, Korea
| | - Sang-Hee Cho
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Ju-Young Song
- Department of Radiation Oncology, Chonnam National University Medical School, Gwangju, Korea
| | - Sung-Ja Ahn
- Department of Radiation Oncology, Chonnam National University Medical School, Gwangju, Korea
| | - Ik-Joo Chung
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Jae-Uk Jeong
- Department of Radiation Oncology, Chonnam National University Medical School, Gwangju, Korea
| | - Woong-Ki Chung
- Department of Radiation Oncology, Chonnam National University Medical School, Gwangju, Korea
| | - Byung-Sik Nah
- Department of Radiation Oncology, Chonnam National University Medical School, Gwangju, Korea
| |
Collapse
|
16
|
Harada H. How can we overcome tumor hypoxia in radiation therapy? JOURNAL OF RADIATION RESEARCH 2011; 52:545-56. [PMID: 21952313 DOI: 10.1269/jrr.11056] [Citation(s) in RCA: 131] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Local recurrence and distant metastasis frequently occur after radiation therapy for cancer and can be fatal. Evidence obtained from radiochemical and radiobiological studies has revealed these problems to be caused, at least in part, by a tumor-specific microenvironment, hypoxia. Moreover, a transcription factor, hypoxia-inducible factor 1 (HIF-1), was identified as pivotal to hypoxia-mediated radioresistance. To overcome the problems, radiation oncologists have recently obtained powerful tools, such as "simultaneous integrated boost intensity-modulated radiation therapy (SIB-IMRT), which enables a booster dose of radiation to be delivered to small target fractions in a malignant tumor", "hypoxia-selective cytotoxins/drugs", and "HIF-1 inhibitors" etc. In order to fully exploit these innovative and interdisciplinary strategies in cancer therapy, it is critical to unveil the characteristics, intratumoral localization, and dynamics of hypoxia/HIF-1-active tumor cells during tumor growth and after radiation therapy. We have performed optical imaging experiments using tumor-bearing mice and revealed that the locations of HIF-1-active tumor cells changes dramatically as tumors grow. Moreover, HIF-1 activity changes markedly after radiation therapy. This review overviews 1) fundamental problems surrounding tumor hypoxia in current radiation therapy, 2) the function of HIF-1 in tumor radioresistance, 3) the dynamics of hypoxic tumor cells during tumor growth and after radiation therapy, and 4) how we should overcome the difficulties with radiation therapy using innovative interdisciplinary technologies.
Collapse
Affiliation(s)
- Hiroshi Harada
- Group of Radiation and Tumor Biology, Career-Path Promotion Unit for Young Life Scientists, Kyoto University, Japan.
| |
Collapse
|
17
|
Teng LS, Jin KT, He KF, Wang HH, Cao J, Yu DC. Advances in combination of antiangiogenic agents targeting VEGF-binding and conventional chemotherapy and radiation for cancer treatment. J Chin Med Assoc 2010; 73:281-8. [PMID: 20603084 DOI: 10.1016/s1726-4901(10)70062-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Accepted: 04/14/2010] [Indexed: 12/27/2022] Open
Abstract
Despite great efforts and resources being devoted to treatment, the incidence and mortality of numerous cancers have not decreased in recent decades. This is a result of the resistance of cancer cells to chemotherapeutic agents and radio-therapy. The development of antiangiogenic agents that target vascular endothelial growth factor (VEGF) provides a new option for treatment of cancer. Major advances have been achieved with cancer therapy based on antiangiogenic VEGF-targeted agents in the past few years, and some of the recently approved therapies are now being used in daily clinical practice. A further challenge is finding a more efficacious combination of antiangiogenic VEGF-targeted therapies and conventional radio- and chemotherapies. This review outlines the current preclinical and clinical cancer treatments using optimized combinations of antiangiogenic VEGF-targeted agents and conventional radiochemotherapy and highlights that better scheduling for the combination of radiochemotherapy and antiangiogenic VEGF-targeted agents should be developed to achieve better treatment outcomes.
Collapse
Affiliation(s)
- Li-Song Teng
- Department of Surgical Oncology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| | | | | | | | | | | |
Collapse
|