1
|
Xia X, Pi W, Chen M, Wang W, Cai D, Wang X, Lan Y, Yang H. Emerging roles of PHLPP phosphatases in lung cancer. Front Oncol 2023; 13:1216131. [PMID: 37576883 PMCID: PMC10414793 DOI: 10.3389/fonc.2023.1216131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 07/12/2023] [Indexed: 08/15/2023] Open
Abstract
Pleckstrin homologous domain leucine-rich repeating protein phosphatases (PHLPPs) were originally identified as protein kinase B (Akt) kinase hydrophobic motif specific phosphatases to maintain the cellular homeostasis. With the continuous expansion of PHLPPs research, imbalanced-PHLPPs were mainly found as a tumor suppressor gene of a variety of solid tumors. In this review, we simply described the history and structures of PHLPPs and summarized the recent achievements in emerging roles of PHLPPs in lung cancer by 1) the signaling pathways affected by PHLPPs including Phosphoinositide 3-kinase (PI3K)/AKT, RAS/RAF/mitogen-activated protein kinase (MEK)/extracellular signal-regulated kinase (ERK) and Protein kinase C (PKC) signaling cascades. 2) function of PHLPPs regulatory factor USP46 and miR-190/miR-215, 3) the potential roles of PHLPPs in disease prognosis, Epidermal growth factor receptors (EGFR)- tyrosine kinase inhibitor (TKI) resistance and DNA damage, 4) and the possible function of PHLPPs in radiotherapy, ferroptosis and inflammation response. Therefore, PHLPPs can be considered as either biomarker or prognostic marker for lung cancer treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Haihua Yang
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| |
Collapse
|
2
|
Ye C, Lin Q, Zheng C. Overexpressed RBPMS-AS1 increased cell radiosensitivity by sponging miR-19a-3p in lung cancer cell lines (A549 and SK-MES-1) via regulating PTEN/AKT axis. Int J Radiat Biol 2023; 99:1352-1363. [PMID: 36912590 DOI: 10.1080/09553002.2023.2181997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 01/14/2023] [Accepted: 02/13/2023] [Indexed: 03/14/2023]
Abstract
PURPOSE This paper intended to study RBPMS-AS1 in lung cancer (LC) radiosensitivity. MATERIALS AND METHODS LC cells were transfected with RBPMS-AS1 overexpression plasmid and miR-19a-3p mimic and treated with radiation. PTEN, AKT, p-AKT, RBPMS-AS1, and miR-19a-3p expressions were detected via Western blot and qRT-PCR. The localization of RBPMS-AS1 in cells was determined through fluorescence in situ hybridization assay. The targeting relationships of RBPMS-AS1 and miR-19a-3p/miR-19a-3p and PTEN were determined through RIP and dual luciferase reporter analysis. Cell survival, viability, and apoptosis were assessed through colony formation, CCK-8, and flow-cytometry assays. RESULTS RBPMS-AS1 was downregulated in LC and mainly distributed in cytoplasm. RBPMS-AS1 targeted miR-19a-3p in LC cells. Radiation suppressed LC cell survival, viability, and induced apoptosis, as overexpressed RBPMS-AS1 performed the similar effects and enhanced those effects induced by radiation. MiR-19a-3p mimic reversed the effect of overexpressed RBPMS-AS1 on enhancing radiation-induced LC cell apoptosis. MiR-19a-3p targeted PTEN and miR-19a-3p mimic reversed the effect of overexpressed RBPMS-AS1 on PTEN and phosphorylation of AKT in LC cells. CONCLUSION Overexpressed RBPMS-AS1 sponged miR-19a-3p to increase cell radiosensitivity in LC via regulating PTEN/AKT axis.
Collapse
Affiliation(s)
- Chengyu Ye
- Department of Radiotherapy, Wenzhou Central Hospital/Theorem Hospital Affiliated of Wenzhou Medical University, Wenzhou, China
| | - Quanbing Lin
- Department of Radiotherapy, Wenzhou Central Hospital/Theorem Hospital Affiliated of Wenzhou Medical University, Wenzhou, China
| | - Cuiping Zheng
- Department of Chemotherapy, Wenzhou Central Hospital/Theorem Hospital Affiliated of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
3
|
Zhou T, Zhang LY, He JZ, Miao ZM, Li YY, Zhang YM, Liu ZW, Zhang SZ, Chen Y, Zhou GC, Liu YQ. Review: Mechanisms and perspective treatment of radioresistance in non-small cell lung cancer. Front Immunol 2023; 14:1133899. [PMID: 36865554 PMCID: PMC9971010 DOI: 10.3389/fimmu.2023.1133899] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 01/31/2023] [Indexed: 02/16/2023] Open
Abstract
Radiotherapy is the major treatment of non-small cell lung cancer (NSCLC). The radioresistance and toxicity are the main obstacles that leading to therapeutic failure and poor prognosis. Oncogenic mutation, cancer stem cells (CSCs), tumor hypoxia, DNA damage repair, epithelial-mesenchymal transition (EMT), and tumor microenvironment (TME) may dominate the occurrence of radioresistance at different stages of radiotherapy. Chemotherapy drugs, targeted drugs, and immune checkpoint inhibitors are combined with radiotherapy to treat NSCLC to improve the efficacy. This article reviews the potential mechanism of radioresistance in NSCLC, and discusses the current drug research to overcome radioresistance and the advantages of Traditional Chinese medicine (TCM) in improving the efficacy and reducing the toxicity of radiotherapy.
Collapse
Affiliation(s)
- Ting Zhou
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China,Experimental & Training Teaching Centers, Gansu University of Chinese Medicine, Lanzhou, China
| | - Li-Ying Zhang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China,College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Jian-Zheng He
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China,College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Zhi-Ming Miao
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yang-Yang Li
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yi-Ming Zhang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
| | - Zhi-Wei Liu
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
| | - Shang-Zu Zhang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yan Chen
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
| | - Gu-Cheng Zhou
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yong-Qi Liu
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China,College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China,Key Laboratory of Dunhuang Medicine and Transformation at Provincial and Ministerial Level, Gansu University of Chinese Medicine, Lanzhou, China,*Correspondence: Yong-Qi Liu,
| |
Collapse
|
4
|
Venugopala KN. Targeting the DNA Damage Response Machinery for Lung Cancer Treatment. Pharmaceuticals (Basel) 2022; 15:ph15121475. [PMID: 36558926 PMCID: PMC9781725 DOI: 10.3390/ph15121475] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 11/18/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Lung cancer is considered the most commonly diagnosed cancer and one of the leading causes of death globally. Despite the responses from small-cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC) patients to conventional chemo- and radiotherapies, the current outcomes are not satisfactory. Recently, novel advances in DNA sequencing technologies have started to take off which have provided promising tools for studying different tumors for systematic mutation discovery. To date, a limited number of DDR inhibition trials have been conducted for the treatment of SCLC and NSCLC patients. However, strategies to test different DDR inhibitor combinations or to target multiple pathways are yet to be explored. With the various biomarkers that have either been recently discovered or are the subject of ongoing investigations, it is hoped that future trials would be designed to allow for studying targeted treatments in a biomarker-enriched population, which is defensible for the improvement of prognosis for SCLC and NSCLC patients. This review article sheds light on the different DNA repair pathways and some of the inhibitors targeting the proteins involved in the DNA damage response (DDR) machinery, such as ataxia telangiectasia and Rad3-related protein (ATR), DNA-dependent protein kinase (DNA-PK), and poly-ADP-ribose polymerase (PARP). In addition, the current status of DDR inhibitors in clinical settings and future perspectives are discussed.
Collapse
Affiliation(s)
- Katharigatta N. Venugopala
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia;
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, Durban 4000, South Africa
| |
Collapse
|
5
|
Genome-wide CRISPR/Cas9 screening identifies CARHSP1 responsible for radiation resistance in glioblastoma. Cell Death Dis 2021; 12:724. [PMID: 34290231 PMCID: PMC8295287 DOI: 10.1038/s41419-021-04000-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 01/09/2023]
Abstract
Glioblastomas (GBM) is the most common primary malignant brain tumor, and radiotherapy plays a critical role in its therapeutic management. Unfortunately, the development of radioresistance is universal. Here, we identified calcium-regulated heat-stable protein 1 (CARHSP1) as a critical driver for radioresistance utilizing genome-wide CRISPR activation screening. This is a protein with a cold-shock domain (CSD)-containing that is highly similar to cold-shock proteins. CARHSP1 mRNA level was upregulated in irradiation-resistant GBM cells and knockdown of CARHSP1 sensitized GBM cells to radiotherapy. The high expression of CARHSP1 upon radiation might mediate radioresistance by activating the inflammatory signaling pathway. More importantly, patients with high levels of CARHSP1 had poorer survival when treated with radiotherapy. Collectively, our findings suggested that targeting the CARHSP1/TNF-α inflammatory signaling activation induced by radiotherapy might directly affect radioresistance and present an attractive therapeutic target for GBM, particularly for patients with high levels of CARHSP1.
Collapse
|
6
|
Haslett K, Koh P, Hudson A, Ryder W, Falk S, Mullan D, Taylor B, Califano R, Blackhall F, Faivre-Finn C. Phase I trial of the MEK inhibitor selumetinib in combination with thoracic radiotherapy in non-small cell lung cancer. Clin Transl Radiat Oncol 2021; 28:24-31. [PMID: 33748440 PMCID: PMC7970011 DOI: 10.1016/j.ctro.2021.02.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/19/2021] [Accepted: 02/20/2021] [Indexed: 12/25/2022] Open
Abstract
Background The RAS/RAF/MEK/ERK signalling pathway has a pivotal role in cancer proliferation and modulating treatment response. Selumetinib inhibits MEK and enhances effects of radiotherapy in preclinical studies. Patients and methods Single-arm, single-centre, open-label phase I trial. Patients with stage III NSCLC unsuitable for concurrent chemo-radiotherapy, or stage IV with dominant thoracic symptoms, were recruited to a dose-finding stage (Fibonacci 3 + 3 design; maximum number = 18) then an expanded cohort (n = 15). Oral selumetinib was administered twice daily (starting dose 50 mg) commencing 7 days prior to thoracic radiotherapy, then with radiotherapy (6-6.5 weeks; 60-66 Gy/30-33 fractions). The primary objective was to determine the recommended phase II dose (RP2D) of selumetinib in combination with thoracic radiotherapy. Results 21 patients were enrolled (06/2010-02/2015). Median age: 62y (range 50-73). M:F ratio 12(57%):9(43%). ECOG PS 0:1, 7(33%):14(67%). Stage III 16(76%); IV 5(24%). Median GTV 64 cm3 (range 1-224 cm3). 15 patients comprised the expanded cohort at starting dose. All 21 patients completed thoracic radiotherapy as planned and received induction chemotherapy. 13 (62%) patients received the full dose of selumetinib.In the starting cohort no enhanced radiotherapy-related toxicity was seen. Two patients had dose-limiting toxicity (1x grade 3 diarrhoea/fatigue and 1x pulmonary embolism). Commonest grade 3-4 adverse events: lymphopaenia (19/21 patients) and hypertension (7/21 patients). One patient developed grade 3 oesophagitis. No patients developed grade ≥3 radiation pneumonitis. Two patients were alive at the time of analysis (24 and 26 months follow-up, respectively). Main cause of first disease progression: distant metastases ± locoregional progression (12/21 [57.1%] patients). Six patients had confirmed/suspected pneumocystis jiroveci pneumonia. Conclusion We report poor outcome and severe lymphopenia in most patients treated with thoracic radiotherapy and selumetinib at RP2D in combination, contributing to confirmed/clinically suspected pneumocystis jiroveci pneumonia. These results suggest that this combination should not be pursued in a phase II trial.ClinicalTrials.gov reference: NCT01146756.
Collapse
Affiliation(s)
- K. Haslett
- The Christie NHS Foundation Trust, United Kingdom
| | - P. Koh
- University of Manchester, United Kingdom
- New Cross Hospital, United Kingdom
| | - A. Hudson
- The Christie NHS Foundation Trust, United Kingdom
| | - W.D. Ryder
- University of Manchester, United Kingdom
| | - S. Falk
- The Christie NHS Foundation Trust, United Kingdom
| | - D. Mullan
- The Christie NHS Foundation Trust, United Kingdom
| | - B. Taylor
- The Christie NHS Foundation Trust, United Kingdom
| | - R. Califano
- The Christie NHS Foundation Trust, United Kingdom
- University of Manchester, United Kingdom
| | - F. Blackhall
- The Christie NHS Foundation Trust, United Kingdom
- University of Manchester, United Kingdom
| | - C. Faivre-Finn
- The Christie NHS Foundation Trust, United Kingdom
- University of Manchester, United Kingdom
- Corresponding author at: The Christie NHS Foundation Trust, United Kingdom.
| |
Collapse
|
7
|
Circulating Exosomal miR-96 as a Novel Biomarker for Radioresistant Non-Small-Cell Lung Cancer. JOURNAL OF ONCOLOGY 2021; 2021:5893981. [PMID: 33727921 PMCID: PMC7937465 DOI: 10.1155/2021/5893981] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 01/24/2021] [Accepted: 01/31/2021] [Indexed: 12/17/2022]
Abstract
Patients with non-small-cell lung cancer (NSCLC) frequently develop radioresistance, resulting in poor response to radiation and unfavourable prognosis. Early detection of radioresistance hence can guide the adjustment of treatment regimens in time. Exosomes are lipid bilayer-enclosed vesicles with sub-micrometer size that are released by various cells. Exosomes contain a tissue-specific signature wherein a variety of proteins and nucleic acids are selectively packaged. Growing evidence shows exosomes are involved in cancer pathophysiology and exosomes as the latest addition to the liquid biopsy portfolio have been used in cancer diagnosis. Compared to cell free RNA, exosomal lipid envelope can effectively protect RNA cargo against degradation. Therefore, exosomes may hold great promise for the identification of radioresistance. Here, we report six plasma exosomal miRNAs could be used to distinguish radioresistant NSCLC patients from radiosensitive NSCLC patients and to evaluate the prognosis of NSCLC. Samples were obtained from 52 NSCLC patients with or without radioresistance and 45 age-matched healthy volunteers. Exosomes in 1 ml plasma were isolated followed by extraction of small RNA. The expression levels of miRNAs were determined by quantitative real-time PCR. Potential miRNA markers were further evaluated in additional 52 NSCLC patients. We found exosomal miR-1246 and miR-96 are significantly overexpressed in NSCLC patients. Moreover, exosomal miR-96 in patients with radioresistant NSCLC is significantly higher than that of controls. Exosomal miR-96 also demonstrates a significant correlation with vascular invasion and poor overall survival. Altogether, our results indicate that exosomal miR-96 could be a non-invasive diagnostic and prognostic marker of radioresistant NSCLC.
Collapse
|
8
|
Chen JJ, Sullivan AJ, Shi DD, Krishnan MS, Hertan LM, Roldan CS, Huynh MA, Spektor A, Fareed MM, Lam TC, Balboni TA. Characteristics and Predictors of Radiographic Local Failure in Patients With Spinal Metastases Treated With Palliative Conventional Radiation Therapy. Adv Radiat Oncol 2021; 6:100665. [PMID: 33817411 PMCID: PMC8010570 DOI: 10.1016/j.adro.2021.100665] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 12/18/2020] [Accepted: 01/24/2021] [Indexed: 11/20/2022] Open
Abstract
Purpose Although local control is an important issue for longer-term survivors of spinal metastases treated with conventional external beam radiation therapy (EBRT), the literature on radiographic local failure (LF) in these patients is sparse. To inform clinical decision-making, we evaluated rates, consequences, and predictors of radiographic LF in patients with spinal metastases managed with palliative conventional EBRT alone. Methods and Materials We retrospectively reviewed 296 patients with spinal metastases who received palliative EBRT at a single institution (2006-2013). Radiographic LF was defined as radiologic progression within the treatment field, with death considered a competing risk. Kaplan-Meier, cumulative incidence, and Cox regression analyses determined overall survival estimates, LF rates, and predictors of LF, respectively. Results There were 182 patients with follow-up computed tomography or magnetic resonance imaging; median overall survival for these patients was 7.7 months. Patients received a median of 30 Gy in 10 fractions to a median of 4 vertebral bodies. Overall, 74 of 182 patients (40.7%) experienced LF. The 6-, 12-, and 18-month LF rates were 26.5%, 33.1%, and 36.5%, respectively, while corresponding rates of death were 24.3%, 38.1%, and 45.9%. Median time to LF was 3.8 months. Of those with LF, 51.4% had new compression fractures, 39.2% were admitted for pain control, and 35.1% received reirradiation; median time from radiation therapy (RT) to each of these events was 3.0, 5.7, and 9.2 months, respectively. Independent predictors of LF included single-fraction RT (8 Gy) (hazard ratio [HR], 2.592; 95% confidence interval [CI], 1.437-4.675; P = .002), lung histology (HR, 3.568; 95% CI, 1.532-8.309; P = .003), and kidney histology (HR, 4.937; 95% CI, 1.529-15.935; P = .008). Conclusions Patients experienced a >30% rate of radiographic LF by 1 year after EBRT. Single-fraction RT and lung or kidney histology predicted LF. Given the high rates of LF for patients with favorable prognosis, assessing the risk of death versus LF is important for clinical decision-making.
Collapse
Affiliation(s)
- Jie Jane Chen
- Department of Radiation Oncology, Dana-Farber Cancer Institute/Brigham and Women’s Cancer Center, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Adam J. Sullivan
- Department of Biostatistics, Brown University, Providence, Rhode Island
| | - Diana D. Shi
- Department of Radiation Oncology, Dana-Farber Cancer Institute/Brigham and Women’s Cancer Center, Boston, Massachusetts
| | - Monica S. Krishnan
- Department of Radiation Oncology, Dana-Farber Cancer Institute/Brigham and Women’s Cancer Center, Boston, Massachusetts
| | - Lauren M. Hertan
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Claudia S. Roldan
- Department of Radiation Oncology, Dana-Farber Cancer Institute/Brigham and Women’s Cancer Center, Boston, Massachusetts
| | - Mai Anh Huynh
- Department of Radiation Oncology, Dana-Farber Cancer Institute/Brigham and Women’s Cancer Center, Boston, Massachusetts
| | - Alexander Spektor
- Department of Radiation Oncology, Dana-Farber Cancer Institute/Brigham and Women’s Cancer Center, Boston, Massachusetts
| | - M. Mohsin Fareed
- Department of Radiation Oncology, Dana-Farber Cancer Institute/Brigham and Women’s Cancer Center, Boston, Massachusetts
| | - Tai Chung Lam
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Tracy A. Balboni
- Department of Radiation Oncology, Dana-Farber Cancer Institute/Brigham and Women’s Cancer Center, Boston, Massachusetts
- Corresponding author: Tracy A. Balboni, MD, MPH
| |
Collapse
|
9
|
Guo G, Li L, Song G, Wang J, Yan Y, Zhao Y. miR‑7/SP1/TP53BP1 axis may play a pivotal role in NSCLC radiosensitivity. Oncol Rep 2020; 44:2678-2690. [PMID: 33125142 PMCID: PMC7640372 DOI: 10.3892/or.2020.7824] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 09/04/2020] [Indexed: 12/19/2022] Open
Abstract
MicroRNA‑7 (miR‑7) has been identified as a tumor suppressor in non‑small cell lung cancer (NSCLC) and a radiosensitivity regulator. Numerous studies have revealed that specific protein 1 (SP1) plays a critical role in the tumorigenesis of various types of cancers and regulates radiosensitivity and tumor suppressor p53‑binding protein 1 (TP53BP1), which plays an essential role in DNA repair. However, it is not clear whether miR‑7 has a regulatory effect on SP1 and TP53BP1 in NSCLC. In the present study it was revealed that miR‑7 directly binds to the 3'UTR of SP1, thereby suppressing SP1 expression to regulate radiosensitivity. Overexpression of miR‑7 and SP1 and knockdown of miR‑7 and SP1 were performed using lentiviral transfection. Protein and mRNA abundance of SP1 and TP53BP1 were determined using western blotting and RT‑qPCR, respectively, while miR‑7 binding to SP1 was validated using a luciferase reporter assay. Biological function analysis indicated that miR‑7 negatively regulated SP1 and inhibited cell proliferation, migration, and invasion when combined with radiation. It was also revealed that the expression of TP53BP1 was positively regulated by SP1 or negatively regulated by miR‑7. In conclusion, SP1 was a target of miR‑7, and the decreased expression of SP1 resulting from miR‑7 overexpression in NSCLC was vital for improving radiosensitivity in NSCLC cells. Moreover, SP1 expression was detected in 95 paired NSCLC and adjacent normal tissues, and it was determined that SP1 was significantly upregulated in NSCLC tissues and that its upregulation was correlated with the degree of tissue differentiation. Thus, SP1 and/or miR‑7 may be potential molecular targets in NSCLC radiotherapy.
Collapse
Affiliation(s)
- Genyan Guo
- Department of Radiation Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, P.R. China
| | - Lingling Li
- Department of Radiation Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, P.R. China
| | - Guanchu Song
- Department of Radiation Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, P.R. China
| | - Jie Wang
- Department of Radiation Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, P.R. China
- Department of Radiation Oncology, Dalian Municipal Central Hospital, Dalian, Liaoning 116033, P.R. China
| | - Ying Yan
- Department of Radiation Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, P.R. China
- Department of Radiation Oncology, The General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, P.R. China
| | - Yuxia Zhao
- Department of Radiation Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, P.R. China
| |
Collapse
|
10
|
Zeng Y, Jie X, Wu B, Wu G, Liu L, Xu S. IQGAP3 interacts with Rad17 to recruit the Mre11-Rad50-Nbs1 complex and contributes to radioresistance in lung cancer. Cancer Lett 2020; 493:254-265. [PMID: 32896617 DOI: 10.1016/j.canlet.2020.08.042] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/11/2020] [Accepted: 08/29/2020] [Indexed: 12/25/2022]
Abstract
IQ motif containing GTPase-activating protein 3 (IQGAP3) has been implicated in diverse cellular processes, including neuronal morphogenesis, cell proliferation and motility, and epithelial-mesenchymal transition. However, its role in cancer radioresistance is completely unknown. Here, we report that IQGAP3 is overproduced in lung cancer patients and correlates with poor clinical outcomes. Functionally, we demonstrate that depletion of IQGAP3 impairs oncogenesis and overcomes radioresistance in lung cancer in vitro and in vivo. Mechanistically, we uncover that IQGAP3 interacts with Rad17 and controls its expression to activate the ATM/Chk2 and ATR/Chk1 signaling pathways by recruiting the Mre11-Rad50-Nbs1 (MRN) complex in response to DNA damage. Moreover, Rad17 is identified as the major downstream effector that mediates the functions of IQGAP3 in lung cancer. Clinically, IQGAP3 overexpression positively correlates with Rad17 upregulation in human lung cancer tissues. Collectively, these data support key role for IQGAP3 in promoting lung cancer radioresistance by interacting with Rad17 and suggest that targeting IQGAP3 may be an attractive strategy for lung cancer radiotherapy.
Collapse
Affiliation(s)
- Yulan Zeng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaohua Jie
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Bian Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Gang Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Li Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Shuangbing Xu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
11
|
Kim MY, Shin JY, Kim JO, Son KH, Kim YS, Jung CK, Kang JH. Anti-tumor efficacy of CKD-516 in combination with radiation in xenograft mouse model of lung squamous cell carcinoma. BMC Cancer 2020; 20:1057. [PMID: 33143663 PMCID: PMC7607852 DOI: 10.1186/s12885-020-07566-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 10/26/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Hypoxic tumors are known to be highly resistant to radiotherapy and cause poor prognosis in non-small cell lung cancer (NSCLC) patients. CKD-516, a novel vascular disrupting agent (VDA), mainly affects blood vessels in the central area of the tumor and blocks tubulin polymerization, thereby destroying the aberrant tumor vasculature with a rapid decrease in blood, resulting in rapid tumor cell death. Therefore, we evaluated the anti-tumor efficacy of CKD-516 in combination with irradiation (IR) and examined tumor necrosis, delayed tumor growth, and expression of proteins involved in hypoxia and angiogenesis in this study. METHODS A xenograft mouse model of lung squamous cell carcinoma was established, and the tumor was exposed to IR 5 days per week. CKD-516 was administered with two treatment schedules (day 1 or days 1 and 5) 1 h after IR. After treatment, tumor tissues were stained with hematoxylin and eosin, and pimonidazole. HIF-1α, Glut-1, VEGF, CD31, and Ki-67 expression levels were evaluated using immunohistochemical staining. RESULTS Short-term treatment with IR alone and CKD-516 + IR (d1) significantly reduced tumor volume (p = 0.006 and p = 0.048, respectively). Treatment with CKD-516 + IR (d1 and d1, 5) resulted in a marked reduction in the number of blood vessels (p < 0.005). More specifically, CKD-516 + IR (d1) caused the most extensive tumor necrosis, which resulted in a significantly large hypoxic area (p = 0.02) and decreased HIF-1α, Glut-1, VEGF, and Ki-67 expression. Long-term administration of CKD-516 + IR reduced tumor volume and delayed tumor growth. This combination also greatly reduced the number of blood vessels (p = 0.0006) and significantly enhanced tumor necrosis (p = 0.004). CKD-516 + IR significantly increased HIF-1α expression (p = 0.0047), but significantly reduced VEGF expression (p = 0.0046). CONCLUSIONS Taken together, our data show that when used in combination, CKD-516 and IR can significantly enhance anti-tumor efficacy compared to monotherapy in lung cancer xenograft mice.
Collapse
Affiliation(s)
- Min-Young Kim
- Laboratory of Medical Oncology, Cancer Research Institute, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jung-Young Shin
- Laboratory of Medical Oncology, Cancer Research Institute, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jeong-Oh Kim
- Laboratory of Medical Oncology, Cancer Research Institute, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kyoung-Hwa Son
- Laboratory of Medical Oncology, Cancer Research Institute, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yeon Sil Kim
- Department of Radiation Oncology, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Chan Kwon Jung
- Department of Pathology, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jin-Hyoung Kang
- Laboratory of Medical Oncology, Cancer Research Institute, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Medical Oncology, Seoul St. Mary’s Hospital, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591 Republic of Korea
| |
Collapse
|
12
|
Ding Z, Kang J, Yang Y. Long non-coding RNA CASC2 enhances irradiation-induced endoplasmic reticulum stress in NSCLC cells through PERK signaling. 3 Biotech 2020; 10:449. [PMID: 33062578 DOI: 10.1007/s13205-020-02443-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 09/15/2020] [Indexed: 12/24/2022] Open
Abstract
Radiotherapy is instrumental in the treatment of inoperable non-small cell lung cancer (NSCLC). Studies have revealed that radiotherapy induces endoplasmic reticulum (ER) stress, which consequently induces apoptosis and sensitization of cancer cells. A recent study has revealed that long non-coding RNA (lncRNA) CASC2 is negatively correlated with the malignancy of NSCLC cells. The present study investigated the effects and molecular mechanisms of CASC2 on radiosensitivity and ER stress in NSCLC cells. The overexpression of CASC2 markedly decreased cell survival and increased apoptosis, expression of PERK, phosphorylated-eIF2α and CHOP in irradiated human NSCLC cells, whereas knocking down PERK reversed these effects. Moreover, CASC2 considerably promoted the stability of PERK mRNA, but had no effect on the activity of PERK gene promoter in irradiated NSCLC cells. Strikingly, CASC2 exhibited no apparent effect on non-irradiated NSCLC cells. This study demonstrated that lncRNA CASC2 increases the stability of PERK mRNA, which consequently triggers the PERK/eIF2α/CHOP ER stress pathway and promotes radiosensitivity or apoptosis in irradiated NSCLC cells. Results of the present study suggest that CASC2 can act as an effective therapeutic target to enhance the efficacy of radiotherapy in the treatment of NSCLC.
Collapse
Affiliation(s)
- Zhouli Ding
- Department of Respiratory, Qingpu Branch, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Jianmei Kang
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 0086-410011 Hunan People's Republic of China
| | - Yu Yang
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 0086-410011 Hunan People's Republic of China
| |
Collapse
|
13
|
Liu YM, Chan YL, Wu TH, Li TL, Hsia S, Chiu YH, Wu CJ. Antitumor, Inhibition of Metastasis and Radiosensitizing Effects of Total Nutrition Formula on Lewis Tumor-Bearing Mice. Nutrients 2019; 11:nu11081944. [PMID: 31426614 PMCID: PMC6723674 DOI: 10.3390/nu11081944] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 08/14/2019] [Accepted: 08/15/2019] [Indexed: 12/14/2022] Open
Abstract
Non-small-cell lung cancer (NSCLC) causes high mortality. Radiotherapy is an induction regimen generally applied to patients with NSCLC. In view of therapeutic efficacy, the outcome is not appealing in addition to bringing about unwanted side effects. Total nutrition is a new trend in cancer therapy, which benefits cancer patients under radiotherapy. Male C57BL/6JNarl mice were experimentally divided into five groups: one control group, one T group (borne with Lewis lung carcinoma but no treatment), and three Lewis lung carcinoma-bearing groups administrated with a total nutrition formula (T + TNuF group), a local radiotherapy plus daily 3 Gy in three fractions (T + R group), or a combination TNuF and radiotherapy (T + R + TNuF group). These mice were assessed for their mean tumor volumes, cachectic symptoms and tumor metastasis. TNuF administration significantly suppressed tumor growth and activated apoptotic cell death in NSCLC-bearing mice under radiation. The body-weight gain was increased, while the radiation-induced cachexia was alleviated. Analysis of mechanisms suggests that TNuF downregulates EGFR and VEGF signaling pathways, inhibiting angiogenesis and metastasis. In light of radiation-induced tumor cell death, mitigation of radiation-induced cachexia and inhibition of tumor cell distant metastasis, the combination of TNuF and radiotherapy synergistically downregulates EGFR and VEGF signaling in NSCLC-bearing mice.
Collapse
Affiliation(s)
- Yu-Ming Liu
- Division of Radiation Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- School of Medicine, National Yang Ming University, Taipei 11221, Taiwan
| | - Yi-Lin Chan
- Department of Life Science, Chinese Culture University, Taipei 11114, Taiwan
| | - Tsung-Han Wu
- Department of Food Science and Center of Excellence for the Oceans, National Taiwan Ocean University, Keelung 20224, Taiwan
- Division of Hemato-oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 33320, Taiwan
| | - Tsung-Lin Li
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Simon Hsia
- Taiwan Nutraceutical Association, Taipei 10596, Taiwan
| | - Yi-Han Chiu
- Department of Nursing, St. Mary's Junior College of Medicine, Nursing and Management, Yilan 26647, Taiwan.
- Institute of Long-Term Care, Mackay Medical College, New Taipei City 25245, Taiwan.
| | - Chang-Jer Wu
- Department of Food Science and Center of Excellence for the Oceans, National Taiwan Ocean University, Keelung 20224, Taiwan.
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan.
- Department of Health and Nutrition Biotechnology, Asia University, Taichung 41354, Taiwan.
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| |
Collapse
|
14
|
Jiang W, Jin G, Cai F, Chen X, Cao N, Zhang X, Liu J, Chen F, Wang F, Dong W, Zhuang H, Hua ZC. Extracellular signal-regulated kinase 5 increases radioresistance of lung cancer cells by enhancing the DNA damage response. Exp Mol Med 2019; 51:1-20. [PMID: 30804322 PMCID: PMC6389946 DOI: 10.1038/s12276-019-0209-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 10/26/2018] [Accepted: 10/29/2018] [Indexed: 12/18/2022] Open
Abstract
Radiotherapy is a frequent mode of cancer treatment, although the development of radioresistance limits its effectiveness. Extensive investigations indicate the diversity of the mechanisms underlying radioresistance. Here, we aimed to explore the effects of extracellular signal-regulated kinase 5 (ERK5) on lung cancer radioresistance and the associated mechanisms. Our data showed that ERK5 is activated during solid lung cancer development, and ectopic expression of ERK5 promoted cell proliferation and G2/M cell cycle transition. In addition, we found that ERK5 is a potential regulator of radiosensitivity in lung cancer cells. Mechanistic investigations revealed that ERK5 could trigger IR-induced activation of Chk1, which has been implicated in DNA repair and cell cycle arrest in response to DNA double-strand breaks (DSBs). Subsequently, ERK5 knockdown or pharmacological inhibition selectively inhibited colony formation of lung cancer cells and enhanced IR-induced G2/M arrest and apoptosis. In vivo, ERK5 knockdown strongly radiosensitized A549 and LLC tumor xenografts to inhibition, with a higher apoptotic response and reduced tumor neovascularization. Taken together, our data indicate that ERK5 is a novel potential target for the treatment of lung cancer, and its expression might be used as a biomarker to predict radiosensitivity in NSCLC patients. Resistance to radiotherapy in patients with lung cancer may be countered by targeting a protein involved in promoting DNA repair. Radiotherapy causes DNA double-stranded breaks in lung cancer cells in order to kill them. However, cancer cells can show improved DNA repair and responses to damage, resulting in resistance to treatment. Zi-Chun Hua, Hongqin Zhuang at Nanjing University in China and co-workers examined the activity of the extracellular signal-related kinase 5 (ERK5) protein in response to the stress of ionizing radiation. They found that after radiation exposure ERK5 increased expression of another protein involved in DNA repair, facilitating cancer cell recovery. Knocking out ERK5 suppressed this resistance to radiotherapy. ERK5 could be a valuable target for treating lung cancer, and ERK5 expression level could be used as a biomarker for patient sensitivity to radiotherapy.
Collapse
Affiliation(s)
- Weiwei Jiang
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, PR China
| | - Guanghui Jin
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, PR China.,Department of Basic Medical Sciences, Medical College, Xiamen University, Xiamen, PR China
| | - Fangfang Cai
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, PR China
| | - Xiao Chen
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, PR China
| | - Nini Cao
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, PR China
| | - Xiangyu Zhang
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, PR China
| | - Jia Liu
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, PR China
| | - Fei Chen
- Department of Nuclear Medicine, The Affiliated Nanjing First Hospital, Nanjing Medical University, Nanjing, PR China
| | - Feng Wang
- Department of Nuclear Medicine, The Affiliated Nanjing First Hospital, Nanjing Medical University, Nanjing, PR China
| | - Wei Dong
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, PR China
| | - Hongqin Zhuang
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, PR China.
| | - Zi-Chun Hua
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, PR China. .,Changzhou High-Tech Research Institute of Nanjing University and Jiangsu Target Pharma Laboratories Inc., Changzhou, 213164, PR China.
| |
Collapse
|
15
|
Kim W, Son B, Lee S, Do H, Youn B. Targeting the enzymes involved in arachidonic acid metabolism to improve radiotherapy. Cancer Metastasis Rev 2018; 37:213-225. [DOI: 10.1007/s10555-018-9742-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
16
|
Lu Z, Tang Y, Luo J, Zhang S, Zhou X, Fu L. Advances in targeting the transforming growth factor β1 signaling pathway in lung cancer radiotherapy. Oncol Lett 2017; 14:5681-5687. [PMID: 29113195 DOI: 10.3892/ol.2017.6991] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 07/21/2017] [Indexed: 02/06/2023] Open
Abstract
Lung cancer was demonstrated to be the most lethal type of malignant tumor amongst humans in the global cancer statistics of 2012. As one of the primary treatments, radiotherapy has been reported to induce remission in, and even cure, patients with lung cancer. However, the side effects of radiotherapy may prove lethal in certain patients. In past decades, the transforming growth factor β1 (TGFB1) signaling pathway has been revealed to serve multiple functions in the control of lung cancer progression and the radiotherapy response. In mammals, this signaling pathway is initiated through activation of the TGFB1 receptor complex, which signals via cytoplasmic SMAD proteins or other downstream signaling pathways. Multiple studies have demonstrated that TGFB1 serves important functions in lung cancer radiotherapy. The present study summarized and reviewed recent progress in elucidating the function of the TGFB1 signaling pathway in predicting radiation pneumonitis, as well as current strategies for targeting the TGFB1 signaling pathway in lung cancer radiotherapy, which may provide potential targets for lung cancer therapy.
Collapse
Affiliation(s)
- Zhonghua Lu
- Department of Radiation Oncology, Changzhou Cancer Hospital, Soochow University, Changzhou, Jiangsu 213001, P.R. China
| | - Yiting Tang
- Department of Radiation Oncology, Changzhou Cancer Hospital, Soochow University, Changzhou, Jiangsu 213001, P.R. China
| | - Judong Luo
- Department of Radiation Oncology, Changzhou Cancer Hospital, Soochow University, Changzhou, Jiangsu 213001, P.R. China
| | - Shuyu Zhang
- Department of Radiation Biology, School of Radiation Medicine and Protection and Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Xifa Zhou
- Department of Radiation Oncology, Changzhou Cancer Hospital, Soochow University, Changzhou, Jiangsu 213001, P.R. China
| | - Lei Fu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| |
Collapse
|
17
|
Marostica LL, de Barros ALB, Oliveira J, Salgado BS, Cassali GD, Leite EA, Cardoso VN, Lang KL, Caro MSB, Durán FJ, Schenkel EP, de Oliveira MC, Simões CMO. Antitumor effectiveness of a combined therapy with a new cucurbitacin B derivative and paclitaxel on a human lung cancer xenograft model. Toxicol Appl Pharmacol 2017; 329:272-281. [DOI: 10.1016/j.taap.2017.06.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Revised: 06/06/2017] [Accepted: 06/09/2017] [Indexed: 02/07/2023]
|
18
|
De Ruysscher D, Faivre-Finn C, Moeller D, Nestle U, Hurkmans CW, Le Péchoux C, Belderbos J, Guckenberger M, Senan S. European Organization for Research and Treatment of Cancer (EORTC) recommendations for planning and delivery of high-dose, high precision radiotherapy for lung cancer. Radiother Oncol 2017; 124:1-10. [PMID: 28666551 DOI: 10.1016/j.radonc.2017.06.003] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 04/25/2017] [Accepted: 06/05/2017] [Indexed: 12/23/2022]
Abstract
PURPOSE To update literature-based recommendations for techniques used in high-precision thoracic radiotherapy for lung cancer, in both routine practice and clinical trials. METHODS A literature search was performed to identify published articles that were considered clinically relevant and practical to use. Recommendations were categorised under the following headings: patient positioning and immobilisation, Tumour and nodal changes, CT and FDG-PET imaging, target volumes definition, radiotherapy treatment planning and treatment delivery. An adapted grading of evidence from the Infectious Disease Society of America, and for models the TRIPOD criteria, were used. RESULTS Recommendations were identified for each of the above categories. CONCLUSION Recommendations for the clinical implementation of high-precision conformal radiotherapy and stereotactic body radiotherapy for lung tumours were identified from the literature. Techniques that were considered investigational at present are highlighted.
Collapse
Affiliation(s)
- Dirk De Ruysscher
- Maastricht University Medical Center+, Department of Radiation Oncology (Maastro Clinic), GROW Research Institute, The Netherlands; KU Leuven, Radiation Oncology, Belgium.
| | - Corinne Faivre-Finn
- Division of Cancer Sciences University of Manchester, Christie NHS Foundation Trust, UK
| | - Ditte Moeller
- Aarhus University Hospital, Department of Oncology, Denmark
| | - Ursula Nestle
- Freiburg University Medical Center (DKTK partner site), Department of Radiation Oncology, Germany; Department of Radiation Oncology, Kliniken Maria Hilf, Moenchengladbach, Germany
| | - Coen W Hurkmans
- Catharina Hospital, Department of Radiation Oncology, Eindhoven, The Netherlands
| | | | - José Belderbos
- Netherlands Cancer Institute, Department of Radiation Oncology, Amsterdam, The Netherlands
| | | | - Suresh Senan
- VU University Medical Center, Department of Radiation Oncology, Amsterdam, The Netherlands
| |
Collapse
|
19
|
Ishihara M, Igawa S, Sasaki J, Otani S, Fukui T, Ryuge S, Katono K, Hiyoshi Y, Kasajima M, Mitsufuji H, Kubota M, Yokoba M, Katagiri M, Sekiguchi A, Soda I, Ishiyama H, Hayakawa K, Masuda N. Evaluation of concurrent chemoradiotherapy for locally advanced NSCLC according to EGFR mutation status. Oncol Lett 2017; 14:885-890. [PMID: 28693247 DOI: 10.3892/ol.2017.6231] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 01/31/2017] [Indexed: 01/19/2023] Open
Abstract
Concurrent chemoradiotherapy (cCRT) is the standard treatment for patients with locally advanced non-small cell lung cancer (LA-NSCLC). However, the efficacy and safety of this treatment has not been compared between patients who possess epidermal growth factor receptor (EGFR) mutations and patients with wild-type EGFR. The objective of the present study was to evaluate the effect of the presence of EGFR gene mutations in patients with LA-NSCLC receiving cCRT. Between January 2007 and December 2013, the records of 64 patients were reviewed retrospectively. The data were statistically analyzed to evaluate the efficacy of cCRT according to EGFR mutation status. In total, 15/64 were revealed to possess EGFR mutations, 23%, and comprised the mutant EGFR group. The progression-free survival time was significantly shorter in the mutant EGFR group compared with the patient group with tumors exhibiting wild-type EGFR, 6.3 and 9.5 months, respectively (P<0.001). The overall survival rate was longer in the mutant EGFR group compared with the wild-type EGFR group, although the difference was not statistically significant, 37.1 and 21.1 months, respectively (P=0.26). The disease recurred in all of the patients of the mutant EGFR group, whilst the recurrence rate in the wild-type EGFR group was 89%. The frequency of distant metastasis was significantly higher in the mutant EGFR group compared with the wild-type EGFR group. In conclusion, these data suggest that additional studies are required to identify strategies for reinforcing the efficacy of cCRT, with a focus on the potential use of EGFR tyrosine kinase inhibitors for patients exhibiting an EGFR mutation.
Collapse
Affiliation(s)
- Mikiko Ishihara
- Department of Respiratory Medicine, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan
| | - Satoshi Igawa
- Department of Respiratory Medicine, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan
| | - Jiichiro Sasaki
- Research and Development Center for New Medical Frontiers, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan
| | - Sakiko Otani
- Department of Respiratory Medicine, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan
| | - Tomoya Fukui
- Department of Respiratory Medicine, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan
| | - Shinichiro Ryuge
- Department of Respiratory Medicine, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan
| | - Ken Katono
- Department of Respiratory Medicine, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan
| | - Yasuhiro Hiyoshi
- Department of Respiratory Medicine, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan
| | - Masashi Kasajima
- Department of Respiratory Medicine, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan
| | - Hisashi Mitsufuji
- Department of Respiratory Medicine, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan
| | - Masaru Kubota
- Department of Respiratory Medicine, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan
| | - Masanori Yokoba
- School of Allied Health Sciences, Kitasato University, Sagamihara, Kanagawa 252-0373, Japan
| | - Masato Katagiri
- School of Allied Health Sciences, Kitasato University, Sagamihara, Kanagawa 252-0373, Japan
| | - Akane Sekiguchi
- Department of Radiology and Radiation Oncology, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan
| | - Itaru Soda
- Department of Radiology and Radiation Oncology, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan
| | - Hiromichi Ishiyama
- Department of Radiology and Radiation Oncology, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan
| | - Kazushige Hayakawa
- Department of Radiology and Radiation Oncology, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan
| | - Noriyuki Masuda
- Department of Respiratory Medicine, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan
| |
Collapse
|
20
|
Design, synthesis and biological activities of N-(furan-2-ylmethyl)-1H-indole-3-carboxamide derivatives as epidemal growth factor receptor inhibitors and anticancer agents. Chem Res Chin Univ 2017. [DOI: 10.1007/s40242-017-7041-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
21
|
Wang Y, He J, Zhang S, Yang Q. Intracellular calcium promotes radioresistance of non-small cell lung cancer A549 cells through activating Akt signaling. Tumour Biol 2017; 39:1010428317695970. [PMID: 28351325 DOI: 10.1177/1010428317695970] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Radiotherapy is a major therapeutic approach in non-small cell lung cancer but is restricted by radioresistance. Although Akt signaling promotes radioresistance in non-small cell lung cancer, it is not well understood how Akt signaling is activated. Since intracellular calcium (Ca2+) could activate Akt in A549 cells, we investigated the relationship between intracellular calcium (Ca2+) and Akt signaling in radioresistant A549 cells by establishing radioresistant non-small cell lung cancer A549 cells. The radioresistant cell line A549 was generated by dose-gradient irradiation of the parental A549 cells. The cell viability, proliferation, and apoptosis were, respectively, assessed using the cell counting kit-8, EdU labeling, and flow cytometry analysis. The phosphorylation of Akt was evaluated by Western blotting, and the intracellular Ca2+ concentration was assessed by Fluo 4-AM. The radioresistant A549 cells displayed mesenchymal morphology. After additional irradiation, the radioresistant A549 cells showed decreased cell viability and proliferation but increased apoptosis. Moreover, the intracellular Ca2+ concentration and the phosphorylation level on the Akt473 site in radioresistant A549 cells were higher than those in original cells, whereas the percentage of apoptosis in radioresistant A549 cells was less. All these results could be reversed by verapamil. In conclusion, our study found that intracellular Ca2+ could promote radioresistance of non-small cell lung cancer cells through phosphorylating of Akt on the 473 site, which contributes to a better understanding on the non-small cell lung cancer radioresistance, and may provide a new target for radioresistance management.
Collapse
Affiliation(s)
- Yiling Wang
- Department of Thoracic Surgery, Shanghai Tenth People's Hospital, Shanghai, China
| | - Jiantao He
- Department of Thoracic Surgery, Shanghai Tenth People's Hospital, Shanghai, China
| | - Shenghui Zhang
- Department of Thoracic Surgery, Shanghai Tenth People's Hospital, Shanghai, China
| | - Qingbo Yang
- Department of Thoracic Surgery, Shanghai Tenth People's Hospital, Shanghai, China
| |
Collapse
|
22
|
Hu S, Fu W, Li T, Yuan Q, Wang F, Lv G, Lv Y, Fan X, Shen Y, Lin F, Tang Y, Ye X, Yang Y, Lei C. Antagonism of EGFR and Notch limits resistance to EGFR inhibitors and radiation by decreasing tumor-initiating cell frequency. Sci Transl Med 2017; 9:9/380/eaag0339. [PMID: 28275151 DOI: 10.1126/scitranslmed.aag0339] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 10/16/2016] [Accepted: 02/11/2017] [Indexed: 12/15/2022]
Abstract
Epidermal growth factor receptor (EGFR) blockade and radiation are efficacious in the treatment of cancer, but resistance is commonly reported. Studies have suggested that dysregulation of Notch signaling and enrichment of the cancer stem cell population underlie these treatment challenges. Our data show that dual targeting of EGFR and Notch2/3 receptors with antibody CT16 not only inhibited signaling mediated by these receptors but also showed a strong anti-stem cell effect both in vitro and in vivo. Treatment with CT16 prevented acquired resistance to EGFR inhibitors and radiation in non-small cell lung cancer (NSCLC) cell line models and patient-derived xenograft tumors. CT16 also had a superior radiosensitizing impact compared with EGFR inhibitors. CT16 in combination with radiation had a larger antitumor effect than the combination of radiation with EGFR inhibitors or tarextumab. Mechanistically, CT16 treatment inhibits the stem cell-like subpopulation, which has a high mesenchymal gene expression and DNA repair activity, and reduces tumor-initiating cell frequency. This finding highlights the capacity of a combined blockade of EGFR and Notch signaling to augment the response to radiation and suggests that CT16 may achieve clinical efficacy when combined with radiation in NSCLC treatment.
Collapse
Affiliation(s)
- Shi Hu
- Department of Biophysics, College of Basic Medical Sciences, Second Military Medical University, Shanghai 200433, China.
| | - Wenyan Fu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200030, China.,Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Collaborative Innovation Center of Cancer Medicine, Fudan University Shanghai Cancer Center, Shanghai 200030, China
| | - Tian Li
- Department of Biophysics, College of Basic Medical Sciences, Second Military Medical University, Shanghai 200433, China
| | - Qingning Yuan
- Department of Biophysics, College of Basic Medical Sciences, Second Military Medical University, Shanghai 200433, China
| | - Feifei Wang
- Department of Biophysics, College of Basic Medical Sciences, Second Military Medical University, Shanghai 200433, China
| | - Gaojian Lv
- Department of Biophysics, College of Basic Medical Sciences, Second Military Medical University, Shanghai 200433, China
| | - Yuanyuan Lv
- Department of Biophysics, College of Basic Medical Sciences, Second Military Medical University, Shanghai 200433, China
| | - Xiaoyan Fan
- Department of Biophysics, College of Basic Medical Sciences, Second Military Medical University, Shanghai 200433, China
| | - Yafeng Shen
- Department of Biophysics, College of Basic Medical Sciences, Second Military Medical University, Shanghai 200433, China
| | - Fangxing Lin
- Department of Biophysics, College of Basic Medical Sciences, Second Military Medical University, Shanghai 200433, China
| | - Ying Tang
- Department of Biophysics, College of Basic Medical Sciences, Second Military Medical University, Shanghai 200433, China
| | - Xuting Ye
- Department of Biophysics, College of Basic Medical Sciences, Second Military Medical University, Shanghai 200433, China
| | - Yongji Yang
- Department of Biophysics, College of Basic Medical Sciences, Second Military Medical University, Shanghai 200433, China
| | - Changhai Lei
- Department of Biophysics, College of Basic Medical Sciences, Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
23
|
Schneider NFZ, Silva IT, Persich L, de Carvalho A, Rocha SC, Marostica L, Ramos ACP, Taranto AG, Pádua RM, Kreis W, Barbosa LA, Braga FC, Simões CMO. Cytotoxic effects of the cardenolide convallatoxin and its Na,K-ATPase regulation. Mol Cell Biochem 2017; 428:23-39. [DOI: 10.1007/s11010-016-2914-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 12/21/2016] [Indexed: 01/08/2023]
|
24
|
Wang WD, Liu Y, Su Y, Xiong XZ, Shang D, Xu JJ, Liu HJ. ANTITUMOR AND APOPTOTIC EFFECTS OF CUCURBITACIN A IN A-549 LUNG CARCINOMA CELLS IS MEDIATED VIA G2/M CELL CYCLE ARREST AND M-TOR/PI3K/AKT SIGNALLING PATHWAY. AFRICAN JOURNAL OF TRADITIONAL, COMPLEMENTARY, AND ALTERNATIVE MEDICINES 2017; 14:75-82. [PMID: 28573224 PMCID: PMC5446468 DOI: 10.21010/ajtcam.v14i2.9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Background: The main aim of this study was to demonstrate the antitumor potential of cucurbitacin A on A-549 NSCLC (non-small cell lung cancer cells). The effects of Cucurbitacin A on apoptotic induction, cell physic, cell cycle failure and m-TOR/PI3K/Akt signalling pathway were also investigated in the present study. Materials and Methods: MTT assay and clonogenic assay were carried out to study effects of this compound on cell cytotoxicity and colony forming tendency in A-549 cells. Moreover, phase and fluorescence microscopic techniques were used to examine the effects on cell morphology and induction of apoptosis. The effects on cell cycle phase distribution were investigated by flow cytometry and effects on m-TOR/PI3K/Akt signalling proteins were assessed by western blot analysis. Results: Results showed that cucurbitacin A induced dose-dependent cytotoxic effects along with suppressing the colony forming tendency in these cells. Cucurbitacin A also induced morphological changes in these cells featuring chromatin condensation, cell shrinkage and apoptotic body formation. G2/M phase cell cycle collapse was also induced by Cucurbitacin A along with inhibition of expression levels of m-TOR/PI3K/Akt proteins. Conclusions: In conclusion, cucurbitacin A inhibits cancer growth in A-549 NSCLC cells by inducing apoptosis, targeting m-TOR/PI3K/Akt signalling pathway and G2/M cell cycle.
Collapse
Affiliation(s)
- Wen-Dong Wang
- Department of Thoracic surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yan Liu
- Department of Respiratory Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yuan Su
- Department of Respiratory Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xian-Zhi Xiong
- Department of Respiratory Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Dan Shang
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Juan-Juan Xu
- Department of Respiratory Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hong-Ju Liu
- Department of Respiratory Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
25
|
Kwon T, Youn H, Son B, Kim D, Seong KM, Park S, Kim W, Youn B. DANGER is involved in high glucose-induced radioresistance through inhibiting DAPK-mediated anoikis in non-small cell lung cancer. Oncotarget 2016; 7:7193-206. [PMID: 26769850 PMCID: PMC4872778 DOI: 10.18632/oncotarget.6887] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Accepted: 01/05/2016] [Indexed: 12/11/2022] Open
Abstract
18F-labeled fluorodeoxyglucose (FDG) uptake during FDG positron emission tomography seems to reflect increased radioresistance. However, the exact molecular mechanism underlying high glucose (HG)-induced radioresistance is unclear. In the current study, we showed that ionizing radiation-induced activation of the MEK-ERK-DAPK-p53 signaling axis is required for anoikis (anchorage-dependent apoptosis) of non-small cell lung cancer (NSCLC) cells in normal glucose media. Phosphorylation of DAPK at Ser734 by ERK was essential for p53 transcriptional activity and radiosensitization. In HG media, overexpressed DANGER directly bound to the death domain of DAPK, thus inhibiting the catalytic activity of DAPK. In addition, inhibition of the DAPK-p53 signaling axis by DANGER promoted anoikis-resistance and epithelial-mesenchymal transition (EMT), resulting in radioresistance of HG-treated NSCLC cells. Notably, knockdown of DANGER enhanced anoikis, EMT inhibition, and radiosensitization in a mouse xenograft model of lung cancer. Taken together, our findings offered evidence that overexpression of DANGER and the subsequent inhibitory effect on DAPK kinase activity are critical responses that account for HG-induced radioresistance of NSCLC.
Collapse
Affiliation(s)
- TaeWoo Kwon
- Department of Integrated Biological Science, Pusan National University, Busan, 609-735, Republic of Korea
| | - HyeSook Youn
- Department of Biological Sciences, Pusan National University, Busan, 609-735, Republic of Korea.,Nuclear Science Research Institute, Pusan National University, Busan, 609-735, Republic of Korea
| | - Beomseok Son
- Department of Integrated Biological Science, Pusan National University, Busan, 609-735, Republic of Korea
| | - Daehoon Kim
- Department of Integrated Biological Science, Pusan National University, Busan, 609-735, Republic of Korea
| | - Ki Moon Seong
- National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Sciences, Seoul, 139-706, Republic of Korea
| | - Sungkyun Park
- Department of Physics, Pusan National University, Busan, 609-735, Republic of Korea
| | - Wanyeon Kim
- Department of Biological Sciences, Pusan National University, Busan, 609-735, Republic of Korea.,Nuclear Science Research Institute, Pusan National University, Busan, 609-735, Republic of Korea
| | - BuHyun Youn
- Department of Integrated Biological Science, Pusan National University, Busan, 609-735, Republic of Korea.,Department of Biological Sciences, Pusan National University, Busan, 609-735, Republic of Korea.,Nuclear Science Research Institute, Pusan National University, Busan, 609-735, Republic of Korea
| |
Collapse
|
26
|
Harrow S, Hanna GG, Faivre-Finn C, McDonald F, Chalmers AJ. The Challenges Faced in Developing Novel Drug Radiation Combinations in Non-small Cell Lung Cancer. Clin Oncol (R Coll Radiol) 2016; 28:720-725. [PMID: 27591000 DOI: 10.1016/j.clon.2016.08.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 07/29/2016] [Accepted: 08/05/2016] [Indexed: 02/07/2023]
Abstract
Lung cancer is the most common cancer diagnosed in the UK. Outcomes for patients with this disease remain poor and new strategies to treat this disease require investigation. One potential option is to combine novel agents with radiotherapy in clinical studies. Here we discuss some of the important issues to consider when combining novel agents with radiotherapy, together with potential solutions as discussed at a recent Clinical Translational Radiotherapy Group (CTRad) workshop.
Collapse
Affiliation(s)
- S Harrow
- Department of Clinical Oncology, Beatson West of Scotland Cancer Centre, Glasgow, UK.
| | - G G Hanna
- Centre for Cancer Research and Cell Biology, Queen's University of Belfast, Belfast, UK
| | - C Faivre-Finn
- The University of Manchester, Manchester Academic Health Science Centre, Institute of Cancer Sciences, Manchester Cancer Research Centre, Manchester, UK
| | | | - A J Chalmers
- Department of Clinical Oncology, Beatson West of Scotland Cancer Centre, Glasgow, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
27
|
Zou N, Xie G, Cui T, Srivastava AK, Qu M, Yang L, Wei S, Zheng Y, Wang QE. DDB2 increases radioresistance of NSCLC cells by enhancing DNA damage responses. Tumour Biol 2016; 37:14183-14191. [PMID: 27553023 DOI: 10.1007/s13277-016-5203-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 07/13/2016] [Indexed: 01/24/2023] Open
Abstract
Radiotherapy resistance is one of the major factors limiting the efficacy of radiotherapy in lung cancer patients. The extensive investigations indicate the diversity in the mechanisms underlying radioresistance. Here, we revealed that DNA damage binding protein 2 (DDB2) is a potential regulator in the radiosensitivity of non-small cell lung cancer (NSCLC) cells. DDB2, originally identified as a DNA damage recognition factor in the nucleotide excision repair, promotes the survival and inhibits the apoptosis of NSCLC cell lines upon ionizing radiation (IR). Mechanistic investigations demonstrated that DDB2 is able to facilitate IR-induced phosphorylation of Chk1, which plays a critical role in the cell cycle arrest and DNA repair in response to IR-induced DNA double-strand breaks (DSBs). Indeed, knockdown of DDB2 compromised the G2 arrest in the p53-proficient A549 cell line and reduced the efficiency of homologous recombination (HR) repair. Taken together, our data indicate that the expression of DDB2 in NSCLC could be used as a biomarker to predict radiosensitivity of the patients. Targeting Chk1 can be used to increase the efficacy of radiotherapy in patients of NSCLC possessing high levels of DDB2.
Collapse
Affiliation(s)
- Ning Zou
- Department of Radiation Oncology, Hubei Cancer Hospital, Wuhan, Hubei, 430079, China
- Department of Radiology, Division of Radiobiology, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Guozhen Xie
- Department of Radiology, Division of Radiobiology, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Dublin Coffman High School, Dublin, OH, 43017, USA
| | - Tiantian Cui
- Department of Radiology, Division of Radiobiology, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Amit Kumar Srivastava
- Department of Radiology, Division of Radiobiology, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Meihua Qu
- Department of Pharmacology, Weifang Medical University, Weifang, Shandong, 261053, China
| | - Linlin Yang
- Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Shaozhong Wei
- Department of Gastrointestinal Oncology, Hubei Cancer Hospital, Wuhan, Hubei, 430079, China
| | - Yanfang Zheng
- Oncology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China.
| | - Qi-En Wang
- Department of Radiology, Division of Radiobiology, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA.
| |
Collapse
|
28
|
Lawler M, French D, Henderson R, Aggarwal A, Sullivan R. Shooting for the Moon or Flying Too Near the Sun? Crossing the Value Rubicon in Precision Cancer Care. Public Health Genomics 2016; 19:132-6. [DOI: 10.1159/000446530] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
29
|
Lehman M. Improving Therapeutic Outcomes in Non-small Cell Lung Cancer not Suitable for Curative Intent Therapy — A Review of the Role of Radiation Therapy in an Era of Increasing Systemic Therapy Options. Clin Oncol (R Coll Radiol) 2016; 28:327-33. [DOI: 10.1016/j.clon.2015.11.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Revised: 10/23/2015] [Accepted: 10/26/2015] [Indexed: 01/27/2023]
|
30
|
Bhardwaj B, Revannasiddaiah S, Bhardwaj H, Balusu S, Shwaiki A. Molecular targeted therapy to improve radiotherapeutic outcomes for non-small cell lung carcinoma. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:50. [PMID: 26904572 DOI: 10.3978/j.issn.2305-5839.2015.10.35] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Effective treatments for non-small cell lung carcinoma (NSCLC) remain elusive. The use of concurrent chemotherapy with radiotherapy (RT) has improved outcomes, but a significant proportion of NSCLC patients are too frail to be able to tolerate an intense course of concurrent chemoradiotherapy. The development of targeted therapies ignited new hope in enhancing radiotherapeutic outcomes. The use of targeted therapies against the epidermal growth factor receptor (EGFR) has offered slight but significant benefits in concurrent use with RT for certain patients in certain situations. However, despite theoretical promise, the use of anti-angiogenics, such as bevacizumab and endostatin, has not proven clinically safe or useful in combination with RT. However, many new targeted agents against new targets are being experimented for combined use with RT. It is hoped that these agents may provide a significant breakthrough in the radiotherapeutic management of NSCLC. The current review provides a brief discussion about the targets, the targeted therapies, the rationale for the use of targeted therapies in combination with RT, and a brief review of the existing data on the subject.
Collapse
Affiliation(s)
- Bhaskar Bhardwaj
- 1 Department of Internal medicine, University of Missouri, Kansas City, MO, USA ; 2 Department of Radiation Oncology, Government Medical College, Haldwani, Nanital, India ; 3 Department of Pulmonary Medicine and Critical Care, University of Oklahoma, Norman, OK, USA ; 4 Department of Internal Medicine, University of Missouri, Kansas City, MO, USA ; 5 Department of Hematology and Oncology, Saint Luke's Hospital, Kansas City, MO, USA
| | - Swaroop Revannasiddaiah
- 1 Department of Internal medicine, University of Missouri, Kansas City, MO, USA ; 2 Department of Radiation Oncology, Government Medical College, Haldwani, Nanital, India ; 3 Department of Pulmonary Medicine and Critical Care, University of Oklahoma, Norman, OK, USA ; 4 Department of Internal Medicine, University of Missouri, Kansas City, MO, USA ; 5 Department of Hematology and Oncology, Saint Luke's Hospital, Kansas City, MO, USA
| | - Himanshu Bhardwaj
- 1 Department of Internal medicine, University of Missouri, Kansas City, MO, USA ; 2 Department of Radiation Oncology, Government Medical College, Haldwani, Nanital, India ; 3 Department of Pulmonary Medicine and Critical Care, University of Oklahoma, Norman, OK, USA ; 4 Department of Internal Medicine, University of Missouri, Kansas City, MO, USA ; 5 Department of Hematology and Oncology, Saint Luke's Hospital, Kansas City, MO, USA
| | - Sree Balusu
- 1 Department of Internal medicine, University of Missouri, Kansas City, MO, USA ; 2 Department of Radiation Oncology, Government Medical College, Haldwani, Nanital, India ; 3 Department of Pulmonary Medicine and Critical Care, University of Oklahoma, Norman, OK, USA ; 4 Department of Internal Medicine, University of Missouri, Kansas City, MO, USA ; 5 Department of Hematology and Oncology, Saint Luke's Hospital, Kansas City, MO, USA
| | - Ali Shwaiki
- 1 Department of Internal medicine, University of Missouri, Kansas City, MO, USA ; 2 Department of Radiation Oncology, Government Medical College, Haldwani, Nanital, India ; 3 Department of Pulmonary Medicine and Critical Care, University of Oklahoma, Norman, OK, USA ; 4 Department of Internal Medicine, University of Missouri, Kansas City, MO, USA ; 5 Department of Hematology and Oncology, Saint Luke's Hospital, Kansas City, MO, USA
| |
Collapse
|
31
|
Cytotoxic effects of natural and semisynthetic cucurbitacins on lung cancer cell line A549. Invest New Drugs 2016; 34:139-48. [DOI: 10.1007/s10637-015-0317-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 12/17/2015] [Indexed: 12/13/2022]
|
32
|
Tang Y, Cui Y, Li Z, Jiao Z, Zhang Y, He Y, Chen G, Cheng G, Zhou Q, Wang W, Zhou X, Luo J, Zhang S. Radiation-induced miR-208a increases the proliferation and radioresistance by targeting p21 in human lung cancer cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2016; 35:7. [PMID: 26754670 PMCID: PMC4710038 DOI: 10.1186/s13046-016-0285-3] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 01/06/2016] [Indexed: 12/14/2022]
Abstract
BACKGROUND Lung cancer has long been the most dangerous malignant tumor among males in both well developed and poorly developed countries. Radiotherapy plays a critical role in the curative management of inoperable non-small cell lung cancer (NSCLC) and is also used as a post-surgical treatment in lung cancer patients. Radioresistance is an important factor that limits the efficacy of radiotherapy for NSCLC patients. Increasing evidence suggests that microRNAs (miRNAs) possess diverse cellular regulatory roles in radiation responses. METHODS In this study, we used miRNA microarray technology to identify serum miRNAs that were differentially expressed before and after radiotherapy in lung cancer patients. We further examined the biological function of miR-208a on cell viability, apoptotic death and cell cycle distribution in human lung cancer cells and explored the probable mechanism. RESULTS Nine miRNAs, including miR-29b-3p, miR-200a-3p, and miR-126-3p were significantly down-regulated, whereas miR-208a was the only miRNA that was up-regulated in the serum of the patients after radiation treatment (P < 0.05). The expression of miR-208a could be induced by X-ray irradiation in lung cancer cells. Forced expression of miR-208a promoted cell proliferation and induced radioresistance via targeting p21 with a corresponding activation of the AKT/mTOR pathway in lung cancer cells, whereas down-regulation of miR-208a resulted in the opposite effects. In addition, down-regulation of miR-208a increased the percentage of cells undergoing apoptosis and inhibited the G1 phase arrest in NSCLC cells. Moreover, miR-208a from the serum exosome fraction of lung cancer patients could shuttle to A549 cells in a time-dependent manner, which was likely to contribute to the subsequent biological effects. CONCLUSIONS The present study provides evidence that miR-208a can affect the proliferation and radiosensitivity of human lung cancer cells by targeting p21 and can be transported by exosomes. Thus, miR-208a may serve as a potential therapeutic target for lung cancer patients.
Collapse
Affiliation(s)
- Yiting Tang
- Department of Radiation Oncology, Changzhou Cancer Hospital, Soochow University, Changzhou, 213001, China.
| | - Yayun Cui
- Department of Radiation Oncology, Anhui Provincial Hospital, Hefei, 213001, China.
| | - Zengpeng Li
- State Key Laboratory Breeding Base of Marine Genetic Resources, Third Institute of Oceanography, State Oceanic Administration, Xiamen, 361005, China.
| | - Zhuqing Jiao
- Department School of Information Science and Engineering, Changzhou University, Changzhou, 213164, China.
| | - Yong Zhang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong University, Jinan, 250117, China.
| | - Yan He
- School of Radiation Medicine and Protection and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China.
| | - Guangxia Chen
- Department of Gastroenterology, First People's Hospital of Xuzhou, Xuzhou, 221002, China.
| | - Guangxia Cheng
- Department of Gastroenterology, First People's Hospital of Xuzhou, Xuzhou, 221002, China.
| | - Qunyan Zhou
- Department of Gastroenterology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, 214002, China.
| | - Wenjie Wang
- School of Radiation Medicine and Protection and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China.
| | - Xifa Zhou
- Department of Radiation Oncology, Changzhou Cancer Hospital, Soochow University, Changzhou, 213001, China.
| | - Judong Luo
- Department of Radiation Oncology, Changzhou Cancer Hospital, Soochow University, Changzhou, 213001, China.
| | - Shuyu Zhang
- School of Radiation Medicine and Protection and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
33
|
Marostica LL, Silva IT, Kratz JM, Persich L, Geller FC, Lang KL, Caro MSB, Durán FJ, Schenkel EP, Simões CMO. Synergistic Antiproliferative Effects of a New Cucurbitacin B Derivative and Chemotherapy Drugs on Lung Cancer Cell Line A549. Chem Res Toxicol 2015; 28:1949-60. [PMID: 26372186 DOI: 10.1021/acs.chemrestox.5b00153] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Nonsmall cell lung cancer (NSCLC) represents an important cause of mortality worldwide due to its aggressiveness and growing resistance to currently available therapy. Cucurbitacins have emerged as novel potential anticancer agents showing strong antiproliferative effects and can be promising candidates for combined treatments with clinically used anticancer agents. This study investigates the synergistic antiproliferative effects of a new semisynthetic derivative of cucurbitacin B (DACE) with three chemotherapy drugs: cisplatin (CIS), irinotecan (IRI), and paclitaxel (PAC) on A549 cells. The most effective combinations were selected for studies of the mechanism of action. Using an in silico tool, DACE seems to act by a different mechanism of action when compared with that of different classes of drugs already used in clinical settings. DACE also showed potent synergic effects with drugs, and the most potent combinations induced G2/M cell cycle arrest by modulating survivin and p53 expression, disruption of F-actin cytoskeleton, and cell death by apoptosis. These treatments completely inhibited the clonogenic potential and did not reduce the proliferation of nontumoral lung cells (MRC-5). DACE also showed relevant antimigratory and anti-invasive effects, and combined treatments modulated cell migration signaling pathways evolved with metastasis progression. The effects of DACE associated with drugs was potentiated by the oxidant agent l-buthionine-sulfoximine (BSO), and attenuated by N-acetilcysteine (NAC), an antioxidant agent. The antiproliferative effects induced by combined treatments were attenuated by a pan-caspase inhibitor, indicating that the effects of these treatments are dependent on caspase activity. Our data highlight the therapeutic potential of DACE used in combination with known chemotherapy drugs and offer important insights for the development of more effective and selective therapies against lung cancer.
Collapse
Affiliation(s)
- Lucas Lourenço Marostica
- Departamento de Ciências Farmacêuticas, Universidade Federal de Santa Catarina , Campus Trindade, CEP 88040-900, Florianópolis, Santa Catarina, Brazil
| | - Izabella Thaís Silva
- Departamento de Ciências Farmacêuticas, Universidade Federal de Santa Catarina , Campus Trindade, CEP 88040-900, Florianópolis, Santa Catarina, Brazil
| | - Jadel Müller Kratz
- Departamento de Ciências Farmacêuticas, Universidade Federal de Santa Catarina , Campus Trindade, CEP 88040-900, Florianópolis, Santa Catarina, Brazil
| | - Lara Persich
- Departamento de Ciências Farmacêuticas, Universidade Federal de Santa Catarina , Campus Trindade, CEP 88040-900, Florianópolis, Santa Catarina, Brazil
| | - Fabiana Cristina Geller
- Departamento de Ciências Farmacêuticas, Universidade Federal de Santa Catarina , Campus Trindade, CEP 88040-900, Florianópolis, Santa Catarina, Brazil
| | - Karen Luise Lang
- Departamento de Ciências Farmacêuticas, Universidade Federal de Santa Catarina , Campus Trindade, CEP 88040-900, Florianópolis, Santa Catarina, Brazil
| | - Miguel Soriano Balparda Caro
- Departamento de Química, Universidade Federal de Santa Catarina , Campus Trindade, CEP 88040-900, Florianópolis, Santa Catarina, Brazil
| | - Fernando Javier Durán
- UMYMFOR-CONICET, Departamento de Química Orgánica, Universidad de Buenos Aires , Buenos Aires, Argentina
| | - Eloir Paulo Schenkel
- Departamento de Ciências Farmacêuticas, Universidade Federal de Santa Catarina , Campus Trindade, CEP 88040-900, Florianópolis, Santa Catarina, Brazil
| | - Cláudia Maria Oliveira Simões
- Departamento de Ciências Farmacêuticas, Universidade Federal de Santa Catarina , Campus Trindade, CEP 88040-900, Florianópolis, Santa Catarina, Brazil
| |
Collapse
|
34
|
Zheng YF, Ge W, Xu HL, Cao DED, Liu L, Ming PP, Li CH, Xu XM, Tao WP, Tao ZZ. Endostar enhances the antitumor effects of radiation by affecting energy metabolism and alleviating the tumor microenvironment in a Lewis lung carcinoma mouse model. Oncol Lett 2015; 10:3067-3072. [PMID: 26722291 DOI: 10.3892/ol.2015.3679] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 03/02/2015] [Indexed: 01/14/2023] Open
Abstract
Lung cancer is a leading cause of morbidity and mortality. Previous studies have identified that an improvement in treatment efficacy was achieved using Endostar; however, the role of Endostar in lung cancer remains poorly understood. The present study investigated whether the enhanced antitumor effects of Endostar in combination with radiation involved changes in the metabolism and microenvironment in non-small cell lung cancer. A Lewis lung carcinoma mouse model was used, including the control, Endostar (ES), radiotherapy (RT) and Endostar plus radiotherapy (ES + RT) groups. The tumor inhibition rates and growth were described based on changes in tumor volume. In addition, ultraviolet enzymatic analysis was performed to determine the lactate level and reverse transcription-polymerase chain reaction was used to measure the mRNA expression of lactate dehydrogenase (LDH). A Meph-3 pH meter was used to detect the ranges of tumor interstitial tissue pH, and immunohistochemical analysis was adopted to examine hypoxia within the tumor microenvironment. The tumor inhibition rate of the ES + RT group was significantly higher compared with the other three groups (P<0.05). Following treatment, the lactate levels decreased in all three treatment groups compared with the control, particularly in the ES + RT group (P<0.05). Reduced LDH expression and hypoxic fraction in the tumor microenvironment were also observed in the ES + RT group (P<0.05). Furthermore, changes from acidic to alkaline pH in the tumor microenvironment were detected in the ES + RT group. The present study suggested that Endostar is involved in the regulation of metabolism and tumor microenvironment hypoxia, which may be responsible for the enhanced antitumor effect of Endostar in combination with radiotherapy.
Collapse
Affiliation(s)
- Yong-Fa Zheng
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Wei Ge
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Hui-Lin Xu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - DE-Dong Cao
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Liang Liu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Ping-Po Ming
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Chang-Hu Li
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xi-Ming Xu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Wei-Ping Tao
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Ze-Zhang Tao
- Department of Otolaryngology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
35
|
Yuan ZQ, Li JZ, Liu Y, Chen WL, Yang SD, Zhang CG, Zhu WJ, Zhou XF, Liu C, Zhang XN. Systemic delivery of micelles loading with paclitaxel using N-succinyl-palmitoyl-chitosan decorated with cRGDyK peptide to inhibit non-small-cell lung cancer. Int J Pharm 2015; 492:141-51. [PMID: 26188316 DOI: 10.1016/j.ijpharm.2015.07.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Revised: 05/26/2015] [Accepted: 07/08/2015] [Indexed: 01/10/2023]
Abstract
This study aimed to prepare efficient cRGDyK peptide-decorated micelles for the targeted therapy of non-small-cell lung cancer (NSCLC). An amphiphilic copolymer N-succinyl-palmitoyl-chitosan (SPCS) was synthesized and characterized. cRGDyK peptide is a ligand that can target tumors via specific binding integrin receptor overexpressed on tumor neovascularization and cells. cRGDyK-functionalized SPCS micelles loaded with paclitaxel (PTX/cRGDyK-SPCS) were prepared by film dispersion method and then characterized according to morphology, size, and zeta potential. PTX/cRGDyK-SPCS micelles presented pH-triggered drug release behavior under acidic conditions. The accumulation of micelles detected by laser confocal fluorescence microscopy and flow cytometry showed that cRGDyK-SPCS micelles were easily taken up by A549 cells marked with the luciferase gene (luc-A549). Meanwhile, co-localization of the micelles and lysosomes was recorded dynamically using a live cell station. MTT assays and cell apoptosis studies revealed that cell viability was significantly inhibited by PTX/cRGDyK-SPCS micelles. More importantly, in vivo animal studies showed that cRGDyK-SPCS micelles mainly accumulated in the orthotopic tumor site. PTX/cRGDyK-SPCS micelles exhibited better anti-tumor activity in subcutaneous and orthotopic lung tumors compared with PTX/SPCS micelles and Taxol(®). These results suggested that PTX/cRGDyK-SPCS micelles had better cancer targeting capacity and superior anti-tumor efficacy. Thus, these micelles have great potential as novel carriers in delivering anti-tumor drugs.
Collapse
Affiliation(s)
- Zhi-qiang Yuan
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, People's Republic of China
| | - Ji-zhao Li
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, People's Republic of China
| | - Yang Liu
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, People's Republic of China
| | - Wei-liang Chen
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, People's Republic of China
| | - Shu-di Yang
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, People's Republic of China
| | - Chun-ge Zhang
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, People's Republic of China
| | - Wen-jing Zhu
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, People's Republic of China
| | - Xiao-feng Zhou
- College of Radiological Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; Changshu Hospital of Traditional Chinese Medicine, Changshu 215500, People's Republic of China
| | - Chun Liu
- The hospital of Suzhou People's Hospital affiliated to Nanjing Medical University, Suzhou 215000, People's Republic of China
| | - Xue-nong Zhang
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, People's Republic of China.
| |
Collapse
|
36
|
Hendriks LE, Schoenmaekers J, Zindler JD, Eekers DB, Hoeben A, De Ruysscher DK, Dingemans AMC. Safety of cranial radiotherapy concurrent with tyrosine kinase inhibitors in non-small cell lung cancer patients: A systematic review. Cancer Treat Rev 2015; 41:634-45. [DOI: 10.1016/j.ctrv.2015.05.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 05/05/2015] [Accepted: 05/06/2015] [Indexed: 12/30/2022]
|
37
|
Kim W, Youn H, Kang C, Youn B. Inflammation-induced radioresistance is mediated by ROS-dependent inactivation of protein phosphatase 1 in non-small cell lung cancer cells. Apoptosis 2015; 20:1242-52. [DOI: 10.1007/s10495-015-1141-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
38
|
Guo W, Xie L, Zhao L, Zhao Y. mRNA and microRNA expression profiles of radioresistant NCI-H520 non-small cell lung cancer cells. Mol Med Rep 2015; 12:1857-67. [PMID: 25873351 PMCID: PMC4464398 DOI: 10.3892/mmr.2015.3600] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 03/09/2015] [Indexed: 01/05/2023] Open
Abstract
To elucidate the mechanism of radioresistance in non-small cell lung cancer (NSCLC) cells and to identify key molecules conferring radioresistance, the radioresistant subclone NCI-H520/R, derived from the NCI-H520 NSCLC cell line, was established with eight rounds of sublethal irradiation. The radioresistant features were subsequently assessed using a clonogenic assay, analysis of apoptosis and an MTT assay, the gene expression levels were examined using an Agilent Whole Human Genome 4×44 k Oligo microarray and Agilent Human miRCURY™ LNA array, and confirmed by reverse transcription-quantitative polymerase chain reaction. Pathway analysis and Gene Ontology (GO) analysis were performed to determine the biological functions of the subset of differentially expressed genes. miRNA-mRNA correlation analysis between the expression levels of each miRNA and all its predicted target genes was performed to further understand the radioresistance in the NCI-H520 cells. Following eight rounds of sublethal irradiation, a total of 2,862 mRNAs were significantly differentially expressed in the NCI-H520/R cells, including 893 upregulated genes and 1,969 downregulated genes. A total of 162 upregulated miRNAs and 274 downregulated miRNAs were significantly deregulated in the NCI-H520/R cells. Multiple core regulatory processes and signaling pathways were identified as being of likely relevance to radioresistance in NCI-H520/R cells, including the mitogen-activated protein kinase signaling pathway and neurotrophin signaling pathway. The expression of genes associated with radioresistance reflects the complex biological processes involved in clinical cancer cell eradication and requires further investigation for future enhancement of therapy.
Collapse
Affiliation(s)
- Wei Guo
- Ultrasound Diagnosis Department, Shandong Cancer Hospital and Institute, Jinan, Shandong 250117, P.R. China
| | - Li Xie
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong 250117, P.R. China
| | - Long Zhao
- Ultrasound Diagnosis Department, Shandong Cancer Hospital and Institute, Jinan, Shandong 250117, P.R. China
| | - Yuehuan Zhao
- Ultrasound Diagnosis Department, Shandong Cancer Hospital and Institute, Jinan, Shandong 250117, P.R. China
| |
Collapse
|
39
|
Tang Y, Geng Y, Luo J, Shen W, Zhu W, Meng C, Li M, Zhou X, Zhang S, Cao J. Downregulation of ubiquitin inhibits the proliferation and radioresistance of non-small cell lung cancer cells in vitro and in vivo. Sci Rep 2015; 5:9476. [PMID: 25820571 PMCID: PMC4377628 DOI: 10.1038/srep09476] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 03/05/2015] [Indexed: 12/26/2022] Open
Abstract
Radioresistance has been an important factor in restricting efficacy of radiotherapy for non-small cell lung cancer (NSCLC) patients and new approaches to inhibit cancer growth and sensitize irradiation were warranted. Despite the important role of ubiquitin/proteasome system (UPS) during cancer progression and treatment, the expression and biological role of ubiquitin (Ub) in human NSCLC has not been characterized. In this study, we found that ubiquitin was significantly overexpressed in 75 NSCLC tissues, compared to their respective benign tissues by immunohistochemistry (P < 0.0001). Knock-down of ubiquitin by mixed shRNAs targeting its coding genes ubiquitin B (UBB) and ubiquitin C (UBC) suppressed the growth and increased the radiosensitivity in NSCLC H1299 cells. Apoptosis and γ H2AX foci induced by X-ray irradiation were enhanced by knock-down of ubiquitin. Western blot and immunostaining showed that knock-down of ubiquitin decreased the expression and translocation of NF-κB to the nucleus by reduced phospho-IκBα after irradiation. Suppression of ubiquitin decreased the proliferation and radioresistance of H1299 transplanted xenografts in nude mice by promoting apoptosis. Taken together, our results demonstrate the critical role of ubiquitin in NSCLC proliferation and radiosensitivity. Targeting ubiquitin may serve as a potentially important and novel approach for NSCLC prevention and therapy.
Collapse
Affiliation(s)
- Yiting Tang
- 1] School of Radiation Medicine and Protection and Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China [2] Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions and School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China [3] Department of Radiotherapy, Changzhou Tumor Hospital, Soochow University, Changzhou. 213001, China
| | - Yangyang Geng
- 1] School of Radiation Medicine and Protection and Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China [2] Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions and School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China
| | - Judong Luo
- Department of Radiotherapy, Changzhou Tumor Hospital, Soochow University, Changzhou. 213001, China
| | - Wenhao Shen
- 1] School of Radiation Medicine and Protection and Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China [2] Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions and School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China
| | - Wei Zhu
- 1] School of Radiation Medicine and Protection and Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China [2] Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions and School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China
| | - Cuicui Meng
- Department of Radiotherapy, Changzhou Tumor Hospital, Soochow University, Changzhou. 213001, China
| | - Ming Li
- 1] School of Radiation Medicine and Protection and Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China [2] Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions and School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China
| | - Xifa Zhou
- Department of Radiotherapy, Changzhou Tumor Hospital, Soochow University, Changzhou. 213001, China
| | - Shuyu Zhang
- 1] School of Radiation Medicine and Protection and Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China [2] Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions and School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China
| | - Jianping Cao
- 1] School of Radiation Medicine and Protection and Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China [2] Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions and School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China
| |
Collapse
|
40
|
Cardenal F, Nadal E, Jové M, Faivre-Finn C. Concurrent systemic therapy with radiotherapy for the treatment of poor-risk patients with unresectable stage III non-small-cell lung cancer: a review of the literature. Ann Oncol 2015; 26:278-88. [PMID: 24942274 DOI: 10.1093/annonc/mdu229] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND There is no consensus on the therapeutic approach to poor-risk patients with unresectable stage III non-small-cell lung cancer (NSCLC), despite the increasing number of these patients in current clinical practice. In terms of survival, the combination of concurrent systemic therapy with standard radiotherapy might be advantageous over radiotherapy alone. The purpose of this review is to ascertain the feasibility, safety and efficacy of the combination of concurrent systemic therapy and standard radiotherapy in these patients. METHODS A computer-based literature search was carried out using PubMed and Science Direct for relevant publications; data reported at major conferences in abstract form were also included. RESULTS In unresectable stage III NSCLC, advanced age, poor performance status, weight loss and comorbidities are factors that influence treatment options and disease outcomes in clinical practice. Prospective studies including poor-risk patients have been reviewed. Trials specifically recruiting poor-risk patients have been separated into those using chemotherapy and those using targeted agents with or without chemotherapy. Only two phase III studies specifically including poor-risk patients have been published. Some recent studies suggested that tolerable radio-sensitizing therapy combined with radiotherapy can provide longer survival outcomes than those reported earlier with chemo-radiotherapy or with radiotherapy alone. CONCLUSIONS There is an unmet need to develop well-designed clinical trials with tolerable combinations of systemic therapy and radiotherapy specifically tailored to this lung cancer population. Such trials should incorporate careful comorbidity measurement and, in older adults, a validated geriatric assessment.
Collapse
Affiliation(s)
- F Cardenal
- Department of Medical Oncology, Catalan Institute of Oncology, L'Hospitalet, Barcelona, Spain
| | - E Nadal
- Division of Thoracic Surgery, Department of Surgery, University of Michigan Comprehensive Cancer Center, Ann Arbor, USA
| | - M Jové
- Department of Medical Oncology, Catalan Institute of Oncology, L'Hospitalet, Barcelona, Spain
| | - C Faivre-Finn
- Radiation Related Research, The Christie NHS Foundation Trust and Institute of Cancer Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
41
|
Traditional Phase 1 and 2 Studies in Thoracic Radiation Oncology Should Be Abandoned. Int J Radiat Oncol Biol Phys 2014; 90:487-9. [DOI: 10.1016/j.ijrobp.2014.05.044] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 05/20/2014] [Accepted: 05/27/2014] [Indexed: 12/25/2022]
|
42
|
Lefebvre L, Doré M, Giraud P. Nouvelles techniques et bénéfices attendus pour la radiothérapie du cancer du poumon. Cancer Radiother 2014; 18:473-9. [DOI: 10.1016/j.canrad.2014.06.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 06/23/2014] [Accepted: 06/27/2014] [Indexed: 12/25/2022]
|
43
|
Abstract
The development of molecular targeted therapeutics in oncology builds on many years of scientific investigation into the cellular mechanics of malignant transformation and progression. The past two decades have brought an accelerating pace to the clinical investigation of new molecular targeted agents, particularly in the setting of metastatic disease. The integration of molecular targeted agents into phase III clinical trial design has lagged in the curative treatment setting, particularly in combination with established therapeutic modalities such as radiation. In this review, we discuss the interaction of radiation and molecular targeted therapeutics. The dynamics of cellular and tumor response to radiation offer unique opportunities for beneficial interplay with molecular targeted agents that may go unrecognized with conventional screening and monotherapy clinical testing of novel agents. By using epidermal growth factor receptor (EGFR) as a primary example, we discuss recent clinical studies that illustrate the potential synergy of molecular targeted agents with radiation and highlight the clinical value of such interactions. For various molecular targeted agents, their greatest clinical impact may rest in combination with radiation, and efforts to facilitate systematic investigation of this approach appear highly warranted.
Collapse
Affiliation(s)
- Zachary S Morris
- All authors: University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Paul M Harari
- All authors: University of Wisconsin School of Medicine and Public Health, Madison, WI.
| |
Collapse
|
44
|
Kim E, Youn H, Kwon T, Son B, Kang J, Yang HJ, Seong KM, Kim W, Youn B. PAK1 Tyrosine Phosphorylation Is Required to Induce Epithelial–Mesenchymal Transition and Radioresistance in Lung Cancer Cells. Cancer Res 2014; 74:5520-31. [DOI: 10.1158/0008-5472.can-14-0735] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
45
|
McDonald F, Popat S. Combining targeted agents and hypo- and hyper-fractionated radiotherapy in NSCLC. J Thorac Dis 2014; 6:356-68. [PMID: 24688780 DOI: 10.3978/j.issn.2072-1439.2013.12.05] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Accepted: 12/03/2013] [Indexed: 12/12/2022]
Abstract
Radical radiotherapy remains the cornerstone of treatment for patients with unresectable locally advanced non small cell lung cancer (NSCLC) either as single modality treatment for poor performance status patients or with sequential or concomitant chemotherapy for good performance status patients. Advances in understanding of tumour molecular biology, targeted drug development and experiences of novel agents in the advanced disease setting have brought targeted agents into the NSCLC clinic. In parallel experience using modified accelerated fractionation schedules in locally advanced disease have demonstrated improved outcomes compared to conventional fractionation in the single modality and sequential chemo-radiotherapy settings. Early studies of targeted agents combined with (chemo-) radiotherapy in locally advanced disease in different clinical settings are discussed below and important areas for future studies are high-lighted.
Collapse
|
46
|
Selzer E, Kornek G. Targeted drugs in combination with radiotherapy for the treatment of solid tumors: current state and future developments. Expert Rev Clin Pharmacol 2014; 6:663-76. [PMID: 24164614 DOI: 10.1586/17512433.2013.841540] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The continuously rising use of novel drugs, especially of molecules belonging to the group of targeted drugs is now shaping the therapeutic landscape. However, treatment combinations of targeted drugs with radiotherapy are still rare. Only the monoclonal antibody cetuximab (Erbitux®) has been approved for the treatment of locally advanced squamous cell cancer of the head and neck in combination with radiotherapy. Several targeted compounds are in advanced stages of clinical development for combination treatments with radiotherapy, of which substances with either anti-EGFR or anti-angiogenic mechanisms, such as trastuzumab, panitumumab, erlotinib, cilengitide and bevacizumab are the most promising. Aim of this article is to provide, mainly from a radio-oncological point of view, an overview about the current state as well as to give an outlook on the near future of the most advanced targeted combined treatment concepts for solid tumors.
Collapse
Affiliation(s)
- Edgar Selzer
- Department of Radiation Oncology, Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
47
|
Zhuang H, Zhao X, Zhao L, Chang JY, Wang P. Progress of clinical research on targeted therapy combined with thoracic radiotherapy for non-small-cell lung cancer. Drug Des Devel Ther 2014; 8:667-75. [PMID: 24936128 PMCID: PMC4047835 DOI: 10.2147/dddt.s61977] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The combination of radiotherapy and targeted therapy is an important approach in the application of targeted therapy in clinical practice, and represents an important opportunity for the development of radiotherapy itself. Numerous agents, including epidermal growth factor receptor, monoclonal antibodies, tyrosine kinase inhibitors, and antiangiogenic therapies, have been used for targeted therapy. A number of studies of radiotherapy combined with targeted therapy in non-small-cell lung carcinoma have been completed or are ongoing. This paper briefly summarizes the drugs involved and the important related clinical research, and indicates that considerable progress has been made with the joint efforts of the two disciplines. Many issues, including drug selection, identification of populations most likely to benefit, timing of administration of medication, and side effects of treatment require further investigation. However, further fundamental research and accumulation of clinical data will provide a more comprehensive understanding of these therapies. Targeted therapy in combination with radiotherapy has a bright future.
Collapse
Affiliation(s)
- Hongqing Zhuang
- Department of Radiotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, and Tianjin Lung Cancer Center, Tianjin, People’s Republic of China
| | - Xianzhi Zhao
- Department of Radiotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, and Tianjin Lung Cancer Center, Tianjin, People’s Republic of China
| | - Lujun Zhao
- Department of Radiotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, and Tianjin Lung Cancer Center, Tianjin, People’s Republic of China
| | - Joe Y Chang
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ping Wang
- Department of Radiotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, and Tianjin Lung Cancer Center, Tianjin, People’s Republic of China
| |
Collapse
|
48
|
Zheng DJ, Yu GH, Gao JF, Gu JD. Concomitant EGFR inhibitors combined with radiation for treatment of non-small cell lung carcinoma. Asian Pac J Cancer Prev 2014; 14:4485-94. [PMID: 24083690 DOI: 10.7314/apjcp.2013.14.8.4485] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) is considered to be one of the key driver genes in non-small cell lung cancer (NSCLC). Several clinical trials have shown great promise of EGFR tyrosine kinase inhibitors (TKIs) in the first-line treatment of NSCLC. Many advances have been made in the understanding of EGFR signal transduction network and the interaction between EGFR and tumor microenvironment in mediating cancer survival and development. The concomitant targeted therapy and radiation is a new strategy in the treatment of NSCLC. A number of preclinical studies have demonstrated synergistic anti-tumor activity in the combination of EGFR inhibitors and radiotherapy in vitro and in vivo. In the present review, we discuss the rationale of the combination of EGFR inhibitors and radiotherapy in the treatment of NSCLC.
Collapse
Affiliation(s)
- De-Jie Zheng
- Department of Clinical Oncology, Weifang People's Hospital, Weifang, China E-mail :
| | | | | | | |
Collapse
|
49
|
李 夏, 朱 广. [Clinical developments for the EGFR-TKI combined with radiotherapy in advanced non-small cell lung cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2014; 17:357-62. [PMID: 24758913 PMCID: PMC6000013 DOI: 10.3779/j.issn.1009-3419.2014.04.12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 12/08/2013] [Indexed: 11/30/2022]
Abstract
Lung cancer is one of the most common malignant tumor in the world, severely threatening human life. Recently, targeted therapy such as the epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) made huge progress in treatment of lung cancer. EGFR-TKIs, with its high selectivity and low toxicity, is the first choice for EGFR-mutated patients in stage IV non-small cell lung cancer (NSCLC). However, secondary drug resistance becomes a clinical problem to be urgently resolved. In recent years, a series of preclinical studies showed that EGFR-TKI can enhance the antitumor activity of ionizing radiation. Therefore, EGFR-TKI combined with radiation is extremely promising therapy pattern for advanced NSCLC. This review will discuss the research status in EGFR-TKI and radiotherapy for advanced NSCLC.
Collapse
Affiliation(s)
- 夏南 李
- 100044 北京,北京大学人民医院Peking University Renmin Hospital, Beijing 100044, China
| | - 广迎 朱
- 100142 北京,北京市肿瘤防治研究所,恶性肿瘤发病机制及转化研究教育部重点实验室,北京大学肿瘤医院Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital, Beijing 100142, China
| |
Collapse
|
50
|
Reymen B, van Baardwijk A, Wanders R, Borger J, Dingemans AMC, Bootsma G, Pitz C, Lunde R, Geraedts W, Lambin P, De Ruysscher D. Long-term survival of stage T4N0-1 and single station IIIA-N2 NSCLC patients treated with definitive chemo-radiotherapy using individualised isotoxic accelerated radiotherapy (INDAR). Radiother Oncol 2014; 110:482-7. [PMID: 24444527 DOI: 10.1016/j.radonc.2013.12.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 12/10/2013] [Accepted: 12/18/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) stage T4N0-1 or single nodal station IIIA-N2 are two stage III sub-groups for which the outcome of non-surgical therapy is not well known. We investigated the results of individualised isotoxic accelerated radiotherapy (INDAR) and chemotherapy in this setting. METHODS Analysis of NSCLC patients included in 2 prospective trials (NCT00573040 and NCT00572325) stage T4N0-1 or IIIA-N2 with 1 pathologic nodal station, treated with chemo-radiotherapy (CRT) using INDAR with concurrent or sequential platinum-based chemotherapy. Overall survival (OS) was updated and calculated from date of diagnosis (Kaplan-Meier). Toxicity was scored following CTCAEv3.0. To allow comparison with other articles the subgroups were also analysed separately for toxicity, progression free and overall survival. RESULTS 83 patients (42 T4N0-1 and 41 IIIA-N2) were identified: the median radiotherapy dose was 65Gy. Thirty-seven percent of patients received sequential CRT and 63% received concurrent CRT. At a median follow-up of 48 months the median OS for T4N0-1 patients was 34 months with 55% 2-year survival and 25% 5-year survival. For stage IIIA-N2 at a median follow-up of 50 months the median OS was 26 months with 2- and 5-year survival rates of 53% and 24%, respectively. CONCLUSION Chemo-radiation using INDAR yields promising survival results in patients with single-station stage IIIA-N2 or T4N0-1 NSCLC.
Collapse
Affiliation(s)
- Bart Reymen
- Department of Radiation Oncology (MAASTRO Clinic), The Netherlands.
| | | | - Rinus Wanders
- Department of Radiation Oncology (MAASTRO Clinic), The Netherlands
| | - Jacques Borger
- Department of Radiation Oncology (MAASTRO Clinic), The Netherlands
| | - Anne-Marie C Dingemans
- Department of Pulmonology, University Medical Centre Maastricht, GROW-School for Oncology and Developmental Biology, The Netherlands
| | - Gerben Bootsma
- Department of Pulmonology, Atrium Medical Centre, Heerlen, The Netherlands
| | - Cordula Pitz
- Department of Pulmonology, Laurentius Hospital, Roermond, The Netherlands
| | - Ragnar Lunde
- Department of Pulmonology, St. Jansgasthuis, Weert, The Netherlands
| | - Wiel Geraedts
- Department of Pulmonology, Orbis Medical Centre, Sittard, The Netherlands
| | - Philippe Lambin
- Department of Radiation Oncology (MAASTRO Clinic), The Netherlands
| | - Dirk De Ruysscher
- Department of Radiation Oncology (MAASTRO Clinic), The Netherlands; University Hospital Leuven/KU Leuven, Belgium
| |
Collapse
|