51
|
Sun Y, Dai H, Chen S, Zhang Y, Wu T, Cao X, Zhao G, Xu A, Wang J, Wu L. Disruption of Chromosomal Architecture of cox2 Locus Sensitizes Lung Cancer Cells to Radiotherapy. Mol Ther 2018; 26:2456-2465. [PMID: 30131302 PMCID: PMC6171098 DOI: 10.1016/j.ymthe.2018.08.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 07/27/2018] [Accepted: 08/01/2018] [Indexed: 11/15/2022] Open
Abstract
Despite treatment of lung cancer with radiotherapy and chemotherapy, the survival rate of lung cancer patients remains poor. Previous studies demonstrated the importance of upregulation of inflammatory factors, such as cyclooxygenase 2 (cox2), in tumor tolerance. In the present study, we investigated the role of cox2 in radiosensitivity of lung cancer. Our results showed that the combination treatment of radiation with aspirin, an anti-inflammatory drug, induced a synergistic reduction of cell survival in A549 and H1299 lung cancer cells. In comparison with normal human lung fibroblasts (NHLFs), the cell viability was significantly decreased and the level of apoptosis was remarkably enhanced in A549 cells. Mechanistic studies revealed that the reduction of cox2 by aspirin in A549 and H1299 was caused by disruption of the chromosomal architecture of the cox2 locus. Moreover, the disruption of chromatin looping was mediated by the inhibition of nuclear translocation of p65 and decreased enrichment of p65 at cox2-regulatory elements. Importantly, disorganization of the chromosomal architecture of cox2 triggered A549 cells sensitive to γ-radiation by the induction of apoptosis. In conclusion, we present evidence of an effective therapeutic treatment targeting the epigenetic regulation of lung cancer and a potential strategy to overcome radiation resistance in cancer cells.
Collapse
Affiliation(s)
- Yuxiang Sun
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China; Key Laboratory of Environmental Toxicology and Pollution Control Technology of Anhui Province, Hefei, Anhui 230031, China
| | - Hui Dai
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China; University of Science and Technology of China, Hefei, Anhui 230026, China; Key Laboratory of Environmental Toxicology and Pollution Control Technology of Anhui Province, Hefei, Anhui 230031, China
| | - Shaopeng Chen
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China; Key Laboratory of Environmental Toxicology and Pollution Control Technology of Anhui Province, Hefei, Anhui 230031, China.
| | - Yajun Zhang
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China; University of Science and Technology of China, Hefei, Anhui 230026, China; Key Laboratory of Environmental Toxicology and Pollution Control Technology of Anhui Province, Hefei, Anhui 230031, China
| | - Tao Wu
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China; University of Science and Technology of China, Hefei, Anhui 230026, China; Key Laboratory of Environmental Toxicology and Pollution Control Technology of Anhui Province, Hefei, Anhui 230031, China
| | - Xianbin Cao
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China; University of Science and Technology of China, Hefei, Anhui 230026, China; Key Laboratory of Environmental Toxicology and Pollution Control Technology of Anhui Province, Hefei, Anhui 230031, China
| | - Guoping Zhao
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China; Key Laboratory of Environmental Toxicology and Pollution Control Technology of Anhui Province, Hefei, Anhui 230031, China
| | - An Xu
- Institute of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China; Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China; Key Laboratory of Environmental Toxicology and Pollution Control Technology of Anhui Province, Hefei, Anhui 230031, China
| | - Jun Wang
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China; Key Laboratory of Environmental Toxicology and Pollution Control Technology of Anhui Province, Hefei, Anhui 230031, China
| | - Lijun Wu
- Institute of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China; Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China; Key Laboratory of Environmental Toxicology and Pollution Control Technology of Anhui Province, Hefei, Anhui 230031, China.
| |
Collapse
|
52
|
Understanding the relationship between cell death and tissue shrinkage via a stochastic agent-based model. J Biomech 2018; 73:9-17. [PMID: 29622482 DOI: 10.1016/j.jbiomech.2018.03.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 03/07/2018] [Accepted: 03/08/2018] [Indexed: 10/17/2022]
Abstract
Cell death, a process which can occur both naturally and in response to insult, is both a complex and diverse phenomenon. Under some circumstances, dying cells actively contract and cause their neighbors to rearrange and maintain tissue integrity. Under other circumstances, dying cells leave behind gaps, which results in tissue separation. A better understanding of how the cellular scale features of cell death manifest on the population scale has implications ranging from morphogenesis to tumor response to treatment. However, the mechanistic relationship between cell death and population scale shrinkage is not well understood, and computational methods for studying these relationships are not well established. Here we propose a mechanically robust agent-based cell model designed to capture the implications of cell death on the population scale. In our agent-based model, algorithmic rules applied on the cellular level emerge on the population scale where their effects are quantified. To better quantify model uncertainty and parameter interactions, we implement a recently developed technique for conducting a variance-based sensitivity analysis on the stochastic model. From this analysis and subsequent investigation, we find that cellular scale shrinkage has the largest influence of all model parameters tested, and that by adjusting cellular scale shrinkage population shrinkage varies widely even across simulations which contain the same fraction of dying cells. We anticipate that the methods and results presented here are a starting point for significant future investigation toward quantifying the implications of different mechanisms of cell death on population and tissue scale behavior.
Collapse
|
53
|
Kolenda T, Przybyła W, Kapałczyńska M, Teresiak A, Zajączkowska M, Bliźniak R, Lamperska KM. Tumor microenvironment - Unknown niche with powerful therapeutic potential. Rep Pract Oncol Radiother 2018; 23:143-153. [PMID: 29760589 PMCID: PMC5948324 DOI: 10.1016/j.rpor.2018.01.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 11/20/2017] [Accepted: 01/20/2018] [Indexed: 12/25/2022] Open
Abstract
Head and neck squamous cell carcinomas (HNSCC) are in a group of cancers that are the most resistant to treatment. The survival rate of HNSCC patients has been still very low since last 20 years. The existence of relationship between oncogenic and surrounding cells is probably the reason for a poor response to treatment. Fibroblasts are an important element of tumor stroma which increases tumor cells ability to proliferate. Another highly resistance, tumorigenic and metastatic cell population in tumor microenvironment are cancer initiating cells (CICs). The population of cancer initiating cells can be found regardless of differentiation status of cancer and they seem to be crucial for HNSCC development. In this review, we describe the current state of knowledge about HNSCC biological and physiological tumor microenvironment.
Collapse
Affiliation(s)
- Tomasz Kolenda
- Laboratory of Cancer Genetic, Greater Poland Cancer Centre, Poznan, Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Poland
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poznan, Poland
| | - Weronika Przybyła
- Laboratory of Cancer Genetic, Greater Poland Cancer Centre, Poznan, Poland
- Department of Pediatric Research, Division of Pediatric and Adolescent Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Marta Kapałczyńska
- Laboratory of Cancer Genetic, Greater Poland Cancer Centre, Poznan, Poland
- Department of Gastroenterology and Hepatology, Charite University Medicine Berlin, Berlin, Germany
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Anna Teresiak
- Laboratory of Cancer Genetic, Greater Poland Cancer Centre, Poznan, Poland
| | - Maria Zajączkowska
- Laboratory of Cancer Genetic, Greater Poland Cancer Centre, Poznan, Poland
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poznan, Poland
| | - Renata Bliźniak
- Laboratory of Cancer Genetic, Greater Poland Cancer Centre, Poznan, Poland
| | | |
Collapse
|
54
|
Basler L, Kowalczyk A, Heidenreich R, Fotin-Mleczek M, Tsitsekidis S, Zips D, Eckert F, Huber SM. Abscopal effects of radiotherapy and combined mRNA-based immunotherapy in a syngeneic, OVA-expressing thymoma mouse model. Cancer Immunol Immunother 2018; 67:653-662. [PMID: 29335856 PMCID: PMC11028190 DOI: 10.1007/s00262-018-2117-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 01/11/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND Tumor metastasis and immune evasion present major challenges of cancer treatment. Radiotherapy can overcome immunosuppressive tumor microenvironments. Anecdotal reports suggest abscopal anti-tumor immune responses. This study assesses abscopal effects of radiotherapy in combination with mRNA-based cancer vaccination (RNActive®). METHODS C57BL/6 mice were injected with ovalbumin-expressing thymoma cells into the right hind leg (primary tumor) and left flank (secondary tumor) with a delay of 4 days. Primary tumors were irradiated with 3 × 2 Gy, while secondary tumors were shielded. RNA and combined treatment groups received mRNA-based RNActive® vaccination. RESULTS Radiotherapy and combined radioimmunotherapy significantly delayed primary tumor growth with a tumor control in 15 and 53% of mice, respectively. In small secondary tumors, radioimmunotherapy significantly slowed growth rate compared to vaccination (p = 0.002) and control groups (p = 0.01). Cytokine microarray analysis of secondary tumors showed changes in the cytokine microenvironment, even in the non-irradiated contralateral tumors after combination treatment. CONCLUSION Combined irradiation and immunotherapy is able to induce abscopal responses, even with low, normofractionated radiation doses. Thus, the combination of mRNA-based vaccination with irradiation might be an effective regimen to induce systemic anti-tumor immunity.
Collapse
Affiliation(s)
- Lucas Basler
- Department of Radiation Oncology, University of Tübingen, Rämistrasse 100, 8091, Tübingen, Germany.
- Department of Radiation Oncology, University Hospital Zürich, Zurich, Switzerland.
| | - Aleksandra Kowalczyk
- CureVac AG, Tübingen, Germany
- Boehringer-Ingelheim, Birkendorferstr. 85, 88397, Biberach an der Riss, Germany
| | | | | | - Savas Tsitsekidis
- Department of Radiation Oncology, University of Tübingen, Rämistrasse 100, 8091, Tübingen, Germany
| | - Daniel Zips
- Department of Radiation Oncology, University of Tübingen, Rämistrasse 100, 8091, Tübingen, Germany
| | - Franziska Eckert
- Department of Radiation Oncology, University of Tübingen, Rämistrasse 100, 8091, Tübingen, Germany
| | - Stephan M Huber
- Department of Radiation Oncology, University of Tübingen, Rämistrasse 100, 8091, Tübingen, Germany
| |
Collapse
|
55
|
The in vitro radiosensitizer potential of resveratrol on MCF-7 breast cancer cells. Chem Biol Interact 2018; 282:85-92. [DOI: 10.1016/j.cbi.2018.01.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 12/11/2017] [Accepted: 01/11/2018] [Indexed: 12/31/2022]
|
56
|
Abstract
Failure to eliminate cancer cells that have been exposed to cytotoxic agents may contribute to the development of resistance to antitumor drugs. A widespread model in present day oncology is that antitumor therapy involves the triggering of tumor cells to undergo apoptosis, and cells that can avoid apoptosis will be resistant to such therapy. Apoptosis is a defined program of cell death that is markedly influenced by the fact that many routes leading to it are mutated or deregulated in human cancer. Mutations in the tumor suppressor protein p53, a common feature of many cancers, may decrease the sensitivity of cells to some antitumor agents. Moreover, it has been increasingly reported that antitumor therapy not only causes apoptosis, but other forms of cell death as well, such as mitotic catastrophe, necrosis and autophagy, or a permanent cell arrest with phenotype characteristics of senescence. Mitotic catastrophe is a form of cell death that results from abnormal mitosis, which does not seem to depend on wild-type p53. Sometimes mitotic catastrophe is used restrictively for faulty mitosis leading to cell death, which may occur via apoptosis or necrosis. We critically review herein how antitumor therapy may elicit the response of human cancers through different cell pathways leading to cell death.
Collapse
|
57
|
Preusser M, Winkler F, Valiente M, Manegold C, Moyal E, Widhalm G, Tonn JC, Zielinski C. Recent advances in the biology and treatment of brain metastases of non-small cell lung cancer: summary of a multidisciplinary roundtable discussion. ESMO Open 2018; 3:e000262. [PMID: 29387475 PMCID: PMC5786916 DOI: 10.1136/esmoopen-2017-000262] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 11/21/2017] [Accepted: 11/29/2017] [Indexed: 12/21/2022] Open
Abstract
This article is the result of a round table discussion held at the European Lung Cancer Conference (ELCC) in Geneva in May 2017. Its purpose is to explore and discuss the advances in the knowledge about the biology and treatment of brain metastases originating from non-small cell lung cancer. The authors propose a series of recommendations for research and treatment within the discussed context.
Collapse
Affiliation(s)
- Matthias Preusser
- Clinical Division of Oncology, Department of Medicine I, Comprehensive Cancer Centre, Medical University Vienna - General Hospital, Vienna, Austria
| | - Frank Winkler
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Neuro-oncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Manuel Valiente
- Brain Metastasis Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Christian Manegold
- Medical Faculty Mannheim, University of Heidelberg, Mannheim, Baden-Württemberg, Germany
| | - Elizabeth Moyal
- Radiation Oncology Department, Radiobiology team 11, UMR1037 INSERM, Institut Universitaire du Cancer de Toulouse Oncopole, Centre de Recherche contre le Cancer, Toulouse, France
| | - Georg Widhalm
- Department of Neurosurgery, Medical University of Vienna (MUV), Vienna, Austria.,Department of Neurosurgery, University of California San Francisco (UCSF), San Francisco, USA.,Comprehensive Cancer Center-Central Nervous System Tumours Unit (CCC-CNS), Medical University Vienna (MUV), Vienna, Austria
| | - Jörg-Christian Tonn
- Department of Neurosurgery, Ludwig-Maximilians University, Munich-Grosshadern, Germany and German Cancer Consortium (DKTK) at the German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Christoph Zielinski
- Clinical Division of Oncology, Department of Medicine I, Comprehensive Cancer Centre, Medical University Vienna - General Hospital, Vienna, Austria
| |
Collapse
|
58
|
Mitotic progression following DNA damage enables pattern recognition within micronuclei. Nature 2017; 548:466-470. [PMID: 28759889 PMCID: PMC5857357 DOI: 10.1038/nature23470] [Citation(s) in RCA: 1077] [Impact Index Per Article: 134.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 07/11/2017] [Indexed: 12/21/2022]
Abstract
Inflammatory gene expression following genotoxic cancer therapy is well documented, yet the events underlying its induction remain poorly understood. Inflammatory cytokines modify the tumour microenvironment by recruiting immune cells and are critical for both local and systemic (abscopal) tumour responses to radiotherapy. A poorly understood feature of these responses is the delayed onset (days), in contrast to the acute DNA-damage responses that occur in minutes to hours. Such dichotomous kinetics implicate additional rate-limiting steps that are essential for DNA-damage-induced inflammation. Here we show that cell cycle progression through mitosis following double-stranded DNA breaks leads to the formation of micronuclei, which precede activation of inflammatory signalling and are a repository for the pattern-recognition receptor cyclic GMP-AMP synthase (cGAS). Inhibiting progression through mitosis or loss of pattern recognition by stimulator of interferon genes (STING)-cGAS impaired interferon signalling. Moreover, STING loss prevented the regression of abscopal tumours in the context of ionizing radiation and immune checkpoint blockade in vivo. These findings implicate temporal modulation of the cell cycle as an important consideration in the context of therapeutic strategies that combine genotoxic agents with immune checkpoint blockade.
Collapse
|
59
|
Amawi H, Karthikeyan C, Pathak R, Hussein N, Christman R, Robey R, Ashby CR, Trivedi P, Malhotra A, Tiwari AK. Thienopyrimidine derivatives exert their anticancer efficacy via apoptosis induction, oxidative stress and mitotic catastrophe. Eur J Med Chem 2017; 138:1053-1065. [PMID: 28759878 DOI: 10.1016/j.ejmech.2017.07.028] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 07/14/2017] [Accepted: 07/16/2017] [Indexed: 11/17/2022]
Abstract
In this study, a series of 13 structural variants of thieno[2,3d]pyrimidine derivatives (6a-6m) were synthesized and screened for cytotoxicity in a panel of colorectal, ovarian, and brain cancer cell lines. The selectivity of the compounds was assessed by determining the cytotoxicity in normal epithelial cell line (CHO). The most potent compound, 6j, was efficacious (with IC50 range of 0.6-1.2 μM) in colon (HCT116 and HCT15), brain (LN-229 and GBM-10) and ovarian (A2780 and OV2008) cancer cell lines. In contrast, in the normal cell line (CHO), the IC50 values for 6j were 14 ± 1.3 μM. Compound 6j significantly inhibited the clonogenic potential of HCT116, OV2008 and A2780 cell lines in concentration - dependent (0.5-4 μM) manner. Also, 6j induced 1) formation of reactive oxygen species; 2) apoptosis and 3) mitotic catastrophe in HCT116 and OV2008 cells (IC50 = 0.5-2 μM). Furthermore, apoptosis was the predominant mechanism of death in A2780 cells. The cytotoxicity of 6j in wild type HCT116 cells was similar to that in HCT116 cells lacking the apoptotic genes for Bax, Bak, or Bak and Bax, indicating that 6j induces mitotic catastrophe as alternative mechanism of death when when certain apoptotic proteins are absent. In summary, this study has identified a lead molecule, 6j, that selectively induces oxidative stress, apoptosis and mitotic catastrophe in specific cancer (colon and ovarian) cell lines.
Collapse
Affiliation(s)
- Haneen Amawi
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, OH, USA
| | - Chandrabose Karthikeyan
- School of Pharmaceutical Sciences, Rajiv Gandhi Proudyogiki Vishwavidyalaya, Airport Bypass Road, Gandhi Nagar, Bhopal MP, India.
| | - Rekha Pathak
- School of Pharmaceutical Sciences, Rajiv Gandhi Proudyogiki Vishwavidyalaya, Airport Bypass Road, Gandhi Nagar, Bhopal MP, India
| | - Noor Hussein
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, OH, USA
| | - Ryann Christman
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, OH, USA
| | - Robert Robey
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Charles R Ashby
- Pharmaceutical Sciences, College of Pharmacy, St. John's University Queens, NY, USA
| | - Piyush Trivedi
- School of Pharmaceutical Sciences, Rajiv Gandhi Proudyogiki Vishwavidyalaya, Airport Bypass Road, Gandhi Nagar, Bhopal MP, India
| | - Ashim Malhotra
- School of Pharmacy, Pacific University, 222 SE 8th Ave, Hillsboro, OR, USA
| | - Amit K Tiwari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, OH, USA.
| |
Collapse
|
60
|
Zhang G, Wang W, Yao C, Ren J, Zhang S, Han M. Salinomycin overcomes radioresistance in nasopharyngeal carcinoma cells by inhibiting Nrf2 level and promoting ROS generation. Biomed Pharmacother 2017; 91:147-154. [DOI: 10.1016/j.biopha.2017.04.095] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 04/20/2017] [Accepted: 04/20/2017] [Indexed: 01/10/2023] Open
|
61
|
Diegeler S, Hellweg CE. Intercellular Communication of Tumor Cells and Immune Cells after Exposure to Different Ionizing Radiation Qualities. Front Immunol 2017. [PMID: 28638385 PMCID: PMC5461334 DOI: 10.3389/fimmu.2017.00664] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Ionizing radiation can affect the immune system in many ways. Depending on the situation, the whole body or parts of the body can be acutely or chronically exposed to different radiation qualities. In tumor radiotherapy, a fractionated exposure of the tumor (and surrounding tissues) is applied to kill the tumor cells. Currently, mostly photons, and also electrons, neutrons, protons, and heavier particles such as carbon ions, are used in radiotherapy. Tumor elimination can be supported by an effective immune response. In recent years, much progress has been achieved in the understanding of basic interactions between the irradiated tumor and the immune system. Here, direct and indirect effects of radiation on immune cells have to be considered. Lymphocytes for example are known to be highly radiosensitive. One important factor in indirect interactions is the radiation-induced bystander effect which can be initiated in unexposed cells by expression of cytokines of the irradiated cells and by direct exchange of molecules via gap junctions. In this review, we summarize the current knowledge about the indirect effects observed after exposure to different radiation qualities. The different immune cell populations important for the tumor immune response are natural killer cells, dendritic cells, and CD8+ cytotoxic T-cells. In vitro and in vivo studies have revealed the modulation of their functions due to ionizing radiation exposure of tumor cells. After radiation exposure, cytokines are produced by exposed tumor and immune cells and a modulated expression profile has also been observed in bystander immune cells. Release of damage-associated molecular patterns by irradiated tumor cells is another factor in immune activation. In conclusion, both immune-activating and -suppressing effects can occur. Enhancing or inhibiting these effects, respectively, could contribute to modified tumor cell killing after radiotherapy.
Collapse
Affiliation(s)
- Sebastian Diegeler
- Division of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), Köln, Germany
| | - Christine E Hellweg
- Division of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), Köln, Germany
| |
Collapse
|
62
|
Baues C, Trommer-Nestler M, Jablonska K, Bröckelmann PJ, Schlaak M, von Bergwelt-Baildon M, Engert A, Semrau R, Marnitz S, Theurich S. Short review of potential synergies of immune checkpoint inhibition and radiotherapy with a focus on Hodgkin lymphoma: radio-immunotherapy opens new doors. Immunotherapy 2017; 9:423-433. [PMID: 28357914 DOI: 10.2217/imt-2017-0002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Radiotherapy is an established local treatment in patients with various malignancies. Systemic responses following local irradiation have been described as abscopal effects. Modern cancer immunotherapy with immune checkpoint inhibitors has shown impressive response rates and prolongation of survival even in heavily pretreated patients with advanced solid malignancies and lymphomas. Radiotherapy has been shown to modulate immune response, and its application in the context of immune checkpoint inhibition has recently evolved into an active field of research. Prospective studies investigating combination treatment are currently ongoing and will answer questions as to the optimal schedule and radiation dosing. This short review focuses on the immunomodulatory role of radiotherapy and the use of immune checkpoint inhibition with a special focus on Hodgkin lymphoma.
Collapse
Affiliation(s)
- Christian Baues
- Department of Radio-Oncology & CyberKnife Center University Hospital Cologne, Cologne, Germany.,Radio Immune-Oncology Consortium (RIO), University Hospital Cologne, Cologne, Germany.,German Hodgkin Study Group, University Hospital Cologne, Cologne, Germany.,Center for Integrated Oncology (CIO) Cologne Bonn, University Hospital Cologne, Cologne, Germany
| | - Maike Trommer-Nestler
- Department of Radio-Oncology & CyberKnife Center University Hospital Cologne, Cologne, Germany.,Radio Immune-Oncology Consortium (RIO), University Hospital Cologne, Cologne, Germany
| | - Karolina Jablonska
- Department of Radio-Oncology & CyberKnife Center University Hospital Cologne, Cologne, Germany
| | - Paul J Bröckelmann
- German Hodgkin Study Group, University Hospital Cologne, Cologne, Germany.,Center for Integrated Oncology (CIO) Cologne Bonn, University Hospital Cologne, Cologne, Germany.,Department I of Internal Medicine, Hematology & Oncology, University Hospital Cologne, Cologne, Germany
| | - Max Schlaak
- Radio Immune-Oncology Consortium (RIO), University Hospital Cologne, Cologne, Germany.,Center for Integrated Oncology (CIO) Cologne Bonn, University Hospital Cologne, Cologne, Germany.,Department of Dermatology & Venerology, University Hospital Cologne, Cologne, Germany
| | - Michael von Bergwelt-Baildon
- Radio Immune-Oncology Consortium (RIO), University Hospital Cologne, Cologne, Germany.,Center for Integrated Oncology (CIO) Cologne Bonn, University Hospital Cologne, Cologne, Germany.,Department I of Internal Medicine, Hematology & Oncology, University Hospital Cologne, Cologne, Germany
| | - Andreas Engert
- German Hodgkin Study Group, University Hospital Cologne, Cologne, Germany.,Center for Integrated Oncology (CIO) Cologne Bonn, University Hospital Cologne, Cologne, Germany.,Department I of Internal Medicine, Hematology & Oncology, University Hospital Cologne, Cologne, Germany
| | - Robert Semrau
- Department of Radio-Oncology & CyberKnife Center University Hospital Cologne, Cologne, Germany.,German Hodgkin Study Group, University Hospital Cologne, Cologne, Germany.,Center for Integrated Oncology (CIO) Cologne Bonn, University Hospital Cologne, Cologne, Germany
| | - Simone Marnitz
- Department of Radio-Oncology & CyberKnife Center University Hospital Cologne, Cologne, Germany.,German Hodgkin Study Group, University Hospital Cologne, Cologne, Germany.,Center for Integrated Oncology (CIO) Cologne Bonn, University Hospital Cologne, Cologne, Germany
| | - Sebastian Theurich
- Radio Immune-Oncology Consortium (RIO), University Hospital Cologne, Cologne, Germany.,Center for Integrated Oncology (CIO) Cologne Bonn, University Hospital Cologne, Cologne, Germany.,Department I of Internal Medicine, Hematology & Oncology, University Hospital Cologne, Cologne, Germany
| |
Collapse
|
63
|
Baghery S, Zolfigol MA. 3,6-Dioxaoctamethylenediamminium trifluoromethanesulfonate [3,6-DOMDA]OTf as a novel ionic liquid catalyst for the synthesis of functionalized 1,4-dihydropyridines. J Mol Liq 2017. [DOI: 10.1016/j.molliq.2017.02.073] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
64
|
Gao SJ, Corso CD, Wang EH, Blasberg JD, Detterbeck FC, Boffa DJ, Decker RH, Kim AW. Timing of Surgery after Neoadjuvant Chemoradiation in Locally Advanced Non-Small Cell Lung Cancer. J Thorac Oncol 2016; 12:314-322. [PMID: 27720827 DOI: 10.1016/j.jtho.2016.09.122] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 08/30/2016] [Accepted: 09/08/2016] [Indexed: 01/09/2023]
Abstract
INTRODUCTION A subset of patients with potentially resectable clinical stage IIIA NSCLC are managed with trimodality therapy. However, little data exist to guide the timing of surgery after neoadjuvant therapy. This study examined whether the time interval between neoadjuvant chemoradiation (NCRT) and surgical resection affects overall survival. METHODS Patients with clinical stage IIIA disease (T1-3 N2) NSCLC who underwent NCRT were identified in the National Cancer Data Base (NCDB) between 2004 and 2012 and categorized on the basis of the interval between chemoradiation and surgery (0 to ≤3, >3 to ≤6, >6 to ≤9, and >9 to ≤12 weeks). Other clinical stages were excluded. The Kaplan-Meier method and log-rank tests were used to compare overall survival rates, and a bootstrapped Cox proportional hazards model was used to determine significant contributors to overall survival. RESULTS Of the 1623 patients identified, 7.9% underwent an operation 0 to 3 weeks or less after NCRT, 50.5% underwent an operation greater than 3 and less than or equal to 6 weeks after NCRT, 31.9% underwent an operation greater than 6 and less than or equal to 9 weeks after NCRT, and 9.6% underwent an operation greater than 9 and less than or equal to 12 weeks after NCRT. Multivariate survival analysis demonstrated no significant difference in survival in those who underwent an operation within 6 weeks of NCRT. However, significant drops in overall survival were observed in those who had an operation greater than 6 and less than or equal to 9 weeks after NCRT (hazard ratio = 1.33, 95% confidence interval: 1.01-1.76, p = 0.043) and greater than 9 and less than or equal to 12 weeks after NCRT (hazard ratio = 1.44, 95% confidence interval: 1.04-2.01, p = 0.030). CONCLUSIONS The findings from this retrospective study suggest that overall survival may be significantly lower in patients with clinical stage IIIA N2 NSCLC who undergo an operation later than 6 weeks after NCRT. These results discourage unnecessary delays in surgery.
Collapse
Affiliation(s)
- Sarah J Gao
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Christopher D Corso
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Elyn H Wang
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Justin D Blasberg
- Department of Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - Frank C Detterbeck
- Department of Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - Daniel J Boffa
- Department of Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - Roy H Decker
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Anthony W Kim
- Department of Surgery, Yale University School of Medicine, New Haven, Connecticut.
| |
Collapse
|
65
|
Mechanistic Modelling of DNA Repair and Cellular Survival Following Radiation-Induced DNA Damage. Sci Rep 2016; 6:33290. [PMID: 27624453 PMCID: PMC5022028 DOI: 10.1038/srep33290] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 08/09/2016] [Indexed: 12/12/2022] Open
Abstract
Characterising and predicting the effects of ionising radiation on cells remains challenging, with the lack of robust models of the underlying mechanism of radiation responses providing a significant limitation to the development of personalised radiotherapy. In this paper we present a mechanistic model of cellular response to radiation that incorporates the kinetics of different DNA repair processes, the spatial distribution of double strand breaks and the resulting probability and severity of misrepair. This model enables predictions to be made of a range of key biological endpoints (DNA repair kinetics, chromosome aberration and mutation formation, survival) across a range of cell types based on a set of 11 mechanistic fitting parameters that are common across all cells. Applying this model to cellular survival showed its capacity to stratify the radiosensitivity of cells based on aspects of their phenotype and experimental conditions such as cell cycle phase and plating delay (correlation between modelled and observed Mean Inactivation Doses R(2) > 0.9). By explicitly incorporating underlying mechanistic factors, this model can integrate knowledge from a wide range of biological studies to provide robust predictions and may act as a foundation for future calculations of individualised radiosensitivity.
Collapse
|
66
|
Liao Y, Chu HP, Hu Z, Merkin JJ, Chen J, Liu Z, Degenhardt K, White E, Ryazanov AG. Paradoxical Roles of Elongation Factor-2 Kinase in Stem Cell Survival. J Biol Chem 2016; 291:19545-57. [PMID: 27466362 DOI: 10.1074/jbc.m116.724856] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Indexed: 11/06/2022] Open
Abstract
Protein synthesis inhibition is an immediate response during stress to switch the composition of protein pool in order to adapt to the new environment. It was reported that this response could be either protective or deleterious. However, how cells choose to live or die upon protein synthesis inhibition is largely unknown. Previously, we have shown that elongation factor-2 kinase (eEF2K), a protein kinase that suppresses protein synthesis during elongation phase, is a positive regulator of apoptosis both in vivo and in vitro Consistently, here we report that knock-out of eEF2K protects mice from a lethal dose of whole-body ionizing radiation at 8 Gy by reducing apoptosis levels in both bone marrow and gastrointestinal tracts. Surprisingly, similar to the loss of p53, eEF2K deficiency results in more severe damage to the gastrointestinal tract at 20 Gy with the increased mitotic cell death in small intestinal stem cells. Furthermore, using epithelial cell lines, we showed that eEF2K is required for G2/M arrest induced by radiation to prevent mitotic catastrophe in a p53-independent manner. Specifically, we observed the elevation of Akt/ERK activity as well as the reduction of p21 expression in Eef2k(-/-) cells. Therefore, eEF2K also provides a protective strategy to maintain genomic integrity by arresting cell cycle in response to stress. Our results suggest that protective versus pro-apoptotic roles of eEF2K depend on the type of cells: eEF2K is protective in highly proliferative cells, such as small intestinal stem cells and cancer cells, which are more susceptible to mitotic catastrophe.
Collapse
Affiliation(s)
- Yi Liao
- From the Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen, Fujian 361102, China, the Department of Pharmacology, Robert Wood Johnson Medical School, and
| | - Hsueh-Ping Chu
- the Department of Pharmacology, Robert Wood Johnson Medical School, and the Department of Molecular Biology, Massachusetts General Hospital, Department of Genetics, Harvard Medical School, Boston, Massachusetts 02114
| | - Zhixian Hu
- the Department of Pharmacology, Robert Wood Johnson Medical School, and
| | - Jason J Merkin
- the Department of Pharmacology, Robert Wood Johnson Medical School, and
| | - Jianmin Chen
- the Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey 08854
| | - Zuguo Liu
- From the Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen, Fujian 361102, China, the Affiliated Xiamen Eye Center of Xiamen University, Xiamen, Fujian 361102, China
| | - Kurt Degenhardt
- the Cancer Institute of New Jersey, New Brunswick, New Jersey 08903, and
| | - Eileen White
- the Cancer Institute of New Jersey, New Brunswick, New Jersey 08903, and
| | - Alexey G Ryazanov
- the Department of Pharmacology, Robert Wood Johnson Medical School, and
| |
Collapse
|
67
|
Deloch L, Derer A, Hartmann J, Frey B, Fietkau R, Gaipl US. Modern Radiotherapy Concepts and the Impact of Radiation on Immune Activation. Front Oncol 2016; 6:141. [PMID: 27379203 PMCID: PMC4913083 DOI: 10.3389/fonc.2016.00141] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 05/23/2016] [Indexed: 12/12/2022] Open
Abstract
Even though there is extensive research carried out in radiation oncology, most of the clinical studies focus on the effects of radiation on the local tumor tissue and deal with normal tissue side effects. The influence of dose fractionation and timing particularly with regard to immune activation is not satisfactorily investigated so far. This review, therefore, summarizes current knowledge on concepts of modern radiotherapy (RT) and evaluates the potential of RT for immune activation. Focus is set on radiation-induced forms of tumor cell death and consecutively the immunogenicity of the tumor cells. The so-called non-targeted, abscopal effects can contribute to anti-tumor responses in a specific and systemic manner and possess the ability to target relapsing tumor cells as well as metastases. The impact of distinct RT concepts on immune activation is outlined and pre-clinical evidence and clinical observations on RT-induced immunity will be discussed. Knowledge on the radiosensitivity of immune cells as well as clinical evidence for enhanced immunity after RT will be considered. While stereotactic ablative body radiotherapy seem to have a beneficial outcome over classical RT fractionation in pre-clinical animal models, in vitro model systems suggest an advantage for classical fractionated RT for immune activation. Furthermore, the optimal approach may differ based on the tumor site and/or genetic signature. These facts highlight that clinical trials are urgently needed to identify whether high-dose RT is superior to induce anti-tumor immune responses compared to classical fractionated RT and in particular how the outcome is when RT is combined with immunotherapy in selected tumor entities.
Collapse
Affiliation(s)
- Lisa Deloch
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen , Germany
| | - Anja Derer
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen , Germany
| | - Josefin Hartmann
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen , Germany
| | - Benjamin Frey
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen , Germany
| | - Rainer Fietkau
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen , Germany
| | - Udo S Gaipl
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen , Germany
| |
Collapse
|
68
|
A Systematic Comparison Identifies an ATP-Based Viability Assay as Most Suitable Read-Out for Drug Screening in Glioma Stem-Like Cells. Stem Cells Int 2016; 2016:5623235. [PMID: 27274737 PMCID: PMC4871979 DOI: 10.1155/2016/5623235] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 02/15/2016] [Accepted: 02/17/2016] [Indexed: 11/18/2022] Open
Abstract
Serum-free culture methods for patient-derived primary glioma cultures, selecting for glioma stem-like cells (GSCs), are becoming the gold standard in neurooncology research. These GSCs can be implemented in drug screens to detect patient-specific responses, potentially bridging the translational gap to personalized medicine. Since numerous compounds are available, a rapid and reliable readout for drug efficacies is required. This can be done using approaches that measure viability, confluency, cytotoxicity, or apoptosis. To determine which assay is best suitable for drug screening, 10 different assays were systematically tested on established glioma cell lines and validated on a panel of GSCs. General applicability was assessed using distinct treatment modalities, being temozolomide, radiation, rapamycin, and the oncolytic adenovirus Delta24-RGD. The apoptosis and cytotoxicity assays did not unequivocally detect responses and were excluded from further testing. The NADH- and ATP-based viability assays revealed comparable readout for all treatments; however, the latter had smaller standard deviations and direct readout. Importantly, drugs that interfere with cell metabolism require alternative techniques such as confluency monitoring to accurately measure treatment effects. Taken together, our data suggest that the combination of ATP luminescence assays with confluency monitoring provides the most specific and reproducible readout for drug screening on primary GSCs.
Collapse
|
69
|
Swartz JE, Pothen AJ, van Kempen PMW, Stegeman I, Formsma FK, Cann EMV, Willems SM, Grolman W. Poor prognosis in human papillomavirus-positive oropharyngeal squamous cell carcinomas that overexpress hypoxia inducible factor-1α. Head Neck 2016; 38:1338-46. [PMID: 27027530 DOI: 10.1002/hed.24445] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/02/2016] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Hypoxia induces stabilization of the transcription factor HIF-1alpha (HIF-1α), associated with (chemo-)radiotherapy resistance in oropharyngeal squamous cell carcinoma (SCC). We investigated the effect of HIF-1α expression on survival in relation to human papillomavirus (HPV) status in oropharyngeal SCC. METHODS We conducted an immunohistochemical analysis of HIF-1α protein expression and downstream targets carbonic anhydrase-IX (CA-IX) and glucose transporter-1 (GLUT-1) in 274 patients with oropharyngeal SCC. Overall survival (OS) was analyzed in total and stratified for HPV status and treatment. RESULTS In HPV-positive tumors (n = 44), HIF-1α overexpression predicted worse OS (hazard ratio [HR] = 6.23; p = .012), whereas TNM classification or treatment modality did not. In HPV-negative tumors (n = 218), advanced T and N classification and HIF-1α overexpression all independently predicted worse OS. However, the effect of HIF-1α overexpression on OS was lower in HPV-negative (HR = 1.50; p = .024) than in HPV-positive tumors. CONCLUSION HIF-1α overexpression is associated with worse OS and characterized a subgroup of patients with HPV-positive oropharyngeal SCC with poor prognosis. Possibly, patients with HIF-1α overexpressing HPV-positive tumors should not be eligible for treatment dose deescalation. © 2016 Wiley Periodicals, Inc. Head Neck 38: 1338-1346, 2016.
Collapse
Affiliation(s)
- Justin Egidius Swartz
- Department of Otorhinolaryngology - Head and Neck Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ajit Joe Pothen
- Department of Otorhinolaryngology - Head and Neck Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Inge Stegeman
- Department of Otorhinolaryngology - Head and Neck Surgery, University Medical Center Utrecht, Utrecht, The Netherlands.,Brain Center Rudolph Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Fleurieke Karlijn Formsma
- Department of Oral and Maxillofacial Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ellen Maria Van Cann
- Department of Oral and Maxillofacial Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Stefan Martin Willems
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wilko Grolman
- Department of Otorhinolaryngology - Head and Neck Surgery, University Medical Center Utrecht, Utrecht, The Netherlands.,Brain Center Rudolph Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
70
|
Simon KA, Mosadegh B, Minn KT, Lockett MR, Mohammady MR, Boucher DM, Hall AB, Hillier SM, Udagawa T, Eustace BK, Whitesides GM. Metabolic response of lung cancer cells to radiation in a paper-based 3D cell culture system. Biomaterials 2016; 95:47-59. [PMID: 27116031 DOI: 10.1016/j.biomaterials.2016.03.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 02/29/2016] [Accepted: 03/02/2016] [Indexed: 02/06/2023]
Abstract
This work demonstrates the application of a 3D culture system-Cells-in-Gels-in-Paper (CiGiP)-in evaluating the metabolic response of lung cancer cells to ionizing radiation. The 3D tissue-like construct-prepared by stacking multiple sheets of paper containing cell-embedded hydrogels-generates a gradient of oxygen and nutrients that decreases monotonically in the stack. Separating the layers of the stack after exposure enabled analysis of the cellular response to radiation as a function of oxygen and nutrient availability; this availability is dictated by the distance between the cells and the source of oxygenated medium. As the distance between the cells and source of oxygenated media increased, cells show increased levels of hypoxia-inducible factor 1-alpha, decreased proliferation, and reduced sensitivity to ionizing radiation. Each of these cellular responses are characteristic of cancer cells observed in solid tumors. With this setup we were able to differentiate three isogenic variants of A549 cells based on their metabolic radiosensitivity; these three variants have known differences in their metastatic behavior in vivo. This system can, therefore, capture some aspects of radiosensitivity of populations of cancer cells related to mass-transport phenomenon, carry out systematic studies of radiation response in vitro that decouple effects from migration and proliferation of cells, and regulate the exposure of oxygen to subpopulations of cells in a tissue-like construct either before or after irradiation.
Collapse
Affiliation(s)
- Karen A Simon
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, MA 02138, USA
| | - Bobak Mosadegh
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, 60 Oxford Street, Cambridge, MA 02138, USA; Dalio Institute of Cardiovascular Imaging, Department of Radiology, Weill Cornell Medicine, 413 E. 69th Street Suite BRB-108, New York, NY 10021, USA
| | - Kyaw Thu Minn
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, MA 02138, USA
| | - Matthew R Lockett
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, MA 02138, USA; Department of Chemistry, University of North Carolina at Chapel Hill, 125 South Road, Chapel Hill, NC 27599, USA
| | - Marym R Mohammady
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, MA 02138, USA
| | - Diane M Boucher
- Vertex Pharmaceuticals Incorporated, 50 Northern Blvd., Boston, MA 02210, USA
| | - Amy B Hall
- Vertex Pharmaceuticals Incorporated, 50 Northern Blvd., Boston, MA 02210, USA
| | - Shawn M Hillier
- Vertex Pharmaceuticals Incorporated, 50 Northern Blvd., Boston, MA 02210, USA
| | - Taturo Udagawa
- Vertex Pharmaceuticals Incorporated, 50 Northern Blvd., Boston, MA 02210, USA
| | - Brenda K Eustace
- Vertex Pharmaceuticals Incorporated, 50 Northern Blvd., Boston, MA 02210, USA.
| | - George M Whitesides
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, 60 Oxford Street, Cambridge, MA 02138, USA.
| |
Collapse
|
71
|
Rice SR, Katz MSJ, Mehta MP. Context for Protons as Adjunctive Therapy in Neovascular Age-Related Macular Degeneration: A Review. Int J Part Ther 2016; 2:555-569. [PMID: 31772967 PMCID: PMC6871634 DOI: 10.14338/ijpt-15-00019.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 12/04/2015] [Indexed: 11/21/2022] Open
Abstract
In the last few years we have witnessed increasing availability of proton therapy in the United States and worldwide. As a result, proton therapy is considered as either a primary or adjunctive approach for numerous indications where conventional radiation therapy shows promise but is accompanied by toxicities. Age-related macular degeneration (AMD) remains the leading cause of adult blindness in industrialized nations, and third worldwide, following cataract and glaucoma. Current standard therapy is intravitreal injection of anti-vascular endothelial growth factor agents. While this treatment shows improvement and stabilization in visual acuity for 40% of patients, 60% still experience disease progression. These injections are costly, necessitate repeated office visits, and carry the risk of endophthalmitis. The pathophysiology underlying neovascular AMD (nAMD) underscores the need to simultaneously target multiple pathways to retain useful vision. Radiation can be antiangiogenic, anti-inflammatory, and antiproliferative. Early photon therapy clinical trials were heterogeneous, and a Cochrane review of data demonstrated usefulness in treatment of nAMD but recommended further studies. Advantages of proton therapy over photon therapy include the ability to deliver a focal dose to the target while minimizing dose to normal structures, which is enhanced by unique treatment planning software that uses fluorescein angiography to verify target location and allows conformation of dose to the irregular shape and thickness characteristic of choroidal neovascular membranes, the pathognomonic finding in nAMD. Preliminary data suggest a potential role for proton therapy in the treatment of nAMD. In this article we review previous treatments for AMD, including those with both photon and proton radiation, and recommend future directions for clinical investigations to evaluate the role of proton therapy as an adjunct to antiangiogenic therapy, the current standard of care in this challenging setting.
Collapse
Affiliation(s)
- Stephanie R. Rice
- Department of Radiation Oncology, University of Maryland Medical Center, Baltimore, MD, USA
| | | | - Minesh P. Mehta
- Department of Radiation Oncology, University of Maryland Medical Center, Baltimore, MD, USA
- Department of Ophthalmology, National Retina Institute, Towson, MD, USA
| |
Collapse
|
72
|
Cellular Pathways in Response to Ionizing Radiation and Their Targetability for Tumor Radiosensitization. Int J Mol Sci 2016; 17:ijms17010102. [PMID: 26784176 PMCID: PMC4730344 DOI: 10.3390/ijms17010102] [Citation(s) in RCA: 293] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 12/22/2015] [Accepted: 12/25/2015] [Indexed: 12/20/2022] Open
Abstract
During the last few decades, improvements in the planning and application of radiotherapy in combination with surgery and chemotherapy resulted in increased survival rates of tumor patients. However, the success of radiotherapy is impaired by two reasons: firstly, the radioresistance of tumor cells and, secondly, the radiation-induced damage of normal tissue cells located in the field of ionizing radiation. These limitations demand the development of drugs for either radiosensitization of tumor cells or radioprotection of normal tissue cells. In order to identify potential targets, a detailed understanding of the cellular pathways involved in radiation response is an absolute requirement. This review describes the most important pathways of radioresponse and several key target proteins for radiosensitization.
Collapse
|
73
|
Oropesa Ávila M, Fernández Vega A, Garrido Maraver J, Villanueva Paz M, De Lavera I, De La Mata M, Cordero MD, Alcocer Gómez E, Delgado Pavón A, Álvarez Córdoba M, Cotán D, Sánchez-Alcázar JA. Emerging roles of apoptotic microtubules during the execution phase of apoptosis. Cytoskeleton (Hoboken) 2015; 72:435-46. [PMID: 26382917 DOI: 10.1002/cm.21254] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 09/01/2015] [Accepted: 09/09/2015] [Indexed: 12/14/2022]
Abstract
Apoptosis is a genetically programmed energy-dependent process of cell demise, characterized by specific morphological and biochemical events in which the activation of caspases has an essential role. During apoptosis the cytoskeleton participates actively in characteristic morphological rearrangements of the dying cell. This reorganisation has been assigned mainly to actinomyosin ring contraction, while microtubule and intermediate filaments are depolymerized at early stages of apoptosis. However, recent reports have showed that microtubules are reformed during the execution phase of apoptosis organizing an apoptotic microtubule network (AMN). AMN is organized behind plasma membrane, forming a cortical structure. Apoptotic microtubules repolymerization takes place in many cell types and under different apoptotic inducers. It has been hypothesized that AMN is critical for maintaining plasma membrane integrity and cell morphology during the execution phase of apoptosis. AMN disorganization leads apoptotic cells to secondary necrosis and the release of potential toxic molecules which can damage neighbor cells and promotes inflammation. Therefore, AMN formation during physiological apoptosis or in pathological apoptosis induced by anti-cancer treatments is essential for tissue homeostasis and the prevention of additional cell damage and inflammation.
Collapse
Affiliation(s)
- Manuel Oropesa Ávila
- Centro Andaluz De Biología Del Desarrollo (CABD), and Centro De Investigación Biomédica En Red: Enfermedades Raras, Instituto De Salud Carlos III, Universidad Pablo De Olavide-Consejo Superior De Investigaciones Científicas, Sevilla, 41013, Spain
| | - Alejandro Fernández Vega
- Centro Andaluz De Biología Del Desarrollo (CABD), and Centro De Investigación Biomédica En Red: Enfermedades Raras, Instituto De Salud Carlos III, Universidad Pablo De Olavide-Consejo Superior De Investigaciones Científicas, Sevilla, 41013, Spain
| | - Juan Garrido Maraver
- Centro Andaluz De Biología Del Desarrollo (CABD), and Centro De Investigación Biomédica En Red: Enfermedades Raras, Instituto De Salud Carlos III, Universidad Pablo De Olavide-Consejo Superior De Investigaciones Científicas, Sevilla, 41013, Spain
| | - Marina Villanueva Paz
- Centro Andaluz De Biología Del Desarrollo (CABD), and Centro De Investigación Biomédica En Red: Enfermedades Raras, Instituto De Salud Carlos III, Universidad Pablo De Olavide-Consejo Superior De Investigaciones Científicas, Sevilla, 41013, Spain
| | - Isabel De Lavera
- Centro Andaluz De Biología Del Desarrollo (CABD), and Centro De Investigación Biomédica En Red: Enfermedades Raras, Instituto De Salud Carlos III, Universidad Pablo De Olavide-Consejo Superior De Investigaciones Científicas, Sevilla, 41013, Spain
| | - Mario De La Mata
- Centro Andaluz De Biología Del Desarrollo (CABD), and Centro De Investigación Biomédica En Red: Enfermedades Raras, Instituto De Salud Carlos III, Universidad Pablo De Olavide-Consejo Superior De Investigaciones Científicas, Sevilla, 41013, Spain
| | - Mario D Cordero
- Facultad De Odontología. Universidad De Sevilla, Sevilla, 41009, Spain
| | - Elizabet Alcocer Gómez
- Centro Andaluz De Biología Del Desarrollo (CABD), and Centro De Investigación Biomédica En Red: Enfermedades Raras, Instituto De Salud Carlos III, Universidad Pablo De Olavide-Consejo Superior De Investigaciones Científicas, Sevilla, 41013, Spain
| | - Ana Delgado Pavón
- Centro Andaluz De Biología Del Desarrollo (CABD), and Centro De Investigación Biomédica En Red: Enfermedades Raras, Instituto De Salud Carlos III, Universidad Pablo De Olavide-Consejo Superior De Investigaciones Científicas, Sevilla, 41013, Spain
| | - Mónica Álvarez Córdoba
- Centro Andaluz De Biología Del Desarrollo (CABD), and Centro De Investigación Biomédica En Red: Enfermedades Raras, Instituto De Salud Carlos III, Universidad Pablo De Olavide-Consejo Superior De Investigaciones Científicas, Sevilla, 41013, Spain
| | - David Cotán
- Centro Andaluz De Biología Del Desarrollo (CABD), and Centro De Investigación Biomédica En Red: Enfermedades Raras, Instituto De Salud Carlos III, Universidad Pablo De Olavide-Consejo Superior De Investigaciones Científicas, Sevilla, 41013, Spain
| | - José Antonio Sánchez-Alcázar
- Centro Andaluz De Biología Del Desarrollo (CABD), and Centro De Investigación Biomédica En Red: Enfermedades Raras, Instituto De Salud Carlos III, Universidad Pablo De Olavide-Consejo Superior De Investigaciones Científicas, Sevilla, 41013, Spain
| |
Collapse
|
74
|
Larochelle A, Bellavance MA, Michaud JP, Rivest S. Bone marrow-derived macrophages and the CNS: An update on the use of experimental chimeric mouse models and bone marrow transplantation in neurological disorders. Biochim Biophys Acta Mol Basis Dis 2015; 1862:310-22. [PMID: 26432480 DOI: 10.1016/j.bbadis.2015.09.017] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 09/17/2015] [Accepted: 09/25/2015] [Indexed: 12/12/2022]
Abstract
The central nervous system (CNS) is a very unique system with multiple features that differentiate it from systemic tissues. One of the most captivating aspects of its distinctive nature is the presence of the blood brain barrier (BBB), which seals it from the periphery. Therefore, to preserve tissue homeostasis, the CNS has to rely heavily on resident cells such as microglia. These pivotal cells of the mononuclear lineage have important and dichotomous roles according to various neurological disorders. However, certain insults can overwhelm microglia as well as compromising the integrity of the BBB, thus allowing the infiltration of bone marrow-derived macrophages (BMDMs). The use of myeloablation and bone marrow transplantation allowed the generation of chimeric mice to study resident microglia and infiltrated BMDM separately. This breakthrough completely revolutionized the way we captured these 2 types of mononuclear phagocytic cells. We now realize that microglia and BMDM exhibit distinct features and appear to perform different tasks. Since these cells are central in several pathologies, it is crucial to use chimeric mice to analyze their functions and mechanisms to possibly harness them for therapeutic purpose. This review will shed light on the advent of this methodology and how it allowed deciphering the ontology of microglia and its maintenance during adulthood. We will also compare the different strategies used to perform myeloablation. Finally, we will discuss the landmark studies that used chimeric mice to characterize the roles of microglia and BMDM in several neurological disorders. This article is part of a Special Issue entitled: Neuro Inflammation edited by Helga E. de Vries and Markus Schwaninger.
Collapse
Affiliation(s)
- Antoine Larochelle
- Neuroscience Laboratory, CHU de Québec Research Center, Department of Molecular Medicine, Faculty of Medicine, Laval University, 2705 Laurier Blvd., Québec G1V 4G2, Canada
| | - Marc-André Bellavance
- Neuroscience Laboratory, CHU de Québec Research Center, Department of Molecular Medicine, Faculty of Medicine, Laval University, 2705 Laurier Blvd., Québec G1V 4G2, Canada
| | - Jean-Philippe Michaud
- Neuroscience Laboratory, CHU de Québec Research Center, Department of Molecular Medicine, Faculty of Medicine, Laval University, 2705 Laurier Blvd., Québec G1V 4G2, Canada
| | - Serge Rivest
- Neuroscience Laboratory, CHU de Québec Research Center, Department of Molecular Medicine, Faculty of Medicine, Laval University, 2705 Laurier Blvd., Québec G1V 4G2, Canada.
| |
Collapse
|
75
|
Krüger M, Pabst AM, Al-Nawas B, Horke S, Moergel M. Paraoxonase-2 (PON2) protects oral squamous cell cancer cells against irradiation-induced apoptosis. J Cancer Res Clin Oncol 2015; 141:1757-66. [PMID: 25708945 DOI: 10.1007/s00432-015-1941-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Accepted: 02/16/2015] [Indexed: 12/14/2022]
Abstract
PURPOSE Patients with oral squamous cell carcinomas (OSCC) often receive radiotherapy to preferentially induce apoptosis of cancer cells through generation of overwhelming DNA damage. This is amplified by generation of reactive oxygen species (ROS), thereby causing oxidative stress and cell death. However, tumors resist through different mechanisms, including upregulation of anti-apoptotic factors and enhanced ROS resistance. We recently reported that the antioxidative enzyme PON2 significantly enhances cellular stress resistance by attenuating mitochondrial ROS-mediated apoptosis. Further, PON2 is often upregulated in cancer. This prompted us to investigate its yet unknown role in the protection of OSCC against irradiation-induced cell death. METHODS PON2 expression was determined after 7 Gy singular irradiation in four OSCC cell lines (PCI-13, PCI-52, SCC-4, SCC-68) accompanied by the detection of caspase 3/7 activity. A direct role of PON2 was tested by siRNA-mediated knockdown. In vivo PON2 expression was tested in five patients with oral carcinoma and compared with healthy mucosa for the evaluation of clinical significance. RESULTS PON2 is variably expressed in OSCC in vitro and in vivo. Compared with the other cell lines, SCC-4 cells showed twofold more basal PON2 (p ≤ 0.05) and the lowest caspase 3/7 activity after singular irradiation (p ≤ 0.05). Contrarily, irradiation led to 1.2-fold induction of PON2 in PCI-13 with no effect on SCC-4 (≤0.05), suggesting that PON2 levels reflect the cells' irradiation sensitivity. In agreement, PON2 knockdown resulted in significant higher apoptosis rates (p ≤ 0.05). CONCLUSION Our findings give first evidence that upregulation of PON2 may protect OSCC against irradiation-induced apoptosis.
Collapse
Affiliation(s)
- Maximilian Krüger
- Department of Oral and Maxillofacial Surgery, University Medical Center of the Johannes Gutenberg-University, Augustusplatz 2, 55131, Mainz, Germany,
| | | | | | | | | |
Collapse
|
76
|
Li H, Yang ZY, Liu C, Zeng YP, Hao YH, Gu Y, Wang WD, Li R. PEGylated ceria nanoparticles used for radioprotection on human liver cells under γ-ray irradiation. Free Radic Biol Med 2015; 87:26-35. [PMID: 26117316 DOI: 10.1016/j.freeradbiomed.2015.06.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 06/03/2015] [Accepted: 06/06/2015] [Indexed: 10/23/2022]
Abstract
Ceria nanoparticles (CNPs) have recently been shown to protect cells and animals from radiation-induced damage. However, most of the CNPs used in previous studies were either naked or weakly protected by surfactants, which inevitably encounter many obstacles in biological applications. Here, alendronate was used as an ideal anchor to graft polyethylene glycol (PEG) onto CNPs, leading to enhanced stability, reduced cytotoxicity, and improved biological properties. Further investigation assessed the protective ability of the nanoparticles against radiation-induced effects for human normal liver cells (L-02), indicating that the PEGylated CNPs (CNPs-AL-PEG) were more efficient than naked CNPs. We determined that enhanced Ce(3+)/Ce(4+) ratios improved intracellular dispersion and that the ameliorated intracellular distribution of CNPs-AL-PEG contributes to the elevated expression of SOD2, which leads to increased protection of normal cells against ROS and reduces the oxidatively generated DNA damage. These studies hold tremendous promise for radioprotection and biological applications.
Collapse
Affiliation(s)
- Hong Li
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Zhang-You Yang
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Cong Liu
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Yi-Ping Zeng
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Yu-Hui Hao
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Ying Gu
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Wei-Dong Wang
- Department of Radiation Oncology, Sichuan Cancer Hospital, Chengdu 610041, China.
| | - Rong Li
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
77
|
Hernández-Corbacho MJ, Canals D, Adada MM, Liu M, Senkal CE, Yi JK, Mao C, Luberto C, Hannun YA, Obeid LM. Tumor Necrosis Factor-α (TNFα)-induced Ceramide Generation via Ceramide Synthases Regulates Loss of Focal Adhesion Kinase (FAK) and Programmed Cell Death. J Biol Chem 2015; 290:25356-73. [PMID: 26318452 DOI: 10.1074/jbc.m115.658658] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Indexed: 11/06/2022] Open
Abstract
Ceramide synthases (CerS1-CerS6), which catalyze the N-acylation of the (dihydro)sphingosine backbone to produce (dihydro)ceramide in both the de novo and the salvage or recycling pathway of ceramide generation, have been implicated in the control of programmed cell death. However, the regulation of the de novo pathway compared with the salvage pathway is not fully understood. In the current study, we have found that late accumulation of multiple ceramide and dihydroceramide species in MCF-7 cells treated with TNFα occurred by up-regulation of both pathways of ceramide synthesis. Nevertheless, fumonisin B1 but not myriocin was able to protect from TNFα-induced cell death, suggesting that ceramide synthase activity is crucial for the progression of cell death and that the pool of ceramide involved derives from the salvage pathway rather than de novo biosynthesis. Furthermore, compared with control cells, TNFα-treated cells exhibited reduced focal adhesion kinase and subsequent plasma membrane permeabilization, which was blocked exclusively by fumonisin B1. In addition, exogenously added C6-ceramide mimicked the effects of TNFα that lead to cell death, which were inhibited by fumonisin B1. Knockdown of individual ceramide synthases identified CerS6 and its product C16-ceramide as the ceramide synthase isoform essential for the regulation of cell death. In summary, our data suggest a novel role for CerS6/C16-ceramide as an upstream effector of the loss of focal adhesion protein and plasma membrane permeabilization, via the activation of caspase-7, and identify the salvage pathway as the critical mechanism of ceramide generation that controls cell death.
Collapse
Affiliation(s)
| | - Daniel Canals
- From the Stony Brook Cancer Center, the Department of Medicine, and
| | - Mohamad M Adada
- From the Stony Brook Cancer Center, the Department of Medicine, and
| | - Mengling Liu
- From the Stony Brook Cancer Center, the Department of Medicine, and
| | - Can E Senkal
- From the Stony Brook Cancer Center, the Department of Medicine, and
| | - Jae Kyo Yi
- From the Stony Brook Cancer Center, the Department of Medicine, and
| | - Cungui Mao
- From the Stony Brook Cancer Center, the Department of Medicine, and
| | - Chiara Luberto
- From the Stony Brook Cancer Center, the Department of Physiology and Biophysics, Stony Brook University, Health Sciences Center, Stony Brook, New York 11794 and
| | - Yusuf A Hannun
- From the Stony Brook Cancer Center, the Department of Medicine, and
| | - Lina M Obeid
- From the Stony Brook Cancer Center, the Department of Medicine, and the Northport Veterans Affairs Medical Center, Northport, New York 11768
| |
Collapse
|
78
|
Tamotsu K, Okumura H, Uchikado Y, Kita Y, Sasaki K, Omoto I, Owaki T, Arigami T, Uenosono Y, Nakajo A, Kijima Y, Ishigami S, Natsugoe S. Correlation of Aurora-A expression with the effect of chemoradiation therapy on esophageal squamous cell carcinoma. BMC Cancer 2015; 15:323. [PMID: 25924824 PMCID: PMC4423148 DOI: 10.1186/s12885-015-1329-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 04/21/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Chemoradiation therapy (CRT) is one of the most useful treatments for esophageal squamous cell carcinoma (ESCC). However, because some patients respond well to CRT and others do not, it is important to be able to predict response to CRT before beginning treatment by using markers. Aurora-A encodes a cell cycle regulated serine/threonine kinase that has essential functions in centrosome maturation and chromosome segregation. In this study, we investigated the relationship between the expression of Aurora-A and the response to CRT in patients with ESCC. METHODS We immunohistochemically investigated the expression of Aurora-A in biopsy specimens of untreated primary tumors of 78 patients with ESCC and determined the relationship between Aurora-A levels and patient responses to CRT, which consisted of 5-fluorouracil plus cisplatin and 40 Gy of radiation. RESULTS Tumors were judged as Aurora-A positive when more than 10% of the cancer cells displayed a distinct positive nuclear anti-Aurora-A immunoreaction by immunohistochemical evaluation. The tumors of 46 of 78 patients (58.9%) displayed positive expression of Aurora-A. In terms of clinical response the percentage of patients showing complete response (CR), incomplete response/stable disease of primary lesion (IR/SD), and progressive disease (PD) was 19.2, 69.2, and 11.5%, respectively. In terms of histological response the tumor grade of the 41 patients who underwent surgery was as follows: grade 1, 48.8%; grade 2, 29.2%; grade 3, 22.0%. CRT was effective for patients who had Aurora-A (+) tumors (clinically: P = 0.0003, histologically: P = 0.036). CONCLUSIONS Our results suggest that Aurora-A expression in biopsy specimens of primary tumors is associated with CRT efficacy in patients with ESCC. Assessment of Aurora-A expression in biopsy specimens maybe useful for regarding the potential utility of CRT therapy for patients with ESCC before treatment.
Collapse
Affiliation(s)
- Kiyokazu Tamotsu
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical Sciences, Kagoshima University, Sakuragaoka 8-35-1, Kagoshima, 890-8520, Japan.
| | - Hiroshi Okumura
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical Sciences, Kagoshima University, Sakuragaoka 8-35-1, Kagoshima, 890-8520, Japan.
| | - Yasuto Uchikado
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical Sciences, Kagoshima University, Sakuragaoka 8-35-1, Kagoshima, 890-8520, Japan.
| | - Yoshiaki Kita
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical Sciences, Kagoshima University, Sakuragaoka 8-35-1, Kagoshima, 890-8520, Japan.
| | - Ken Sasaki
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical Sciences, Kagoshima University, Sakuragaoka 8-35-1, Kagoshima, 890-8520, Japan.
| | - Itaru Omoto
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical Sciences, Kagoshima University, Sakuragaoka 8-35-1, Kagoshima, 890-8520, Japan.
| | - Tetsuhiro Owaki
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical Sciences, Kagoshima University, Sakuragaoka 8-35-1, Kagoshima, 890-8520, Japan.
| | - Takaaki Arigami
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical Sciences, Kagoshima University, Sakuragaoka 8-35-1, Kagoshima, 890-8520, Japan.
| | - Yoshikazu Uenosono
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical Sciences, Kagoshima University, Sakuragaoka 8-35-1, Kagoshima, 890-8520, Japan.
| | - Akihiro Nakajo
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical Sciences, Kagoshima University, Sakuragaoka 8-35-1, Kagoshima, 890-8520, Japan.
| | - Yuko Kijima
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical Sciences, Kagoshima University, Sakuragaoka 8-35-1, Kagoshima, 890-8520, Japan.
| | - Sumiya Ishigami
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical Sciences, Kagoshima University, Sakuragaoka 8-35-1, Kagoshima, 890-8520, Japan.
| | - Shoji Natsugoe
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical Sciences, Kagoshima University, Sakuragaoka 8-35-1, Kagoshima, 890-8520, Japan.
| |
Collapse
|
79
|
Swartz JE, Pothen AJ, Stegeman I, Willems SM, Grolman W. Clinical implications of hypoxia biomarker expression in head and neck squamous cell carcinoma: a systematic review. Cancer Med 2015; 4:1101-16. [PMID: 25919147 PMCID: PMC4529348 DOI: 10.1002/cam4.460] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 03/05/2015] [Accepted: 03/10/2015] [Indexed: 02/06/2023] Open
Abstract
Awareness increases that the tumor biology influences treatment outcome and prognosis in cancer. Tumor hypoxia is thought to decrease sensitivity to radiotherapy and some forms of chemotherapy. Presence of hypoxia may be assessed by investigating expression of endogenous markers of hypoxia (EMH) using immunohistochemistry (IHC). In this systematic review we investigated the effect of EMH expression on local control and survival according to treatment modality in head and neck cancer (head and neck squamous cell carcinoma [HNSCC]). A search was performed in MEDLINE and EMBASE. Studies were eligible for inclusion that described EMH expression in relation to outcome in HNSCC patients. Quality was assessed using the Quality in Prognosis Studies (QUIPS) tool. Hazard ratios for locoregional control and survival were extracted. Forty studies of adequate quality were included. HIF-1a, HIF-2a, CA-IX, GLUT-1, and OPN were identified as the best described EMHs. With exception of HIF-2a, all EMHs were significantly related to adverse outcome in multiple studies, especially in studies where patients underwent single-modality treatment. Positive expression was often correlated with adverse clinical characteristics, including disease stage and differentiation grade. In summary, EMH expression was common in HNSCC patients and negatively influenced their prognosis. Future studies should investigate the effect of hypoxia-modified treatment schedules in patients with high In summary, EMH expression. These may include ARCON, treatment with nimorazole, or novel targeted therapies directed at hypoxic tissue. Also, the feasibility of surgical removal of the hypoxic tumor volume prior to radiotherapy should be investigated.
Collapse
Affiliation(s)
- Justin E Swartz
- Department of Otorhinolaryngology - Head and Neck Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ajit J Pothen
- Department of Otorhinolaryngology - Head and Neck Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Inge Stegeman
- Department of Otorhinolaryngology - Head and Neck Surgery, University Medical Center Utrecht, Utrecht, The Netherlands.,Brain Center Rudolph Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Stefan M Willems
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wilko Grolman
- Department of Otorhinolaryngology - Head and Neck Surgery, University Medical Center Utrecht, Utrecht, The Netherlands.,Brain Center Rudolph Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
80
|
Talamo G, Dimaio C, Abbi KKS, Pandey MK, Malysz J, Creer MH, Zhu J, Mir MA, Varlotto JM. Current role of radiation therapy for multiple myeloma. Front Oncol 2015; 5:40. [PMID: 25741475 PMCID: PMC4332323 DOI: 10.3389/fonc.2015.00040] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 02/04/2015] [Indexed: 11/25/2022] Open
Abstract
Background: Radiation therapy (RT) is a treatment modality traditionally used in patients with multiple myeloma (MM), but little is known regarding the role and effectiveness of RT in the era of novel agents, i.e., immunomodulatory drugs and proteasome inhibitors. Methods: We retrospectively reviewed data from 449 consecutive MM patients seen at our institute in 2010–2012 to assess indications for RT as well as its effectiveness. Pain response was scored similarly to RTOG 0631 and used the Numerical Rating Pain Scale. Results: Among 442 evaluable patients, 149 (34%) patients and 262 sites received RT. The most common indication for RT was palliation of bone pain (n = 109, 42%), followed by prevention/treatment of pathological fractures (n = 73, 28%), spinal cord compression (n = 26, 10%), and involvement of vital organs/extramedullary disease (n = 25, 10%). Of the 55 patients evaluable for pain relief, complete and partial responses were obtained in 76.4 and 7.2%, respectively. Prior RT did not significantly decrease the median number of peripheral blood stem cells collected for autologous transplant, even when prior RT was given to both the spine and pelvis. Inadequacy of stem cell collection for autologous stem cell transplant (ASCT) was not significantly different and it occurred in 9 and 15% of patients receiving no RT and spine/pelvic RT, respectively. None of the three cases of therapy-induced acute myelogenous leukemia/MDS occurred in the RT group. Conclusion: Despite the introduction of novel effective agents in the treatment of MM, RT remains a major therapeutic component for the management in 34% of patients, and it effectively provides pain relief while not interfering with successful peripheral blood stem cell collection for ASCT.
Collapse
Affiliation(s)
| | | | - Kamal K S Abbi
- Penn State Milton S. Hershey Medical Center , Hershey, PA , USA
| | - Manoj K Pandey
- Penn State Milton S. Hershey Medical Center , Hershey, PA , USA
| | - Jozef Malysz
- Penn State Milton S. Hershey Medical Center , Hershey, PA , USA
| | - Michael H Creer
- Penn State Milton S. Hershey Medical Center , Hershey, PA , USA
| | - Junjia Zhu
- Penn State Milton S. Hershey Medical Center , Hershey, PA , USA
| | - Muhammad A Mir
- Penn State Milton S. Hershey Medical Center , Hershey, PA , USA
| | - John M Varlotto
- Penn State Milton S. Hershey Medical Center , Hershey, PA , USA
| |
Collapse
|
81
|
Meng MB, Wang HH, Guo WH, Wu ZQ, Zeng XL, Zaorsky NG, Shi HS, Qian D, Niu ZM, Jiang B, Zhao LJ, Yuan ZY, Wang P. Targeting pyruvate kinase M2 contributes to radiosensitivity of non-small cell lung cancer cells in vitro and in vivo. Cancer Lett 2014; 356:985-93. [PMID: 25444918 DOI: 10.1016/j.canlet.2014.11.016] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 09/17/2014] [Accepted: 11/09/2014] [Indexed: 02/05/2023]
Abstract
Aerobic glycolysis, a metabolic hallmark of cancer, is associated with radioresistance in non-small cell lung cancer (NSCLC). Pyruvate kinase M2 isoform (PKM2), a key regulator of glycolysis, is expressed exclusively in cancers. However, the impact of PKM2 silencing on the radiosensitivity of NSCLC has not been explored. Here, we show a plasmid of shRNA-PKM2 for expressing a short hairpin RNA targeting PKM2 (pshRNA-PKM2) and demonstrate that treatment with pshRNA-PKM2 effectively inhibits PKM2 expression in NSCLC cell lines and xenografts. Silencing of PKM2 expression enhanced ionizing radiation (IR)-induced apoptosis and autophagy in vitro and in vivo, accompanied by inhibiting AKT and PDK1 phosphorylation, but enhanced ERK and GSK3β phosphorylation. These results demonstrated that knockdown of PKM2 expression enhances the radiosensitivity of NSCLC cell lines and xenografts as well as may aid in the design of new therapies for the treatment of NSCLC.
Collapse
Affiliation(s)
- Mao-Bin Meng
- Department of Radiation Oncology, CyberKnife Center, and Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China.
| | - Huan-Huan Wang
- Department of Radiation Oncology, CyberKnife Center, and Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
| | - Wen-Hao Guo
- Department of Abdominal Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, West China Clinical Medicine School, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhi-Qiang Wu
- Department of Radiation Oncology, CyberKnife Center, and Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
| | - Xian-Liang Zeng
- Department of Radiation Oncology, CyberKnife Center, and Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
| | - Nicholas G Zaorsky
- Department of Radiation Oncology, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | - Hua-Shan Shi
- Department of Abdominal Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, West China Clinical Medicine School, Sichuan University, Chengdu, Sichuan 610041, China
| | - Dong Qian
- Department of Radiation Oncology, CyberKnife Center, and Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
| | - Zhi-Min Niu
- Department of Radiation Oncology, CyberKnife Center, and Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
| | - Bo Jiang
- Department of Radiation Oncology, CyberKnife Center, and Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
| | - Lu-Jun Zhao
- Department of Radiation Oncology, CyberKnife Center, and Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
| | - Zhi-Yong Yuan
- Department of Radiation Oncology, CyberKnife Center, and Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
| | - Ping Wang
- Department of Radiation Oncology, CyberKnife Center, and Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
| |
Collapse
|
82
|
Radiosensitization by a novel Bcl-2 and Bcl-XL inhibitor S44563 in small-cell lung cancer. Cell Death Dis 2014; 5:e1423. [PMID: 25232677 PMCID: PMC4540189 DOI: 10.1038/cddis.2014.365] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 06/06/2014] [Accepted: 06/26/2014] [Indexed: 12/14/2022]
Abstract
Radiotherapy has a critical role in the treatment of small-cell lung cancer (SCLC). The effectiveness of radiation in SCLC remains limited as resistance results from defects in apoptosis. In the current study, we investigated whether using the Bcl-2/Bcl-XL inhibitor S44563 can enhance radiosensitivity of SCLC cells in vitro and in vivo. In vitro studies confirmed that S44563 caused SCLC cells to acquire hallmarks of apoptosis. S44563 markedly enhanced the sensitivity of SCLC cells to radiation, as determined by a clonogenic assay. The combination of S44563 and cisplatin-based chemo-radiation showed a significant tumor growth delay and increased overall survival in mouse xenograft models. This positive interaction was greater when S44563 was given after the completion of the radiation, which might be explained by the radiation-induced overexpression of anti-apoptotic proteins secondary to activation of the NF-κB pathway. These data underline the possibility of combining IR and Bcl-2/Bcl-XL inhibition in the treatment of SCLC as they underscore the importance of administering conventional and targeted therapies in an optimal sequence.
Collapse
|
83
|
Keta O, Todorović D, Popović N, Korićanac L, Cuttone G, Petrović I, Ristić-Fira A. Radiosensitivity of human ovarian carcinoma and melanoma cells to γ-rays and protons. Arch Med Sci 2014; 10:578-86. [PMID: 25097591 PMCID: PMC4107263 DOI: 10.5114/aoms.2014.43751] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 12/14/2012] [Accepted: 02/24/2013] [Indexed: 01/16/2023] Open
Abstract
INTRODUCTION Proton radiation offers physical advantages over conventional radiation. Radiosensitivity of human 59M ovarian cancer and HTB140 melanoma cells was investigated after exposure to γ-rays and protons. MATERIAL AND METHODS Irradiations were performed in the middle of a 62 MeV therapeutic proton spread out Bragg peak with doses ranging from 2 to 16 Gy. The mean energy of protons was 34.88 ±2.15 MeV, corresponding to the linear energy transfer of 4.7 ±0.2 keV/µm. Irradiations with γ-rays were performed using the same doses. Viability, proliferation and survival were assessed 7 days after both types of irradiation while analyses of cell cycle and apoptosis were performed 48 h after irradiation. RESULTS Results showed that γ-rays and protons reduced the number of viable cells for both cell lines, with stronger inactivation achieved after irradiation with protons. Surviving fractions for 59M were 0.91 ±0.01 for γ-rays and 0.81 ±0.01 for protons, while those for HTB140 cells were 0.93 ±0.01 for γ-rays and 0.86 ±0.01 for protons. Relative biological effectiveness of protons, being 2.47 ±0.22 for 59M and 2.08 ±0.36 for HTB140, indicated that protons provoked better cell elimination than γ-rays. After proton irradiation proliferation capacity of the two cell lines was slightly higher as compared to γ-rays. Proliferation was higher for 59M than for HTB140 cells after both types of irradiation. Induction of apoptosis and G2 arrest detected after proton irradiation were more prominent in 59M cells. CONCLUSIONS The obtained results suggest that protons exert better antitumour effects on ovarian carcinoma and melanoma cells than γ-rays. The dissimilar response of these cells to radiation is related to their different features.
Collapse
Affiliation(s)
- Otilija Keta
- Vinča Institute of Nuclear Sciences, University of Belgrade, Belgrade, Serbia
| | | | - Nataša Popović
- Vinča Institute of Nuclear Sciences, University of Belgrade, Belgrade, Serbia
| | - Lela Korićanac
- Vinča Institute of Nuclear Sciences, University of Belgrade, Belgrade, Serbia
| | - Giacomo Cuttone
- Istituto Nazionale di Fisica Nucleare, Laboratori Nazionali del Sud, Catania, Italy
| | - Ivan Petrović
- Vinča Institute of Nuclear Sciences, University of Belgrade, Belgrade, Serbia
| | | |
Collapse
|
84
|
Claro S, Oshiro MEM, Mortara RA, Paredes-Gamero EJ, Pereira GJS, Smaili SS, Ferreira AT. γ-Rays-generated ROS induce apoptosis via mitochondrial and cell cycle alteration in smooth muscle cells. Int J Radiat Biol 2014; 90:914-27. [DOI: 10.3109/09553002.2014.911988] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
85
|
Li WF, Zhang L, Li HY, Zheng SS, Zhao L. WISP-1 contributes to fractionated irradiation-induced radioresistance in esophageal carcinoma cell lines and mice. PLoS One 2014; 9:e94751. [PMID: 24728101 PMCID: PMC3984255 DOI: 10.1371/journal.pone.0094751] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 03/19/2014] [Indexed: 12/22/2022] Open
Abstract
Cancer cells that survive fractionated irradiation can be radioresistant and cause tumor recurrence. However, the molecular mechanisms underlying the development of radioresistance in cancer cells remain elusive. The aim of this study was to investigate the role of WISP-1 in the development of radioresistance in esophageal carcinoma during fractionated irradiation. Radioresistant esophageal cancer cells were generated from normal esophageal cancer cells via fractionated irradiation, and expression levels of related proteins were determined by Western blot. Radiosensitivity of cells was established by clonogenic cell survival assays, and cell cycle distribution was evaluated by flow cytometry. Protein distributions were determined by immunofluorescence, and cell toxicity was evaluated by cell counting kit-8 assays. In vivo validations were performed in a xenograft transplantation mouse model. Our data indicate that WISP-1 plays an important role in the development of radioresistance in esophageal cancer cells during fractionated irradiation. The overexression of WISP-1 in esophageal cancer cells was associated with radioresistance. Depletion of extracellular WISP-1 by antibody neutralizing reversed radioresistance and directly induced mitotic catastrophe resulting in cell death. WISP-1 may be a candidate therapeutic target in the treatment of recurrent esophageal carcinoma after radiotherapy.
Collapse
Affiliation(s)
- Wen-Feng Li
- Department of Radiation Oncology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Li Zhang
- Department of Radiation Oncology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hai-Ying Li
- Laboratory of Internal Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Si-Si Zheng
- Division of PET/CT, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Liang Zhao
- Division of PET/CT, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- * E-mail:
| |
Collapse
|
86
|
Genotoxic anti-cancer agents and their relationship to DNA damage, mitosis, and checkpoint adaptation in proliferating cancer cells. Int J Mol Sci 2014; 15:3403-31. [PMID: 24573252 PMCID: PMC3975345 DOI: 10.3390/ijms15033403] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 01/22/2014] [Accepted: 02/14/2014] [Indexed: 12/19/2022] Open
Abstract
When a human cell detects damaged DNA, it initiates the DNA damage response (DDR) that permits it to repair the damage and avoid transmitting it to daughter cells. Despite this response, changes to the genome occur and some cells, such as proliferating cancer cells, are prone to genome instability. The cellular processes that lead to genomic changes after a genotoxic event are not well understood. Our research focuses on the relationship between genotoxic cancer drugs and checkpoint adaptation, which is the process of mitosis with damaged DNA. We examine the types of DNA damage induced by widely used cancer drugs and describe their effects upon proliferating cancer cells. There is evidence that cell death caused by genotoxic cancer drugs in some cases includes exiting a DNA damage cell cycle arrest and entry into mitosis. Furthermore, some cells are able to survive this process at a time when the genome is most susceptible to change or rearrangement. Checkpoint adaptation is poorly characterised in human cells; we predict that increasing our understanding of this pathway may help to understand genomic instability in cancer cells and provide insight into methods to improve the efficacy of current cancer therapies.
Collapse
|
87
|
Emery SM, Alotaibi MR, Tao Q, Selley DE, Lichtman AH, Gewirtz DA. Combined antiproliferative effects of the aminoalkylindole WIN55,212-2 and radiation in breast cancer cells. J Pharmacol Exp Ther 2014; 348:293-302. [PMID: 24259678 PMCID: PMC4859093 DOI: 10.1124/jpet.113.205120] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 11/19/2013] [Indexed: 01/08/2023] Open
Abstract
The potential antitumor activity of cannabinoid receptor agonists, such as the aminoalklylindole WIN55,212-2 (WIN2), has been studied extensively, but their potential interaction with conventional cancer therapies, such as radiation, remains unknown. In the present work, the influence of WIN2 on the antiproliferative activity of radiation in human (MCF-7 and MDA-MB231) and murine (4T1) breast cancer cells was investigated. The antiproliferative effects produced by combination of WIN2 and radiation were more effective than either agent alone. The stereoisomer of WIN2, WIN55,212-3 (WIN3), failed to inhibit growth or potentiate the growth-inhibitory effects of radiation, indicative of stereospecificity. Two other aminoalkylindoles, pravadoline and JWH-015 [(2-methyl-1-propyl-1H-indol-3-yl)-1-naphthalenyl-methanone], also enhanced the antiproliferative effects of radiation, but other synthetic cannabinoids (i.e., nabilone, CP55,940 [(+)-rel-5-(1,1-dimethylheptyl)-2-[(1R,2R,5R)-5-hydroxy-2-(3-hydroxypropyl)cyclohexyl]-phenol], and methanandamide) or phytocannabinoids [i.e., Δ⁹-tetrahydrocannabinol (THC) and cannabidiol] did not. The combination treatment of WIN2 + radiation promoted both autophagy and senescence but not apoptosis or necrosis. WIN2 also failed to alter radiation-induced DNA damage or the apparent rate of DNA repair. Although the antiproliferative actions of WIN2 were mediated through noncannabinoid receptor-mediated pathways, the observation that WIN2 interfered with growth stimulation by sphingosine-1-phosphate (S1P) implicates the potential involvement of S1P/ceramide signaling pathways. In addition to demonstrating that aminoalkylindole compounds could potentially augment the effectiveness of radiation treatment in breast cancer, the present study suggests that THC and nabilone are unlikely to interfere with the effectiveness of radiation therapy, which is of particular relevance to patients using cannabinoid-based drugs to ameliorate the toxicity of cancer therapies.
Collapse
Affiliation(s)
- Sean M Emery
- Departments of Pharmacology and Toxicology (S.M.E., M.R.A., Q.T., D.E.S., A.H.T., D.A.G.), Massey Cancer Center (D.A.G.), Virginia Commonwealth University, Richmond, Virginia
| | | | | | | | | | | |
Collapse
|
88
|
Kahandal SS, Kale SR, Gawande MB, Jayaram RV. A mild route for one pot synthesis of 5,6-unsubstituted 1,4-dihydropyridines catalyzed by sulphated mixed metal oxides. Catal Sci Technol 2014. [DOI: 10.1039/c3cy00651d] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
89
|
A combination of radiosurgery and soluble tissue factor enhances vascular targeting for experimental glioblastoma. BIOMED RESEARCH INTERNATIONAL 2013; 2013:390714. [PMID: 24307995 PMCID: PMC3838847 DOI: 10.1155/2013/390714] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2013] [Accepted: 09/25/2013] [Indexed: 11/17/2022]
Abstract
Radiosurgery for glioblastoma is limited to the development of resistance, allowing tumor cells to survive and initiate tumor recurrence. Based on our previous work that coadministration of tissue factor and lipopolysaccharide following radiosurgery selectively induced thrombosis in cerebral arteriovenous malformations, achieving thrombosis of 69% of the capillaries and 39% of medium sized vessels, we hypothesized that a rapid and selective shutdown of the capillaries in glioblastoma vasculature would decrease the delivery of oxygen and nutrients, reducing tumor growth, preventing intracranial hypertension, and improving life expectancy. Glioblastoma was formed by implantation of GL261 cells into C57Bl/6 mouse brain. Mice were intravenously injected tissue factor, lipopolysaccharide, a combination of both, or placebo 24 hours after radiosurgery. Control mice received both agents after sham irradiation. Coadministration of tissue factor and lipopolysaccharide led to the formation of thrombi in up to 87 ± 8% of the capillaries and 46 ± 4% of medium sized vessels within glioblastoma. The survival rate of mice in this group was 80% versus no survivor in placebo controls 30 days after irradiation. Animal body weight increased with time in this group (r = 0.88, P = 0.0001). Thus, radiosurgery enhanced treatment with tissue factor, and lipopolysaccharide selectively induces thrombosis in glioblastoma vasculature, improving life expectancy.
Collapse
|
90
|
Mechanisms of radiation toxicity in transformed and non-transformed cells. Int J Mol Sci 2013; 14:15931-58. [PMID: 23912235 PMCID: PMC3759894 DOI: 10.3390/ijms140815931] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 07/19/2013] [Accepted: 07/22/2013] [Indexed: 12/31/2022] Open
Abstract
Radiation damage to biological systems is determined by the type of radiation, the total dosage of exposure, the dose rate, and the region of the body exposed. Three modes of cell death—necrosis, apoptosis, and autophagy—as well as accelerated senescence have been demonstrated to occur in vitro and in vivo in response to radiation in cancer cells as well as in normal cells. The basis for cellular selection for each mode depends on various factors including the specific cell type involved, the dose of radiation absorbed by the cell, and whether it is proliferating and/or transformed. Here we review the signaling mechanisms activated by radiation for the induction of toxicity in transformed and normal cells. Understanding the molecular mechanisms of radiation toxicity is critical for the development of radiation countermeasures as well as for the improvement of clinical radiation in cancer treatment.
Collapse
|
91
|
Wei F, Liu Y, Guo Y, Xiang A, Wang G, Xue X, Lu Z. miR-99b-targeted mTOR induction contributes to irradiation resistance in pancreatic cancer. Mol Cancer 2013; 12:81. [PMID: 23886294 PMCID: PMC3726417 DOI: 10.1186/1476-4598-12-81] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 07/22/2013] [Indexed: 02/08/2023] Open
Abstract
Background Radiation exerts direct antitumor effects and is widely used in clinics, but the efficacy is severely compromised by tumor resistance. Therefore uncovering the mechanism of radioresistance might promote the development of new strategies to overcome radioresistance by manipulating activity of the key molecules. Methods Immunohistochemistry were used to find whether mTOR were over-activated in radioresistant patients’ biopsies. Then Western blot, real-time PCR and transfection were used to find whether radiotherapy regulates the expression and activity of mTOR by modulating its targeting microRNA in human pancreatic cancer cell lines PANC-1, Capan-2 and BxPC-3. Finally efficacy of radiation combined with mTOR dual inhibitor AZD8055 was assessed in vitro and in vivo. Results Ionizing radiation promoted mTOR expression and activation in pancreatic cancer cells through reducing miR-99b expression, which negatively regulated mTOR. Novel mTOR inhibitor, AZD8055 (10 nM, 100 nM, 500 nM) synergistically promoted radiation (0–10 Gy) induced cell growth inhibition and apoptosis. In human pancreatic cancer xenografts, fractionated radiation combined with AZD8055 treatment further increased the anti-tumor effect, the tumor volume was shrinked to 278 mm3 after combination treatment for 3 weeks compared with single radiation (678 mm3) or AZD8055 (708 mm3) treatment (P < 0.01). Conclusions Our data provide a rationale for overcoming radio-resistance by combined with mTOR inhibitor AZD8055 in pancreatic cancer therapy.
Collapse
|
92
|
Andzans Z, Krauze A, Adlere I, Krasnova L, Duburs G. Synthesis and enantioselective lipase-catalyzed kinetic resolution of methyl 6-(methoxycarbonyl-methyl)sulfanyl-1,4-dihydropyridine-3-carboxylates. Chem Heterocycl Compd (N Y) 2013. [DOI: 10.1007/s10593-013-1263-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
93
|
Lanvin O, Monferran S, Delmas C, Couderc B, Toulas C, Cohen-Jonathan-Moyal E. Radiation-induced mitotic cell death and glioblastoma radioresistance: a new regulating pathway controlled by integrin-linked kinase, hypoxia-inducible factor 1 alpha and survivin in U87 cells. Eur J Cancer 2013; 49:2884-91. [PMID: 23747271 DOI: 10.1016/j.ejca.2013.05.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 04/22/2013] [Accepted: 05/09/2013] [Indexed: 01/03/2023]
Abstract
We have previously shown that integrin-linked kinase (ILK) regulates U87 glioblastoma cell radioresistance by modulating the main radiation-induced cell death mechanism in solid tumours, the mitotic cell death. To decipher the biological pathways involved in these mechanisms, we constructed a U87 glioblastoma cell model expressing an inducible shRNA directed against ILK (U87shILK). We then demonstrated that silencing ILK enhanced radiation-induced centrosome overduplication, leading to radiation-induced mitotic cell death. In this model, ionising radiations induce hypoxia-inducible factor 1 alpha (HIF-1α) stabilisation which is inhibited by silencing ILK. Moreover, silencing HIF-1α in U87 cells reduced the surviving fraction after 2 Gy irradiation by increasing cell sensitivity to radiation-induced mitotic cell death and centrosome amplification. Because it is known that HIF-1α controls survivin expression, we then looked at the ILK silencing effect on survivin expression. We show that survivin expression is decreased in U87shILK cells. Furthermore, treating U87 cells with the specific survivin suppressor YM155 significantly increased the percentage of giant multinucleated cells, centrosomal overduplication and thus U87 cell radiosensitivity. In consequence, we decipher here a new pathway of glioma radioresistance via the regulation of radiation-induced centrosome duplication and therefore mitotic cell death by ILK, HIF-1α and survivin. This work identifies new targets in glioblastoma with the intention of radiosensitising these highly radioresistant tumours.
Collapse
Affiliation(s)
- Olivia Lanvin
- Institut National de Santé et de Recherche Médicale (INSERM), UMR 1037, Cancer Research Center of Toulouse, Toulouse F-31000, France
| | | | | | | | | | | |
Collapse
|
94
|
Kumar P, Bolshette NB, Jamdade VS, Mundhe NA, Thakur KK, Saikia KK, Lahkar M. Breast cancer status in India: An overview. ACTA ACUST UNITED AC 2013. [DOI: 10.1016/j.bionut.2013.03.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
95
|
Inhibition of Ephrin B3-mediated survival signaling contributes to increased cell death response of non-small cell lung carcinoma cells after combined treatment with ionizing radiation and PKC 412. Cell Death Dis 2013; 4:e454. [PMID: 23303128 PMCID: PMC3563978 DOI: 10.1038/cddis.2012.188] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Radiation therapy is frequently used to treat non-small cell lung cancers (NSCLCs). We have previously shown that a combination of ionizing radiation (IR) and the staurosporine analog PKC 412, but not Ro 31–8220, increases cell death in NSCLC cells. To identify genes involved in the enhancement of cell death, a total gene profiling in response to co-administration of (i) PKC 412 with IR, or (ii) Ro 31–8220 with IR was implemented. These combined treatments caused upregulation of 140 and 179 genes and downregulation of 253 and 425 genes, respectively. Certain genes were selected and verified by real-time quantitative PCR and, of these genes, robust suppression of Ephrin B3 expression was suggested as a possible cell death-inducing mechanism of combined treatment with IR and PKC 412. Indeed, silencing of Ephrin B3 using siRNA in NSCLC cells resulted in a major alteration of their morphology with an elongated phenotype, decreased proliferation and increased cell death signaling. Moreover, silencing of Ephrin B3 in combination with IR caused a decrease in IR-mediated G2-arrest, induced cellular senescence, inhibited MAPK ERK and p38 phosphorylation, and caused an upregulation of p27kip1 expression. Finally, silencing of Ephrin B3 in combination with IR sensitized U-1810 cells to IR-induced apoptosis. In conclusion, we identify and describe Ephrin B3 as a putative signaling molecule involved in the response of NSCLC cells to combined treatment with PKC 412 and ionizing radiation.
Collapse
|
96
|
Ahmadianpour MR, Abdolmaleki P, Mowla SJ, Hosseinkhani S. Gamma radiation alters cell cycle and induces apoptosis in p53 mutant E6.1 Jurkat cells. Appl Radiat Isot 2013; 71:29-33. [DOI: 10.1016/j.apradiso.2012.09.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Revised: 08/05/2012] [Accepted: 09/07/2012] [Indexed: 01/09/2023]
|
97
|
Therapeutic efficacy of 188Re-liposome in a C26 murine colon carcinoma solid tumor model. Invest New Drugs 2012; 31:801-11. [DOI: 10.1007/s10637-012-9906-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 11/12/2012] [Indexed: 11/26/2022]
|
98
|
Mukherjee D, Coates PJ, Rastogi S, Lorimore SA, Wright EG. Radiation-induced bone marrow apoptosis, inflammatory bystander-type signaling and tissue cytotoxicity. Int J Radiat Biol 2012; 89:139-46. [PMID: 23078404 DOI: 10.3109/09553002.2013.741280] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE A study of irradiated (0.25-2 Gy) murine bone marrow has investigated the relationships between apoptotic responses of cells exposed in vivo and in vitro and between in vivo apoptosis and tissue cytotoxicity. MATERIALS AND METHODS The time course of reduction in bone marrow cellularity in vivo was determined by femoral cell counts and apoptosis measurements obtained using three commonly used assays. Inflammatory pro-apoptotic cytokine production at 24 h post-exposure in vivo was investigated using a bystander protocol. RESULTS In vivo, there is a dose- and time-dependent non-linear reduction in bone marrow cellularity up to 24 h post- irradiation not directly represented by apoptosis measurements. The majority of cells are killed within 6 h but there is on-going cell loss in vivo up to 24 h post-irradiation in the absence of elevated levels of apoptosis and associated with the induction of cytokines produced in response to the initial tumor protein 53 (p53)-dependent apoptosis. CONCLUSION The results demonstrate that small increases in measured apoptosis can reflect significant intramedullary cell death and with apoptotic processes being responsible for pro-inflammatory mechanisms that can contribute to additional on-going cell death. The findings demonstrate the importance of studying tissue responses when considering the mechanisms underlying the consequences of radiation exposures.
Collapse
Affiliation(s)
- Debayan Mukherjee
- Centre for Oncology and Molecular Medicine University of Dundee Medical School, Dundee, Scotland, UK
| | | | | | | | | |
Collapse
|
99
|
Oakley MS, Gerald N, Anantharaman V, Gao Y, Majam V, Mahajan B, Pham PT, Lotspeich-Cole L, Myers TG, McCutchan TF, Morris SL, Aravind L, Kumar S. Radiation-induced cellular and molecular alterations in asexual intraerythrocytic Plasmodium falciparum. J Infect Dis 2012; 207:164-74. [PMID: 23100570 DOI: 10.1093/infdis/jis645] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND γ-irradiation is commonly used to create attenuation in Plasmodium parasites. However, there are no systematic studies on the survival, reversion of virulence, and molecular basis for γ-radiation-induced cell death in malaria parasites. METHODS The effect of γ-irradiation on the growth of asexual Plasmodium falciparum was studied in erythrocyte cultures. Cellular and ultrastructural changes within the parasite were studied by fluorescence and electron microscopy, and genome-wide transcriptional profiling was performed to identify parasite biomarkers of attenuation and cell death. RESULTS γ-radiation induced the death of P. falciparum in a dose-dependent manner. These parasites had defective mitosis, sparse cytoplasm, fewer ribosomes, disorganized and clumped organelles, and large vacuoles-observations consistent with "distressed" or dying parasites. A total of 185 parasite genes were transcriptionally altered in response to γ-irradiation (45.9% upregulated, 54.1% downregulated). Loss of parasite survival was correlated with the downregulation of genes encoding translation factors and with upregulation of genes associated with messenger RNA-sequestering stress granules. Genes pertaining to cell-surface interactions, host-cell remodeling, and secreted proteins were also altered. CONCLUSIONS These studies provide a framework to assess the safety of γ-irradiation attenuation and promising targets for genetic deletion to produce whole parasite-based attenuated vaccines.
Collapse
Affiliation(s)
- Miranda S Oakley
- Division of Bacterial, Center for Biologics Evaluation and Research, Food and Drug Administration, Rockville, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Abstract
The strategy of clinically targeting cancerous cells at their most vulnerable state during mitosis has instigated numerous studies into the mitotic cell death (MCD) pathway. As the hallmark of cancer revolves around cell-cycle deregulation, it is not surprising that antimitotic therapies are effective against the abnormal proliferation of transformed cells. Moreover, these antimitotic drugs are also highly selective and sensitive. Despite the robust rate of discovery and the development of mitosis-selective inhibitors, the unpredictable complexities of the human body's response to these drugs still herald the biggest challenge towards clinical success. Undoubtedly, the need to bridge the gap between promising preclinical trials and effective translational bedside treatment prompts further investigations towards mapping out the mechanistic pathways of MCD, understanding how these drugs work as medicine in the body and more comprehensive target validations. In this review, current antimitotic agents are summarized with particular emphasis on the evaluation of their clinical efficacy as well as their limitations. In addition, we discuss the basis behind the lack of activity of these inhibitors in human trials and the potential and future directions of mitotic anticancer strategies.
Collapse
|