301
|
Zhang M, Qiu Q, Li Z, Sachdeva M, Min H, Cardona DM, DeLaney TF, Han T, Ma Y, Luo L, Ilkayeva OR, Lui K, Nichols AG, Newgard CB, Kastan MB, Rathmell JC, Dewhirst MW, Kirsch DG. HIF-1 Alpha Regulates the Response of Primary Sarcomas to Radiation Therapy through a Cell Autonomous Mechanism. Radiat Res 2015; 183:594-609. [PMID: 25973951 PMCID: PMC4800000 DOI: 10.1667/rr14016.1] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Hypoxia is a major cause of radiation resistance, which may predispose to local recurrence after radiation therapy. While hypoxia increases tumor cell survival after radiation exposure because there is less oxygen to oxidize damaged DNA, it remains unclear whether signaling pathways triggered by hypoxia contribute to radiation resistance. For example, intratumoral hypoxia can increase hypoxia inducible factor 1 alpha (HIF-1α), which may regulate pathways that contribute to radiation sensitization or radiation resistance. To clarify the role of HIF-1α in regulating tumor response to radiation, we generated a novel genetically engineered mouse model of soft tissue sarcoma with an intact or deleted HIF-1α. Deletion of HIF-1α sensitized primary sarcomas to radiation exposure in vivo. Moreover, cell lines derived from primary sarcomas lacking HIF-1α, or in which HIF-1α was knocked down, had decreased clonogenic survival in vitro, demonstrating that HIF-1α can promote radiation resistance in a cell autonomous manner. In HIF-1α-intact and -deleted sarcoma cells, radiation-induced reactive oxygen species, DNA damage repair and activation of autophagy were similar. However, sarcoma cells lacking HIF-1α had impaired mitochondrial biogenesis and metabolic response after irradiation, which might contribute to radiation resistance. These results show that HIF-1α promotes radiation resistance in a cell autonomous manner.
Collapse
Affiliation(s)
- Minsi Zhang
- Department of Pharmacology and Cancer Biology, Duke University
| | - Qiong Qiu
- Department of Radiation Oncology, Duke University Medical Center
| | - Zhizhong Li
- Department of Radiation Oncology, Duke University Medical Center
| | - Mohit Sachdeva
- Department of Radiation Oncology, Duke University Medical Center
| | - Hooney Min
- Department of Radiation Oncology, Duke University Medical Center
| | | | | | - Tracy Han
- Department of Radiation Oncology, Duke University Medical Center
| | - Yan Ma
- Department of Radiation Oncology, Duke University Medical Center
| | - Lixia Luo
- Department of Radiation Oncology, Duke University Medical Center
| | - Olga R. Ilkayeva
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine
| | - Ki Lui
- Department of Pharmacology and Cancer Biology, Duke University
| | | | - Christopher B. Newgard
- Department of Pharmacology and Cancer Biology, Duke University
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine
| | - Michael B. Kastan
- Department of Pharmacology and Cancer Biology, Duke University
- Department of Pediatrics, Duke University Medical Center
| | - Jeffrey C. Rathmell
- Department of Pharmacology and Cancer Biology, Duke University
- Department of Immunology, Duke University Medical Center
| | - Mark W. Dewhirst
- Department of Radiation Oncology, Duke University Medical Center
- Department of Pathology, Duke University Medical Center
| | - David G. Kirsch
- Department of Pharmacology and Cancer Biology, Duke University
- Department of Radiation Oncology, Duke University Medical Center
| |
Collapse
|
302
|
Hatano K, Kumar B, Zhang Y, Coulter JB, Hedayati M, Mears B, Ni X, Kudrolli TA, Chowdhury WH, Rodriguez R, DeWeese TL, Lupold SE. A functional screen identifies miRNAs that inhibit DNA repair and sensitize prostate cancer cells to ionizing radiation. Nucleic Acids Res 2015; 43:4075-86. [PMID: 25845598 PMCID: PMC4417178 DOI: 10.1093/nar/gkv273] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 03/17/2015] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs (miRNAs) have been implicated in DNA repair pathways through transcriptional responses to DNA damaging agents or through predicted miRNA regulation of DNA repair genes. We hypothesized that additional DNA damage regulating miRNAs could be identified by screening a library of 810 miRNA mimetics for the ability to alter cellular sensitivity to ionizing radiation (IR). A prostate cancer Metridia luciferase cell model was applied to examine the effects of individual miRNAs on IR sensitivity. A large percentage of miRNA mimetics were found to increase cellular sensitivity to IR, while a smaller percentage were protective. Two of the most potent IR sensitizing miRNAs, miR-890 and miR-744–3p, significantly delayed IR induced DNA damage repair. Both miRNAs inhibited the expression of multiple components of DNA damage response and DNA repair. miR-890 directly targeted MAD2L2, as well as WEE1 and XPC, where miR-744–3p directly targeted RAD23B. Knock-down of individual miR-890 targets by siRNA was not sufficient to ablate miR-890 radiosensitization, signifying that miR-890 functions by regulating multiple DNA repair genes. Intratumoral delivery of miR-890 mimetics prior to IR therapy significantly enhanced IR therapeutic efficacy. These results reveal novel miRNA regulation of DNA repair and identify miR-890 as a potent IR sensitizing agent.
Collapse
Affiliation(s)
- Koji Hatano
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Binod Kumar
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yonggang Zhang
- The Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jonathan B Coulter
- The Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mohammad Hedayati
- The Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Brian Mears
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xiaohua Ni
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA Shanghai Institute of Planned Parenthood Research, National Population & Family Planning Key Laboratory of Contraceptive Drugs and Devices. 2140 Xietu Rd., Shanghai 200032, China
| | - Tarana A Kudrolli
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Wasim H Chowdhury
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA The Department of Urology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Ronald Rodriguez
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA The Department of Urology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Theodore L DeWeese
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA The Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shawn E Lupold
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA The Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
303
|
Mantovani A, Allavena P. The interaction of anticancer therapies with tumor-associated macrophages. ACTA ACUST UNITED AC 2015; 212:435-45. [PMID: 25753580 PMCID: PMC4387285 DOI: 10.1084/jem.20150295] [Citation(s) in RCA: 501] [Impact Index Per Article: 50.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 02/17/2015] [Indexed: 11/19/2022]
Abstract
Mantovani and Allavena provide a comprehensive review on the effects of conventional anticancer therapies on tumor-associated macrophages. Macrophages are essential components of the inflammatory microenvironment of tumors. Conventional treatment modalities (chemotherapy and radiotherapy), targeted drugs, antiangiogenic agents, and immunotherapy, including checkpoint blockade, all profoundly influence or depend on the function of tumor-associated macrophages (TAMs). Chemotherapy and radiotherapy can have dual influences on TAMs in that a misdirected macrophage-orchestrated tissue repair response can result in chemoresistance, but in other circumstances, TAMs are essential for effective therapy. A better understanding of the interaction of anticancer therapies with innate immunity, and TAMs in particular, may pave the way to better patient selection and innovative combinations of conventional approaches with immunotherapy.
Collapse
Affiliation(s)
- Alberto Mantovani
- A. Mantovani and P. Allavena are at the IRCCS Humanitas Clinical and Research Center and Humanitas University, 20089 Rozzano, Milano, Italy
| | - Paola Allavena
- A. Mantovani and P. Allavena are at the IRCCS Humanitas Clinical and Research Center and Humanitas University, 20089 Rozzano, Milano, Italy
| |
Collapse
|
304
|
Leiker AJ, DeGraff W, Choudhuri R, Sowers AL, Thetford A, Cook JA, Van Waes C, Mitchell JB. Radiation Enhancement of Head and Neck Squamous Cell Carcinoma by the Dual PI3K/mTOR Inhibitor PF-05212384. Clin Cancer Res 2015; 21:2792-801. [PMID: 25724523 DOI: 10.1158/1078-0432.ccr-14-3279] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 02/23/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Radiation remains a mainstay for the treatment of nonmetastatic head and neck squamous cell carcinoma (HNSCC), a malignancy characterized by a high rate of PI3K/mTOR signaling axis activation. We investigated the ATP-competitive dual PI3K/mTOR inhibitor, PF-05212384, as a radiosensitizer in preclinical HNSCC models. EXPERIMENTAL DESIGN Extent of radiation enhancement of two HNSCC cell lines (UMSCC1-wtP53 and UMSCC46-mtP53) and normal human fibroblast (1522) was assessed by in vitro clonogenic assay with appropriate target inhibition verified by immunoblotting. Radiation-induced DNA damage repair was evaluated by γH2AX Western blots with the mechanism of DNA double-strand break repair abrogation investigated by cell cycle analysis, immunoblotting, and RT-PCR. PF-05212384 efficacy in vivo was assessed by UMSCC1 xenograft tumor regrowth delay, xenograft lysate immunoblotting, and tissue section immunohistochemistry. RESULTS PF-05212384 effectively inhibited PI3K and mTOR, resulting in significant radiosensitization of exponentially growing and plateau-phase cells with 24-hour treatment following irradiation, and variable radiation enhancement with 24-hour treatment before irradiation. Tumor cells radiosensitized to a greater extent than normal human fibroblasts. Postirradiation PF-05212384 treatment delays γH2AX foci resolution. PF-05212384 24-hour exposure resulted in an evident G1-S phase block in p53-competent cells. Fractionated radiation plus i.v. PF-05212384 synergistically delayed nude mice bearing UMSCC1 xenograft regrowth, with potential drug efficacy biomarkers identified, including pS6, pAkt, p4EBP1, and Ki67. CONCLUSIONS Taken together, our results of significant radiosensitization both in vitro and in vivo validate the PI3K/mTOR axis as a radiation modification target and PF-05212384 as a potential clinical radiation modifier of nonmetastatic HNSCC.
Collapse
Affiliation(s)
- Andrew J Leiker
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland. Medical Research Scholars Program, NIH, Bethesda, Maryland
| | - William DeGraff
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Rajani Choudhuri
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Anastasia L Sowers
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Angela Thetford
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - John A Cook
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Carter Van Waes
- Head and Neck Surgery Branch, NIDCD, NIH, Bethesda, Maryland
| | - James B Mitchell
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland.
| |
Collapse
|
305
|
Hodzic J, Dingjan I, Maas MJ, van der Meulen-Muileman IH, de Menezes RX, Heukelom S, Verheij M, Gerritsen WR, Geldof AA, van Triest B, van Beusechem VW. A cell-based high-throughput screening assay for radiation susceptibility using automated cell counting. Radiat Oncol 2015; 10:55. [PMID: 25888875 PMCID: PMC4355372 DOI: 10.1186/s13014-015-0355-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 02/13/2015] [Indexed: 01/04/2023] Open
Abstract
Background Radiotherapy is one of the mainstays in the treatment for cancer, but its success can be limited due to inherent or acquired resistance. Mechanisms underlying radioresistance in various cancers are poorly understood and available radiosensitizers have shown only modest clinical benefit. There is thus a need to identify new targets and drugs for more effective sensitization of cancer cells to irradiation. Compound and RNA interference high-throughput screening technologies allow comprehensive enterprises to identify new agents and targets for radiosensitization. However, the gold standard assay to investigate radiosensitivity of cancer cells in vitro, the colony formation assay (CFA), is unsuitable for high-throughput screening. Methods We developed a new high-throughput screening method for determining radiation susceptibility. Fast and uniform irradiation of batches up to 30 microplates was achieved using a Perspex container and a clinically employed linear accelerator. The readout was done by automated counting of fluorescently stained nuclei using the Acumen eX3 laser scanning cytometer. Assay performance was compared to that of the CFA and the CellTiter-Blue homogeneous uniform-well cell viability assay. The assay was validated in a whole-genome siRNA library screening setting using PC-3 prostate cancer cells. Results On 4 different cancer cell lines, the automated cell counting assay produced radiation dose response curves that followed a linear-quadratic equation and that exhibited a better correlation to the results of the CFA than did the cell viability assay. Moreover, the cell counting assay could be used to detect radiosensitization by silencing DNA-PKcs or by adding caffeine. In a high-throughput screening setting, using 4 Gy irradiated and control PC-3 cells, the effects of DNA-PKcs siRNA and non-targeting control siRNA could be clearly discriminated. Conclusions We developed a simple assay for radiation susceptibility that can be used for high-throughput screening. This will aid the identification of molecular targets for radiosensitization, thereby contributing to improving the efficacy of radiotherapy.
Collapse
Affiliation(s)
- Jasmina Hodzic
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1118, 1081HV, Amsterdam, The Netherlands.
| | - Ilse Dingjan
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1118, 1081HV, Amsterdam, The Netherlands.
| | - Mariëlle Jp Maas
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1118, 1081HV, Amsterdam, The Netherlands.
| | - Ida H van der Meulen-Muileman
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1118, 1081HV, Amsterdam, The Netherlands.
| | - Renee X de Menezes
- Department of Epidemiology and Biostatistics, VU University Medical Center, De Boelelaan 1118, 1081HV, Amsterdam, The Netherlands.
| | - Stan Heukelom
- Department of Radiology & Nuclear Medicine, VU University Medical Center, De Boelelaan 1118, 1081HV, Amsterdam, The Netherlands.
| | - Marcel Verheij
- Department of Radiotherapy, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
| | - Winald R Gerritsen
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1118, 1081HV, Amsterdam, The Netherlands. .,Present address: Department of Medical Oncology, The Radboud University Medical Center, Comeniuslaan 4, 6525 HP, Nijmegen, The Netherlands.
| | - Albert A Geldof
- Department of Radiology & Nuclear Medicine, VU University Medical Center, De Boelelaan 1118, 1081HV, Amsterdam, The Netherlands. .,Department of Urology, VU University Medical Center, De Boelelaan 1118, 1081HV, Amsterdam, The Netherlands.
| | - Baukelien van Triest
- Department of Radiotherapy, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
| | - Victor W van Beusechem
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1118, 1081HV, Amsterdam, The Netherlands.
| |
Collapse
|
306
|
McQuade C, Zaki AA, Desai Y, Vido M, Sakhuja T, Cheng Z, Hickey RJ, Joh D, Park SJ, Kao G, Dorsey JF, Tsourkas A. A multifunctional nanoplatform for imaging, radiotherapy, and the prediction of therapeutic response. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2015; 11:834-43. [PMID: 25264301 PMCID: PMC4329028 DOI: 10.1002/smll.201401927] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 08/22/2014] [Indexed: 05/26/2023]
Abstract
Gold nanoparticles have garnered interest as both radiosensitzers and computed tomography (CT) contrast agents. However, the extremely high concentrations of gold required to generate CT contrast is far beyond that needed for meaningful radiosensitization, which limits their use as combined therapeutic-diagnostic (theranostic) agents. To establish a theranostic nanoplatform with well-aligned radiotherapeutic and diagnostic properties for better integration into standard radiation therapy practice, a gold- and superparamagnetic iron oxide nanoparticle (SPION)-loaded micelle (GSM) is developed. Intravenous injection of GSMs into tumor-bearing mice led to selective tumoral accumulation, enabling magnetic resonance (MR) imaging of tumor margins. Subsequent irradiation leads to a 90-day survival of 71% in GSM-treated mice, compared with 25% for irradiation-only mice. Furthermore, measurements of the GSM-enhanced MR contrast are highly predictive of tumor response. Therefore, GSMs may not only guide and enhance the efficacy of radiation therapy, but may allow patients to be managed more effectively.
Collapse
Affiliation(s)
- Casey McQuade
- Department of Bioengineering, University of Pennsylvania, 210 S. 33 Street, 240 Skirkanich Hall, Philadelphia, PA 19104, USA
| | - Ajlan Al Zaki
- Department of Bioengineering, University of Pennsylvania, 210 S. 33 Street, 240 Skirkanich Hall, Philadelphia, PA 19104, USA
| | - Yaanik Desai
- Department of Bioengineering, University of Pennsylvania, 210 S. 33 Street, 240 Skirkanich Hall, Philadelphia, PA 19104, USA
| | - Michael Vido
- Department of Bioengineering, University of Pennsylvania, 210 S. 33 Street, 240 Skirkanich Hall, Philadelphia, PA 19104, USA
| | - Timothy Sakhuja
- Department of Bioengineering, University of Pennsylvania, 210 S. 33 Street, 240 Skirkanich Hall, Philadelphia, PA 19104, USA
| | - Zhiliang Cheng
- Department of Bioengineering, University of Pennsylvania, 210 S. 33 Street, 240 Skirkanich Hall, Philadelphia, PA 19104, USA
| | - Robert J. Hickey
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel Joh
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - So-Jung Park
- Department of Chemistry and Nano Science, Global Top 5 Program, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 120-750, Korea
| | - Gary Kao
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jay F. Dorsey
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrew Tsourkas
- Department of Bioengineering, University of Pennsylvania, 210 S. 33 Street, 240 Skirkanich Hall, Philadelphia, PA 19104, USA
| |
Collapse
|
307
|
Buckel L, Savariar EN, Crisp JL, Jones KA, Hicks AM, Scanderbeg DJ, Nguyen QT, Sicklick JK, Lowy AM, Tsien RY, Advani SJ. Tumor radiosensitization by monomethyl auristatin E: mechanism of action and targeted delivery. Cancer Res 2015; 75:1376-1387. [PMID: 25681274 DOI: 10.1158/0008-5472.can-14-1931] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 02/04/2015] [Indexed: 12/19/2022]
Abstract
Intrinsic tumor resistance to radiotherapy limits the efficacy of ionizing radiation (IR). Sensitizing cancer cells specifically to IR would improve tumor control and decrease normal tissue toxicity. The development of tumor-targeting technologies allows for developing potent radiosensitizing drugs. We hypothesized that the anti-tubulin agent monomethyl auristatin E (MMAE), a component of a clinically approved antibody-directed conjugate, could function as a potent radiosensitizer and be selectively delivered to tumors using an activatable cell-penetrating peptide targeting matrix metalloproteinases and RGD-binding integrins (ACPP-cRGD-MMAE). We evaluated the ability of MMAE to radiosensitize both established cancer cells and a low-passage cultured human pancreatic tumor cell line using clonogenic and DNA damage assays. MMAE sensitized colorectal and pancreatic cancer cells to IR in a schedule- and dose-dependent manner, correlating with mitotic arrest. Radiosensitization was evidenced by decreased clonogenic survival and increased DNA double-strand breaks in irradiated cells treated with MMAE. MMAE in combination with IR resulted in increased DNA damage signaling and activation of CHK1. To test a therapeutic strategy of MMAE and IR, PANC-1 or HCT-116 murine tumor xenografts were treated with nontargeted free MMAE or tumor-targeted MMAE (ACPP-cRGD-MMAE). While free MMAE in combination with IR resulted in tumor growth delay, tumor-targeted ACPP-cRGD-MMAE with IR produced a more robust and significantly prolonged tumor regression in xenograft models. Our studies identify MMAE as a potent radiosensitizer. Importantly, MMAE radiosensitization can be localized to tumors by targeted activatable cell-penetrating peptides.
Collapse
Affiliation(s)
- Lisa Buckel
- Department of Radiation Medicine and Applied Sciences
| | | | | | | | - Angel M Hicks
- Department of Radiation Medicine and Applied Sciences
| | | | | | | | | | - Roger Y Tsien
- Department of Pharmacology.,Howard Hughes Medical Institute
| | - Sunil J Advani
- Department of Radiation Medicine and Applied Sciences.,Center for Advanced Radiotherapy Technologies University of California San Diego
| |
Collapse
|
308
|
CHO HYUNJI, AHN KWANGCHUL, CHOI JAEYEON, HWANG SANGGU, KIM WUNJAE, UM HONGDUCK, PARK JONGKUK. Luteolin acts as a radiosensitizer in non-small cell lung cancer cells by enhancing apoptotic cell death through activation of a p38/ROS/caspase cascade. Int J Oncol 2015; 46:1149-58. [DOI: 10.3892/ijo.2015.2831] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 12/09/2014] [Indexed: 11/06/2022] Open
|
309
|
Deng Z, Yu L, Cao W, Zheng W, Chen T. A selenium-containing ruthenium complex as a cancer radiosensitizer, rational design and the important role of ROS-mediated signalling. Chem Commun (Camb) 2015; 51:2637-40. [DOI: 10.1039/c4cc07926d] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
We have described the rational design of selenium-containing ruthenium complexes and their use as cancer radiosensitizers through regulating ROS-mediated pathways.
Collapse
Affiliation(s)
- Zhiqin Deng
- Department of Chemistry
- Jinan University
- Guangzhou 510632
- China
| | - Lianling Yu
- Department of Chemistry
- Jinan University
- Guangzhou 510632
- China
| | - Wenqiang Cao
- Department of Chemistry
- Jinan University
- Guangzhou 510632
- China
| | - Wenjie Zheng
- Department of Chemistry
- Jinan University
- Guangzhou 510632
- China
| | - Tianfeng Chen
- Department of Chemistry
- Jinan University
- Guangzhou 510632
- China
| |
Collapse
|
310
|
Abstract
Deletion of prolyl hydroxylase domain proteins or overexpression of hypoxia-inducible factor 2α (HIF2α) in the gastrointestinal epithelium improves survival of mice after abdominal irradiation (Taniguchi et al., this issue).
Collapse
Affiliation(s)
- Chang-Lung Lee
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | - Everett J Moding
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - David G Kirsch
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA. Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
311
|
Korpela E, Liu SK. Endothelial perturbations and therapeutic strategies in normal tissue radiation damage. Radiat Oncol 2014; 9:266. [PMID: 25518850 PMCID: PMC4279961 DOI: 10.1186/s13014-014-0266-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 11/18/2014] [Indexed: 02/08/2023] Open
Abstract
Most cancer patients are treated with radiotherapy, but the treatment can also damage the surrounding normal tissue. Radiotherapy side-effects diminish patients’ quality of life, yet effective biological interventions for normal tissue damage are lacking. Protecting microvascular endothelial cells from the effects of irradiation is emerging as a targeted damage-reduction strategy. We illustrate the concept of the microvasculature as a mediator of overall normal tissue radiation toxicity through cell death, vascular inflammation (hemodynamic and molecular changes) and a change in functional capacity. Endothelial cell targeted therapies that protect against such endothelial cell perturbations and the development of acute normal tissue damage are mostly under preclinical development. Since acute radiation toxicity is a common clinical problem in cutaneous, gastrointestinal and mucosal tissues, we also focus on damage in these tissues.
Collapse
Affiliation(s)
- Elina Korpela
- Biological Sciences, Sunnybrook Research Institute and Odette Cancer Centre, Sunnybrook Health Sciences Centre, 2075 Bayview Ave., Toronto, M4N 3M5, Canada. .,Department of Medical Biophysics, University of Toronto, 101 College St., Toronto, M5G 1L7, Canada.
| | - Stanley K Liu
- Biological Sciences, Sunnybrook Research Institute and Odette Cancer Centre, Sunnybrook Health Sciences Centre, 2075 Bayview Ave., Toronto, M4N 3M5, Canada. .,Department of Medical Biophysics, University of Toronto, 101 College St., Toronto, M5G 1L7, Canada. .,Department of Radiation Oncology, University of Toronto, 149 College St., Toronto, M5T 1P5, Canada.
| |
Collapse
|
312
|
De Ruysscher D, Faivre-Finn C, Le Pechoux C, Peeters S, Belderbos J. High-dose re-irradiation following radical radiotherapy for non-small-cell lung cancer. Lancet Oncol 2014; 15:e620-e624. [PMID: 25456380 DOI: 10.1016/s1470-2045(14)70345-6] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
As the prognosis of lung cancer patients improves, more patients are at risk of developing local recurrence or a new primary tumour in previously irradiated areas. Technological advances in radiotherapy and imaging have made treatment of patients with high-dose re-irradiation possible, with the aim of long-term disease-free survival and even cure. However, high-dose re-irradiation with overlapping volumes of previously irradiated tissues is not without risks. Late, irreversible, and potentially serious normal tissue damage may occur because of injury to surrounding thoracic structures and organs at risk. In this Review, we aimed to report the efficacy and toxic effects of high-dose re-irradiation for locoregional recurrent non-small-cell lung cancer. Our findings indicate that high-dose re-irradiation might be beneficial in selected patients; however, patients and physicians should be aware of the scarcity of high-quality data when considering this treatment.
Collapse
Affiliation(s)
- Dirk De Ruysscher
- Radiation Oncology, University Hospitals Leuven/KU Leuven, Leuven, Belgium.
| | - Corinne Faivre-Finn
- Radiation Related Research, The Christie NHS Foundation Trust and University of Manchester, Manchester, UK
| | - Cecile Le Pechoux
- Department of Radiation Oncology, Institut Gustave Roussy, Villejuif, France
| | - Stéphanie Peeters
- Radiation Oncology, University Hospitals Leuven/KU Leuven, Leuven, Belgium
| | - José Belderbos
- Department of Radiation Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| |
Collapse
|
313
|
Baskar R, Dai J, Wenlong N, Yeo R, Yeoh KW. Biological response of cancer cells to radiation treatment. Front Mol Biosci 2014; 1:24. [PMID: 25988165 PMCID: PMC4429645 DOI: 10.3389/fmolb.2014.00024] [Citation(s) in RCA: 388] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 10/31/2014] [Indexed: 12/15/2022] Open
Abstract
Cancer is a class of diseases characterized by uncontrolled cell growth and has the ability to spread or metastasize throughout the body. In recent years, remarkable progress has been made toward the understanding of proposed hallmarks of cancer development, care, and treatment modalities. Radiation therapy or radiotherapy is an important and integral component of cancer management, mostly conferring a survival benefit. Radiation therapy destroys cancer by depositing high-energy radiation on the cancer tissues. Over the years, radiation therapy has been driven by constant technological advances and approximately 50% of all patients with localized malignant tumors are treated with radiation at some point in the course of their disease. In radiation oncology, research and development in the last three decades has led to considerable improvement in our understanding of the differential responses of normal and cancer cells. The biological effectiveness of radiation depends on the linear energy transfer (LET), total dose, number of fractions and radiosensitivity of the targeted cells or tissues. Radiation can either directly or indirectly (by producing free radicals) damages the genome of the cell. This has been challenged in recent years by a newly identified phenomenon known as radiation induced bystander effect (RIBE). In RIBE, the non-irradiated cells adjacent to or located far from the irradiated cells/tissues demonstrate similar responses to that of the directly irradiated cells. Understanding the cancer cell responses during the fractions or after the course of irradiation will lead to improvements in therapeutic efficacy and potentially, benefitting a significant proportion of cancer patients. In this review, the clinical implications of radiation induced direct and bystander effects on the cancer cell are discussed.
Collapse
Affiliation(s)
- Rajamanickam Baskar
- Division of Cellular and Molecular Research, Department of Radiation Oncology, National Cancer Centre Singapore, Singapore
| | - Jiawen Dai
- Division of Cellular and Molecular Research, Department of Radiation Oncology, National Cancer Centre Singapore, Singapore
| | - Nei Wenlong
- Division of Cellular and Molecular Research, Department of Radiation Oncology, National Cancer Centre Singapore, Singapore
| | - Richard Yeo
- Division of Cellular and Molecular Research, Department of Radiation Oncology, National Cancer Centre Singapore, Singapore
| | - Kheng-Wei Yeoh
- Division of Cellular and Molecular Research, Department of Radiation Oncology, National Cancer Centre Singapore, Singapore
| |
Collapse
|
314
|
Pettersen EO, Ebbesen P, Gieling RG, Williams KJ, Dubois L, Lambin P, Ward C, Meehan J, Kunkler IH, Langdon SP, Ree AH, Flatmark K, Lyng H, Calzada MJ, Peso LD, Landazuri MO, Görlach A, Flamm H, Kieninger J, Urban G, Weltin A, Singleton DC, Haider S, Buffa FM, Harris AL, Scozzafava A, Supuran CT, Moser I, Jobst G, Busk M, Toustrup K, Overgaard J, Alsner J, Pouyssegur J, Chiche J, Mazure N, Marchiq I, Parks S, Ahmed A, Ashcroft M, Pastorekova S, Cao Y, Rouschop KM, Wouters BG, Koritzinsky M, Mujcic H, Cojocari D. Targeting tumour hypoxia to prevent cancer metastasis. From biology, biosensing and technology to drug development: the METOXIA consortium. J Enzyme Inhib Med Chem 2014; 30:689-721. [PMID: 25347767 DOI: 10.3109/14756366.2014.966704] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 09/15/2014] [Indexed: 01/06/2023] Open
Abstract
The hypoxic areas of solid cancers represent a negative prognostic factor irrespective of which treatment modality is chosen for the patient. Still, after almost 80 years of focus on the problems created by hypoxia in solid tumours, we still largely lack methods to deal efficiently with these treatment-resistant cells. The consequences of this lack may be serious for many patients: Not only is there a negative correlation between the hypoxic fraction in tumours and the outcome of radiotherapy as well as many types of chemotherapy, a correlation has been shown between the hypoxic fraction in tumours and cancer metastasis. Thus, on a fundamental basis the great variety of problems related to hypoxia in cancer treatment has to do with the broad range of functions oxygen (and lack of oxygen) have in cells and tissues. Therefore, activation-deactivation of oxygen-regulated cascades related to metabolism or external signalling are important areas for the identification of mechanisms as potential targets for hypoxia-specific treatment. Also the chemistry related to reactive oxygen radicals (ROS) and the biological handling of ROS are part of the problem complex. The problem is further complicated by the great variety in oxygen concentrations found in tissues. For tumour hypoxia to be used as a marker for individualisation of treatment there is a need for non-invasive methods to measure oxygen routinely in patient tumours. A large-scale collaborative EU-financed project 2009-2014 denoted METOXIA has studied all the mentioned aspects of hypoxia with the aim of selecting potential targets for new hypoxia-specific therapy and develop the first stage of tests for this therapy. A new non-invasive PET-imaging method based on the 2-nitroimidazole [(18)F]-HX4 was found to be promising in a clinical trial on NSCLC patients. New preclinical models for testing of the metastatic potential of cells were developed, both in vitro (2D as well as 3D models) and in mice (orthotopic grafting). Low density quantitative real-time polymerase chain reaction (qPCR)-based assays were developed measuring multiple hypoxia-responsive markers in parallel to identify tumour hypoxia-related patterns of gene expression. As possible targets for new therapy two main regulatory cascades were prioritised: The hypoxia-inducible-factor (HIF)-regulated cascades operating at moderate to weak hypoxia (<1% O(2)), and the unfolded protein response (UPR) activated by endoplasmatic reticulum (ER) stress and operating at more severe hypoxia (<0.2%). The prioritised targets were the HIF-regulated proteins carbonic anhydrase IX (CAIX), the lactate transporter MCT4 and the PERK/eIF2α/ATF4-arm of the UPR. The METOXIA project has developed patented compounds targeting CAIX with a preclinical documented effect. Since hypoxia-specific treatments alone are not curative they will have to be combined with traditional anti-cancer therapy to eradicate the aerobic cancer cell population as well.
Collapse
|
315
|
Wong P, Houghton P, Kirsch DG, Finkelstein SE, Monjazeb AM, Xu-Welliver M, Dicker AP, Ahmed M, Vikram B, Teicher BA, Coleman CN, Machtay M, Curran WJ, Wang D. Combining targeted agents with modern radiotherapy in soft tissue sarcomas. J Natl Cancer Inst 2014; 106:dju329. [PMID: 25326640 DOI: 10.1093/jnci/dju329] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Improved understanding of soft-tissue sarcoma (STS) biology has led to better distinction and subtyping of these diseases with the hope of exploiting the molecular characteristics of each subtype to develop appropriately targeted treatment regimens. In the care of patients with extremity STS, adjunctive radiation therapy (RT) is used to facilitate limb and function, preserving surgeries while maintaining five-year local control above 85%. In contrast, for STS originating from nonextremity anatomical sites, the rate of local recurrence is much higher (five-year local control is approximately 50%) and a major cause of death and morbidity in these patients. Incorporating novel technological advancements to administer accurate RT in combination with novel radiosensitizing agents could potentially improve local control and overall survival. RT efficacy in STS can be increased by modulating biological pathways such as angiogenesis, cell cycle regulation, cell survival signaling, and cancer-host immune interactions. Previous experiences, advancements, ongoing research, and current clinical trials combining RT with agents modulating one or more of the above pathways are reviewed. The standard clinical management of patients with STS with pretreatment biopsy, neoadjuvant treatment, and primary surgery provides an opportune disease model for interrogating translational hypotheses. The purpose of this review is to outline a strategic vision for clinical translation of preclinical findings and to identify appropriate targeted agents to combine with radiotherapy in the treatment of STS from different sites and/or different histology subtypes.
Collapse
Affiliation(s)
- Philip Wong
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - Peter Houghton
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - David G Kirsch
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - Steven E Finkelstein
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - Arta M Monjazeb
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - Meng Xu-Welliver
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - Adam P Dicker
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - Mansoor Ahmed
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - Bhadrasain Vikram
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - Beverly A Teicher
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - C Norman Coleman
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - Mitchell Machtay
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - Walter J Curran
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - Dian Wang
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW).
| |
Collapse
|
316
|
Coleman CN, Lawrence TS, Kirsch DG. Enhancing the efficacy of radiation therapy: premises, promises, and practicality. J Clin Oncol 2014; 32:2832-5. [PMID: 25113766 PMCID: PMC4152710 DOI: 10.1200/jco.2014.57.3865] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
317
|
Shin JS, Tut TG, Ho V, Lee CS. Predictive markers of radiotherapy-induced rectal cancer regression. J Clin Pathol 2014; 67:859-64. [DOI: 10.1136/jclinpath-2014-202494] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
318
|
Moding EJ, Lee CL, Castle KD, Oh P, Mao L, Zha S, Min HD, Ma Y, Das S, Kirsch DG. Atm deletion with dual recombinase technology preferentially radiosensitizes tumor endothelium. J Clin Invest 2014; 124:3325-38. [PMID: 25036710 DOI: 10.1172/jci73932] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 05/15/2014] [Indexed: 02/06/2023] Open
Abstract
Cells isolated from patients with ataxia telangiectasia are exquisitely sensitive to ionizing radiation. Kinase inhibitors of ATM, the gene mutated in ataxia telangiectasia, can sensitize tumor cells to radiation therapy, but concern that inhibiting ATM in normal tissues will also increase normal tissue toxicity from radiation has limited their clinical application. Endothelial cell damage can contribute to the development of long-term side effects after radiation therapy, but the role of endothelial cell death in tumor response to radiation therapy remains controversial. Here, we developed dual recombinase technology using both FlpO and Cre recombinases to generate primary sarcomas in mice with endothelial cell-specific deletion of Atm to determine whether loss of Atm in endothelial cells sensitizes tumors and normal tissues to radiation. Although deletion of Atm in proliferating tumor endothelial cells enhanced the response of sarcomas to radiation, Atm deletion in quiescent endothelial cells of the heart did not sensitize mice to radiation-induced myocardial necrosis. Blocking cell cycle progression reversed the effect of Atm loss on tumor endothelial cell radiosensitivity. These results indicate that endothelial cells must progress through the cell cycle in order to be radiosensitized by Atm deletion.
Collapse
|
319
|
Nieder C, Andratschke NH, Grosu AL. Normal tissue studies in radiation oncology: A systematic review of highly cited articles and citation patterns. Oncol Lett 2014; 8:972-976. [PMID: 25120644 PMCID: PMC4114634 DOI: 10.3892/ol.2014.2252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Accepted: 05/15/2014] [Indexed: 11/25/2022] Open
Abstract
Radiation therapy is one of the cornerstones of modern multidisciplinary cancer treatment. Normal tissue tolerance is critical as radiation-induced side effects may compromise organ function and quality of life. The importance of normal tissue research is reflected by the large number of scientific articles, which have been published between 2006 and 2010. The present study identified important areas of research as well as seminal publications. The article citation rate is among the potential indicators of scientific impact. Highly cited articles, arbitrarily defined as those with ≥15 citations, were identified via a systematic search of the citation database, Scopus. Up to 608 articles per year were published between 2006 and 2010, however, <10% of publications in each year accumulated ≥15 citations. This figure is notably low, when compared with other oncology studies. A large variety of preclinical and clinical topics, including toxicity prediction, the dose-volume relationship and radioprotectors, accumulated ≥15 citations. However, clinical prevention or mitigation studies were underrepresented. The following conclusion may be drawn from the present study; despite the improved technology that has resulted in superior dose distribution, clinical prevention or mitigation studies are critical and must receive higher priority, funding and attention.
Collapse
Affiliation(s)
- Carsten Nieder
- Department of Oncology and Palliative Medicine, Nordland Hospital, Bodø 8092, Norway ; Institute of Clinical Medicine, Faculty of Health Sciences, University of Tromsø, Tromsø 9038, Norway
| | | | - Anca L Grosu
- Department of Radiation Oncology, University Hospital Freiburg, Freiburg D-79106, Germany
| |
Collapse
|
320
|
Weigel C, Schmezer P, Plass C, Popanda O. Epigenetics in radiation-induced fibrosis. Oncogene 2014; 34:2145-55. [PMID: 24909163 DOI: 10.1038/onc.2014.145] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 04/17/2014] [Accepted: 04/23/2014] [Indexed: 02/06/2023]
Abstract
Radiotherapy is a major cancer treatment option but dose-limiting side effects such as late-onset fibrosis in the irradiated tissue severely impair quality of life in cancer survivors. Efforts to explain radiation-induced fibrosis, for example, by genetic variation remained largely inconclusive. Recently published molecular analyses on radiation response and fibrogenesis showed a prominent role of epigenetic gene regulation. This review summarizes the current knowledge on epigenetic modifications in fibrotic disease and radiation response, and it points out the important role for epigenetic mechanisms such as DNA methylation, microRNAs and histone modifications in the development of this disease. The synopsis illustrates the complexity of radiation-induced fibrosis and reveals the need for investigations to further unravel its molecular mechanisms. Importantly, epigenetic changes are long-term determinants of gene expression and can therefore support those mechanisms that induce and perpetuate fibrogenesis even in the absence of the initial damaging stimulus. Future work must comprise the interconnection of acute radiation response and long-lasting epigenetic effects in order to assess their role in late-onset radiation fibrosis. An improved understanding of the underlying biology is fundamental to better comprehend the origin of this disease and to improve both preventive and therapeutic strategies.
Collapse
Affiliation(s)
- C Weigel
- Department of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - P Schmezer
- Department of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - C Plass
- Department of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - O Popanda
- Department of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
321
|
Gollavelli G, Ling YC. Magnetic and fluorescent graphene for dual modal imaging and single light induced photothermal and photodynamic therapy of cancer cells. Biomaterials 2014; 35:4499-507. [DOI: 10.1016/j.biomaterials.2014.02.011] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 02/07/2014] [Indexed: 12/23/2022]
|
322
|
Pawar SA, Shao L, Chang J, Wang W, Pathak R, Zhu X, Wang J, Hendrickson H, Boerma M, Sterneck E, Zhou D, Hauer-Jensen M. C/EBPδ deficiency sensitizes mice to ionizing radiation-induced hematopoietic and intestinal injury. PLoS One 2014; 9:e94967. [PMID: 24747529 PMCID: PMC3991713 DOI: 10.1371/journal.pone.0094967] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 03/21/2014] [Indexed: 12/20/2022] Open
Abstract
Knowledge of the mechanisms involved in the radiation response is critical for developing interventions to mitigate radiation-induced injury to normal tissues. Exposure to radiation leads to increased oxidative stress, DNA-damage, genomic instability and inflammation. The transcription factor CCAAT/enhancer binding protein delta (Cebpd; C/EBPδ is implicated in regulation of these same processes, but its role in radiation response is not known. We investigated the role of C/EBPδ in radiation-induced hematopoietic and intestinal injury using a Cebpd knockout mouse model. Cebpd−/− mice showed increased lethality at 7.4 and 8.5 Gy total-body irradiation (TBI), compared to Cebpd+/+ mice. Two weeks after a 6 Gy dose of TBI, Cebpd−/− mice showed decreased recovery of white blood cells, neutrophils, platelets, myeloid cells and bone marrow mononuclear cells, decreased colony-forming ability of bone marrow progenitor cells, and increased apoptosis of hematopoietic progenitor and stem cells compared to Cebpd+/+ controls. Cebpd−/− mice exhibited a significant dose-dependent decrease in intestinal crypt survival and in plasma citrulline levels compared to Cebpd+/+ mice after exposure to radiation. This was accompanied by significantly decreased expression of γ-H2AX in Cebpd−/− intestinal crypts and villi at 1 h post-TBI, increased mitotic index at 24 h post-TBI, and increase in apoptosis in intestinal crypts and stromal cells of Cebpd−/− compared to Cebpd+/+ mice at 4 h post-irradiation. This study uncovers a novel biological function for C/EBPδ in promoting the response to radiation-induced DNA-damage and in protecting hematopoietic and intestinal tissues from radiation-induced injury.
Collapse
Affiliation(s)
- Snehalata A. Pawar
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- * E-mail:
| | - Lijian Shao
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Jianhui Chang
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Wenze Wang
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Rupak Pathak
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Xiaoyan Zhu
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Junru Wang
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Howard Hendrickson
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Marjan Boerma
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Esta Sterneck
- Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
| | - Daohong Zhou
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Martin Hauer-Jensen
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- Surgical Service, Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, United States of America
| |
Collapse
|
323
|
Novel nitroimidazole alkylsulfonamides as hypoxic cell radiosensitisers. Bioorg Med Chem 2014; 22:2123-32. [PMID: 24650701 DOI: 10.1016/j.bmc.2014.02.039] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 02/07/2014] [Accepted: 02/18/2014] [Indexed: 12/31/2022]
Abstract
A novel class of nitroimidazole alkylsulfonamides have been prepared and evaluated as hypoxia-selective cytotoxins and radiosensitisers. The sulfonamide side chain markedly influences the physicochemical properties of the analogues: lowering aqueous solubility and raising the electron affinity of the nitroimidazole group. The addition of hydroxyl or basic amine groups increased aqueous solubility, with charged amine groups contributing to increased electron affinity. The analogues covered the range of electron affinity for effective radiosensitisation with one-electron reduction potentials ranging from -503 to -342mV. Cytotoxicity under normoxia or anoxia against a panel of human tumour cell lines was determined using a proliferation assay. 2-Nitroimidazole sulfonamides displayed significant hypoxia-selective cytotoxicity (6 to 64-fold), while 4- and 5-nitroimidazole analogues did not display hypoxia-selective cytotoxicity. All analogues sensitised anoxic HCT-116 human colorectal cells to radiation at non-toxic concentrations. 2-Nitroimidazole analogues provided modest sensitisation due to the relatively low concentrations used while several 5-nitroimidazole analogues provided equivalent sensitisation to misonidazole and etanidazole at similar molar concentrations.
Collapse
|
324
|
Hematulin A, Ingkaninan K, Limpeanchob N, Sagan D. Ethanolic Extract from Derris scandens Benth Mediates Radiosensitzation via Two Distinct Modes of Cell Death in Human Colon Cancer HT-29 Cells. Asian Pac J Cancer Prev 2014; 15:1871-7. [DOI: 10.7314/apjcp.2014.15.4.1871] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
325
|
López Alfonso JC, Jagiella N, Núñez L, Herrero MA, Drasdo D. Estimating dose painting effects in radiotherapy: a mathematical model. PLoS One 2014; 9:e89380. [PMID: 24586734 PMCID: PMC3935877 DOI: 10.1371/journal.pone.0089380] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Accepted: 01/20/2014] [Indexed: 12/25/2022] Open
Abstract
Tumor heterogeneity is widely considered to be a determinant factor in tumor progression and in particular in its recurrence after therapy. Unfortunately, current medical techniques are unable to deduce clinically relevant information about tumor heterogeneity by means of non-invasive methods. As a consequence, when radiotherapy is used as a treatment of choice, radiation dosimetries are prescribed under the assumption that the malignancy targeted is of a homogeneous nature. In this work we discuss the effects of different radiation dose distributions on heterogeneous tumors by means of an individual cell-based model. To that end, a case is considered where two tumor cell phenotypes are present, which we assume to strongly differ in their respective cell cycle duration and radiosensitivity properties. We show herein that, as a result of such differences, the spatial distribution of the corresponding phenotypes, whence the resulting tumor heterogeneity can be predicted as growth proceeds. In particular, we show that if we start from a situation where a majority of ordinary cancer cells (CCs) and a minority of cancer stem cells (CSCs) are randomly distributed, and we assume that the length of CSC cycle is significantly longer than that of CCs, then CSCs become concentrated at an inner region as tumor grows. As a consequence we obtain that if CSCs are assumed to be more resistant to radiation than CCs, heterogeneous dosimetries can be selected to enhance tumor control by boosting radiation in the region occupied by the more radioresistant tumor cell phenotype. It is also shown that, when compared with homogeneous dose distributions as those being currently delivered in clinical practice, such heterogeneous radiation dosimetries fare always better than their homogeneous counterparts. Finally, limitations to our assumptions and their resulting clinical implications will be discussed.
Collapse
Affiliation(s)
- Juan Carlos López Alfonso
- Department of Applied Mathematics, Faculty of Mathematics, Universidad Complutense de Madrid, Madrid, Spain
| | - Nick Jagiella
- Institut National de Recherche en Informatique et en Automatique (INRIA), Domaine de Voluceau - Rocquencourt, Paris, France
- Institute of Computational Biology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Luis Núñez
- Radiophysics Department, Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain
| | - Miguel A. Herrero
- Department of Applied Mathematics, Faculty of Mathematics, Universidad Complutense de Madrid, Madrid, Spain
- * E-mail:
| | - Dirk Drasdo
- Institut National de Recherche en Informatique et en Automatique (INRIA), Domaine de Voluceau - Rocquencourt, Paris, France
- University of Paris 6 (UPMC), CNRS UMR 7598, Laboratoire Jacques-Louis Lions, Paris, France
- Interdisciplinary Center for Bioinformatics (IZBI), University of Leipzig, Leipzig, Germany
| |
Collapse
|
326
|
Zereshkian A, Leyton JV, Cai Z, Bergstrom D, Weinfeld M, Reilly RM. The human polynucleotide kinase/phosphatase (hPNKP) inhibitor A12B4C3 radiosensitizes human myeloid leukemia cells to Auger electron-emitting anti-CD123 ¹¹¹In-NLS-7G3 radioimmunoconjugates. Nucl Med Biol 2014; 41:377-83. [PMID: 24637100 DOI: 10.1016/j.nucmedbio.2014.02.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Revised: 02/06/2014] [Accepted: 02/07/2014] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Leukemia stem cells (LSCs) are believed to be responsible for initiating and propagating acute myeloid leukemia (AML) and for causing relapse after treatment. Radioimmunotherapy (RIT) targeting these cells may improve the treatment of AML, but is limited by the low density of target epitopes. Our objective was to study a human polynucleotide kinase/phosphatase (hPNKP) inhibitor that interferes with DNA repair as a radiosensitizer for the Auger electron RIT agent, ¹¹¹In-NLS-7G3, which recognizes the CD123⁺/CD131⁻ phenotype uniquely displayed by LSCs. METHODS The surviving fraction (SF) of CD123⁺/CD131⁻ AML-5 cells exposed to ¹¹¹In-NLS-7G3 (33-266 nmols/L; 0.74MBq/μg) or to γ-radiation (0.25-5Gy) was determined by clonogenic assays. The effect of A12B4C3 (25 μmols/L) combined with ¹¹¹In-NLS-7G3 (16-66 nmols/L) or with γ-radiation (0.25-2Gy) on the SF of AML-5 cells was assessed. The density of DNA double-strand breaks (DSBs) in the nucleus was measured using the γ-H2AX assay. Cellular dosimetry was estimated based on the subcellular distribution of ¹¹¹In-NLS-7G3 measured by cell fractionation. RESULTS Binding of (111)In-NLS-7G3 to AML-5 cells was reduced by 2.2-fold in the presence of an excess (1μM) of unlabeled NLS-7G3, demonstrating specific binding to the CD123⁺/CD131⁻ epitope. ¹¹¹In-NLS-7G3 reduced the SF of AML-5 cells from 86.1 ± 11.0% at 33 nmols/L to 10.5 ± 3.6% at 266 nmols/L. Unlabeled NLS-7G3 had no significant effect on the SF. Treatment of AML-5 cells with γ-radiation reduced the SF from 98.9 ± 14.9% at 0.25Gy to 0.03 ± 0.1% at 5 Gy. A12B4C3 combined with ¹¹¹In-NLS-7G3 (16-66 nmols/L) enhanced the cytotoxicity up to 1.7-fold compared to treatment with radioimmunoconjugates alone and was associated with a 1.6-fold increase in DNA DSBs in the nucleus. A12B4C3 enhanced the cytotoxicity of γ-radiation (0.25-0.5Gy) on AML-5 cells by up to 1.5-fold, and DNA DSBs were increased by 1.7-fold. Exposure to ¹¹¹In-NLS-7G3 (66 nmols/L) delivered up to 0.6Gy to AML-5 cells. CONCLUSIONS We conclude that A12B4C3 radiosensitized AML cells to the DNA damaging effects of ¹¹¹In-NLS-7G3. Combination treatment may increase the effectiveness for Auger electron RIT of AML targeting the LSC subpopulation.
Collapse
Affiliation(s)
- Arman Zereshkian
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | - Jeffrey V Leyton
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada; Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Zhongli Cai
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | - Dane Bergstrom
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | | | - Raymond M Reilly
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada; Department of Medical Imaging, University of Toronto, Toronto, ON, Canada; Toronto General Research Institute, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
327
|
Kim JH, Thimmulappa RK, Kumar V, Cui W, Kumar S, Kombairaju P, Zhang H, Margolick J, Matsui W, Macvittie T, Malhotra SV, Biswal S. NRF2-mediated Notch pathway activation enhances hematopoietic reconstitution following myelosuppressive radiation. J Clin Invest 2014; 124:730-41. [PMID: 24463449 PMCID: PMC3904618 DOI: 10.1172/jci70812] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 10/31/2013] [Indexed: 12/13/2022] Open
Abstract
A nuclear disaster may result in exposure to potentially lethal doses of ionizing radiation (IR). Hematopoietic acute radiation syndrome (H-ARS) is characterized by severe myelosuppression, which increases the risk of infection, bleeding, and mortality. Here, we determined that activation of nuclear factor erythroid-2-related factor 2 (NRF2) signaling enhances hematopoietic stem progenitor cell (HSPC) function and mitigates IR-induced myelosuppression and mortality. Augmenting NRF2 signaling in mice, either by genetic deletion of the NRF2 inhibitor Keap1 or by pharmacological NRF2 activation with 2-trifluoromethyl-2'-methoxychalone (TMC), enhanced hematopoietic reconstitution following bone marrow transplantation (BMT). Strikingly, even 24 hours after lethal IR exposure, oral administration of TMC mitigated myelosuppression and mortality in mice. Furthermore, TMC administration to irradiated transgenic Notch reporter mice revealed activation of Notch signaling in HSPCs and enhanced HSPC expansion by increasing Jagged1 expression in BM stromal cells. Administration of a Notch inhibitor ablated the effects of TMC on hematopoietic reconstitution. Taken together, we identified a mechanism by which NRF2-mediated Notch signaling improves HSPC function and myelosuppression following IR exposure. Our data indicate that targeting this pathway may provide a countermeasure against the damaging effects of IR exposure.
Collapse
Affiliation(s)
- Jung-Hyun Kim
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.
Laboratory of Synthetic Chemistry, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA.
Department of Radiation Oncology, School of Medicine, University of Maryland, Baltimore, Maryland, USA.
Department of Microbiology and Molecular Immunology, Johns Hopkins Bloomberg School of Public Heath, Baltimore, Maryland, USA.
Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Baltimore, Maryland, USA
| | - Rajesh K. Thimmulappa
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.
Laboratory of Synthetic Chemistry, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA.
Department of Radiation Oncology, School of Medicine, University of Maryland, Baltimore, Maryland, USA.
Department of Microbiology and Molecular Immunology, Johns Hopkins Bloomberg School of Public Heath, Baltimore, Maryland, USA.
Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Baltimore, Maryland, USA
| | - Vineet Kumar
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.
Laboratory of Synthetic Chemistry, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA.
Department of Radiation Oncology, School of Medicine, University of Maryland, Baltimore, Maryland, USA.
Department of Microbiology and Molecular Immunology, Johns Hopkins Bloomberg School of Public Heath, Baltimore, Maryland, USA.
Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Baltimore, Maryland, USA
| | - Wanchang Cui
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.
Laboratory of Synthetic Chemistry, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA.
Department of Radiation Oncology, School of Medicine, University of Maryland, Baltimore, Maryland, USA.
Department of Microbiology and Molecular Immunology, Johns Hopkins Bloomberg School of Public Heath, Baltimore, Maryland, USA.
Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Baltimore, Maryland, USA
| | - Sarvesh Kumar
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.
Laboratory of Synthetic Chemistry, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA.
Department of Radiation Oncology, School of Medicine, University of Maryland, Baltimore, Maryland, USA.
Department of Microbiology and Molecular Immunology, Johns Hopkins Bloomberg School of Public Heath, Baltimore, Maryland, USA.
Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Baltimore, Maryland, USA
| | - Ponvijay Kombairaju
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.
Laboratory of Synthetic Chemistry, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA.
Department of Radiation Oncology, School of Medicine, University of Maryland, Baltimore, Maryland, USA.
Department of Microbiology and Molecular Immunology, Johns Hopkins Bloomberg School of Public Heath, Baltimore, Maryland, USA.
Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Baltimore, Maryland, USA
| | - Hao Zhang
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.
Laboratory of Synthetic Chemistry, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA.
Department of Radiation Oncology, School of Medicine, University of Maryland, Baltimore, Maryland, USA.
Department of Microbiology and Molecular Immunology, Johns Hopkins Bloomberg School of Public Heath, Baltimore, Maryland, USA.
Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Baltimore, Maryland, USA
| | - Joseph Margolick
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.
Laboratory of Synthetic Chemistry, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA.
Department of Radiation Oncology, School of Medicine, University of Maryland, Baltimore, Maryland, USA.
Department of Microbiology and Molecular Immunology, Johns Hopkins Bloomberg School of Public Heath, Baltimore, Maryland, USA.
Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Baltimore, Maryland, USA
| | - William Matsui
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.
Laboratory of Synthetic Chemistry, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA.
Department of Radiation Oncology, School of Medicine, University of Maryland, Baltimore, Maryland, USA.
Department of Microbiology and Molecular Immunology, Johns Hopkins Bloomberg School of Public Heath, Baltimore, Maryland, USA.
Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Baltimore, Maryland, USA
| | - Thomas Macvittie
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.
Laboratory of Synthetic Chemistry, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA.
Department of Radiation Oncology, School of Medicine, University of Maryland, Baltimore, Maryland, USA.
Department of Microbiology and Molecular Immunology, Johns Hopkins Bloomberg School of Public Heath, Baltimore, Maryland, USA.
Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Baltimore, Maryland, USA
| | - Sanjay V. Malhotra
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.
Laboratory of Synthetic Chemistry, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA.
Department of Radiation Oncology, School of Medicine, University of Maryland, Baltimore, Maryland, USA.
Department of Microbiology and Molecular Immunology, Johns Hopkins Bloomberg School of Public Heath, Baltimore, Maryland, USA.
Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Baltimore, Maryland, USA
| | - Shyam Biswal
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.
Laboratory of Synthetic Chemistry, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA.
Department of Radiation Oncology, School of Medicine, University of Maryland, Baltimore, Maryland, USA.
Department of Microbiology and Molecular Immunology, Johns Hopkins Bloomberg School of Public Heath, Baltimore, Maryland, USA.
Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
328
|
The Molecular Crosstalk between the MET Receptor Tyrosine Kinase and the DNA Damage Response-Biological and Clinical Aspects. Cancers (Basel) 2013; 6:1-27. [PMID: 24378750 PMCID: PMC3980615 DOI: 10.3390/cancers6010001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 12/06/2013] [Accepted: 12/06/2013] [Indexed: 02/07/2023] Open
Abstract
Radiation therapy remains an imperative treatment modality for numerous malignancies. Enduring significant technical achievements both on the levels of treatment planning and radiation delivery have led to improvements in local control of tumor growth and reduction in healthy tissue toxicity. Nevertheless, resistance mechanisms, which presumably also involve activation of DNA damage response signaling pathways that eventually may account for loco-regional relapse and consequent tumor progression, still remain a critical problem. Accumulating data suggest that signaling via growth factor receptor tyrosine kinases, which are aberrantly expressed in many tumors, may interfere with the cytotoxic impact of ionizing radiation via the direct activation of the DNA damage response, leading eventually to so-called tumor radioresistance. The aim of this review is to overview the current known data that support a molecular crosstalk between the hepatocyte growth factor receptor tyrosine kinase MET and the DNA damage response. Apart of extending well established concepts over MET biology beyond its function as a growth factor receptor, these observations directly relate to the role of its aberrant activity in resistance to DNA damaging agents, such as ionizing radiation, which are routinely used in cancer therapy and advocate tumor sensitization towards DNA damaging agents in combination with MET targeting.
Collapse
|
329
|
Corre I, Guillonneau M, Paris F. Membrane signaling induced by high doses of ionizing radiation in the endothelial compartment. Relevance in radiation toxicity. Int J Mol Sci 2013; 14:22678-96. [PMID: 24252908 PMCID: PMC3856084 DOI: 10.3390/ijms141122678] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 11/01/2013] [Accepted: 11/06/2013] [Indexed: 01/30/2023] Open
Abstract
Tumor areas can now be very precisely delimited thanks to technical progress in imaging and ballistics. This has also led to the development of novel radiotherapy protocols, delivering higher doses of ionizing radiation directly to cancer cells. Despite this, radiation toxicity in healthy tissue remains a major issue, particularly with dose-escalation in these new protocols. Acute and late tissue damage following irradiation have both been linked to the endothelium irrigating normal tissues. The molecular mechanisms involved in the endothelial response to high doses of radiation are associated with signaling from the plasma membrane, mainly via the acid sphingomyelinase/ceramide pathway. This review describes this signaling pathway and discusses the relevance of targeting endothelial signaling to protect healthy tissues from the deleterious effects of high doses of radiation.
Collapse
Affiliation(s)
- Isabelle Corre
- CRCNA-UMR Inserm U892-CNRS 6299-Institut de Recherche en Santé de l'Université de Nantes, Nantes 44007, France.
| | | | | |
Collapse
|