The Pathway from Radiation to Fibrosis: LET Dependence

A B S T R A C T

It is well-known that Radiation-induced fibrosis (RIF) is a late event occurring months to years after the initial radiation exposure. Fibrotic lesions have been shown to manifest in many tissues including the skin, heart, lung, liver and kidney. Fibrosis occurs due to abnormal accumulation of extracellular matrix (ECM) proteins that result in loss of normal tissue and organ function. The cell type involved in RIF is myofibroblasts, which do not undergo apoptosis after healing but instead continue to accumulate, producing excessive amounts of ECM proteins, thereby damaging the tissues and organs. Reactive oxygen species, generated in response to radiation, is one signal that helps maintain the myofibroblast phenotype. In this review, we discuss molecular mechanisms leading to this late radiation event, known biomarkers for prediction, preclinical animal models of radiation-induced toxicity and current clinical trials designed for mitigation and treatment of radiation-induced fibrosis. We also discuss other physical properties such as linear energy transfer (LET) than the ones used in the clinics today which may have the potential to change our understanding on this inevitable pathway from radiation treatment to organ fibrosis.

Keywords

Radiation-induced fibrosis (RIF), myofibroblasts, reactive oxygen species (ROS), transforming growth factor-β (TGF-β), linear energy transfer (LET)

Introduction

Radiation-induced Fibrosis (RIF) develops several months or years following radiotherapy [1-3]. Also, RIF occurs due to differentiation of normal fibroblasts into myofibroblasts, which in turn deposit aberrant amounts of extracellular matrix protein instead of undergoing apoptosis. Persistence of active myofibroblasts occurs through autocrine and paracrine signals and influx of inflammatory cells [4, 5]. In the context of radiation exposure, a major trigger towards development of RIF is reactive oxygen species (ROS), generated immediately after radiation exposure. Radiation-induced bystander effects (RIBE), where unirradiated cells exhibit effects of irradiated cells due to signals received from nearby irradiated cells, contribute to amplifying the ROS levels [6-10]. Generation of ROS is believed to induce epigenetic changes that cause differentiation of fibroblasts to myofibroblasts [11, 12]. Ionizing radiation sources used in cancer therapy include gamma rays and X-rays, which possess sufficient energy to displace electrons from atoms. When these energy waves interact with water molecules, it leads to excitation and ionization of water molecules to form free radicals and ROS. Generation of ROS takes place in three cellular compartments: mitochondria, endoplasmic reticulum (ER) and cytosol.

The ROS generating processes at all three locations, mutually affects each other while also being influenced by the exogenous ROS, thereby amplifying the production of ROS from initial levels. Maintenance of homeostasis is achieved by biochemical mechanisms in place in the normal physiological setting, to counteract the damaging effects of free radical damage by ROS. These include action of enzymes such as super oxide dismutase (SOD) and DNA methyltransferases, p53 interplay and its regulation to restore the cell to its normal redox state. Ionizing radiation induced oxidative stress depends on radiation dose, dose rate, and linear energy transfer (LET) [13]. High LET radiation also causes persistent ROS, DNA damage, genomic instability in long term effect. Failure to maintain the ROS levels at physiological concentrations help maintain the myofibroblasts phenotype leading to radiation-induced fibrosis. While RIF is an unnecessary complication that occurs months to years following radiation, advances in understanding the molecular mechanisms have identified biomarkers that can predict patients at increased or decreased risk of treatment-related injury, with a goal toward improving the therapeutic ratio in order to enable physicians to optimize & individualize therapy for patients.

The TGF-β cytokine that stimulates fibroblast proliferation while upregulating ECM production has now been identified as a serum marker of developing RIF [14, 15]. Studies from pre-clinical animal models indicate Pravastatin to play a role in inhibition of rho kinase, the modulator of the Rho/ CCN2/extracellular matrix cascade and thereby improving radiation-induced fibrosis [16-19]. Clinical trials with Pravastatin have shown some reversal in radiation-induced fibrosis [20]. Charged particle radiation such as proton and helium, and heavy ion carbon radiotherapy with its unique properties to minimize scatter as particulate beams pass through the tissue, depositing energy at precise depths shows promise for reducing the molecular events leading to late radiation-induced fibrotic tissue damage. In this review, we summarize and discuss this pathway from initial radiotherapy to the tissue fibrotic endpoint with an aim to shed light into the importance of careful planning and evaluation on an individualized basis in order to reduce unwanted complications of radiation with severe consequences.

I Molecular Signaling in Radiation-Induced Fibrosis
i TGF-β Signaling Through SMAD Proteins

Transforming growth factor-β has a central role in the radiation-induced pathway to fibrosis [21]. Upon activation, either by irradiation via reactive oxygen species or other mechanisms, signaling is initiated by ligand binding to Type I and Type II TGF-β receptor complexes on the cell surface, inducing trans-phosphorylation of the glycine and serine residue-rich region (GS) in type I receptor by type II receptor kinases [21, 22-24]. The activated type I receptors phosphorylate SMADs 2 and 3 at the C terminal of the receptor. SMAD 2 and SMAD3 form a complex with a common SMAD 4. The activated SMAD trimer then enters the nucleus to activate gene transcription and promote cell growth and survival.

ii Smad3 Predominance in Radiation Induced Fibrosis

Given the predominant role of Smad3 in controlling downstream gene expression the TGF-β/Smad3 pathway is implicated in specific TGF-β associated pathologic fibrosis, since it controls the synthesis of proteins of the extracellular matrix such as Collagens 1, 5 and 6 and its role in epithelial to mesenchymal cell transition [21, 25, 26, 27,28]. Clinical data shows enhanced expression of TGF-β years after initial exposure [29].

iii Role of NF-κB in Radiation-Induced Response

NF-κB is a transcription factor that has an active role in the radiation-induced adaptive response [30]. The onset of radiation activates a group of NF-κB regulated cytokines including TNF-α, which contributes to enhanced sensitivity of a cell to radiation [31, 32]. TNF-α activates NF-κB via receptor activation and regulates the expression of several genes involved in the immune and inflammatory response TNF-α activated protein kinases activate I κB kinase and c-jun N-terminal kinase,that are involved in the NF-κB mediated radiation response [33-39].

iv Reactive Oxygen Species (ROS)

Radiation interacts with molecules of water in cells, causing them to ionize and produce ROS which includes superoxide (O2-), hydrogen peroxide (H2O2) and hydroxyl radical (OH-), all of which result in progressive damage to cells and consequently cell death [40]. The majority of radiation-induced cell injury is a consequence of the hydroxyl (OH-) radical [41, 42]. Increased production of ROS lead to toxicity of cells of parenchymal origin, which initiate a cascade, altering the mileu of cytokines in the microenvironment, leading to peroxidation of lipids, oxidation of DNA and protein and activation of proinflammatory cytokines both in vitro and in vivo [43-46]. Lipid peroxidation products activate fibrogenic cytokines that function as chemoattractants, mitogens and smooth muscle cell differentiation inducers in the blood vessel wall [47]. A summary of effectors of fibrosis and the biochemical mechanisms involved are illustrated in (Table 1).

Table 1:  Modulators of radiation-induced fibrosis.

Modulator

Biochemical signaling pathway

Fibrotic manifestation

TGFβ

Smad dependent pathways

a SMA collagen

Cytokines

Smad, NFκB

EMT profibrotic responses

TNFα

NFκB

profibrotic responses

ROS/RNS

DNA oxidation, secretion of MMPs

ECM


II Effects of High LET and low LET Radiation on Fibrosis

High LET radiation is known to result in lower survival of the cell per absorbed dose, as compared to low LET radiation. Pro-fibrotic gene, plasminogen activator inhibitor I (PAI-1) is involved in radiation-induced tissue remodeling via p53, TGF-β and Smad pathway. Although high LET radiation induced more apoptosis than low LET induction of the pro-fibrotic gene, plasminogen activator inhibitor I (PAI-1) was similar with high LET and low LET [48]. Since expression of PAI-1 is regulated by p53, specifically by phosphorylation of the latter at serine 315, a correlation of phosphorylation at serine 315 of p53 and levels of PAI-1 protein expression were observed on Western blot and luciferase functional assays with high and low LET [48]. Functional assays did not show a correlation between phosphorylation of p53 at serine 37 and PAI-1 induction. There is a direct correlation between Serine 37 phosphorylation and apoptosis however, with higher apoptosis observed after high LET radiation [48]. Since overexpression of post-mitotic fibroblasts is the hallmark cellular phenomenon leading to fibrosis, another group of investigators studied the differentiation pattern of human fibroblasts along with quantifying the production of extracellular matrix components after induction of high and low LET radiation. Similar differentiation was demonstrated for 195MeVu -1carbon ions with low LET compared with X-rays. Low energy carbon ions were observed to be more efficient than X-rays, versus nickel ions, which showed a lesser effect on induction of fibroblast differentiation [49]. For ECM protein production, a similar pattern of LET dependence was observed as was seen for differentiation [49].

III Dose Effects

A variety of factors affect radiation-induced fibrosis. Among these, total dose and dose per fraction, treated volume, and time course of radiation delivery are the primary factors known to have a significant impact on development of fibrosis in treated tissue [50-54]. However, the NTD50 values vary among different studies depending on variables besides radiation, and the effect of dose rate on the development of fibrosis may become arguable [50]. The dose effect also depends on radiation quality. Carbon ion radiation induces persistent lung injury and inflammation in a mouse model at high doses (over 2 Gy) [55].

IV Biomarkers for Prediction of RIF
i Biomarkers in Blood

TGF-β1 levels in the serum serve as an early biomarker for development of moderate to severe RIF after IR [56-58]. Since, radiation induced damage is the consequence of an early activation of an inflammatory response leading to significant expression in the bloodstream of a cascade of cytokines besides TGF-β1, such as IL-6, KL-6, surfactant proteins and IL-1ra, the latter have potential to be used clinically as early response markers for radiation induced damage [59-61].

ii Genetic Biomarkers

Studies of molecular mechanisms of radiation sensitivity show associations between common variants in DNA damage and repair genes and development of adverse reactions to radiotherapy. Polymorphisms in the XRCC1, ATM, hHR21 and TGFβ1 correlate to an increased risk of developing an adverse tissue reaction to radiotherapy, whilst one variant of the ATM gene has been reported to be radioprotective [62-70].

V Preclinical Animal Models for Treatment of RIF
A TGF-β/Smad Pathway Inhibitors

Since the TGF-β/Smad pathway is a major player in the development of fibrosis, its inhibition has been implicated as a possible therapeutic intervention. SM16, an inhibitor of TGF- βR1, has been demonstrated to be effective in reducing radiation-related lung damage in an animal model [71]. LY2109761, inhibitor of TGF- βI serine/threonine kinase, known to reduce p-Smad 2 and p-Smad I expression, has been shown to suppress and reduce pulmonary fibrosis [72]. Neutralizing antibodies against TGF- β inhibited both proliferation of rat lung fibroblasts and terminal differentiation of progenitor fibroblasts to post-mitotic fibrocytes [73]. SB203580 and WP631 are inhibitors of the Smad signal transduction pathway, have been shown to abrogate the excessive proliferation and reduced expression of p21 (WAF1/CIP1) and PAI-1 (a TGF- β/Smad-responsive profibrotic gene induced by gamma rays and TGF-β1 [74, 75]. MyD88 (an intracellular adaptor for TLR signaling) regulates innate immunity and NF- κB activated responses, attenuates long-term radiation-induced lung injury and protects against fibrosis by alleviating chronic lung injury [76]. Fluorofenidone (1-(3-fluorophenyl)-5-methyl-2-(1H)-pyridone, AKF-PD), a novel pyridine antifibrotic agent, reduced cardiac and kidney fibrosis by inhibiting CTGF (connective tissue growth factor) expression [77, 78].

B Targeting Chronic ROS/RNS Production

Targeting chronic generation of ROS/RNS post-radiation with long-term administration of free-radical scavengers such as superoxide dismutase (SOD) and/or catalytic manganese (Mn) porphyrin-based superoxide dismutase (SOD) mimetics alleviates oxidative stress, organ hypoxia, production of cytokines and injury to heart & lung. The recombinant enzyme SOD-TAT combats radiation-induced lung injury in mice [79]. SOD has also been used in a porcine animal model to successfully treat RIF [80]. Molecular hydrogen is an antioxidant that diffuses through cell membranes, reduces levels of ROS and decreases oxidative stress-induced injury in several organs [81]. The radioprotective drug Amifostine is used clinically for its properties of scavenging free radicals, DNA protection, and acceleration of repair and this drug has potential as a therapeutic for treatment of radiation-induced lung damage [82, 83].

C Anti-TNF Antibodies

TNF-α is known to play a major role in the pathogenesis of post-radiation tissue injury, causing cachexia in addition to tissue and organ damage and shock effects that are irreverible. It is therefore implied that Fibrosis could be prevented with antibodies to TNF. TNF-α receptors I and II in soluble form have potential for their application as inhibitors of radiation-induced tissue injury [84].

D Restoring Immunological Balance

Glucocorticosteroids have value to restore immunological balance in several affected organs and tissues [85]. Sivelestat, a neutrophil elastase inhibitor, significantly decreased deposition of collagen and accumulation of neutrophils in the lung parenchymal cells and showed improvement in static lung compliance of radiation-treated lung [86].

VI Clinical Trials

Since preclinical studies showed the possibility of the anti-oxidant SOD as a good candidate for clinical studies, a clinical trial using Lipsod (a Cu/Zn SOD in liposomal form) was used in a clinical study at the Necker Hospital in Paris in 1984. 34 patients manifesting RIF in skin and underlying tissues were treated from May 1984 to January 1986 and followed for a time frame of 5 years. All patients exhibited clinical regression of fibrosis at varying degrees [87]. In another study, involving 24 women previously treated with radiation for breast cancer, a 6-month treatment with a combination of pentoxifylline and alpha-tocopherol was shown to successfully treat superficial radiation-induced fibrosis [88, 89].

VII Future Directions
i Safer Radiation Protocols

Proton beam therapy (PBT) with unique properties to deliver proton beams with minimal dose deposition beyond the treatment target has the potential to minimize exposure to the heart, lungs, muscle and bone [90]. Historically, a relative biological effectiveness of protons in clinical practice has been established as 1.1, based upon in vitro and in vivo data of the biological effect of protons in the middle of the therapeutic radiation distribution or SOBP [91]. However, data also exists that the RBE of protons is greater at the distal end of the range. If not accounted for, this could result in a greater biologic effect and as a toxicity beyond the target at the ribs, lungs and heart. Modeling and accounting for the optimal physical and biological parameters at the end of the treatment range remains as an important area for laboratory and clinical investigation. Close clinical follow-up and adopting a ‘data pooling culture’ will enable better correlation of individual treatments with long-term outcome of a large number of patients, so as to better understand the impact of variations in radiation treatment protocols [92].

ii Close Monitoring for Timely Intervention

Cardiac damage occurs in more than 50% of patients treated with radiation therapy. Close monitoring of post-radiation effects and clinical intervention in the ischemic stage of RICAD might be valuable since this is a reversible and treatable, thereby suppressing the subsequent progression of damage to irreversible events.

iii Therapeutic Invention Towards the TGF-β Inhibition

Since TGF-β1 is considered the central player in the switch towards fibrosis, future therapeutics should be focused towards inhibition of TGF β1 receptor activity [93]. Promising results have been achieved using small-molecule inhibitors of the TGF-β receptor I kinase in preclinical models to treat radiation-induced lung fibrosis [94]. Inhibition of integrin receptors which have an important role in cell-matrix interactions, serve as another promising new approach for antifibrosis therapy [95-97].

Conclusion

Fibrosis is a consequence of various converging paths including inflammation, oxidative stress and chronic alterations in gene expression. The complexity of the interplay between these pathways may present a varied number of therapeutic targets for combating fibrosis. On the other hand, since some of these targets have pleiotrophic effects, caution should be exerted to ensure knocking down one fibrotic pathway does not lead to toxicity from another interconnected pathway and thereby reduce the overall clinical benefit. This communication attempts to discuss the end-point of current radiation protocols (heart and lung damage), an inevitable consequence of radiation, clinically manifesting as hardening of usually soft organs such as heart and lung by deposition of extracellular matrix components which should have undergone apoptosis in the normal setting. This phenomenon is referred to as Radiation-induced Fibrosis (RIF). RIF has been traditionally considered an irreversible process.

Although there is no known treatment for this inevitable and undesirable side-effect of radiation, contemporary research studies indicate that regression of fibrosis can occur using the anti-oxidant Superoxide dismutase (SOD) and a combination of pentoxifylline and Vitamin E which work synergistically with each other to cause clinical regression and successfully treat fibrosis at different levels [87-90]. Even though, the end-point of Fibrosis is not the scope of our lab, as a social responsibility to the community at large, we have discussed this, since it points to the fact that an even higher regression can be achieved with the safer radiation beams. It opens up future research possibilities with proton beams and charged particles (known to reduce non-targeted effects to normal cells and tissue) and points to the fact radiation-induced fibrosis may be treatable with a combination of safer forms of radiation as well as mild drugs. Our recent studies explore the possibilities to understand and mitigate the radiation related fibrosis [98-100].

Conflicts of interest

The authors whose names are listed below certify that they have no affiliations with or involvement in any organization or entity with any financial interest or non-financial interest in the subject material discussed in this manuscript.

Article Info

Article Type
Review Article
Publication history
Received: Wed 27, Nov 2019
Accepted: Fri 06, Dec 2019
Published: Mon 16, Dec 2019
Copyright
© 2023 Premkumar B. Saganti. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. Hosting by Science Repository.
DOI: 10.31487/j.COCB.2019.01.02

Author Info

Corresponding Author
Premkumar B. Saganti
Texas A&M Chancellor's Research Initiative, Radiation Institute for Science and Engineering, Prairie View A&M University, Texas, United States

Figures & Tables

Table 1:  Modulators of radiation-induced fibrosis.

Modulator

Biochemical signaling pathway

Fibrotic manifestation

TGFβ

Smad dependent pathways

a SMA collagen

Cytokines

Smad, NFκB

EMT profibrotic responses

TNFα

NFκB

profibrotic responses

ROS/RNS

DNA oxidation, secretion of MMPs

ECM


References

  1. Murros Karl E, James F Toole (1989) "The effect of radiation on carotid arteries: a review article." Arch Neurol 46: 449-455. [Crossref]
  2. Schultz-Hector S (1992) "Radiation-induced heart disease: review of experimental data on dose reponse and pathogenesis." Int J Radiat Biol 61: 149-160. [Crossref]
  3. Yi Ann, Hak Hee Kim, Hee Jung Shin, Mi Ock Huh, Seung Do Ahn et al. (2009) "Radiation-induced complications after breast cancer radiation therapy: a pictorial review of multimodality imaging findings." Korean J Radiol 10: 496-507. [Crossref]
  4. Bomb Ritin, Mark R Heckle, Yao Sun, Salvatore Mancarella, Guntaka RV et al. (2016) "Myofibroblast secretome and its auto-/paracrine signaling." Expert review of cardiovascular therapy 14: 591-598. [Crossref]
  5. Mittal Manish, Mohammad Rizwan Siddiqui, Khiem Tran, Sekhar P Reddy, Malik AB (2014) "Reactive oxygen species in inflammation and tissue injury." Antioxid Redox Signal 20: 1126-1167. [Crossref]
  6. Azzam Edouard I, Sonia M De Toledo, Douglas R Spitz, John B Little (2002) "Oxidative metabolism modulates signal transduction and micronucleus formation in bystander cells from α-particle-irradiated normal human fibroblast cultures." Cancer Res 62: 5436-5442. [Crossref]
  7. Nikjoo Hooshang, Igor K Khvostunov (2003) "Biophysical model of the radiation-induced bystander effect." Int J Radiat Biol 79: 43-52. [Crossref]
  8. Kadhim Munira A, Stephen R Moore, Edwin H Goodwin (2004) "Interrelationships amongst radiation-induced genomic instability, bystander effects, and the adaptive response." Mutat Res 568: 21-32. [Crossref]
  9. Mikkelsen Ross (2004) "Redox signaling mechanisms and radiationinduced bystander effects." Hum Exp Toxicol 23: 75-79. [Crossref]
  10. Fakir H, Hofmann W, Tan WY, Sachs RK (2009) "Triggering-response model for radiation-induced bystander effects." Radiat Res 171: 320-331. [Crossref]
  11. Shrishrimal S, Kosmacek EA, Oberley-Deegan RE (2019) "Reactive Oxygen Species Drive Epigenetic Changes in Radiation-Induced Fibrosis." Oxid Med Cell Longev 2019: 4278658. [Crossref]
  12. Weigel C, P Schmezer, Christoph Plass, O Popanda (2015) "Epigenetics in radiation-induced fibrosis." Oncogene 34: 2145-2155. [Crossref]
  13. Azzam EI, Jay-Gerin JP, Pain D (2012) "Ionizing radiation-induced metabolic oxidative stress and prolonged cell injury." Cancer lett 327: 48-60. [Crossref]
  14. Barcellos-Hoff MH (1998) "How do tissues respond to damage at the cellular level? The role of cytokines in irradiated tissues." Radiat res 150: S109-S120. [Crossref]
  15. Boothe DL, Coplowitz S, Greenwood E, Barney CL, Christos PJ et al. (2013) "Transforming growth factor β-1 (TGF-β1) is a serum biomarker of radiation induced fibrosis in patients treated with intracavitary accelerated partial breast irradiation: preliminary results of a prospective study." Int J Radiat Oncol Biol Phys 87: 1030-1036. [Crossref]
  16. Haydont V, Gilliot O, Rivera S, Bourgier C, François A et al. (2007) "Successful mitigation of delayed intestinal radiation injury using pravastatin is not associated with acute injury improvement or tumor protection." Int J Radiat Oncol Biol Phys 68: 1471-1482. [Crossref]
  17. Haydont V, Bourgier C, Pocard M, Lusinchi A, Aigueperse J et al. (2007) "Pravastatin Inhibits the Rho/CCN2/extracellular matrix cascade in human fibrosis explants and improves radiation-induced intestinal fibrosis in rats." Clin Cancer Res 13: 5331-5340. [Crossref]
  18. Bourgier C, V Haydont, F Milliat, A Francois, V Holler et al. (2005) "Inhibition of Rho kinase modulates radiation induced fibrogenic phenotype in intestinal smooth muscle cells through alteration of the cytoskeleton and connective tissue growth factor expression." Gut 54: 336-343. [Crossref]
  19. Monceau V, Pasinetti N, Schupp C, Pouzoulet F, Opolon P et al. (2010) "Modulation of the Rho/ROCK pathway in heart and lung after thorax irradiation reveals targets to improve normal tissue toxicity." Curr Drug Targets 11: 1395-1404. [Crossref]
  20. Bourgier Celine, Anne Auperin, Sofia Rivera, Pierre Boisselier, Benoit Petit et al. (2019) "Pravastatin Reverses Established Radiation-Induced Cutaneous and Subcutaneous Fibrosis in Patients with Head and Neck Cancer: Results of the Biology-Driven Phase 2 Clinical Trial Pravacur." Int J Radiat Oncol Biol Phys 104: 365-373. [Crossref]
  21. Rodemann H Peter, Michael Bamberg (1995) "Cellular basis of radiation-induced fibrosis." Radiother Oncol 35: 83-90. [Crossref]
  22. Derynck Rik, Feng XH (1997) "TGF-β receptor signaling." Biochim Biophys Acta 1333: F105-F150. [Crossref]
  23. Massagué Joan (2000) "How cells read TGF-β signals." Nat Rev Mol Cell Biol 1: 169-178. [Crossref]
  24. Derynck Rik, Ying E Zhang (2003) "Smad-dependent and Smad-independent pathways in TGF-β family signalling." Nature 425: 577-584. [Crossref]
  25. Yang Junwei, Xianghong Zhang, Yingjian Li, Youhua Liu (2003) "Downregulation of Smad transcriptional corepressors SnoN and Ski in the fibrotic kidney: an amplification mechanism for TGF-β1 signaling." J Am Soc Nephrol 14: 3167-3177. [Crossref]
  26. Verrecchia Franck, Mon-Li Chu, Alain Mauviel (2001) "Identification of novel TGF-β/Smad gene targets in dermal fibroblasts using a combined cDNA microarray/promoter transactivation approach." J Biol Chem 276: 17058-17062. [Crossref]
  27. Kalluri Raghu, Eric G Neilson (2003) "Epithelial-mesenchymal transition and its implications for fibrosis." J Clin Invest 112: 1776-1784. [Crossref]
  28. Thiery Jean Paul (2003) "Epithelial–mesenchymal transitions in development and pathologies." Curr Opin Cell Biol 15: 740-746. [Crossref]
  29. Martin Michèle, Jean-Louis Lefaix, Sylvie Delanian (2000) "TGF-β1 and radiation fibrosis: a master switch and a specific therapeutic target?" Int J Radiat Oncol Biol Phys 47: 277-290. [Crossref]
  30. Ahmed Kazi Mokim, Jian Jian Li (2008) "NF-κB-mediated adaptive resistance to ionizing radiation." Free Radic Biol Med 44: 1-13. [Crossref]
  31. Hallahan Dennis E, David R Spriggs, Michael A Beckett, Donald W Kufe  (1989) "Increased tumor necrosis factor alpha mRNA after cellular exposure to ionizing radiation." Proc Natl Acad Sci U S A 86: 10104-10107. [Crossref]
  32. Chen Mei, Jose Quintans, Zvi Fuks, Craig Thompson, Donald W Kufe et al. (1995) "Suppression of Bcl-2 messenger RNA production may mediate apoptosis after ionizing radiation, tumor necrosis factor α, and ceramide." Cancer res 55: 991-994. [Crossref]
  33. Osborn Laurelee, Steven Kunkel, Gary J Nabel (1989) "Tumor necrosis factor alpha and interleukin 1 stimulate the human immunodeficiency virus enhancer by activation of the nuclear factor kappa B." Proc Natl Acad Sci U S A 86: 2336-2340. [Crossref]
  34. Blonska Marzenna, Yun You, Romas Geleziunas, Xin Lin (2004) "Restoration of NF-κB activation by tumor necrosis factor alpha receptor complex-targeted MEKK3 in receptor-interacting protein-deficient cells." Mol Cell Biol 24: 10757-10765. [Crossref]
  35. Baeuerle Patrick A, Thomas Henkel (1994) "Function and activation of NF-kappaB in the immune system." Annu Rev Immunol 12: 141-179. [Crossref]
  36. Baeuerle PA, RA Rupec, HL Pahl (1996) "Reactive oxygen intermediates as second messengers of a general pathogen response." Pathol Biol (Paris) 44: 29-35. [Crossref]
  37. Bradbury C Matthew, Stephanie Markovina, S Jack Wei, Luis M Rene, Imran Zoberi et al. (2001) "Indomethacin-induced radiosensitization and inhibition of ionizing radiation-induced NF-κB activation in HeLa cells occur via a mechanism involving p38 MAP kinase." Cancer res 61: 7689-7696. [Crossref]
  38. Dent Paul, Adly Yacoub, Paul B Fisher, Michael P Hagan, Steven Grant (2003) "MAPK pathways in radiation responses." Oncogene 22: 5885-5896. [Crossref]
  39. Wang Tieli, Yu-Chang Hu, Shaozhong Dong, Ming Fan, Daniel Tamae et al. (2005) "Co-activation of ERK, NF-κB, and GADD45β in response to ionizing radiation." J Biol Chem 280: 12593-12601. [Crossref]
  40. Riley PA (1994) "Free radicals in biology: oxidative stress and the effects of ionizing radiation." Int J Radiat Biol 65: 27-33. [Crossref]
  41. Zhao Feng, Zai‐Qun Liu (2009) "The protective effect of hydroxyl‐substituted Schiff bases on the radical‐induced oxidation of DNA." Journal of Physical Organic Chemistry 22: 791-798.
  42. Terasaki Yasuhiro, Ikuroh Ohsawa, Mika Terasaki, Mikiko Takahashi, Shinobu Kunugi et al. (2011) "Hydrogen therapy attenuates irradiation-induced lung damage by reducing oxidative stress." Am J Physiol Lung Cell Mol Physiol 301: L415-L426. [Crossref]
  43. Ghafoori Paiman, Lawrence B Marks, Zeljko Vujaskovic, Christopher R Kelsey (2008) "Radiation-induced lung injury: Assessment, management, and prevention." Oncology (Williston Park) 22: 37-47 [Crossref]
  44. Zhao Weiling, Mike EC Robbins (2009) "Inflammation and chronic oxidative stress in radiation-induced late normal tissue injury: therapeutic implications." Curr Med Chem 16: 130-143. [Crossref]
  45. Fleckenstein Katharina, Larisa Zgonjanin, Liguang Chen, Zahid Rabbani, Isabel L Jackson et al. (2007) "Temporal onset of hypoxia and oxidative stress after pulmonary irradiation." Int J Radiat Oncol Biol Phys 68: 196-204. [Crossref]
  46. Robbins ME, W Zhao (2004) "Chronic oxidative stress and radiation‐induced late normal tissue injury: a review." Int J Radiat Biol 80: 251-259. [Crossref]
  47. Madamanchi Nageswara R, Aleksandr Vendrov, Marschall S Runge (2005) "Oxidative stress and vascular disease." Arterioscler Thromb Vasc Biol 25: 29-38. [Crossref]
  48. Niemantsverdriet Maarten, Marc-Jan van Goethem, Reinier Bron, Wytse Hogewerf, Sytze Brandenburg, et al. (2012) "High and low LET radiation differentially induce normal tissue damage signals." Int J Radiat Oncol Biol Phys 83: 1291-1297. [Crossref]
  49. Fournier C, M Scholz, WK Weyrather, HP Rodemann, G Kraft (2001) "Changes of fibrosis-related parameters after high-and low-LET irradiation of fibroblasts." Int J Radiat Biol 77: 713-722. [Crossref]
  50. Borger Jacques H, Harry Kemperman, Henk Sillevis Smitt, Augustinus Hart, Joop van Dongen et al. (1994) "Dose and volume effects on fibrosis after breast conservation therapy." Int J Radiat Oncol Biol Phys 30: 1073-1081. [Crossref]
  51. Barendsen GW (1982) "Dose fractionation, dose rate and iso-effect relationships for normal tissue responses." Int J Radiat Oncol Biol Phys 8: 1981-1997. [Crossref]
  52. Gerszten Peter C, John J Moossy, Sanjeev Bahri, Andre Kalend, A Julio Martínez (1999) "Inhibition of peridural fibrosis after laminectomy using low-dose external beam radiation in a rat model." Neurosurgery 44: 597-602. [Crossref]
  53. Gerszten Peter C, John J Moossy, John C Flickinger, Kristina Gerszten, Andre Kalend et al. (2000) "Inhibition of peridural fibrosis after laminectomy using low-dose external beam radiation in a dog model." Neurosurgery 46: 1478-1485.
  54. Straub Jeffrey M, Jacob New, Chase D Hamilton, Chris Lominska, Yelizaveta Shnayder et al. (2015) "Radiation-induced fibrosis: mechanisms and implications for therapy." J Cancer Res Clin Oncol 141: 1985-1994. [Crossref]
  55. Wu Zhenhua, Xinyu Wang, Rong Yang, Yang Liu, Weiping Zhao et al. (2013) "Effects of carbon ion beam irradiation on lung injury and pulmonary fibrosis in mice." Exp Ther Med 5: 771-776. [Crossref]
  56. Boothe Dustin L, Shana Coplowitz, Eleni Greenwood, Christian L Barney, Paul J Christos et al. (2013) "Transforming growth factor β-1 (TGF-β1) is a serum biomarker of radiation induced fibrosis in patients treated with intracavitary accelerated partial breast irradiation: preliminary results of a prospective study." Int J Radiat Oncol Biol Phys 87: 1030-1036. [Crossref]
  57. Barcellos-Hoff MH (1998) "How do tissues respond to damage at the cellular level? The role of cytokines in irradiated tissues." Radiat Res 150: S109-S120. [Crossref]
  58. Li Jingxia, Shuangfeng Mu, Lixiang Mu, Xiaohui Zhang, Ranran Pang et al. (2015) "Transforming growth factor-beta-1 is a serum biomarker of radiation-induced pneumonitis in esophageal cancer patients treated with thoracic radiotherapy: preliminary results of a prospective study." Onco Targets  ther 8: 1129-1136. [Crossref]
  59. Rubin Philip, Carl J Johnston, Jacqueline P Williams, Sandra McDonald, Jacob N Finkelstein (1995) "A perpetual cascade of cytokines postirradiation leads to pulmonary fibrosis." Int J Radiat Oncol Biol Phys 33: 99-109.  [Crossref]
  60. Fleckenstein Katharina, Benjamin Gauter-Fleckenstein, Isabel L Jackson, Zahid Rabbani, Mitchell Anscher et al. (2007) "Using biological markers to predict risk of radiation injury." Semin Radiat Oncol 17: 89-98. [Crossref]
  61. Kong Feng-Ming, Xiaoping Ao, Li Wang, Theodore S Lawrence (2008) "The use of blood biomarkers to predict radiation lung toxicity: a potential strategy to individualize thoracic radiation therapy." Cancer Control 15: 140-150. [Crossref]
  62. Thompson Larry H, Mary G West (2000) "XRCC1 keeps DNA from getting stranded." Mutat Res 459: 1-18. [Crossref]
  63. Moullan Norman, David G Cox, Sandra Angele, Pascale Romestaing, Jean-Pierre Gérard et al. (2003) "Polymorphisms in the DNA repair gene XRCC1, breast cancer risk, and response to radiotherapy." Cancer Epidemiol Biomarkers Prev 12: 1168-1174. [Crossref]
  64. Wang Yunfei, Margaret R Spitz, Yong Zhu, Qiong Dong, Sanjay Shete et al. (2003) "From genotype to phenotype: correlating XRCC1 polymorphisms with mutagen sensitivity." DNA Repair (Amst) 2: 901-908. [Crossref]
  65. Severin Diane Marie, Trevor Leong, Barry Cassidy, Hany Elsaleh, Lester Peters et al. (2001) "Novel DNA sequence variants in the hHR21 DNA repair gene in radiosensitive cancer patients." Int J Radiat Oncol Biol Phys 50: 1323-1331. [Crossref]
  66. Birkenbihl Rainer P, Suresh Subramani (1992) "Cloning and characterization of rad21 an essential gene of Schizosaccharomyces pombe involved in DNA double-strand-break repair." Nucleic Acids Res 20: 6605-6611. [Crossref]
  67. Guacci Vincent, Douglas Koshland, Alexander Strunnikov (1997) "A direct link between sister chromatid cohesion and chromosome condensation revealed through the analysis of MCD1 in S. cerevisiae." Cell 91: 47-57. [Crossref]
  68. Quarmby S, H Fakhoury, Edward Levine, J Barber, J Wylie et al. (2003) "Association of transforming growth factor beta-1 single nucleotide polymorphisms with radiation-induced damage to normal tissues in breast cancer patients." Int J Radiat Biol 79: 137-143. [Crossref]
  69. Bakkenist Christopher J, Michael B Kastan (2003) "DNA damage activates ATM through intermolecular autophosphorylation and dimer dissociation." Nature 421: 499-506. [Crossref]
  70. Shiloh Yosef (2003) "ATM and related protein kinases: safeguarding genome integrity." Nat Rev Cancer 3: 155-168. [Crossref]
  71. Anscher Mitchell S, Bradley Thrasher, Larisa Zgonjanin, Zahid N Rabbani, Michael J Corbley et al. (2008) "Small molecular inhibitor of transforming growth factor-β protects against development of radiation-induced lung injury." Int J Radiat Oncol Biol Phys 71: 829-837. [Crossref]
  72. Flechsig Paul, Monika Dadrich, Sebastian Bickelhaupt, Jürgen Jenne, Kai Hauser et al. (2012) "LY2109761 attenuates radiation-induced pulmonary murine fibrosis via reversal of TGF-β and BMP-associated proinflammatory and proangiogenic signals." Clin Cancer Res 18: 3616-3627. [Crossref]
  73. Hakenjos L, M Bamberg, HP Rodemann  (2000) "TGF-beta1-mediated alterations of rat lung fibroblast differentiation resulting in the radiation-induced fibrotic phenotype." Int J Radiat Biol 76: 503-509. [Crossref]
  74. Dennler Sylviane, Susumu Itoh, Denis Vivien, Peter ten Dijke, Stéphane Huet et al. (1998) "Direct binding of Smad3 and Smad4 to critical TGFβ‐inducible elements in the promoter of human plasminogen activator inhibitor‐type 1 gene." EMBO J 17: 3091-3100. [Crossref]
  75. Li Yang, Liang‑Wen Song, Rui‑Yun Peng, De‑Wen Wang, Mei‑Hong Jin et al. (2008) "Effects of SB203580 and WP631 on Smad signal transduction pathway in lung fibroblasts after irradiation." Ai Zheng 27: 698-702. [Crossref]
  76. Brickey Willie J, Isabel P Neuringer, William Walton, Xiaoyang Hua, Ellis Y Wang et al. (2012) "MyD88 provides a protective role in long-term radiation-induced lung injury." Int J Radiat Biol 88: 335-347. [Crossref]
  77. Wang Ling Hao, Ji Shi Liu, Wang Bin Ning, Qiong Jing Yuan, Fang Fang Zhang et al. (2011) "Fluorofenidone attenuates diabetic nephropathy and kidney fibrosis in db/db mice." Pharmacology 88: 88-99. [Crossref]
  78. Chen Ling-Xi, Kan Kan, Ming Ming, Qiong Chen, Zhao-He Wang et al. (2012) "Fluorofenidone inhibits transforming growth factor-β1-induced cardiac myofibroblast differentiation." Die Pharmazie-An International Journal of Pharmaceutical Sciences 67: 452-456.
  79. Pan Jianru, Ying Su, Xiaojun Hou, Huocong He, Shutao Liu et al. (2012) "Protective effect of recombinant protein SOD-TAT on radiation-induced lung injury in mice." Life Sci 91: 89-93. [Crossref]
  80. Lefaix Jean-Louis, Sylvie Delanian, Jean-Jacques Leplat, Yves Tricaud, Michèle Martin et al. (1996) "Successful treatment of radiation-induced fibrosis using CuZn-SOD and Mn-SOD: an experimental study." Int J Radiat Oncol Biol Phys 35: 305-312. [Crossref]
  81. Terasaki Yasuhiro, Ikuroh Ohsawa, Mika Terasaki, Mikiko Takahashi, Shinobu Kunugi et al. (2011) "Hydrogen therapy attenuates irradiation-induced lung damage by reducing oxidative stress." Am J Physiol Lung Cell Mol Physiol 301: L415-L426. [Crossref]
  82. Kouvaris John R, Vassilis E Kouloulias, Lampros J Vlahos (2007) "Amifostine: the first selective-target and broad-spectrum radioprotector." Oncologist 12: 738-747. [Crossref]
  83. Koukourakis Michael I, Marianthi Panteliadou, Ioannis M Abatzoglou, Kyriaki Sismanidou, Efthimios Sivridis et al. (2013) "Postmastectomy hypofractionated and accelerated radiation therapy with (and without) subcutaneous amifostine cytoprotection." Int J Radiat Oncol Biol Phys 85: e7-e13. [Crossref]
  84. Wolthuis Esther K, Alexander PJ Vlaar, Goda Choi, Joris JTH Roelofs, Jack J Haitsma et al. (2009) "Recombinant human soluble tumor necrosis factor-alpha receptor fusion protein partly attenuates ventilator-induced lung injury." Shock 31: 262-266. [Crossref]
  85. Sekine Yusuke, Kohsuke Takeda, Hidenori Ichijo (2006) "The ASK1-MAP kinase signaling in ER stress and neurodegenerative diseases." Curr Mol Med 6: 87-97. [Crossref]
  86. Fujino Naoya, Hiroshi Kubo, Tomoko Suzuki, Mei He, Takaya Suzuki et al. (2012) "Administration of a specific inhibitor of neutrophil elastase attenuates pulmonary fibrosis after acute lung injury in mice." Exp Lung Res 38: 28-36. [Crossref]
  87. Delanian S, F Baillet, J Huart, J-L Lefaix, C Maulard et al. (1994) "Successful treatment of radiation-induced fibrosis using liposomal CuZn superoxide dismutase: clinical trial." Radiother Oncol 32: 12-20. [Crossref]
  88. Delanian Sylvie, Raphael Porcher, Saida Balla-Mekias, Jean-Louis Lefaix (2003) "Randomized, placebo-controlled trial of combined pentoxifylline and tocopherol for regression of superficial radiation-induced fibrosis." J Clin Oncol 21: 2545-2550. [Crossref]
  89. Chiao Teresa B, Audrey J Lee (2005) "Role of pentoxifylline and vitamin E in attenuation of radiation-induced fibrosis." Ann Pharmacother 39: 516-522. [Crossref]
  90. Corbin Kimberly S, Robert W Mutter (2018) "Proton therapy for breast cancer: progress & pitfalls." Breast Cancer Management 7: BMT06.
  91. Paganetti Harald, Andrzej Niemierko, Marek Ancukiewicz, Leo E Gerweck, Michael Goitein et al. (2002) "Relative biological effectiveness (RBE) values for proton beam therapy." Int J Radiat Oncol Biol Phys 53: 407-421. [Crossref]
  92. Deasy Joseph O, Søren M Bentzen, Andrew Jackson, Randall K Ten Haken, Ellen D Yorke et al. (2010) "Improving normal tissue complication probability models: the need to adopt a “data-pooling” culture." Int J Radiat Oncol Biol Phys 76: S151-S154. [Crossref]
  93. Martin M, S Delanian, V Sivan, MC Vozenin-Brotons, P Reisdorf et al. (2000) "Radiation-induced superficial fibrosis and TGF-alpha 1." Cancer Radiother 4: 369-384. [Crossref]
  94. Flechsig Paul, Monika Dadrich, Sebastian Bickelhaupt, Jürgen Jenne, Kai Hauser et al. (2012) "LY2109761 attenuates radiation-induced pulmonary murine fibrosis via reversal of TGF-β and BMP-associated proinflammatory and proangiogenic signals." Clin Cancer Res 18: 3616-3627. [Crossref]
  95. Puthawala Khalid, Nicos Hadjiangelis, Steven C Jacoby, Emmanuel Bayongan, Zhicheng Zhao et al. (2008) "Inhibition of integrin αvβ6, an activator of latent transforming growth factor-β, prevents radiation-induced lung fibrosis." Am J Respir Crit Care Med 177: 82-90. [Crossref]
  96. Horton Jason A, Fei Li, Eun Joo Chung, Kathryn Hudak, Ayla White et al. (2013) "Quercetin inhibits radiation-induced skin fibrosis." Radiat Res 180: 205-215. [Crossref]
  97. Xavier Sandhya, Ester Piek, Makiko Fujii, Delphine Javelaud, Alain Mauviel et al. (2004) "Amelioration of radiation-induced fibrosis inhibition of transforming growth factor-β signaling by halofuginone." J Biol Chem 279: 15167-15176. [Crossref]
  98. Menezes, Kareena M, Huichen Wang, Megumi Hada, Premkumar B. Saganti (2018) Radiation matters of the heart: a mini review. Front Cardiovasc Med 5 83-88. [Crossref]
  99. Li Z, KK Jella, L Jaafar, S Li, S Park et al. (2018) Exposure to galactic cosmic radiation compromises DNA repair and increases the potential for oncogenic chromosomal rearrangement in bronchial epithelial cells. Sci rep 8: 11038. [Crossref]
  100. Cucinotta, Francis A, Eliedonna Cacao, Myung-Hee Y Kim, Premkumar B (2020) Saganti. Cancer and circulatory disease risks for a human mission to Mars: Private mission considerations. Acta Astronautica166: 529-536.