1755-1536-5-S1-S131755-1536 Proceedings <p>Therapeutic management of intestinal fibrosis induced by radiation therapy: from molecular profiling to new intervention strategies <it>et vice et versa</it> </p> HamamaSaadsaad.hamama@igr.fr DelanianSylviesylvie.delanian@sls.aphp.fr MonceauVirginievirginie.monceau@igr.fr VozeninMarie-Catherinevozenin@igr.fr

INSERM U-1030 "Molecular Radiotherapy" Institut Gustave Roussy, Villejuif, France

"Molecular Radiotherapy", Université Paris Sud Paris XI, France

Unité de Radiopathologie, Service Oncologie-Radiothérapie, Hôpital Saint-Louis, APHP, Paris, France

Fibrogenesis & Tissue Repair <p>Proceedings of Fibroproliferative disorders: from biochemical analysis to targeted therapies</p>Petro E Petrides and David BrennerProceedings<p>Fibroproliferative disorders: from biochemical analysis to targeted therapies</p>Frauenchiemsee, Germany25-30 September 2010www.innova-med.de1755-1536 2012 5 Suppl 1 S13 http://www.fibrogenesis.com/content/5/S1/S13 10.1186/1755-1536-5-S1-S13
662012 2012Hamama et al; licensee BioMed Central Ltd.This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Chronic toxicities of locoregional and systemic oncological treatments commonly develop in long-term cancer survivors. Amongst these toxicities, post-radiotherapeutic complications alter patient's quality of life. Reduction of exposure of normal tissues can be achieved by optimization of radiotherapy. Furthermore, understanding of the fibrogenic mechanisms has provided targets to prevent, mitigate, and reverse late radiation-induced damages. This mini-review shows how (i) global molecular studies using gene profiling can provide tools to develop new intervention strategies and (ii) how successful clinical trials, conducted in particular with combined pentoxifylline-vitamin E, can take benefice of biological and molecular evidences to improve our understanding of fibrogenic mechanisms, enhance the robustness of proposed treatments, and lead ultimately to better treatments for patient's benefice.

Introduction

Therapeutic management of cancer improved during the past decade and is characterized today by a significant increase in patient's survival rates. Although effective on cure rates, both locoregional and systemic oncological treatments present some concerns related to development of chronic toxicities that alter patient's quality of life, while results of combined therapies suggest that normal tissue toxicity will become a major concern within the next years. Amongst these toxicities, radiation enteropathy is a significant delayed side effect of lumbar and pelvic radiotherapy.

The risk, severity and nature of these radiation-induced toxicities depend on several factors including radiotherapy-related factors (total dose, dose per fraction and volume exposure) and patient-related factors (comorbidities) 1 . Accordingly, a real effort has been made to reduce normal tissue exposure by ballistic and imaging optimization of radiotherapy. Besides technological tools, understanding the fibrogenic mechanisms and targeting profibrotic factors has provided alternate and promising approaches to prevent, mitigate or even reverse late radiation-induced damages 1 2 .

If today's clinical practice always aims to limit aggravating factors, current management of radiation-induced damages involves 3 : (i) anti-inflammatory treatments including corticosteroids; (ii) vascular therapy including pentoxifylline (PTX) or hyperbaric oxygen; (iii) antioxidant treatment including superoxide dismutase in the nineties and then combined pentoxifylline-vitamin E (PTX-VitE) in the last decade.

The first part of this mini-review shows how global molecular studies using gene profiling can provide tools to develop new intervention strategies with old molecules or new compounds. The second part shows how successful clinical research done with well-known but low potent old drugs takes benefice of biological and molecular evidences to improve its robustness for patient's benefice.

From molecular profiling to new intervention strategies

Cellular and molecular mechanisms involved in the persistence of radiation fibrosis

In all irradiated tissues and especially when vital organs like the heart, lung or intestine are affected 4 , the most concerning aspect of radiation fibrosis is its progressive and seemingly irreversible evolution. Thus, the development of curative anti-fibrotic strategies is nowadays highly anticipated by both patients and physicians 5 . Definition of new biologically-based anti-fibrotic strategies is therefore an attractive option to be achieved by characterization of the cellular and molecular mechanisms involved in the persistence of radiation fibrosis.

In human radiation enteropathy, fibrosis is the main histopathological hallmark 6 . Fibrosis contributes to the loss of intestinal compliance and impaired intestinal function and we showed that it was associated with heavy deposition of Connective Tissue Growth Factor (CTGF/CCN2) 7 . CTGF gene regulation is known to be under the control of TGF-β via a Smad consensus sequence and TGF-β RE/BCE-1 binding sites located in the CTGF promoter region 8 9 . We showed that, surprisingly, TGF-β1 expression in fibrotic area was low during the onset of radiation enteropathy 7 . The molecular basis of this paradox was investigated using (i) a high-throughput biological approach by cDNA array profiling and (ii) a classical biochemical approach with recombinant TGF-β1 and CTGF. These studies were performed with unique and physiologically relevant cell models, employing primary smooth muscle cells and sub-epithelial myofibroblasts derived from radiation enteropathy. These cells mimic fibrosis in vitro, as they maintain their fibrogenic features in long term culture (6-8 passages) i.e. altered contractile function, modification of the actin cytoskeleton and increased secretory activity 10 11 .

The comprehensive cDNA approach showed activation of the Rho/ROCK pathway 12 . Further functional in vitro experiments showed that this intercellular signaling pathway controls CTGF expression in intestinal smooth muscle cells and in subepithelial myofibroblasts derived from radiation enteropathy 10 13 . In addition, our gene profiling studies showed that radiation enteropathy was associated with a global deregulation of the extracellular matrix remodeling with increased ECM deposition, MMPs and TIMPs activity 14 . Whether this dynamic remodeling process was the cause or the consequence of the phenotypic alteration of the resident mesenchymal cells is currently under investigation.

Cell response to increasing concentrations of recombinant TGF-β1 was investigated. Activation of cell-specific signaling pathways by low TGF-β1 concentrations was demonstrated with a prominent activation of the Rho/ROCK pathway in fibrosis-derived cells, whereas the Smad pathway was predominantly activated in normal cells. This differential fibrogenic response identified in normal versus fibrosis-derived cells opened new therapeutic opportunities for targeted anti-fibrotic therapy. In addition, we showed that recombinant CTGF was able to trigger its auto-induction in fibrosis-derived cells, an effect which was further enhanced by TGF-β1. These results thus identify specific and combinatorial roles of low TGF-β1 doses and CTGF for the maintenance of tissue fibrosis 15 .

From physiopathological mechanisms to clinical transfer

Rho GTPases regulate fundamental cellular processes including cell motility, cell cycle progression, cell survival, transcription, membrane trafficking and cytokinesis via their downstream effectors the Rho-associated kinases (ROCKs) 16 17 . Many Rho functions have been elucidated using pharmacological inhibitors, the most prominent ones being Statins, molecules which inhibit isoprenoid intermediates production and Rho activation. In order to investigate whether the Rho/ROCK cascade regulates radiation-induced fibrogenic program in intestinal mesenchymal cells, pharmacological inhibition of Rho and ROCK activation was performed in vitro using pravastatin and Y-27632, a pyrimidine derivative inhibitor of ROCK. We showed that both agents modulated radiation-induced fibrogenic differentiation and the expression of CTGF, TGF-β1, and collagen Iα2 genes (Figure 1), most likely via NF-κB inhibition 10 11 18 . Next, therapeutic experiments were conducted in pre-clinical models. Pravastatin was chosen as, in the case of convincing results, it would be easy to accelerate the transfer of this drug to the clinic, given the fact that the drug is safe and well tolerated 19 . Remarkably, we showed that pravastatin administration with curative intent improves radiation enteropathy in rats, inhibits Rho and ROCK activity in human samples, and subsequently inhibits CTGF production in-vivo, ex-vivo, and in-vitro. In addition, inhibition of type-I-collagen and fibronectin occurred, indicating that pravastatin modulates the secretory phenotype of mesenchymal cells, probably by inhibition of the Rho/ROCK/CTGF/ECM cascade 13 . Mitigation experiments with pravastatin, relevant to clinically well-established fibrosis, improved delayed radiation enteropathy in rats and decreased both CTGF expression and collagen deposition. Interestingly, pravastatin's protective effect was differential, as no tumor protection occurred 20 . Similar results were obtained by others using Simvastatin 21 and anti-fibrotic efficacy of statins was shown in model of radiation-induced lung 22 23 . These pre-clinical findings encouraged us to propose a phase II randomized clinical trial, which received approval from the local ethics committee and started at Institut Gustave Roussy in January 2010 with the support of the French Ministry of Health (PHRC 2010).

<p>Figure 1</p>

A. Effect of Pravastatin on mRNA expression of TGFβ1, CTGF and Collagen in a kinetic manner

A. Effect of Pravastatin on mRNA expression of TGFβ1, CTGF and Collagen in a kinetic manner: Twenty four hours kinetics of mRNA expression of TGFβ1, CTGF, and Col Iα2 in control and Pravastatin treated cells show that Pravastatin treatment with a dose of 0.1 mM and up reduces levels of mRNAs of these genes with maximum efficiency at six hours post-treatment; C: control, P: Pravastatin treatment. 0.1, 0.5, and 1 refer to treatment dose in mM. *: p < 0.05, **: p < 0.01, ***: p < 0.005 according to kruskal-Wallis test. B.Protein expression of CTGF in control and Pravastatin treated cells: Pravastatin treatment for 24 hours inhibits CTGF protein expression showing also a dose-response relationship. GAPDH is used as a housekeeping gene. C: control, P: Pravastatin treatment. 0.1, 0.5 refer to treatment dose in mM.

Searching for anti-fibrotic treatment using molecular targets lead to the development of targeted drugs. Amongst them anti-CTGF antibody 24 , imatinib (anti-PDGF antibody) 25 26 27 , statin (Rho/Rock-inhibitors) 20 21 22 23 28 have demonstrated efficacy in pre-clinical studies and need now to be validated in clinical trials. Two targeted drugs have recently showed interesting results in recent trials: the small molecule inhibitor of TGF-β1, pirfenidone, and the tyrosine kinase inhibitor, BIBF 1120. Both of them halt progression of idiopathic pulmonary fibrosis 29 30 . Still others, such as combined pentoxifylline-vitamin E has a proven clinical efficacy with an unknown mechanism of action 31 32 33 34 35 36 37 38 .

From clinical trials to understanding the molecular mechanism

In the nineties an antioxidant compound, the liposomal Cu/Zn superoxide dismutase (SOD), was the first drug to half reduce established radiation fibrosis in patients and experimental models (reviewed in 3 ). Biological studies showed that SOD inhibited TGF-β1 and TIMPs and lead to a subsequent reversion of the myofibroblastic phenotype in vitro 39 40 . Later, Delanian et al. showed that combined PTX-Vit.E administered to patients, half reduced superficial radiation fibrosis in 6 months 31 . Results were confirmed in a randomized clinical trial 32 and by long-term clinical results (3 years) 33 . A first experimental study using skin fibrosis in pig as model showed that the histopathological recovery after PTX/VitE treatment was associated with significant reduction of TGF-β1 deposition 41 . Since then, numerous clinical trials were successful and showed improvement of osteoradionecrosis 34 , superficial fibrosis 36 , proctitis/enteritis 37 , pelvic complications 38 , uterine fibro-atrophy 35 42 . Moreover, combined PTX-Vit.E in rat experiment had beneficial effects on radiation-induced myocardial fibrosis and left ventricular function 43 . Recent report using a triple combination PENTOCLO: PTX-VitE and clodronate; a biphosphonate active against macrophages also allows successful healing of severe osteoradionecrosis 44 and reduction of neurological symptoms associated with radiation-induced plexopathies 45 . The efficacy of this triple combination is currently validated in a phase III randomized clinical trial that has started at Saint Louis Hospital for radiation-induced plexitis (support by the French Ministry of Health, PHRC 2009). Therefore, understanding the molecular mechanism of PTX-Vit.E combination solicit much interest for better understanding of radiation-induced fibrosis, to improve treatment robustness and will pave the way toward new more efficient treatments.

Vitamin E (α-tocopherol) is an antioxidant drug known to modulate the expression of several genes such as ICAM-1 integrins and PPAR-gamma 46 . α-tocopherol was reported to decrease the expression of genes known to be involved in the fibrotic process such as (MMP-1) in human skin fibroblast 47 , and IL-1β in THP-1 cells 48 . In mice, D-α-tocopherol supplementation decreased collagen mRNA in the liver by 70% 49 . In type 2 diabetic patients, α-tocopherol supplementation lowered plasma levels of PAI-1 and P-selectin 50 . In other case, α-tocopherol induces the expression of CTGF in human smooth muscle cells while neither β-tocopherol nor N-acetylcysteine do 51 . It was suggested that this modulation of CTGF was unique as it was not triggered by structurally related antioxidant molecules, suggesting occurrence of a non-antioxidant mechanism in the transcriptional regulation of several genes.

Pentoxifylline, PTX is a methylxanthine derivative used to treat vascular disease such as intermittent claudication. In vivo, it has been reported to have anti-TNF-α effect, increase erythrocyte flexibility, vasodilate, and inhibit inflammatory reactions. In vitro studies have indicated that PTX has also antioxidant properties 52 , inhibits human dermal fibroblast proliferation and extracellular matrix production 53 54 55 and increases collagenase activity 53 . However, the doses of pentoxifylline required to produce these effects in vitro are high, and reached 1000 μg/ml in some cases 54 rendering in vivo use of PTX unsuitable. In addition, PTX is known as a non-specific phosphodiesterases inhibitor that subsequently increases intracellular levels of cAMP. Like other cAMP elevating agents PTX could activate protein kinase A (PKA) which would phosphorylate transcription factors, such as cAMP response element binding protein (CREB). Activated CREB recruits the coactivators CBP and P300 that also act as transcriptional co-activators for SMADs 56 57 . Therefore, the sequestration of CBP/P300 by activated CREB could inhibit SMAD-dependent transcription 58 and constitute one molecular mechanism to explain PTX anti-fibrotic action that remains to be demonstrated in vivo. For example, in a model RIF-induced in pigs no clinical or histological changes were observed in RIF after 6 months of treatment with PTX alone using maximum tolerated dose 41 . Extrapolation from in vitro studies would however suggest that higher concentration of PTX would be required to achieve effective suppression of collagen synthesis or to increase collagenase activity. Used at this dosage, PTX might be extremely toxic and suggests that administration of PTX alone does not constitute an anti-fibrotic treatment.

Studies about the mechanism of action of combined pentoxifylline-vitamin E in radiation fibrosis are awfully limited. An in vitro study was conducted in dermal fibroblast using the water-soluble analogue of vitamin E, trolox, to investigate the effects of combined pentoxifylline-trolox on irradiated cells. This study showed reduction in acute and late ROS formation in cells after irradiation, decrease in DNA strand breaks whenever the drugs were added i.e. before or after irradiation supporting an immediate anti-oxidant action that interfere with the DNA repair process 59 . However, the relevance of this study to fibrosis is unclear since only short term response was investigated, therefore we aimed at investigating the role of combined pentoxifylline-vitamin E on two well know fibrogenic pathway, i.e. TGF-β1 and Rho/Rock using an in vitro model of radiation-induced fibrosis consisting of primary smooth muscle cells derived from human radiation enteropathy samples (RE-SMC). The hydrophilic analogous of α-tocopherol, trolox, was used. Incubation of the cells with combined pentoxifylline-trolox didn't regulate RhoB mRNA expression (Figure 2A) nor influence Actin cytoskeleton in RE-SMC (data not shown) but interestingly negatively modulated TGF-β1 mRNA expression at early time point (one hour post-treatment) and subsequently decrease the expression of TGF-β1 targets such as PAI-1 both at mRNA and protein levels (twenty four hours post-treatment) (Figure 2A and 2B). This suggested that the anti-fibrotic effects of combined pentoxifylline-trolox could be mediated by inhibition of the TGF-β1 pathway. Interestingly, pentoxifylline and trolox appear to enhance the activity of each other; and the effect of the combination was more potent than any of the individual treatments, which is the definition of drugs synergy. Combined pentoxifylline-trolox with a dose of 10 μg/ml decreases protein expression of PAI-1 more effectively than trolox alone (10 μg/ml) or pentoxifylline alone (10 μg/ml) (Figure 2B). Thus, the synergy between the elements of this combination at low concentration could constitute the basis of its efficacy conferring a more appropriate therapeutic window. This study offers for the first time, to our knowledge, a molecular base for rationalization of the clinical use of combined Pentoxifylline-vitamin E in radiation fibrosis. Nevertheless, inhibition of TGF-β1 pathway is unlikely to be the sole anti-fibrotic mechanism of action of combined pentoxifylline-trolox and other novel candidates are actually under investigations.

<p>Figure 2</p>

A. Effect of combined pentoxifylline-trolox on mRNA expression of profibrotic genes in a kinetic manner

A. Effect of combined pentoxifylline-trolox on mRNA expression of profibrotic genes in a kinetic manner: Twenty four hours kinetics of mRNA expression of TGF-β1, PAI-1, Col Iα1, and RhoB. Pentoxifylline and trolox synergize to inhibit TGFβ1 at early point time, subsequent inhibition of TGFβ1 targets such as PAI-1 and Col Iα1. Treatment doesn't affect RhoB mRNA expression. P: Pentoxifylline treatment, T: Trolox treatment, PT: Pentoxifylline-trolox treatment. 10 refers to treatment dose in μg/ml. *: p < 0.05, **: p < 0.01, ***: p < 0.005 according to kruskal-Wallis test. B.Protein expression of PAI-1 in control and pentoxifylline-trolox treated cells: Combined pentoxifylline-trolox treatment for 24 hours inhibits PAI-1 protein expression more effectively than pentoxifylline alone or trolox alone; GAPDH is used as a housekeeping gene. C: control, P: Pentoxifylline treatment, T: Trolox treatment, PT: Pentoxifylline-trolox treatment. 10 refers to treatment dose in μg/ml.

Conclusion

From molecular profiling to clinical trials in a bottom-up manner or from the clinical trials to the molecular understanding in a top-down approach, these studies have optimized our understanding of radiation-induced fibrogenesis. Combining these information could ultimately lead to improve management of fibrosis.

In a model of intestinal fibrosis, Statins act principally via inhibition of Rho/Rock pathway decreasing subsequently the expression of CTGF. In contrast, combined pentoxifylline-trolox inhibits TGF-β1 pathway while it appears to have no influence on Rho/Rock pathway. Therefore combining these two medications to generate a more efficient triple therapy seemed a logical follow up. While emerging new combinations might be promising, heavy investigations are needed to prove their safety and efficacy over already solid candidates as Statins or combined pentoxifylline-vitamin E.

Competing interests

The authors declare that they have no competing interests.

Acknowledgements

This article has been published as part of Fibrogenesis & Tissue Repair Volume 5 Supplement 1, 2012: Proceedings of Fibroproliferative disorders: from biochemical analysis to targeted therapies. The full contents of the supplement are available online at http://www.fibrogenesis.com/supplements/5/S1.

<p>The radiation-induced fibroatrophic process: therapeutic perspective via the antioxidant pathway</p>DelanianSLefaixJLRadiother Oncol200473211913110.1016/j.radonc.2004.08.02115542158<p>Molecular aspects of intestinal radiation-induced fibrosis</p>GervazPMorelPVozenin-BrotonsMCCurr Mol Med20099327328010.2174/15665240978784716419355909<p>Current management for late normal tissue injury: radiation-induced fibrosis and necrosis</p>DelanianSLefaixJLSemin Radiat Oncol20071729910710.1016/j.semradonc.2006.11.00617395040<p>Bowel injury: current and evolving management strategies</p>Hauer-JensenMWangJDenhamJWSemin Radiat Oncol200313335737112903023<p>The irreversibility of radiation-induced fibrosis: fact or folklore?</p>AnscherMSJ Clin Oncol200523348551855210.1200/JCO.2005.03.619416260690<p>The pathology of ionizing radiation as defined by morphologic patterns</p>FajardoLFActa Oncol2005441132210.1080/0284186051000744015848902<p>Fibrogenic signals in patients with radiation enteritis are associated with increased connective tissue growth factor expression</p>Vozenin-BrotonsMCMilliatFSabourinJCde GouvilleACFrancoisALasserPMoricePHaie-MederCLusinchiAAntounSInt J Radiat Oncol Biol Phys200356256157210.1016/S0360-3016(02)04601-112738334<p>A novel transforming growth factor beta response element controls the expression of the connective tissue growth factor gene</p>GrotendorstGROkochiHHayashiNCell Growth Differ1996744694809052988<p>Connective tissue growth factor gene regulation. Requirements for its induction by transforming growth factor-beta 2 in fibroblasts</p>LeaskAHolmesABlackCMAbrahamDJJ Biol Chem200327815130081301510.1074/jbc.M21036620012571253<p>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</p>BourgierCHaydontVMilliatFFrancoisAHollerVLasserPBourhisJMatheDVozenin-BrotonsMCGut200554333634310.1136/gut.2004.051169177441815710979<p>Induction of CTGF by TGF-beta1 in normal and radiation enteritis human smooth muscle cells: Smad/Rho balance and therapeutic perspectives</p>HaydontVMatheDBourgierCAbdelaliJAigueperseJBourhisJVozenin-BrotonsMCRadiother Oncol200576221922510.1016/j.radonc.2005.06.02916046020<p>Gene expression profile in human late radiation enteritis obtained by high-density cDNA array hybridization</p>Vozenin-BrotonsMCMilliatFLinardCStrupCFrancoisASabourinJCLasserPLusinchiADeutschEGirinskyTRadiat Res2004161329931110.1667/RR312814982484<p>Rho/ROCK pathway as a molecular target for modulation of intestinal radiation-induced toxicity</p>HaydontVBourgierCVozenin-BrotonsMCBr J Radiol200780Spec No 1S324017704324<p>Global gene expression profiles reveal an increase in mRNA levels of collagens, MMPs, and TIMPs in late radiation enteritis</p>Strup-PerrotCMatheDLinardCViolotDMilliatFFrancoisABourhisJVozenin-BrotonsMCAm J Physiol Gastrointest Liver Physiol20042874G87588510.1152/ajpgi.00088.200415178550<p>Specific signals involved in the long-term maintenance of radiation-induced fibrogenic differentiation: a role for CCN2 and low concentration of TGF-beta1</p>HaydontVRiserBLAigueperseJVozenin-BrotonsMCAm J Physiol Cell Physiol20082946C1332134110.1152/ajpcell.90626.200718400984<p>Rho GTPases and cell migration</p>RidleyAJJ Cell Sci2001114Pt 152713272211683406<p>Actin's latest act: polymerizing to facilitate transcription?</p>VieuEHernandezNNat Cell Biol20068765065110.1038/ncb0706-65016820773<p>Comparative gene expression profiling in three primary human cell lines after treatment with a novel inhibitor of Rho kinase or atorvastatin</p>BoermaMFuQWangJLooseDSBartolozziAEllisJLMcGonigleSParadiseESweetnamPFinkLMBlood Coagul Fibrinolysis200819770971810.1097/MBC.0b013e32830b2891271368118832915<p>Statin therapy and autoimmune disease: from protein prenylation to immunomodulation</p>GreenwoodJSteinmanLZamvilSSNat Rev Immunol20066535837010.1038/nri183916639429<p>Successful mitigation of delayed intestinal radiation injury using pravastatin is not associated with acute injury improvement or tumor protection</p>HaydontVGilliotORiveraSBourgierCFrancoisAAigueperseJBourhisJVozenin-BrotonsMCInt J Radiat Oncol Biol Phys20076851471148210.1016/j.ijrobp.2007.03.04417674977<p>Simvastatin ameliorates radiation enteropathy development after localized, fractionated irradiation by a protein C-independent mechanism</p>WangJBoermaMFuQKulkarniAFinkLMHauer-JensenMInt J Radiat Oncol Biol Phys20076851483149010.1016/j.ijrobp.2007.03.036200070117674978<p>Effect of administration of lovastatin on the development of late pulmonary effects after whole-lung irradiation in a murine model</p>WilliamsJPHernadyEJohnstonCJReedCMFentonBOkunieffPFinkelsteinJNRadiat Res2004161556056710.1667/RR316815161367<p>Modulation of the Rho/ROCK pathway in heart and lung after thorax irradiation reveals targets to improve normal tissue toxicity</p>MonceauVPasinettiNSchuppCPouzouletFOpolonPVozeninMCCurr Drug Targets201011111395140420583978<p>Neutralizing monoclonal antibody to human connective tissue growth factor ameliorates transforming growth factor-beta-induced mouse fibrosis</p>IkawaYNgPSEndoKKondoMChujoSIshidaWShirasakiFFujimotoMTakeharaKJ Cell Physiol2008216368068710.1002/jcp.2144918481257<p>Inhibition of platelet-derived growth factor signaling attenuates pulmonary fibrosis</p>AbdollahiALiMPingGPlathowCDomhanSKiesslingFLeeLBMcMahonGGroneHJLipsonKEJ Exp Med2005201692593510.1084/jem.20041393221309115781583<p>Imatinib mesylate inhibits fibrogenesis in asbestos-induced interstitial pneumonia</p>VuorinenKGaoFOuryTDKinnulaVLMyllarniemiMExp Lung Res200733735737310.1080/01902140701634827265268517849262<p>Targeting tyrosine kinases: a novel therapeutic strategy for systemic sclerosis</p>GordonJKSpieraRFCurr Opin Rheumatol201022669069510.1097/BOR.0b013e32833f110520827202<p>Pravastatin limits endothelial activation after irradiation and decreases the resulting inflammatory and thrombotic responses</p>GauglerMHVereycken-HollerVSquibanCVandammeMVozenin-BrotonsMCBenderitterMRadiat Res2005163547948710.1667/RR330215850408<p>Pirfenidone in patients with idiopathic pulmonary fibrosis (CAPACITY): two randomised trials</p>NoblePWAlberaCBradfordWZCostabelUGlassbergMKKardatzkeDKingTEJrLancasterLSahnSASzwarcbergJLancet201137797791760176910.1016/S0140-6736(11)60405-421571362<p>Efficacy of a tyrosine kinase inhibitor in idiopathic pulmonary fibrosis</p>RicheldiLCostabelUSelmanMKimDSHansellDMNicholsonAGBrownKKFlahertyKRNoblePWRaghuGN Engl J Med2011365121079108710.1056/NEJMoa110369021992121<p>Striking regression of chronic radiotherapy damage in a clinical trial of combined pentoxifylline and tocopherol</p>DelanianSBalla-MekiasSLefaixJLJ Clin Oncol199917103283329010506631<p>Randomized, placebo-controlled trial of combined pentoxifylline and tocopherol for regression of superficial radiation-induced fibrosis</p>DelanianSPorcherRBalla-MekiasSLefaixJLJ Clin Oncol200321132545255010.1200/JCO.2003.06.06412829674<p>Major healing of refractory mandible osteoradionecrosis after treatment combining pentoxifylline and tocopherol: a phase II trial</p>DelanianSDepondtJLefaixJLHead Neck200527211412310.1002/hed.2012115641107<p>Kinetics of response to long-term treatment combining pentoxifylline and tocopherol in patients with superficial radiation-induced fibrosis</p>DelanianSPorcherRRudantJLefaixJLJ Clin Oncol200523348570857910.1200/JCO.2005.02.472916260695<p>Uterine restoration by radiation sequelae regression with combined pentoxifylline-tocopherol: a phase II study</p>Letur-KonirschHGuisFDelanianSFertil Steril20027761219122610.1016/S0015-0282(02)03120-512057732<p>Pentoxifylline and vitamin E combination for superficial radiation-induced fibrosis: a phase II clinical trial</p>HaddadPKalaghchiBAmouzegar-HashemiFRadiother Oncol200577332432610.1016/j.radonc.2005.09.01416236376<p>Effect of pentoxifylline and tocopherol on radiation proctitis/enteritis</p>HilleAChristiansenHPradierOHermannRMSiekmeyerBWeissEHilgersRHessCFSchmidbergerHStrahlenther Onkol2005181960661410.1007/s00066-005-1390-y16170489<p>Phase II study of vitamin E and pentoxifylline in patients with late side effects of pelvic radiotherapy</p>GothardLCornesPBrookerSEarlJGleesJHallEPeckittCTaitDYarnoldJRadiother Oncol200575333434110.1016/j.radonc.2005.02.00216086914<p>Cu/Zn superoxide dismutase modulates phenotypic changes in cultured fibroblasts from human skin with chronic radiotherapy damage</p>DelanianSMartinMBravardALuccioniCLefaixJLRadiother Oncol200158332533110.1016/S0167-8140(00)00332-711230895<p>Antifibrotic action of Cu/Zn SOD is mediated by TGF-beta1 repression and phenotypic reversion of myofibroblasts</p>Vozenin-BrotonsMCSivanVGaultNRenardCGeffrotinCDelanianSLefaixJLMartinMFree Radic Biol Med2001301304210.1016/S0891-5849(00)00431-711134893<p>Striking regression of subcutaneous fibrosis induced by high doses of gamma rays using a combination of pentoxifylline and alpha-tocopherol: an experimental study</p>LefaixJLDelanianSVozeninMCLeplatJJTricaudYMartinMInt J Radiat Oncol Biol Phys199943483984710.1016/S0360-3016(98)00419-210098440<p>Combined treatment by pentoxifylline and tocopherol for recipient women with a thin endometrium enrolled in an oocyte donation programme</p>Ledee-BatailleNOlivennesFLefaixJLChaouatGFrydmanRDelanianSHum Reprod20021751249125310.1093/humrep/17.5.124911980747<p>Prevention and treatment of functional and structural radiation injury in the rat heart by pentoxifylline and alpha-tocopherol</p>BoermaMRobertoKAHauer-JensenMInt J Radiat Oncol Biol Phys200872117017710.1016/j.ijrobp.2008.04.042257460818632215<p>Complete restoration of refractory mandibular osteoradionecrosis by prolonged treatment with a pentoxifylline-tocopherol-clodronate combination (PENTOCLO): a phase II trial</p>DelanianSChatelCPorcherRDepondtJLefaixJLInt J Radiat Oncol Biol Phys201180383283910.1016/j.ijrobp.2010.03.02920638190<p>Significant clinical improvement in radiation-induced lumbosacral polyradiculopathy by a treatment combining pentoxifylline, tocopherol, and clodronate (Pentoclo)</p>DelanianSLefaixJLMaisonobeTSalachasFPradatPFJ Neurol Sci20082751-216416610.1016/j.jns.2008.08.00418804790<p>Vitamin E mediates cell signaling and regulation of gene expression</p>AzziAGysinRKempnaPMunteanuANegisYVillacortaLVisariusTZinggJMAnn N Y Acad Sci20041031869510.1196/annals.1331.00915753136<p>Age-dependent increase of collagenase expression can be reduced by alpha-tocopherol via protein kinase C inhibition</p>RicciarelliRMaroniPOzerNZinggJMAzziAFree Radic Biol Med1999277-872973710.1016/S0891-5849(99)00007-610515576<p>Inhibition of IL-1 beta expression in THP-1 cells by probucol and tocopherol</p>AkesonALWoodsCWMosherLBThomasCEJacksonRLAtherosclerosis1991862-326127010.1016/0021-9150(91)90222-O1872919<p>Long- and short-term D-alpha-tocopherol supplementation inhibits liver collagen alpha1(I) gene expression</p>ChojkierMHouglumKLeeKSBuckMAm J Physiol19982756 Pt 1G148014859843787<p>alpha-Tocopherol supplementation decreases plasminogen activator inhibitor-1 and P-selectin levels in type 2 diabetic patients</p>DevarajSChanAVJrJialalIDiabetes Care200225352452910.2337/diacare.25.3.52411874941<p>Alpha-tocopherol induces expression of connective tissue growth factor and antagonizes tumor necrosis factor-alpha-mediated downregulation in human smooth muscle cells</p>VillacortaLGraca-SouzaAVRicciarelliRZinggJMAzziACirc Res200392110411010.1161/01.RES.0000049103.38175.1B12522127<p>In vitro antioxidant properties of pentoxifylline, piracetam, and vinpocetine</p>HorvathBMartonZHalmosiRAlexyTSzaparyLVekasiJBiroZHabonTKesmarkyGTothKClin Neuropharmacol2002251374210.1097/00002826-200201000-0000711852295<p>Pentoxifylline inhibits normal human dermal fibroblast in vitro proliferation, collagen, glycosaminoglycan, and fibronectin production, and increases collagenase activity</p>BermanBDuncanMRJ Invest Dermatol198992460561010.1111/1523-1747.ep127121402539414<p>Pentoxifylline inhibits the proliferation of human fibroblasts derived from keloid, scleroderma and morphoea skin and their production of collagen, glycosaminoglycans and fibronectin</p>BermanBDuncanMRBr J Dermatol1990123333934610.1111/j.1365-2133.1990.tb06294.x2206972<p>Pentoxifylline, pentifylline, and interferons decrease type I and III procollagen mRNA levels in dermal fibroblasts: evidence for mediation by nuclear factor 1 down-regulation</p>DuncanMRHasanABermanBJ Invest Dermatol1995104228228610.1111/1523-1747.ep126128197530274<p>TGF-beta-stimulated cooperation of smad proteins with the coactivators CBP/p300</p>JanknechtRWellsNJHunterTGenes Dev199812142114211910.1101/gad.12.14.21143170129679056<p>Physical and functional interaction of SMADs and p300/CBP</p>PouponnotCJayaramanLMassagueJJ Biol Chem199827336228652286810.1074/jbc.273.36.228659722503<p>Cyclic adenosine 3',5'-monophosphate-elevating agents inhibit transforming growth factor-beta-induced SMAD3/4-dependent transcription via a protein kinase A-dependent mechanism</p>SchillerMVerrecchiaFMauvielAOncogene200322558881889010.1038/sj.onc.120687114654784<p>Modulation of DNA damage by pentoxifylline and alpha-tocopherol in skin fibroblasts exposed to Gamma rays</p>LaurentCPougetJPVoisinPRadiat Res20051641637210.1667/RR338315966766