Targeting the TGF-beta1 pathway to prevent normal tissue injury after cancer therapy

Mitchell S Anscher, Mitchell S Anscher

Abstract

With >10,000,000 cancer survivors in the U.S. alone, the late effects of cancer treatment are a significant public health issue. Over the past 15 years, much work has been done that has led to an improvement in our understanding of the molecular mechanisms underlying the development of normal tissue injury after cancer therapy. In many cases, these injuries are characterized at the histologic level by loss of parenchymal cells, excessive fibrosis, and tissue atrophy. Among the many cytokines involved in this process, transforming growth factor (TGF)-beta1 is thought to play a pivotal role. TGF-beta1 has a multitude of functions, including both promoting the formation and inhibiting the breakdown of connective tissue. It also inhibits epithelial cell proliferation. TGF-beta1 is overexpressed at sites of injury after radiation and chemotherapy. Thus, TGF-beta1 represents a logical target for molecular therapies designed to prevent or reduce normal tissue injury after cancer therapy. Herein, the evidence supporting the critical role of TGF-beta1 in the development of normal tissue injury after cancer therapy is reviewed and the results of recent research aimed at preventing normal tissue injury by targeting the TGF-beta1 pathway are presented.

Conflict of interest statement

Disclosures: Mitchell S. Anscher: Consultant/advisory role: Civa Tech.

The content of this article has been reviewed by independent peer reviewers to ensure that it is balanced, objective, and free from commercial bias. No financial relationships relevant to the content of this article have been disclosed by the independent peer reviewers.

Figures

Figure 1.
Figure 1.
TGF-β expression in radiation-induced lung injury. (A): Radiation (RT)-induced lung injury in a rat model. Fischer 344 rats were irradiated to 28 Gy in one fraction to the right hemithorax and sacrificed at 6 weeks or 6 months after RT. Slides were stained with either hematoxylin & eosin (top row) or Masson's trichrome (bottom row). A nonirradiated control lung (left column) demonstrates the normal honeycomb architecture of the lung. At 6 weeks after irradiation (middle column), one begins to see thickening of the alveolar walls resulting from edema, corresponding to the inflammatory phase, but with little fibrosis. At 6 months (right column), there is complete loss of normal alveolar architecture, with extensive fibrosis (green staining in bottom right panel). Reprinted from Stone HB, Coleman CN, Anscher MS et al. Effects of radiation on normal tissue: Consequences and mechanisms. Lancet Oncol 2003;4:529–536, with permission from Elsevier. (B): Fischer 344 rats were irradiated to 28 Gy in one fraction to the right hemithorax, sacrificed 6 months after radiation, and stained for transforming growth factor (TGF)-β1. There is very little staining for TGF-β1 in the control lung (nonirradiated) in contrast to the irradiated lung, which demonstrates greater expression of TGF-β1 in regions of fibrosis.
Figure 2.
Figure 2.
TGF-β expression in chemotherapy-induced liver injury. Section of normal liver (A) and liver from patient that died of hepatic veno-occlusive disease after high-dose chemotherapy (B). In (B), evidence of extensive fibrosis around the central veins (C) is noted (arrow) and there is greater staining for transforming growth factor β1 (reddish brown areas) immediately adjacent to regions of fibrosis than in the normal liver in (A). P, portal vein. Reprinted from Anscher MS, Kong FM, Jirtle RL. The relevance of transforming growth factor beta 1 in pulmonary injury after radiation therapy. Lung Cancer 1998;19:109–120, with permission from Elsevier.
Figure 3.
Figure 3.
Altering dose on the basis of an individual patient's sensitivity to toxicity can affect the therapeutic ratio. Dosing of chemotherapy or radiation is based on sensitivity to toxicity that is based on population averages (solid line). In reality, any population also contains individuals that will be either more sensitive (dashed line) or more resistant (dot-dash line) to treatment toxicity than the average population. The sensitive patients, which probably comprise ≤5%–10% of the overall population [1, 2], nonetheless drive the dosing schemes. In this example, by holding the acceptable complication incidence at 10%, a resistant patient (b) could receive an approximately 5% greater dose than an average patient (a) and a 10% greater dose than a sensitive patient. Dose differences of this magnitude have been associated with differences in outcome.
Figure 4.
Figure 4.
Correlation of plasma transforming growth factor (TGF)-β1 concentration and the risk for pulmonary drug toxicity (PDT) or hepatic veno-occlusive disease (VOD) after high-dose chemotherapy for advanced breast cancer. Blood samples were taken from patients after induction chemotherapy but prior to administration of high-dose chemotherapy (day −6) and bone marrow transplant. There is a strong correlation between the plasma TGF-β1 concentration at day −6 and the risk for either PDT or VOD developing after subsequent high-dose chemotherapy. Reprinted from Anscher MS, Kong FM, Jirtle RL. The relevance of transforming growth factor beta 1 in pulmonary injury after radiation therapy. Lung Cancer 1998;19:109–120, with permission from Elsevier.
Figure 5.
Figure 5.
Potential targets for intervention in the TGF-β1 pathway (partial list) [17, 74, 114]. The inactive form of TGF-β1 can be activated through the action of several factors (green ovals), including ROS, proteases, integrins, and thrombospondin-1. The active form of TGF-β1 (yellow) can then signal through either Smad-dependent (light blue) or Smad-independent pathways (orange, pink, gray). Any point along these pathways might be targeted by potential inhibitors. Abbreviations: ERK, extracellular signal–related kinase; MAPK, mitogen-activated protein kinase; mTOR, mammalian target of rapamycin; PI3K, phosphatidylinositol 3′ kinase; ROS, reactive oxygen species; TGF, transforming growth factor.

References

    1. Emami B, Lyman J, Brown A, et al. Tolerance of normal tissue to therapeutic irradiation. Int J Radiat Oncol Biol Phys. 1991;21:109–122.
    1. Milano MT, Constine LS, Okunieff P. Normal tissue tolerance dose metrics for radiation therapy of major organs. Semin Radiat Oncol. 2007;17:131–140.
    1. Mehta V. Radiation pneumonitis and pulmonary fibrosis in non-small-cell lung cancer: Pulmonary function, prediction, and prevention. Int J Radiat Oncol Biol Phys. 2005;63:5–24.
    1. Kong FM, Pan C, Eisbruch A, et al. Physical models and simpler dosimetric descriptors of radiation late toxicity. Semin Radiat Oncol. 2007;17:108–120.
    1. Tsoutsou PG, Koukourakis MI. Radiation pneumonitis and fibrosis: Mechanisms underlying its pathogenesis and implications for future research. Int J Radiat Oncol Biol Phys. 2006;66:1281–1293.
    1. Brush J, Lipnick SL, Phillips T, et al. Molecular mechanisms of late normal tissue injury. Semin Radiat Oncol. 2007;17:121–130.
    1. Rodemann HP, Blaese MA. Responses of normal cells to ionizing radiation. Semin Radiat Oncol. 2007;17:81–88.
    1. Travis EL. Genetic susceptibility to late normal tissue injury. Semin Radiat Oncol. 2007;17:149–155.
    1. Anscher MS, Chen L, Rabbani Z, et al. Recent progress in defining mechanisms and potential targets for prevention of normal tissue injury after radiation therapy. Int J Radiat Oncol Biol Phys. 2005;62:255–259.
    1. Laiho M, Keski-Oja J. Transforming growth factors-beta as regulators of cellular growth and phenotype. Crit Rev Oncog. 1992;3:1–26.
    1. Massagué J, Cheifetz S, Laiho M, et al. Transforming growth factor-beta. Cancer Surv. 1992;12:81–103.
    1. Massagué J, Blain SW, Lo RS. TGFβ signaling in growth control, cancer, and heritable disorders. Cell. 2000;103:295–309.
    1. Lamouille S, Derynck R. Cell size and invasion in TGF-β-induced epithelial to mesenchymal transition is regulated by activation of the mTOR pathway. J Cell Biol. 2007;178:437–451.
    1. Derynck R, Akhurst RJ. Differentiation plasticity regulated by TGF-β family proteins in development and disease. Nat Cell Biol. 2007;9:1000–1004.
    1. Massagué J. TGFβ in Cancer. Cell. 2008;134:215–230.
    1. Taylor IW, Wrana JL. SnapShot: The TGFβ pathway interactome. Cell. 2008;133:378.e1.
    1. Elliott RL, Blobe GC. Role of transforming growth factor beta in human cancer. J Clin Oncol. 2005;23:2078–2093.
    1. Chen C, Wang XF, Sun L. Expression of transforming growth factor beta (TGFβ) type III receptor restores autocrine TGFβ1 activity in human breast cancer MCF-7 cells. J Biol Chem. 1997;272:12862–12867.
    1. Grady WM, Myeroff LL, Swinler SE, et al. Mutational inactivation of transforming growth factor beta receptor type II in microsatellite stable colon cancers. Cancer Res. 1999;59:320–324.
    1. Villanueva A, Garcia C, Paules AB, et al. Disruption of the antiproliferative TGF-β signaling pathways in human pancreatic cancer cells. Oncogene. 1998;17:1969–1978.
    1. Yan Z, Deng X, Friedman E. Oncogenic Ki-ras confers a more aggressive colon cancer phenotype through modification of transforming growth factor-beta receptor III. J Biol Chem. 2001;276:1555–1563.
    1. Derynck R, Akhurst RJ, Balmain A. TGF-β signaling in tumor suppression and cancer progression. Nat Genet. 2001;29:117–129.
    1. Branton MH, Kopp JB. TGF-β and fibrosis. Microbes Infect. 1999;1:1349–1365.
    1. Border WA, Brees D, Noble NA. Transforming growth factor-beta and extracellular matrix deposition in the kidney. Contrib Nephrol. 1994;107:140–145.
    1. Munger J, Huang X, Kawakatsu H, et al. The integrin αvß6 binds and activates latent TGFβ1: A mechanism for regulating pulmonary inflammation and fibrosis. Cell. 1999;96:319–328.
    1. Lawrence D. Identification and activation of latent transforming growth factor β. Methods Enzymol. 1991;198:327–336.
    1. Koli K, Saharinen J, Hyytiäinen M, et al. Latency, activation, and binding proteins of TGF-β. Microsc Res Tech. 2001;52:354–362.
    1. Godár S, Horejsi V, Weidle UH, et al. M6P/IGFII-receptor complexes urokinase receptor and plasminogen for activation of transforming growth factor-ß1. Eur J Immunol. 1999;29:1004–1013.
    1. Blakytny R, Ludlow A, Martin GE, et al. Latent TGF-β1 activation by platelets. J Cell Physiol. 2004;199:67–76.
    1. Barcellos-Hoff MH, Dix TA. Redox-mediated activation of latent transforming growth factor-β1. Mol Endocrinol. 1996;10:1077–1083.
    1. Annes JP, Munger JS, Rifkin DB. Making sense of latent TGFβ activation. J Cell Sci. 2003;116:217–224.
    1. Ashcroft GS. Bidirectional regulation of macrophage function by TGF-β. Microbes Infect. 1999;1:1275–1282.
    1. Boyd F, Massagué J. Transforming growth factor-β inhibition of epithelial cell proliferation linked to the expression of a 53-kDa membrane receptor. J Biol Chem. 1989;264:2272–2278.
    1. Hakenjos L, Bamberg M, Rodemann H. TGF-ß1-mediated alterations of rat lung fibroblast differentiation resulting in the radiation-induced fibrotic phenotype. Int J Radiat Biol. 2000;76:503–509.
    1. Fajardo LF, Prionas SD, Kwan HH, et al. Transforming growth factor β1 induces angiogenesis in vivo with a threshold pattern. Lab Invest. 1996;74:600–608.
    1. Phillips GD, Whitehead RA, Stone AM, et al. Transforming growth factor beta (TGF-B) stimulation of angiogenesis: An electron microscopic study. J Submicroscop Cytol Pathol. 1993;25:149–155.
    1. Roberts AB, McCune BK, Sporn MB. TGFβ regulation of extracellular matrix. Kidney Int. 1992;41:557–559.
    1. Peters H, Border WA, Noble NA. Targeting TGF-β overexpression in renal disease: Maximizing the antifibrotic action of angiotensin II blockade. Kidney Int. 1998;54:1570–1580.
    1. Border WA, Ruoslahti E. Transforming growth factor-ß in disease: The dark side of tissue repair. J Clin Invest. 1992;90:1–7.
    1. Bartram U, Speer CP. The role of transforming growth factor beta in lung development and disease. Chest. 2004;125:754–765.
    1. Anscher MS, Kong FM, Jirtle RL. The relevance of transforming growth factor beta 1 in pulmonary injury after radiation therapy. Lung Cancer. 1998;19:109–120.
    1. Isaka Y, Brees DK, Ikegaya K, et al. Gene therapy by skeletal muscle expression of decorin prevents fibrotic disease in rat kidney. Nat Med. 1996;2:418–423.
    1. Akagi Y, Isaka Y, Arai M, et al. Inhibition of TGF-β 1 expression by antisense oligonucleotides suppressed extracellular matrix accumulation in experimental glomerulonephritis. Kidney Int. 1996;50:148–155.
    1. Tsushima H, Kawata S, Tamura S, et al. Reduced plasma transforming growth factor-ß1 levels in patients with chronic hepatitis C after interferon-α therapy: Association with regression of hepatic fibrosis. J Hepatol. 1999;30:1–7.
    1. Border W, Okuda S, Languino LR, et al. Suppression of experimental glomerulonephritis by antiserum against transforming growth factor beta 1. Nature. 1990;346:371–374.
    1. Rabbani ZN, Batinic-Haberle I, Anscher MS, et al. Long-term administration of a small molecular weight catalytic metalloporphyrin antioxidant, AEOL 10150, protects lungs from radiation-induced injury. Int J Radiat Oncol Biol Phys. 2007;67:573–580.
    1. Rabbani ZN, Anscher MS, Zhang X, et al. Soluble TGFβ type II receptor gene therapy ameliorates acute radiation-induced pulmonary injury in rats. Int J Radiat Oncol Biol Phys. 2003;57:563–572.
    1. Rabbani ZN, Anscher MS, Golson ML, et al. Overexpression of extracellular superoxide dismutase reduces severity of radiation-induced lung toxicity through downregulation of the TGF-β signal transduction pathway. Int J Radiat Oncol Biol Phys. 2003;57:S158–S159.
    1. Nishioka A, Ogawa Y, Mima T, et al. Histopathologic amelioration of fibroproliferative change in rat irradiated lung using soluble transforming growth factor-beta (TGF-β) receptor mediated by adenoviral vector. Int J Radiat Oncol Biol Phys. 2004;58:1235–1241.
    1. Nakao A, Fujii M, Matsumura R, et al. Transient gene transfer and expression of Smad7 prevents bleomycin-induced lung fibrosis in mice. J Clin Invest. 1999;104:5–11.
    1. Kang S, Rabbani Z, Folz R, et al. Overexpression of extracellular superoxide dismutase protects mice from radiation induced lung injury. Int J Radiat Oncol Biol Phys. 2002;54:78.
    1. Giri SN, Hyde DM, Hollinger MA. Effect of antibody to transforming growth factor ß on bleomycin induced accumulation of lung collagen in mice. Thorax. 1993;48:959–966.
    1. Daniels CE, Wilkes MC, Edens M, et al. Imatinib mesylate inhibits the profibrogenic activity of TGF-β and prevents bleomycin-mediated lung fibrosis. J Clin Invest. 2004;114:1308–1316.
    1. Anscher MS, Crocker IR, Jirtle RL. Transforming growth factor-beta 1 expression in irradiated liver. Radiat Res. 1990;122:77–85.
    1. Franko AJ, Sharplin J, Ghahary A, et al. Immunohistochemical localization of transforming growth factor beta and tumor necrosis factor alpha in the lungs of fibrosis-prone and “non-fibrosing” mice during the latent period and early phase after irradiation. Radiat Res. 1997;147:245–256.
    1. Liguang C, Larrier N, Rabbani ZN, et al. Assessment of the protective effect of keratinocyte growth factor on radiation-induced pulmonary toxicity in rats. Int J Radiat Oncol Biol Phys. 2003;57:S162.
    1. Andarawewa KL, Paupert J, Pal A, et al. New rationales for using TGFβ inhibitors in radiotherapy. Int J Radiat Biol. 2007;83:803–811.
    1. Portess DI, Bauer G, Hill MA, et al. Low-dose irradiation of nontransformed cells stimulates the selective removal of precancerous cells via intercellular induction of apoptosis. Cancer Res. 2007;67:1246–1253.
    1. Flanders KC, Sullivan CD, Fujii M, et al. Mice lacking Smad3 are protected against cutaneous injury induced by ionizing radiation. Am J Pathol. 2002;160:1057–1068.
    1. Peters CA, Stock RG, Cesaretti JA, et al. TGFB1 single nucleotide polymorphisms are associated with adverse quality of life in prostate cancer patients treated with radiotherapy. Int J Radiat Oncol Biol Phys. 2008;70:752–759.
    1. Anscher MS, Peters WP, Reisenbichler H, et al. Transforming growth factor beta as a predictor of liver and lung fibrosis after autologous bone marrow transplantation for advanced breast cancer. N Engl J Med. 1993;328:1592–1598.
    1. Kong FM, Anscher MS, Murase T, et al. Elevated plasma transforming growth factor beta 1 levels in breast cancer patients decrease after surgical removal of the tumor. Ann Surg. 1995;222:155–162.
    1. Kong F-M, Anscher M, Xiong Z, et al. Elevated circulating transforming growth factor β1 levels decreased after radiotherapy in patients with lung cancer, cervical cancer and Hodgkin's disease: A possible tumor marker. Int J Radiat Oncol Biol Phys. 1995;32(suppl 1):239.
    1. Novakova-Jiresova A, Van Gameren MM, Coppes RP, et al. Transforming growth factor-beta plasma dynamics and post-irradiation lung injury in lung cancer patients. Radiother Oncol. 2004;71:183–189.
    1. De Jaeger K, Seppenwoolde Y, Kampinga HH, et al. Significance of plasma transforming growth factor-beta levels in radiotherapy for non-small-cell lung cancer. Int J Radiat Oncol Biol Phys. 2004;58:1378–1387.
    1. Anscher MS, Kong FM, Andrews K, et al. Plasma transforming growth factor β1 as a predictor of radiation pneumonitis. Int J Radiat Oncol Biol Phys. 1998;41:1029–1035.
    1. Anscher MS, Kong FM, Marks LB, et al. Changes in plasma transforming growth factor beta during radiotherapy and the risk of symptomatic radiation-induced pneumonitis. Int J Radiat Oncol Biol Phys. 1997;37:253–258.
    1. Anscher MS, Murase T, Prescott DM, et al. Changes in plasma TGF beta levels during pulmonary radiotherapy as a predictor of the risk of developing radiation pneumonitis. Int J Radiat Oncol Biol Phys. 1994;30:671–676.
    1. Kong FM, Ao X, Wang L, et al. The use of blood biomarkers to predict radiation lung toxicity: A potential strategy to individualize thoracic radiation therapy. Cancer Control. 2008;15:140–150.
    1. Fu XL, Huang H, Bentel G, et al. Predicting the risk of symptomatic radiation-induced lung injury using both the physical and biologic parameters V(30) and transforming growth factor beta. Int J Radiat Oncol Biol Phys. 2001;50:899–908.
    1. Anscher MS, Marks LB, Shafman TD, et al. Using plasma transforming growth factor beta-1 during radiotherapy to select patients for dose escalation. J Clin Oncol. 2001;19:3758–3765.
    1. Anscher MS, Marks LB, Shafman TD, et al. Risk of long-term complications after TGF-β1-guided very-high-dose thoracic radiotherapy. Int J Radiat Oncol Biol Phys. 2003;56:988–995.
    1. Teicher BA. Malignant cells, directors of the malignant process: Role of transforming growth factor-beta. Cancer Metastasis Rev. 2001;20:133–143.
    1. Bierie B, Moses HL. Tumour microenvironment: TGFβ: The molecular Jekyll and Hyde of cancer. Nat Rev Cancer. 2006;6:506–520.
    1. Beck C, Schreiber H, Rowley D. Role of TGF-β in immune-evasion of cancer. Microsc Res Tech. 2001;52:387–395.
    1. Shariat SF, Kattan MW, Traxel E, et al. Association of pre- and postoperative plasma levels of transforming growth factor beta(1) and interleukin 6 and its soluble receptor with prostate cancer progression. Clin Cancer Res. 2004;10:1992–1999.
    1. Shariat SF, Walz J, Roehrborn CG, et al. Early postoperative plasma transforming growth factor-β1 is a strong predictor of biochemical progression after radical prostatectomy. J Urol. 2008;179:1593–1597.
    1. Wikström P, Damber J, Bergh A. Role of transforming growth factor-β1 in prostate cancer. Microsc Res Tech. 2001;52:411–419.
    1. Muraoka-Cook RS, Kurokawa H, Koh Y, et al. Conditional overexpression of active transforming growth factor β1 in vivo accelerates metastases of transgenic mammary tumors. Cancer Res. 2004;64:9002–9011.
    1. Muraoka RS, Dumont N, Ritter CA, et al. Blockade of TGF-β inhibits mammary tumor cell viability, migration, and metastases. J Clin Invest. 2002;109:1551–1559.
    1. Bandyopadhyay A, Agyin JK, Wang L, et al. Inhibition of pulmonary and skeletal metastasis by a transforming growth factor-beta type I receptor kinase inhibitor. Cancer Res. 2006;66:6714–6721.
    1. Teicher BA, Ikebe M, Ara G, et al. Transforming growth factor-beta 1 overexpression produces drug resistance in vivo: Reversal by decorin. In Vivo. 1997;11:463–472.
    1. Teicher BA, Holden SA, Ara G, et al. Transforming growth factor-beta in in vivo resistance. Cancer Chemother Pharmacol. 1996;37:601–609.
    1. Kong F, Anscher M, Sporn T, et al. Loss of mannose 6-phosphate/insulin-like growth factor receptor (M6P/IGF2R) may lead to an elevated transforming growth factor β1 (TGFβ1) in circulation. Presented at the 45th Annual Meeting of the Radiation Research Society; May 3–7, 1997; Providence, RI.
    1. Dennis PA, Rifkin DB. Cellular activation of latent transforming growth factor ß requires binding to the cation-independent mannose 6-phosphate/insulin-like growth factor type II receptor. Proc Natl Acad Sci U S A. 1991;88:580–584.
    1. Anscher M, Kong F, Sporn T, et al. Loss of a normally functioning mannose 6-phosphate/insulin-like growth factor 2 receptor may lead to an elevated plasma transforming growth factor ß1 level: Clinical implications. Presented at the 9th Lorne Cancer Conference; February 13–16, 1997; Victoria, Australia.
    1. Wrana JL, Attisano L, Cárcamo J, et al. TGF β signals through a heteromeric protein kinase receptor complex. Cell. 1992;71:1003–1014.
    1. Chen RH, Ebner R, Derynck R. Inactivation of the type II receptor reveals two receptor pathways for the diverse TGF-β activities. Science. 1993;260:1335–1338.
    1. Yang L, Tredget EE, Ghahary A. Activation of latent transforming growth factor-β1 is induced by mannose 6-phosphate/insulin-like growth factor-II receptor. Wound Repair Regen. 2000;8:538–546.
    1. Hankins GR, De Souza AT, Bentley RC, et al. M6P/IGF2 receptor: A candidate breast tumor suppressor gene. Oncogene. 1996;12:2003–2009.
    1. Dahms NM, Lobel P, Kornfeld S. Mannose-6 phosphate receptors and lysosomal enzyme targeting. J Biol Chem. 1989;264:12115–12118.
    1. Kong F, Pulford D, Bandera M, et al. M6P/IGF2R tumor suppressor gene is mutated in lung cancer. Proc Am Assoc Cancer Res. 1999;40:733.
    1. Kong FM, Anscher MS, Sporn TA, et al. Loss of heterozygosity at the mannose 6-phosphate insulin-like growth factor 2 receptor (M6P/IGF2R) locus predisposes patients to radiation-induced lung injury. Int J Radiat Oncol Biol Phys. 2001;49:35–41.
    1. Li Y-Q, Ballinger JR, Nordal RA, et al. Hypoxia in radiation-induced blood-spinal cord barrier breakdown. Cancer Res. 2001;61:3348–3354.
    1. Vujaskovic Z, Anscher MS, Feng Q-F, et al. Radiation-induced hypoxia may perpetuate late normal tissue injury. Int J Radiat Oncol Biol Phys. 2001;50:851–855.
    1. Anscher MS, Thrasher B, Rabbani Z, et al. Antitransforming growth factor-beta antibody 1D11 ameliorates normal tissue damage caused by high-dose radiation. Int J Radiat Oncol Biol Phys. 2006;65:876–881.
    1. Falanga V, Zhou L, Yufit T. Low oxygen tension stimulates collagen synthesis and COL1A1 transcription through the action of TGF-β1. J Cell Physiol. 2002;191:42–50.
    1. Norman JT, Clark IM, Garcia PL. Hypoxia promotes fibrogenesis in human renal fibroblasts. Kidney Int. 2000;58:2351–2366.
    1. Westbury CB, Pearson A, Nerurkar A, et al. Hypoxia can be detected in irradiated normal human tissue: A study using the hypoxic marker pimonidazole hydrochloride. Br J Radiol. 2007;80:934–938.
    1. Haydont V, Mathe D, Bourgier C, et al. Induction of CTGF by TGF-β1 in normal and radiation enteritis human smooth muscle cells: Smad/Rho balance and therapeutic perspectives. Radiother Oncol. 2005;76:219–225.
    1. Haydont V, Riser BL, Aigueperse J, et al. Specific signals involved in the long-term maintenance of radiation-induced fibrogenic differentiation: A role for CCN2 and low concentration of TGF-β1. Am J Physiol Cell Physiol. 2008;294:C1332–C1341.
    1. Anscher MS, Thrasher B, Zgonjanin L, et al. Small molecular inhibitor of transforming growth factor-beta protects against development of radiation-induced lung injury. Int J Radiat Oncol Biol Phys. 2008;71:829–837.
    1. Riley PA. Free radicals in biology: Oxidative stress and the effects of ionizing radiation. Int J Radiat Biol. 1994;65:27–33.
    1. Kang SK, Rabbani ZN, Folz RJ, et al. Overexpression of extracellular superoxide dismutase protects mice from radiation-induced lung injury. Int J Radiat Oncol Biol Phys. 2003;57:1056–1066.
    1. Vujaskovic Z, Batinic-Haberle I, Rabbani ZN, et al. A small molecular weight catalytic metalloporphyrin antioxidant with superoxide dismutase (SOD) mimetic properties protects lungs from radiation-induced injury. Free Radic Biol Med. 2002;33:857–863.
    1. Inomata S, Takahashi H, Nagata M, et al. Acute lung injury as an adverse event of gefitinib. Anticancer Drugs. 2004;15:461–467.
    1. Haston CK, Travis EL. Murine susceptibility to radiation-induced pulmonary fibrosis is influenced by a genetic factor implicated in susceptibility to bleomycin-induced pulmonary fibrosis. Cancer Res. 1997;57:5286–5291.
    1. Nethery DE, Moore BB, Minowada G, et al. Expression of mutant human epidermal receptor 3 attenuates lung fibrosis and improves survival in mice. J Appl Physiol. 2005;99:298–307.
    1. Faress JA, Nethery DE, Kern EF, et al. Bleomycin-induced pulmonary fibrosis is attenuated by a monoclonal antibody targeting HER2. J Appl Physiol. 2007;103:2077–2083.
    1. Cho HJ, Kang JH, Kim T, et al. Suppression of PAI-1 expression through inhibition of the EGFR-mediated signaling cascade in rat kidney fibroblast by ascofuranone. J Cell Biochem. 2009;107:335–344.
    1. Wang Q, Wang Y, Hyde DM, et al. Reduction of bleomycin induced lung fibrosis by transforming growth factor beta soluble receptor in hamsters. Thorax. 1999;54:805–812.
    1. Kolb M, Margetts PJ, Galt T, et al. Transient transgene expression of decorin in the lung reduces the fibrotic response to bleomycin. Am J Respir Crit Care Med. 2001;163:770–777.
    1. Stone HB, Coleman CN, Anscher MS, et al. Effects of radiation on normal tissue: Consequences and mechanisms. Lancet Oncol. 2003;4:529–536.
    1. Koli K, Myllärniemi M, Keski-Oja J, et al. Transforming growth factor-beta activation in the lung: Focus on fibrosis and reactive oxygen species. Antioxid Redox Signal. 2008;10:333–342.

Source: PubMed

3
Subscribe