Radiol Oncol 2002; 36(2): 109-19. review Radiotherapy- and chemotherapy-induced normal tissue damage: the role of cytokines and adhesion molecules Pavlína Plevová Department of Radiotherapy, University Hospital, Ostrava, Czech Republic Background. Ionising radiation and cytostatic agents used in cancer therapy exert damaging effects on normal tissues and induce a complex response at the cellular and molecular levels. Cytokines and adhesion molecules are involved in this response. Methods. Published data on the given topic have been reviewed. Results and conclusions. Various cytokines and adhesion molecules, including tumor necrosis factor a, in-terleukins-1,-2,-4, and -6, interferon y, granulocyte macrophage- and macrophage- colony stimulating factors, transforming growth factor ß, platelet-derived growth factor, insulin-like growth factor I, fibroblast and epidermal growth factors, platelet-activating factor, intercellular adhesion molecule-1, vascular cell adhesion molecule-1, E- and P-selectins are involved in the response of normal tissues to ionizing radiation- and chemotherapy-induced normal tissues damage and are co-responsible for some side effects of these treatment modalities, including fever, anorexia and fatigue, suppression of hematopoiesis, both acute and late local tissue response. Key words: cytokines - radiation effects - drug effects; antineoplatic agents - adverse effects; neoplasms - drug therapy - radiotherapy; cell adhesion molecules - radiation effects - drug effects Received 25 February 2002 Accepted 2 April 2002 Correspondence to: Pavlína Plevová, M.D., Fr. Lyska 8, Ostrava-Belsky les, 700 30 Czech Republic, Phone: +420 69 6717841 or +420 69 6984370; Fax: +420 69 6919010; E-mail: pavlina.plevova@volny.cz Acknowledgements: I am indebted to Pavel Vodvárka and Lenka Zivcakova for their helpful comments. 110 Plevová P / Radiotherapy, chemotherapy and cytokines Introduction Injury of human tissues and generally of mammalian organism ones, activates a nonspecific, but highly complex immune response at the intracellular and intercellular levels, with the aim to protect the tissues and the whole organism against exogenous damage and to regenerate the damaged tissues. Cyto-kines and adhesion molecules are released during this response and mediate intercellular interactions among effectors of immune and other systems.1-3 Cytokines are soluble polypeptides regulating and determining the character of immune response.1,2 The main source of cytokines are macrophages, but neutrophils, lymphocytes, platelets, endothelial cells, fibroblasts, and microglia, acting as the macrophage of the central nervous system (CNS), are able to release cytokines as well.3-5 Cytokines are components of a large, complex signalling network. The great variety of cell types that are able to release cytokines and the great diversity of biological effects of each cytokine is confusing. The ability of individual cytokines to induce or inhibit the synthesis of other cytokines and often of its own further complicates the specification of biological functions of individual cytokines.1, 2, 6 Adhesion molecules mediate the adherence of leukocytes to the molecules on other cells or to extracellular matrix ligands and are thus involved in leukocyte activation, circulation and localization to inflammatory sites.7 Both radiotherapy and chemotherapy exert damaging effects on normal tissues in cancer patients and, consequently, induce an immune response in these tissues. The role of cyto-kines in this response and the possibilities to modulate it in order to lower the risk of side effects of these treatment modalities are reviewed in this article. Radiol Oncol 2002; 36(2): 109-19. Ionizing radiation- and chemotherapy-induced cytokines and adhesion molecules The production of cytokines may result from either DNA damage in cells leading to inhibition in cell cycle progression and resulting in cell death 8,9 or from biochemical changes in cellular environment and metabolism induced by the interaction of ionizing radiation (or chemotherapy) with the target cell (Figure 1).3 The cytokines and adhesion molecules that have been observed to be produced in response to ionizing radiation at the mRNA or protein levels in various human or other mammalian cells or tissues both in vitro and vivo are summarized in Table 1. The mediators have been shown to respond to irradiation in a dose-dependent manner.22, 25, 31, 40, 44, 51, 52 The threshold dose of irradiation ranges from 0.5 to 2 Gy for different proteins, except murine brain cells where the threshold dose of 7 Gy has been found.21 Their production is also time-dependent, peaking usually at 4-24 hours after irradiation with subsequent decrease to normal levels within 24 hours to a few days.21 Increased intercellular adhesion molecule-1 (ICAM-1) expression persists for at least several days; 46, 47, 51 expression of transforming growth factor-ß (TGF-ß) is often delayed to weeks or months after irradiation and persists for months; 38, 40 in the murine lung and small intestine, increased levels of interleu-kin-1 (IL-1), IL-4, tumor necrosis factor-a (TNF-a), and platelet derived growth factor (PDGF) also persist for weeks and months after irradiation; 18, 29, 34 reelevation of TNF-a at 2-3 months and its continued overexpression more than half a year after irradiation has been observed in brain cells.21 Although the immune response to chemo-therapeutic drugs has not been studied as extensively as that to irradiation, it is highly probable that the administration of such toxic and aggressive agents as anticancer drugs induces the protective acute phase response in the human organism. Increased producti- Plevová P / Radiotherapy, chemotherapy and cytokines Ill Cytokine Cytokine receptor Cytokine Inflammatory/ protective proteins: A • DNA repair genes • immune receptors • inflammatory enzyms • adhesion molecules • chemokines •cytokines mRNA Activation signals Cell membrane Figure 1. The mechanism of the acute phase response to irradiation at the cellular level. The interaction of ionizing radiation with the target cell induces biochemical changes in the cellular environment and metabolism, generation of free oxygen radicals and arachidonic acid derivatives (a). These changes activate kinase cascades (b) in macrophages and other responsive cells, leading to the activation of transcription factors, such as nuclear factor-?B (NF-?B), c-jun, c-fos (c). Active transcription factors bind to specific recognition elements of DNA in the nucleus and upregulate transcription of genes coding proteins involved in the immune and inflammatory responses (d), leading to an increased formation of messenger RNA (mRNA) and protein (e, f). The proteins released as the end result of this response include cytokines, chemokines, inflammatory enzymes, adhesion molecules, immune receptors and deoxyribonucleic acid repair genes.7, 9, 10 Some of the cytokines are able to reactivate this cascade through the reactivation of particular transcription factors thus amplifying the response (g). The released cytokines also activate similar response in other cells through binding to the cytokine receptors (h).3, 7, 9, 11-15 Other mediators of inflammation such as serotonine, histamine, bradykinine, and nitric oxide, and other acute phase proteins are also released. All of this leads to clinical inflammatory manifestations, both at the local level with vasodilation, erythema, edema and pain, and at the systemic level with various behavioral, biochemical and nutritional changes. A part of this response is the release of mediators that attenuate it and cause it to resolve.3, 6, 11 on of inflammatory cytokines induced by various anticancer agents has been demonstrated both in vitro and in vivo (Table 1). Chemotherapy-induced immune system activation in vivo is known especially from allogeneic bone marrow transplantation (BMT); conditioning regimens including total body irradiation and high-dose chemotherapy can contribute to the activation of host immune cells with inflammatory cytokine release and upregulation of adhesion molecules. 69, 70 It is highly probable that a wide range of other known cytokines, chemokines, adhesion molecules and other mediators of inflammation not studied in this model so far are released in response to ionizing irradiation and chemotherapy. Radiol Oncol 2002; 36(2): 109-19. h 112 Plevová P / Radiotherapy, chemotherapy and cytokines Table 1. Cytokine and adhesion molecule expression in irradiated tissues Mediator Irradiated tissue or cell type – animal model Irradiated human Induced by CT tissue or cells in vitro Induced by CT in vivo m. macrophages, lung cells, in vitro (16-19) TNF-alpha m. lung, BM, spleen, brain, in vivo (17-21) m. BMT model, serum (22) IL-1 alpha or beta IL-6 PB MNCs, in vitro (23-25) BAL cells, in vivo (25) serum, BMT pts. (26) paclitaxol, CTX, doxorubicine (MNCs) (55-57) bleomycine (lung) (58) CTX (+TBI), CTX+busulfan (serum, BMT CR) (59) m. spleen cells, in vitro (27) lung macrophages, paclitaxol (MNCs) m. lung, BM, spleen, small gut, brain, in vivo (18, 20, 21, 27-30) m. BMT model, serum (22) in vitro (31) (55, 56) serum after brain irradiation, in vivo (32) CTX (+TBI, BMT CR; serum) (60) IL-2 doxorubicine (MNCs) (61, 62) IL-2r PB lymphocytes, in vitro (33) IL-4 m. lung, in vivo (34) m. BM, spleen, in vivo (20) m. BMT model, serum (10) macrophages, MTX, ARA-C (MNCs) epithelial cells, lung (63) fibroblasts, in vitro (35-37) IFNgamma CTX (serum, BMT CR) (65, 66) m. lung, small gut, liver, in vivo TGF-beta (17, 29, 38-40) pig skin, in vivo (41) PB MNCs, in vitro (23) 5-FU (fibroblasts, bleomycine, CTX ECs) (64) (lung) (67,68) colon, small gut, CTX (+TBI, BMT CR; in vivo (42, 43) serum) (60) PDGF m. small gut, in vivo (29) MNCs, BAL cells, in vitro (25) IGF-I MNCs, in vitro (25) FGF bovine ECs, in vitro (44) EGF serum after brain irradiation, in vivo (32) PAF saliva, in vivo (45) ICAM-1 m. lung, brain, in vivo (46-48) ECs, in vitro, in vivo (46, 49-51) skin, in vitro (49, 52) VCAM-1 skin, ECs, in vitro (52) E-selectin m. lung, in vivo (47) ECs, skin, in vitro (51-54) P-selectin m. lung, in vivo (47) Abbreviations: CT, chemotherapy; TNF, tumor necrosis factor; m., murine; BM, bone marrow; BMT, bone marrow transplantation; PB, peripheral blood; MNCs, mononuclear cells; BAL, bronchoalveolar lavage; pts., patients; CTX, cyclophosamide; TBI, total body irradiation; BMT CR, bone marrow transplantation conditioning regimen; IL, in-terleukin; MTX, methotrexate; ARA-C, cytosinarabinoside; IFN, interferon; GM-CSF, granulocyte macrophage-colony stimulating factor; M-CSF, macrophage-colony stimulating factor; TGF, transforming growth factor; 5-FU, 5-fluorouracil; ECs, endothelial cells; PDGF, platelet-derived growth factor; BAL, bronchoalveolar lavage; IGF-I, insulin-like growth factor-I; FGF, fibroblast growth factor; EGF, epidermal growth factor; PAF, platelet-activating factor; ICAM-1, intercellular adhesion molecule-1; VCAM-1, vascular cell adhesion molecule-1. Radiol Oncol 2002; 36(2): 109-19. Plevová P / Radiotherapy, chemotherapy and cytokines 113 Role of cytokines in pathogenesis of radiotherapy- and chemotherapy-induced side effects A simplified list of immune effects of cytokines and adhesion molecules is involved in Table 2. A variety of changes in normal tissues are induced by ionizing radiation, depending on the total dose, fractionation schedule, and volume treated.9 Most normal tissue effects can be attributed to cell killing (cytokines mediate repair processes here); some cannot, though. For instance, the nausea and vomiting that can occur within hours after irradiation of the upper abdomen; the acute edema or erythema that results from radiation-induced acute inflammation and associated vascular leakage; the fatigue in patients receiving irradiation to a large volume, especially within the abdomen; the somnolence and headache after cranial irradiation. These are most likely mediated by radiation-induced inflammatory cytokines.79 Similar symptoms can be observed after chemotherapy. Nausea, vomiting and fever occurring immediately after chemotherapy and the associated anorexia and fatigue are likely to be also mediated by inflammatory cytokines, such as TNF-cc, IL-1, and IL-6. Similar symptoms are associated with infectious diseases; immune response is nonspecific.3 The hematotoxicity of chemotherapy and radiotherapy has been generally attributed to the direct damage of rapidly dividing hematopoietic progenitor cells.79, 80 However, several apparently physiological inhibitors of hema-topoesis have been identified that directly or indirectly suppress the proliferative response of progenitor cells to stimulating cytokines; these include TGF-ß, macrophage inhibitory protein-1a (MIP-1a), TNF-cc, and interferon-y (IFN-y).81-84 Irradiation can induce IFN-y, TGF-ß and TNF-cc release, and these and other cytokines might be responsible for hemato-poiesis suppression after local irradiation not involving large volumes of bone marrow.66 Cell-mediated immune response plays a key role in the pathogenesis of the so-called “anemia of chronic disease”.85 This response may also be involved in the pathogenesis of chemotherapy- and radiotherapy-induced anemia. TNF-cc and IL-1 reduce proliferation of erythroid progenitor cells by exerting either a direct inhibitory effect or an indirect effect via the action of IFN-cc or IFN-ß.86 TNF-cc, IL-1 and IL-6 are able to induce hypoferremia by increasing iron uptake into monocytes/macrophages and synthesis of ferritin, thus contributing to efficient storage of the acquired iron;87, 88 IFN-y and IL-2 enhance strongly the expression of the transferrin receptor, the essential protein for iron uptake.89, 90 Iron deprivation enhances the activity of cytokines such as IFN-cc or TNF-cc and cytotoxic effects of macrophages in order to produce a protective response as efficient as possible. However, hypoferremia reduces hem synthesis in erythroid progenitor cells.85 Proinflammatory cytokines are released immediately after CNS irradiation. The basis of demyelination is the interplay of cytokines between endothelial cells, oligodendrocytes, astrocytes and microglia.21 The disruption of the endothelium leads to the infiltration of lymphocytes into the tissue and initiation of immunologic mechanisms involved in the pathogenesis of encephalopathy and myelopathy.21, 28, 91, 92 Apoptosis, i.e. programmed cell death, is a common mechanism of cell death in response to ionizing radiation and anticancer drug exposure.93-95 The transmembrane forms of TNF-cc and TGF-ß released by peripheral blood mononuclear cells have been shown to be involved in the radiation-induced apoptosis of the endothelial cells.24, 72 Basic fibroblast growth factor (FGF), on the other hand, protects endothelial cells from radiation-induced apoptosis in vitro.93 The induction of apoptosis is co-responsible for normal tissue damage by irradiation and probably by chemotherapy, too. Radiol Oncol 2002; 36(2): 109-19. 114 Plevová P / Radiotherapy, chemotherapy and cytokines Table 2. Biologic effects of cytokines and adhesion molecules. Mediator Effects principal proinflammatory cytokines with profound effects on the processing of the acute phase response (1, 11); activation of neutrophils, T-, B-lymphocytes, TNF-alpha macrophages, ECs, fibroblasts; induction of cytokines and other inflammatory IL-1, IL-6 protein release, upregulation of AM expression, activation of the hypothalamus-hypophysis-adrenal gland synthesis, TNF-alpha induced apoptosis in ECs (1, 69, 71-73) IL-2 activation of lymphocytes and monocytes (74) growth factor of B-lymphocytes; inhibition of the release of mediators of inflammation (1) IL-4 IFN gamma stimulation of phagocytic abilities of macrophages, differentiation of T-lymphocytes, cytotoxic efects (1) suppression of the inflammatory response, stimulation of fibroblast proliferation (40) TGF-beta PDGF, FGF, IGF-I stimulation of proliferation of fibroblasts (25, 38) G-CSF, GM-CSF, M-CSF hematopoietic growth factors playing a pivotal role in regulation of BM progenitor cell proliferation (75) EGF stimulation of epithelial proliferation, and differentiation (76) involved in transmigration of leukocytes into the site of inflammation in PAF cooperation with adhesion molecules; mediator of angiogenesis induced by inflammatory cytokines (77) SCF stimulation of hematopoietic stem cells (78) AMs of the immunoglobulin family (e.g. ICAM-1, VCAM-1) mediate firm adherence of leukocytes to ECs with subsequent extravasation (7) Selectins (E-, P-selectins) mediate loose contact between leukocytes and ECs, i.e. leukocyte rolling (7) Fibrosis is a delayed result of radiation-and chemotherapy-associated tissue damage. It represents a reparation process at the time when the damaging insult does not act on the tissue.79 Fibrosis is more than a mark of tissue damage; it is damaging in itself.96 In association with fibrosis development, increased expression of TGFs-ß have been found in the irradiated animal lung, liver and skin tissue and in the lungs of bleomycin- and cyclophos-phamide-treated mice.17, 38-41, 60, 67, 68 Their expression is increased both in early and late stages of tissue reaction.41 TGFs-ß have che-moattractive effects on fibroblasts and inflammatory cells and promote cell proliferation; they regulate expression, synthesis and storage of components of extracellular ma- trix.40, 97 Other cytokines, such as TNF-a, IL-1, IL-4, PDGF, FGF, insulin-like growth factor-I (IGF-I) are also likely to play an important role in fibrosis development due to fibroblast stimulation.18, 25, 29, 34, 38 Proinflammatory cytokines and adhesion molecules are involved in the pathophysio-logy of BMT-related complications. In an experimental model, the intensification of the conditioning regimen by increasing the total body irradiation dose results in an increased graft-versus-host disease severity. Total body irradiation and allogeneic immune cells appear to act synergistically to damage the gastrointestinal tract, thereby permitting an increased translocation of bacterial endotoxin (lipopolysaccharide) into the systemic circula- Radiol Oncol 2002; 36(2): 109-19. Plevová P / Radiotherapy, chemotherapy and cytokines 115 tion. As total body irradiation increases macrophage and endothelial cell priming to lipo-polysaccharide resulting in higher systemic levels of inflammatory cytokines, adhesion molecules, and probably other mediators of inflammation, an increase in the severity of graft-versus-host-disease results.22, 50, 60, 69 Conclusions Although the interpretation of cytokine concentrations has to be made with caution with respect to the used method of analysis,98 there is no doubt that cytokines are released in response to ionizing radiation and chemotherapy. They are involved in the development of side effects of these treatment modalities-that, however, have a protective role in the organism and tissues. References 1. Borish L, Rosenwasser LJ. Update on cytokines. J Allergy Clin Immunol 1996; 97: 719-34. 2. Thalmeier K, Meissner P, Reisbach G, Hültner L, Mortensen BT, Brechtel A, et al. Constitutive and modulated cytokine expression in two permanent human bone marrow stromal cell lines. Exp Hema-tol 1996; 24: 1-10. 3. Koj A. Initiation of acute phase response and synthesis of cytokines. Biochim Biophys Acta 1996; 1317: 84-94. 4. Laskin DL, Pendino KJ. Macrophages and inflammatory mediators in tissue injury. Annu Rev Pharmacol Toxicol 1995; 35: 655-77. 5. Collins CE, Rampton DS. Review article: platelets in inflammatory bowel disease – pathogenetic role and therapeutic implications. Aliment Pharmacol Ther 1997; 11: 237-47. 6. Gabay C, Kushner I. Acute-phase proteins and other systemic responses to inflammation. N Eng J Med 1999; 340: 448-54. 7. Luster AD. Chemokines – chemotactic cytokines that mediate inflammation. N Engl J Med 1998; 338: 436-45. 8. Iliakis G. Cell cycle regulation in irradiated and nonirradiated cells. Semin Oncol 1997; 24: 602-15. 9. Perez CA, Brady LW, Roti Roti JL. Overview. In: Perez CA, Brady LW, editors. Principles and practice of radiation oncology. 3rd ed. Philadelphia: Lip-pincott- Raven Publishers; 1998. p. 1-78. 10. Xun CQ, Thompson JS, Jennings CD, Brown SA, Widmer MB. Effect of total body irradiation, bu-sulfan-cyclophosphamide, or cyclophosphamide conditionin on inflammatory cytokine release and development of acute and chronic graft- versus-host disease in H-2-incompatible transplanted SCID mice. Blood 1994; 83: 2360-7. 11. Baumann H, Gauldie J. The acute phase response. Immunol Today 1994; 15: 74-80. 12. Weichselbaum RR, Hallahan DE, Fuks Z, Kufe D. Radiation induction of immediate early genes: effectors of the radiation-stress response. Int J Radiat Oncol Biol Phys 1994; 30: 229-34. 13. Kyriakis JM, Avruch J. Protein kinase cascades activated by stress and inflammatory cytokines. Bioessays 1996; 18: 567-77. 14. Ransohoff RM: Cellular responses to interferons and other cytokines: The JAK- STAT paradigm. N Engl J Med 1998; 338: 616-8. 15. Barnes PJ, Karin M. Nuclear factor-(B – a pivotal transcription factor in chronic inflammatory diseases. N Engl J Med 1997; 336: 1066-71. 16. Iwamoto KS, McBride WH. Production of 13-hydroxyoctadecadienoic acid and tumor necrosis factor-a by murine peritoneal macrophages in response to irradiation. Radiat Res 1994; 139: 103-8. 17. Franko AJ, Sharplin J, Ghahary A, Barcellos-Hoff MH. Immunohistochemical localization of transforming growth factor a and tumor necrosis factor ß in the lungs of fibrosis-prone and “non-fibro-sing” mice during latent and early phase after irradiation. Radiat Res 1997; 147: 245-56. 18. Johnston CJ, Piedboeuf B, Rubin P, Williams JP, Baggs R, Finkelstein JN. Early and persistent alterations in the expression of interleukin-1 alpha, in-terleukin-1 beta and tumor necrosis factor alpha mRNA levels in fibrosis-resistant and sensitive mice after thoracic irradiation. Radiat Res 1996; 145: 762-7. 19. Redlich CA, Gao X, Rockwell S, Kelley M, Elias JA. IL-11 enhances survival and decreases TNF production after radiation-induced thoracic injury. J Immunol 1996; 157: 1705-10. 20. Chang CM, Limanni A, Baker WH, Dobson ME, Kalinich JF, Patchen ML. Sublethal gamma irradi- Radiol Oncol 2002; 36(2): 109-19. 116 Plevová P / Radiotherapy, chemotherapy and cytokines ation increases IL-1a, IL-6, and TNF-a mRNA levels in murine hematopoietic tissues. J Interferon Cytokine Res 1997; 17: 567-72. 21. Hong JH, Chiang CS, Campbell IL, Sun JR, Withers HR, McBride WH. Induction of acute phase gene expression by brain irradiation. Int J Radiat Oncol Biol Phys 1995; 33: 619-26. 22. Hill GR, Crawford JM, Cooke KR, Brinson YS, Pan L, Ferrara JL. Total body irradiation and acute graft-versus-host disease: the role of gastrointestinal damage and inflammatory cytokines. Blood 1997; 90: 3204-13. 23. Krivenko S, Dryk S, Komarovskaya M, Karkanitsa L. Ionizing radiation increases TNF/cachectin production by human peripheral blood mononuclear cells in vitro. Int J Hematol 1992; 55: 127-30. 24. Lindner H, Holler E, Ertl B, Multhoff G, Schre-glmann M, Klauke I, et al. Peripheral blood mono-nuclear cells induce programmed cell death in human endothelial cells and may prevent repair: role of cytokines. Blood 1997; 89: 1931-8. 25. Thornton SC, Walsh BJ, Bennett S, Robbins JM, Foulcher E, Morgan GW, et al. Both in vitro and in vivo irradiation are associated with induction of macrophage-derived fibroblast growth factors. Clin Exp Immunol 1996; 103: 67-73. 26. Girinsky TA, Pallardy M, Comoy E, Benassi T, Roger R, Ganem G, et al. Peripheral blood corticotro-pin-releasing factor, adrenocorticotropic hormone and cytokine (interleukin beta, inerleukin 6, tumor necrosis factor alpha) levels after high- and low-dose total-body irradiation in humans. Radiat Res 1994; 139: 360-3. 27. Ishihara H, Tsuneoka K, Dimchev AB, Shikita M. Induction of the expression of the interleukin-1 ß gene in mouse spleen by ionizing radiation. Radiat Res 1993; 133: 321-6. 28. Selmaj KW, Raine CS. Tumor necrosis factor mediates myelin and oligodendrocyte damage in vitro. Ann Neurol 1988; 23: 339-46. 29. Lanberg CW, Hauer-Jensen M, Sung CC, Kane CJM. Expression of fibrogenic cytokines in rat small intestine after fractionated irradiation. Radi-other Oncol 1994; 32: 29-36. 30. Richter KK, Fagerhol MK, Carr JC, Winkler JM, Sung CC, Hauer-Jensen M. Association of granu-locyte transmigration with structural and cellular parameters of injury in experimental radiation en-teropathy. Radiat Oncol Investig 1997; 5: 275-82. 31. O’Brien-Ladner A, Nelson ME, Kimler BF, Wesse-lius LJ. Release of interleukin-1 by human alveolar macrophages after in vitro irradiation. Radiat Res 1993; 136: 37-41. 32. Gridley DS, Loredo LN, Slater JD, Archambeau JO, Bedros AA, Andres ML, et al. Pilot evaluation of cytokine levels in patients undergoing radiotherapy for brain tumor. Cancer Detect Prev 1998; 22: 20-9. 33. Xu Y, Greenstock CL, Trivedi A, Mitchel RE. Occupational levels of radiation exposure induce surface expression of interleukin-2 receptors in stimulated human peripheral blood lymphocytes. Radiat Environ Biophys 1996; 35: 89-93. 34. Buttner C, Skupin A, Reimann T, Rieber EP, Unte-regger G, Geyer P, et al. Local production of inter-leukin-4 during radiation-induced pneumonitis and pulmonary fibrosis in rats: macrophages as a prominent source of interleukin-4. Am J Respir Cell Mol Biol 1997; 17: 315-25. 35. Pons I, Gras G, Courberand S, Benveniste O, Dor-mont D. Consequences of gamma-irradiation on inflammatory cytokine regulation in human mo-nocytes/macrophages. Int J Radiat Biol 1997; 71: 157-66. 36. Beetz A, Messer G, Oppel T, van Beuningen D, Peter RU, Kind P. Induction of interleukin 6 by ionizing radiation in a human epithelial cell line: control by corticosteroids. Int J Radiat Biol 1997; 72: 33-43. 37. Brach MA, Gruss HJ, Kaisho T, Asano Y, Hirano T, Herrmann F. Ionizing radiation induces expression of interleukin 6 by human fibroblasts involving activation of nuclear factor-?B. J Biol Chem 1993; 268: 8466-72. 38. Rubin P, Johnston CJ, Williams JP, McDonald S, Finkelstein JN. A perpetual cascade of cytokines postirradiation leads to pulmonary fibrosis. Int J Radiat Oncol Biol Phys 1995; 33: 99-109. 39. Johnston CJ, Piedboeuf B, Baggs R, Rubin P, Fin-kelstein JN. Differences in correlation of mRNA gene expression in mice sensitive and resistant to radiation-induced pulmonary fibrosis. Radiat Res 1995; 142: 197-203. 40. Anscher MS, Crocker IR, Jirtle RL. Transforming growth factor-ß1 expression in irradiated liver. Radiat Res 1990; 122: 77-85. 41. Martin M, Lefaix JL, Pinton P, Crechet F, Daburon F. Temporal modulation of TGF-ß1 and ß-actin gene expression in pig skin and muscular fibrosis after ionizing radiation. Radiat Res 1993; 134: 63-70. 42. Canney PA, Dean S. Transforming growth factor beta: A promotor of late connective tissue injury following radiotherapy? Br J Radiol 1990; 63: 620-3. Radiol Oncol 2002; 36(2): 109-19. Plevová P / Radiotherapy, chemotherapy and cytokines 117 43. Richter KK, Fink LM, Hughes BM, Sung CC, Hau-er-Jensen M. Is the loss of endothelial thrombomo-dulin involved in the mechanism of chronicity in late radiation enteropathy? Radiother Oncol 1997; 44: 65-71. 44. Haimovitz-Friedman A, Vlodavsky I, Chaudhuri A, Witte L, Fuks Z. Autocrine effects of fibroblast growth factor in repair of radiation damage in en-dothelial cells. Radiat Res 1991; 51: 2552-8. 45. Mc-Manus LM, Ostrom KK, Lear C, Luce EB, Gander DL, Pinckard RN, et al. Radiation-induced increased platelet-activating factor activity in mixed saliva. Lab Invest 1993; 68: 118-24. 46. Hallahan DE, Virudachalam S. Intercellular adhesion molecule 1 knockout abrogates radiation induced pulmonary inflammation. Proc Natl Acad Sci USA 1997; 94: 6432-7. 47. Hallahan DE, Virudachalam S. Ionizing radiation mediates expression of cell adhesion molecules in distinct histological patterns within the lung. Cancer Res 1997; 57: 2096-9. 48. Olschowska JA, Kyrkanides S, Harvey BK, O’Bani-on MK, Williams JP, Rugin P, et al. ICAM-1 induction in the mouse CNS following irradiation. Brain Behav Immun 1997; 11: 273-85. 49. Behrends U, Peter RU, Hintermeier-Knabe R, Eis-sner G, Holler E, Bornkamm GW, et al. Ionizing radiation induces human intercellular adhesion molecule-1 in vitro. J Invest Dermatol 1994; 103: 726-30. 50. Eissner G, Lindner H, Behrends U, Kölch W, Hie-ke A, Klauke I, et al. Influence of bacterial endoto-xin on radiation-induced activation of human en-dothelial cells in vitro and in vivo: protective role of IL-10. Transplantation 1996; 62: 819-27. 51. Hallahan D, Kuchibhotla J, Wyble C. Cell adhesion molecules mediate radiation-induced leukocyte adhesion to the vascular endothelium. Cancer Res 1996; 56: 5150-5. 52. Heckmann M, Douwes K, Peter R, Degitz K. Vascular activation of adhesion molecule mRNA and cell surface expression by ionizing radiation. Exp Cell Res 1998; 238: 148-54. 53. Hallahan DE, Clark ET, Kuchibhotla J, Gewertz B, Collins T. E-selectin gene induction by ionizing radiation. Biochem Biophys Res Commun 1995; 217: 784-95. 54. Hallahan DE, Kuchibhotla J, Wyble C. Sialyl Lewis X mimetics attenuate E-selectin-mediated adhesion of leukocytes to irradiated human endothelial cells. Radiat Res 1997; 147: 41-7. 55. Ding AH, Porteu F, Sanchez E, Nathan CF. Shared actions of endotoxin and Taxol on TNF receptors and TNF release. Science 1990; 248: 370-2. 56. Bogdan C, Ding A. Taxol, a microtubule-stabilizing antineoplastic agent, induces expression of tumor necrosis factor alpha and interleukin-1 in macrophages. J Leukoc Biol 1992; 52: 119-22. 57. Shi F, MacEwen EG, Kurzman ID. In vitro and in vivo effect of doxorubicin combined with liposo-me-encapsulated muramyl tripeptide on canine monocyte activation. Cancer Res 1993; 53: 3986-91. 58. Piguet PF, Collart MA, Grau GE, Kapanchi Y, Vassalli P. Tumor necrosis factor/cachectin plays a key role in bleomycin-induced pneumopathy and fibrosis. J Exp Med 1989; 170: 655-63. 59. Holler E, Kolb HJ, Mittermüller J, Kaul M, Ledde-rose G, Duell T, et al. Modulation of acute graft-versus-host disease after allogeneic BMT by tumor necrosis factor a (TNFa) release in the course of pretransplant conditioning: Role of conditioning regimens and prophylactic application of a mono-lonal antibody neutralizing human TNFa (MAK 195F). Blood 1995; 86: 890-9. 60. Panoskaltsis-Mortari A, Taylor PA, Yaeger TM, Wangesteen OD, Bitterman PB, Ingbar DH, et al. The critical early proinflammatory events associated with idiopathic pneumonia syndrome in irradiated murine allogeneic recipients are due to donor T cell infusion and potentiated by cyclophospha-mide. J Clin Invest 1997; 100: 1015-27. 61. Ehrke MJ, Maccubbin D, Ryoyama K, Cohen SA, Mihich E. Correlation between adriamycin-indu-ced augmentation of interleukin 2 production and cell-mediated cytotoxicity in mice. Cancer Res 1986; 46: 54-60. 62. Abdul Hamied TA, Parker D, Turk JL. Effects of adriamycin, 4-hydroperoxycyclophoshamide and ASTA Z 7557 (INN mafosfamide) on the release of IL-2 and IL-1 in vitro. Int J Immunopharmacol 1987; 9: 355-61. 63. Dryk SI, Karkanitsa LV, Komarovskaya ME. Effects of cytostatic drugs on interleukin-6 production by human peripheral blood mononuclear cells in vitro. Ann Hematol 1993; 66 Suppl II: A93. 64. Clarke E, Rice GC, Weeks RS, Jenkins N, Nelson R, Bianco JA, et al. Lisofylline inhibits transforming growth factor ß release and enhances triline-age hematopoietic recovery afer 5-fluorouracil treatment in mice. Cancer Res 1996; 56: 105-12. 65. Niederwieser D, Herold M, Woloszczuk W. Endogenous IFN-gamma during human bone marrow transplantation. Transplantation 1990; 50: 620-5. Radiol Oncol 2002; 36(2): 109-19. 118 Plevová P / Radiotherapy, chemotherapy and cytokines 66. Schwaighofer H, Kernan NA, O’Reilly RJ, Brankova J, Nachbaur D, Herold M, et al. Serum levels of cytokines and secondary messages after T-cell-de-pleted and non-T-cell-depleted bone marrow transplantation: influence of conditioning and he-matopoietic reconstitution. Transplantation 1996; 62: 947-53. 67. Hoyt DG, Lazo JS. Alterations in pulmonary mR-NA encoding procollagens, fibronectin and transforming growth factor-beta precede bleomycin-in-duced pulmonary fibrosis. J Pharmacol Exp Ther 1988; 246: 765-71. 68. Hoyt DG, Lazo JH. Early increases in pulmonary mRNA encoding procollagens and transforming growth factor-ß in mice sensitive to cyclophospha-mide-induced pulmonary fibrosis. J Pharmacol Exp Ther 1989; 249: 38-43. 69. Antin JH, Ferrara JLM. Cytokine dysregulation and acute graft-versus-host disease. Blood 1992; 80: 2964-8. 70. Xun CQ, Thompson JS, Jenings CD, Brown SA, Widmer MB. Effect of total body irradiation, bu-sulfan-cyclophosphamide, or cyclophosphamide conditioning on inflammatory cytokine release and development of acute and chronic graft-ver-sus-host disease in H-2-incompatible tranplanted SCID mice. Blood 1994; 83: 2360-7 71. Wong GG, Giannotti JW, Hewick RM, Clark SC, Ogawa M. Interleukin 6: identification as a hema-topoietic colony-stimulating factor. Behring Inst Mitt 1988; 83: 40-7. 72. Eissner G, Kohlhuber F, Grell M, Ueffing M, Sche-urich P, Hieke A, et al. Critical involvement of transmembrane tumor necrosis factor-a in endo-thelial programmed cell death mediated by ionizing radiation and bacterial endotoxin. Blood 1995; 86: 4184-93. 73. Eigler A, Sinha B, Hartmann G, Endres S. Taming TNF: strategies to restrain this proinflammatory cytokine. Immunol Today 1997; 18: 487-92. 74. Théze J, Alzari PM, Bertoglio J. Interleukin 2 and its receptors: recent advances and new immunolo-gical fuctions. Immunol Today 1996; 17: 481-6. 75. Griffin JC. Clinical applications of colony stimulation factors. Oncology 1988; 2: 15-23. 76. Cohen S. The epidermal growth factor (EGF). Cancer 1983; 51: 1787-91. 77. Mantovani A, Bussolino F, Introna M. Cytokine regulation of endothelial cell function: from molecular level to the bedside. Immunol Today 1997; 18: 231-40. 78. Kaushansky K. Thrombopoietin and the hematopoietic stem cell. Blood 1998; 92: 1-3. 79. Withers HR, McBride WH. Biologic basis of radiation. In: Perez CA, Brady LW, eds. Principles and practice of radiation oncology. Philadelphia:Lippin-cott-Raven Publishers; 1998. p. 79-118. 80. Cadman EC, Durivage HJ. Cancer chemotherapy. In: Wilson JD, Braunwald E, Isselbacher KJ, Petersdorf RG, Martin JB, Fauci AS, Root RK, editors. Harrinson’s Principals of Internal Medicine. 12th ed. New York: McGraw-Hill, Inc.; 1991. p. 1587-99. 81. Schall TS. Biology of the RANTES/SIS cytokine family. Cytokine 1991; 3: 165-83. 82. Wolpe SD, Cerami A. Macrophage inflammatory proteins 1 and 2: members of a novel superfamily of cytokines. FASEB J 1989; 3: 2565-73. 83. Miller-Graziano CL, Szabo G, Griffey K, Mehta B, Kodys K, Catalano D. Role of elevated monocyte transforming growth factor (TGF-cc) production in post-trauma immunosuppression. J Clin Immunol 1992; 11: 95-102. 84. Ruscetti FW, Jacobsen SE, Birchenall-Roberts M, Broxmeyer HE, Engelman GL, Dubois C, et al. Role of transforming growth factor ß 1 in regulation of hematopoiesis. Negative regulators of hemato-poiesis. Ann NY Acad Sci 1991; 628: 31-43. 85. Weiss, Wachter H, Fuchs D. Linkage of cell-mediated immunity to iron metabolism. Immunol Today 1995; 16: 495-500. 86. Means RT Jr., Krantz SB. Progress in understanding the pathogenesis of the anemia of chronic disease. Blood 1992; 80: 1639-47. 87. Alvarez-Hernández X, Licéaga J, McKay IC, Brock JH. Induction of hypoferremia and modulation of macrophage iron metabolism by tumor necrosis factor. Lab Invest 1989; 61: 319-22. 88. Kobune M, Kohgo Y, Kato J, Miyazaki E, Niitsu Y. Interleukin-6 enhances hepatic transferrin uptake and ferritin expression in rats. Hepatology 1994; 19: 1468-75. 89. Taetle R, Honeysett JM. y-interferon modulates human monocyte/macrophage transferrin receptor expression. Blood 1988; 71: 1590-5. 90. Seiser C, Teixeira S, Kühn LC. Interleukin-2-de-pendent transcriptional regulation of transferrin receptor mRNA. J Biol Chem 1993; 268: 13074-80. 91. Merrill JE. Effects of interleukin-1 and tumor necrosis factor-alpha on astrocytes, microglia, oligodendrocytes, and glial precursors in vitro. Dev Ne-urosci 1991; 13: 130-7. Radiol Oncol 2002; 36(2): 109-19. Plevová P / Radiotherapy, chemotherapy and cytokines 119 92. Rubin P, Constine LS, Williams JP. Late effects of cancer treatment: radiation and drug toxicity. In: Perez CA, Brady LW, editors. Principles and practice of radiation oncology. 3rd ed. Philadelphia: Lip-pincott-Raven Publishers; 1998. p. 155-212. 93. Fuks Z, Persaud RS, Alfieri A, McLoughlin M, Ehleiter D, Schwartz SL, et al. Basic fibroblast growth factor protects endothelial cells against radiation-induced programmed cell death in vitro and in vivo. Cancer Res 1994; 54: 2582-90. 94. Langley RE, Bump EA, Quartuccio SG, Medeiros D, Braunhut SJ. Radiation-induced apoptosis in microvascular endothelial cells. Br J Cancer 1997; 75: 666-72. 95. Debatin KM. Cytotoxic drugs, programmed cell death, and the immune system: defining new roles in an old play. J Natl Cancer Inst 1997; 89: 750-1. 96. Fajardo LF L-G. Morphology of radiation effects on normal tissues. In: Perez CA, Brady LW, editors. Principles and practice of radiation oncology. 3rd ed. Philadelphia: Lippincott-Raven Publishers; 1998. p. 143-54. 97. Chegini N. The role of growth factors in peritoneal healing: transforming growth factor ß (TGF-ß). Eur J Surg 1997; Suppl. 577: 17-23. 98. Barnes A. Measurement of serum cytokines. Lancet 1998; 352: 324-5. Radiol Oncol 2002; 36(2): 109-19.