Curr Gastroenterol Rep (2014) 16:383 DOI 10.1007/s11894-014-0383-3

SMALL INTESTINE (J SELLIN, SECTION EDITOR)

Radiation Enteritis Ali H. Harb & Carla Abou Fadel & Ala I. Sharara

# Springer Science+Business Media New York 2014

Abstract Radiation enteritis continues to be a major health concern in recipients of radiation therapy. The incidence of radiation enteritis is expected to continue to rise during the coming years paralleling the unprecedented use of radiotherapy in pelvic cancers. Radiation enteritis can present as either an acute or chronic syndrome. The acute form presents within hours to days of radiation exposure and typically resolves within few weeks. The chronic form may present as early as 2 months or as long as 30 years after exposure. Risk factors can be divided into patient and treatment-related factors. Chronic radiation enteritis is characterized by progressive obliterative endarteritis with exaggerated submucosal fibrosis and can manifest by stricturing, formation of fistulae, local abscesses, perforation, and bleeding. In the right clinical context, diagnosis can be confirmed by cross-sectional imaging, flexible or video capsule endoscopy. Present treatment strategies are directed primarily towards symptom relief and management of emerging complications. Recently, however, there has been a shift towards rational drug design based on improved understanding of the molecular basis of disease in an effort to limit the fibrotic process and prevent organ damage. Keywords Radiation . Enteritis . Enteropathy . Radiotherapy

radiation energy exposure. Radiation enteritis is a term commonly used to describe both small as well as large intestinal injury after radiation exposure. This, by definition, excludes injury to the rectum, referred to separately as radiation proctitis. The purpose of this review is to assess the current state of knowledge on radiation enteritis by addressing the burden, presentation, risk factors, pathogenesis, prevention, diagnosis, and management of this emerging problem. This article deals primarily with chronic radiation enteritis and the small intestinal response to radiation exposure. Acute radiation enteritis is briefly discussed while radiation proctitis is not covered. Historical Background The first report of radiation-induced gut injury was described in 1897 [1] when experiments revealed radium-induced death of diseased cells [2], only 1 year after the introduction of radiation as a treatment modality and 2 years after the discovery of the X-ray itself by Roentgen [3]. Walsh described the phenomenon as ‘a direct inflammation of the gastrointestinal mucous membranes’ [1]. A quarter of a century later, Warren and Whipple conducted the first systematic study on dogs to characterize the effects of radiation on intestinal structure and function [4].

Introduction Clinical Burden Definition and Scope Radiation enteritis is by simple definition an inflammatory process occurring at the level of the intestines as a response to This article is part of the Topical Collection on Small Intestine A. H. Harb : C. Abou Fadel : A. I. Sharara Division of Gastroenterology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon A. I. Sharara (*) Division of Gastroenterology, American University of Beirut Medical Center, P.O. Box 11-0236/16-B, Beirut, Lebanon e-mail: [email protected]

Recent reports indicate that as many as 70 % of cancer patients receive radiotherapy during the course of their treatment [5]. Estimates indicate that around 300,000 patients received radiotherapy for pelvic malignancy worldwide in 2007 alone [6]. This widespread use of radiation in cancer therapy [5], along with the high incidence of radiation-induced side effects affecting up to 75 % of radiotherapy receivers [7], has led to a tremendous increase in the incidence of radiation enteritis. The concomitant use of chemotherapy [8] as well as the continued improvement in cancer prognosis are expected to increase this even further [9]. Still, it is believed that the true burden of radiation enteritis may even be larger due to under

383, Page 2 of 9

diagnosis and underreporting [10]. Lastly, the significant morbidity and mortality carried by RE in the post-therapy period is not the only thing to be feared [11]. Collateral damage to normal tissue is the main reason for dose limitation, a potential hurdle to treatment [12].

Presentation Radiation enteritis can present as either an acute or chronic syndrome. The acute form presents within hours to days of radiation exposure and typically resolves within few weeks. The classical presentation includes nausea, vomiting, abdominal pain, diarrhea, and tenesmus. The chronic form may present as early as 2 months or as long as 30 years after exposure [13, 14]. Clinical manifestations differ but may include abdominal pain, malabsorption, diarrhea, cachexia, intestinal bleeding, obstruction, and even perforation [15]. Risk Factors Risk factors for RE can be divided into patient- and treatmentrelated factors. Patient factors include previous abdominal or pelvic surgical operations, low body mass index, concomitant cardiovascular disease, atherosclerosis, diabetes, older age, female sex, and tobacco abuse [16, 17]. Risk factors related to treatment include the dose of radiation used and its fractionation schedule, as well as the volume of small bowel in the radiation field [18] and the concomitant use of chemotherapy [19].

Pathophysiology Acute Radiation Enteritis Acute radiation enteritis is thought to be a consequence of the direct exposure of the rapidly dividing intestinal epithelium, especially crypt stem cells, to the cytotoxic effects of radiation [20]. Apoptosis [21] mediated by the overexpression of p53 [22] and the down-regulation of Bcl-2 [23] plays a central role in the process. The resultant mucositis interferes with the intestinal barrier function and may lead to translocation of luminal bacteria [24, 25]. Recently, however, an increasing body of evidence indicates that injury to the endothelium of the microvasculature supplying the intestines might also be contributory [26–28]. A complex interplay between radiation-induced reduction of thrombomodulin in endothelial cells [29], excess activation of thrombin, and alteration of platelet adhesion properties [30] leads to a more thrombophilic endothelium [31], which is thought to be behind not only acute radiation enteritis but also chronic radiation enteritis if perpetuated [32, 33]. The

Curr Gastroenterol Rep (2014) 16:383

diarrhea experienced by most patients is thought to result from a combined suppression of the Na+/K+ pump, lactose intolerance, and intestinal dysmotility leading to bacterial overgrowth [34–36]. Lebrun et al showed a 40 % decrease in Na-K pump activity in rats 4 days following irradiation. A similar decrease in pump activity was described by Sandle in animal models of colitis. This resultant impairment in sodium and chloride absorption caused watery diarrhea [37]. Chronic Radiation Enteritis Chronic radiation enteritis (CRE), on the other hand, is characterized by progressive obliterative endarteritis [38]. This occlusive vasculitis causes tissue ischemia that in turn leads to an exaggerated submucosal fibrosis [39]. Fibrosis is mostly mediated by TGF-β1 [40–42] and its downstream mediator, connective tissue growth factor (CTGF) [9]. Fibrosis worsens the existing ischemia and sustains the problem. Telangiectatic vessels form in response [43], which, along with lymphatic dilation, complete the microscopic picture [18]. Macroscopically, CRE is manifested by stricturing, formation of fistulae, local abscesses, perforation, and bleeding [44]. Altered motility [45] and malabsorption [46] are additional features. Lately, accumulating evidence suggests that luminal microbiota might have a game-changing influence on human response to radiation [47]. This influence is thought to be mediated by toll-like receptors [48]. Experiments carried out on germ-free mice support an enhanced resistance to radiation-induced colitis [49, 50], while other studies point to microbial dysbiosis as a risk factor for RE [51]. A small study comparing patients who developed diarrhea post-radiotherapy to those who did not found that patients with diarrhea had a different microbial profile at baseline in comparison to the other group. Patients with diarrhea, in addition, developed an alteration in their fecal microbial profile post radiation. These findings suggest a role of gut microbiota in determining diarrhea in the host post radiation exposure [52].

Diagnosis The diagnosis of RE might be challenging especially that its symptoms are nonspecific and sometimes multifactorial [15]. A history of radiation exposure and an appropriate combination of symptoms should not prevent the clinician from first excluding recurrent neoplasia [15]. Cross-sectional imaging with either computed tomography enterography (CTE) (Fig. 1) or magnetic resonance imaging (MRI) might be prudent especially when the patient suffers weight loss [18]. Magnetic resonance enterography (MRE) has the additional

Curr Gastroenterol Rep (2014) 16:383

Page 3 of 9, 383

Fig. 1 a, b Videocapsule endoscopy showing segmental mucosal edema, erythema, superficial erosions, and functional narrowing in a patient with chronic radiation enteritis presenting with recurrent unexplained abdominal pain and negative abdominal CT. c CT enterography of a 72-year-old woman who received pelvic radiotherapy for cervical carcinoma. Enteric phase images show ileal luminal narrowing and a thickened enhanced bowel wall (arrow) consistent with radiation enteritis [138]. Source of (c): Graça BM, Freire PA, Brito JB, et al. Radiographics 2010; 30: 235–252. With permission of the Radiological Society of North America (RSNA®)

ability to show luminal irregularities [53] without further radiation exposure. Findings are largely non-specific and indicate segmental inflammation. These include bowel thickening, mucosal hyperenhancement, mesenteric stranding, and luminal stricturing [54]. Non-invasive biological markers are unreliable. C-reactive protein is not useful in uncomplicated CRE [55], and data on fecal lactoferrin [56] and calprotectin remain inconclusive [57]. Serum citrulline, a metabolic by-product of intestinal cells, holds the promise of being used to assess the extent of damage as it was found to correlate with the viable enterocyte mass [58–60]. A symptom-directed approach is thus used to guide minimally invasive techniques to investigate symptoms. CTE and MRE are the preferred initial tests when chronic radiation enteritis is suspected. Esophagogastroduodenoscopy may be helpful to exclude other possibilities, and small intestinal contrast studies may disclose subtle strictures, while ileocolonoscopy or balloon-assisted enteroscopy offer the added value of direct visualization and diagnosis of radiation enteritis [18]. When all studies are negative, video capsule endoscopy (VCE) (Fig. 1) remains one of the best methods to reach the diagnosis [61–63]. Because of the high incidence of capsule impaction, a patency capsule should be considered prior to VCE in order to exclude subtle, yet important, luminal narrowing(s) [64, 65]. Bile salt malabsorption [66], bacterial overgrowth [67], and carbohydrate malabsorption [68] often accompany radiation enteritis and are diagnosed with selenium75 homocholic acid conjugated with taurine (SeHCAT) scanning [69], xylose breath test [70], and hydrogen breath test [71], respectively.

Prevention Several studies have validated the use of small bowel studies to determine the amount of small intestines in the pelvis and use the information to modify radiation fields [72–74]. Technical strategies have succeeded by directing the radiation away from normal tissues through improved planning and delivery systems, physical maneuvers [5] like the use of “belly boards” [75], and the use of physical shields. The use of surgery to displace the small bowels from the radiation field has also been reported [13]. Different techniques have been developed including reperitonealization, the use of omental transposition flaps [76], abdominopelvic omentopexy [77], introducing absorbable mesh slings [78–80], and occluding the pelvis with prosthetics [81, 82] or the bladder, or retroverting the uterus [83]. Radioprotective compounds have been used with some success. Cysteine, in mice [84], and amifostine, in a human trial, have been shown to reduce mucosal toxicity [85] of radiation including radiation enteritis [86]. When administered prior to irradiation, L-carnitine resulted in reduction of ileal mucosal damage severity [87].

Management Present treatment strategies are directed mostly towards symptomatic relief. Recently, however, there has been a shift towards rational drug design that utilizes our improved understanding of the molecular basis of disease to curtail the fibrotic process and prevent organ damage.

383, Page 4 of 9

Treatment Dietary modification might be tried to alleviate symptoms: for example, a high-residue and fatty diet may be eliminated with some symptomatic improvement [88, 89]. Patients who develop lactose intolerance must follow a lactose-free diet [71]. Antidiarrheal agents, such as loperamide, increase transit time and may offer relief [68]. Cholestyramine remains the treatment of choice for patients with bile acid malabsorption [15, 90, 91], while antibiotics may be prescribed to treat confirmed bacterial overgrowth [36]. Antidepressants warrant trial in select patients bearing in mind the strong association of CRE with depression [92, 93]. Parenteral nutrition is also an acceptable adjunct when daily energy requirements cannot be met with oral nutrition alone [94]. However, care must be taken due to the higher incidence of cirrhosis and portal hypertension in patients with short bowel syndrome with a history of radiation exposure [95]. Strictures [96], fistulae, perforations, and obstruction, are best treated surgically [97]. However, surgery in this patient population carries a high complication rate, particularly anastomotic leaks [98], making bypass a favored option over resection-anastomosis [99]. Other studies have found that high-risk patients are better managed either medically or endoscopically, and so should avoid surgery due to the high associated morbidity and mortality [100]. Despite improvements in the medical prevention and treatment of radiation enteritis, the need for surgical treatment has not decreased [101]. Probiotics The idea of using probiotics in RE stems from their antioxidant role given that free-radical production initiates injury [102]. Studied species include Lactobacillus plantarum and lactis [103], Lactobacillus gasseri [104], Lactobacillus fermentum E-3 and E-18 [105], and Streptococcus thermophilus [106], all of which were found to have antiinflammatory effects. Probiotics are thought to act as delivery vehicles for superoxide dismutase [102], an enzyme shown to reduce oxidative stress and downregulate adhesion molecule in mice that had undergone abdominal irradiation [107]. Probiotics can also promote the intestinal release of glutathione [102], an important molecule that helps maintain the mucosal integrity and improve oxidative damage in colitis [108]. In addition, probiotics produce exopolysaccharides which act to reduce the intestinal oxidative stress [109]. With few exceptions like pouchitis and infectious diarrhea, the use of probiotics to manage different conditions has generally been disappointing. Nevertheless, trials evaluating the use in RE have been somewhat supportive. A study of almost 500 patients demonstrated that patients receiving the VSL#3 probiotic mixture prophylactically had a decrease in both severity and frequency of post-radiation diarrhea compared

Curr Gastroenterol Rep (2014) 16:383

to placebo [110]. Similarly, patients receiving Lactobacillus rhamnosus required less anti-diarrheals at a later stage compared to placebo and had more consistent stools [111]. The use of probiotics in the prevention of radiation-induced diarrhea was classified as Recommendation Grade C in 2008, meaning there are some positive studies, but more studies are needed to confirm this beneficial role in radiation enteritis [112]. Experimental Therapies Table 1 lists the experimental therapies used in the treatment or prevention of radiation enteritis. The strongest evidence to date has been for the pentoxyfilline–tocopherol combination shown in two cohort studies to cause relief of symptoms [113, 114] and to attenuate fibrosis [115, 116]. Other studies suggest that anti-inflammatory drugs such as sulfasalazine [117] and methylprednisolone [118] might be beneficial. Additionally, there have been case reports of the successful use of hyperbaric oxygen [119] to enhance recovery. Intraperitoneallyadministered octreotide was shown to protect against oxidative injury and preserve mucosal structure in animal models [120]. Similarly, heparin-binding epidermal growth factor (HB-EGF) was successful in decreasing pro-inflammatory cytokines [121], oxygen-free radical production [122], and intestinal histologic injury, preserving gut barrier function when tested on rats [123]. Statins, shown to decrease radiation pneumonitis [124] have been studied in vitro with results indicating the inhibition of CTGF and collagen production [125, 126]. Cyclooxygenase 2 inhibitors have also been shown to decrease radiation injury in rats [127]. As the role of the endothelium in the pathogenesis of RE unfolds, more endothelial-based therapies are being tested in rats. Direct thrombin inhibitors like hirudin successfully improved radiation enteropathy. In comparison, clopidogrel, an ADP receptor antagonist, had an even higher efficacy in ameliorating enteropathy [128, 129]. Unexpectedly, a trial testing the value of heparin in reducing RE showed a paradoxical worsening in radiation injury [130]. One explanation is offered by another study demonstrating that heparin can decrease the affinity between thrombin and thrombomodulin, causing a further decrease in APC levels in this altered endothelial medium [131]. Rational Drug Design Some drugs remain untested in the context of radiation enteritis despite some apparent theoretical significance. Recombinant thrombomodulin, with proven efficacy in disseminated intravascular coagulation, warrants testing in RE due to its potential in restoring the lost thromboresistance in the endothelial milieu [132, 133]. Recombinant activated protein C, with proven efficacy in severe sepsis, may also be able to achieve that goal [134]. The inhibition of proteinase-activated receptor 1, the

Curr Gastroenterol Rep (2014) 16:383

Page 5 of 9, 383

Table 1 Experimental therapies in the treatment and/or prevention of radiation enteritis Experimental therapy

Study group

End point

Mechanism of action

Cysteine [134]

Animals (rats)

Prevention of acute RE

Repletion of glutathione

Amifostine [82]

Humans

L-carnitine [84]

Animals (rats)

Pentoxyfilline-tocopherol [110]

Humans

Sulfasalazine [114]

Humans

Hyperbaric oxygen [116]

Humans

Octreotide [117]

Animals (rats)

Heparin-binding epidermal growth factor (HB-EGF) [120]

Animals (mice)

Statins [122]

Human explants (in vitro) Animals (rats)

Cyclooxygenase-2 Animals (rats) inhibitors (COX-2) [124] Hirudin [125]

Animals (rats)

Clopidogrel [126]

Animals (rats)

Comments

Oxothiazolidine carboxylate (OTZ), a cysteine prodrug, was given orally to rats with sarcoma before and following abdominal radiation → at day 4 there was a significant increase in glutathione in jejunum. Prevention of acute and Antioxidant Patients with pelvic malignancies randomized chronic radiation injury to radiotherapy ± IV amifostine 15 min before radiotherapy → reduction in acute lower GI toxicity at weeks 3–7. Lesser protection observed at 12 month follow-up Prevention of acute RE Antioxidant Whole-body radiation followed by intraperitoneal L-carnitine × 4 days → significant ↓ in serum MCP-1, IFN- γ and reduced severity of mucosal damage. Apoptotic cells of ileal crypt ↑ early after irradiation and ↓ by 4th day Symptomatic relief Antioxidant 21 patients with radiation proctitis/enteritis Inhibit TGF-β1/Smad treated with pentoxifylline and tocopherol targets → 15/21 patients (71 %) experienced relief of symptoms Diarrhea and acute RE Anti-inflammatory Significant ↓ diarrhea with oral sulphasalazine symptoms vs. placebo GI complications of Promotes angiogenesis Improved complete and partial response chronic RE rates (43 and 25 %, respectively) Prevention of acute RE Antioxidant Irradiation ↑ intestinal myeloperoxidase activities, intestinal malondialdehyde levels and NFκ-B expression. Octreotide preserved mucosal structure, ↓ NFκ-B expression, end organ damage and inflammation Acute radiation injury Antioxidant Intraperitoneal HB-EGF led to increase in proliferative crypts and decrease in severe histologic injury and intestinal permeability Attenuate endothelial cell TNF-α antagonist Inhibits Rho activity, type I collagen, and injury and fibrosis fibronectin in vitro. Pravastatin improves radiation enteropathy in rats with decreased deposition of CCN2 and extracellular matrix deposition Decrease in COX-2 Anti-inflammatory Treatment with rofecoxib significantly ↓ overexpression radiation-induced overexpression of COX-2 Acute radiation injury Direct thrombin inhibitor Attenuates radiation-induced mucosal damage, reactive intestinal wall thickening, TGF-β immunoreactivity levels, and collagen III deposition Acute radiation injury Platelet ADP receptor Attenuates the severity of post-radiation antagonist vascular sclerosis and loss of mucosal surface area

downstream mediator of thrombin’s rheological effect through a specific antagonist that is currently being developed [135] is also very appealing [32]. At the other end of the spectrum, IFN-γ, an antagonist of TGF-β, might prevent the fibrotic cascade that leads to damage of irradiated organs [136].

Conclusion Radiation enteritis is an old yet emerging problem spurred by the rise of cancer radiotherapy. Radiation enteritis usually affects patients with some predisposing characteristics, but is

383, Page 6 of 9

largely influenced by treatment-related factors. The modification of these factors has, so far, been key to prevention. A complex interplay between intestinal epithelium, capillary endothelium, and luminal bacteria is thought to underlie its pathogenesis. Cross-sectional imaging and endoscopy are the mainstay of diagnosis. Treatment is usually palliative, but some experimental therapies and rational drug design hold promise at curtailing the inflammatory process and preventing organ damage. Effective treatment and prevention are critical in minimizing the burden of this daunting disease and at preserving radiation as an effective therapy in combating abdominal cancers. Compliance with Ethics Guidelines Conflict of Interest Ala Sharara, Ali Harb, and C. Abou Fadel disclose no conflicts of interest. Human and Animal Rights and Informed Consent This article does not contain any studies with human or animal subjects performed by any of the authors [137].

References 1. Walsh D. Deep tissue traumatism from roentgen ray exposure. Br Med J. 1897;2:272–3. 2. Grubbe EH. The origin and birth of x-ray therapy. Urol Cutan Rev. 1947;51:375–9. 3. Goldstein HI. The discovery of the roentgen rays; the contribution of Wilhelm Konrad Roentgen to science. J Int Coll Surg. 1946;9: 688–93. 4. Warren SL, Whipple GH. Roentgen Ray intoxication: Iv. Intestinal lesions and acute intoxication produced by radiation in a variety of animals. J Exp Med. 1923;38:741–52. 5. Shadad AK, Sullivan FJ, Martin JD, Egan LJ. Gastrointestinal radiation injury: prevention and treatment. World J Gastroenterol. 2013;19:199–208. 6. Andreyev HJ. Gastrointestinal problems after pelvic radiotherapy: the past, the present and the future. Clin Oncol. 2007;19:790–9. 7. Zimmerer T, Bocker U, Wenz F, Singer MV. Medical prevention and treatment of acute and chronic radiation induced enteritis–is there any proven therapy? A short review. Z Gastroenterol. 2008;46: 441–8. 8. Gerard JP, Conroy T, Bonnetain F, et al. Preoperative radiotherapy with or without concurrent fluorouracil and leucovorin in T3-4 rectal cancers: results of FFCD 9203. J Clin Oncol Off J Am Soc Clin Oncol. 2006;24:4620–5. 9. Zhu Y, Zhou J, Tao G. Molecular aspects of chronic radiation enteritis. Clin Investig Med Med Clin Exp. 2011;34:E119–24. 10. Abayomi JC, Kirwan J, Hackett AF. Coping mechanisms used by women in an attempt to avoid symptoms of chronic radiation enteritis. J Hum Nutr Diet Off J Br Diet Assoc. 2009;22:310–6. 11. Hungness ES, Fischer JE. Radiation enteritis: sometimes the treatment is worse than the disease. Curr Surg. 2000;57:573–7. 12. Seal M, Naito Y, Barreto R, Lorenzetti A, Safran P, Marotta F. Experimental radiotherapy-induced enteritis: a probiotic interventional study. J Dig Dis. 2007;8:143–7. 13. Waddell BE, Rodriguez-Bigas MA, Lee RJ, Weber TK, Petrelli NJ. Prevention of chronic radiation enteritis. J Am Coll Surg. 1999;189: 611–24.

Curr Gastroenterol Rep (2014) 16:383 14. Sher ME, Bauer J. Radiation-induced enteropathy. Am J Gastroenterol. 1990;85:121–8. 15. Andreyev HJ, Vlavianos P, Blake P, Dearnaley D, Norman AR, Tait D. Gastrointestinal symptoms after pelvic radiotherapy: role for the gastroenterologist? Int J Radiat Oncol Biol Phys. 2005;62:1464–71. 16. Cox JD, Byhardt RW, Wilson JF, Haas JS, Komaki R, Olson LE. Complications of radiation therapy and factors in their prevention. World J Surg. 1986;10:171–88. 17. Hauer-Jensen M, Wang J, Denham JW. Bowel injury: current and evolving management strategies. Semin Radiat Oncol. 2003;13: 357–71. 18. Theis VS, Sripadam R, Ramani V, Lal S. Chronic radiation enteritis. Clin Oncol. 2010;22:70–83. 19. Ooi BS, Tjandra JJ, Green MD. Morbidities of adjuvant chemotherapy and radiotherapy for resectable rectal cancer: an overview. Dis Colon Rectum. 1999;42:403–18. 20. Langley RE, Bump EA, Quartuccio SG, Medeiros D, Braunhut SJ. Radiation-induced apoptosis in microvascular endothelial cells. Br J Cancer. 1997;75:666–72. 21. Leadon SA. Repair of DNA damage produced by ionizing radiation: a minireview. Semin Radiat Oncol. 1996;6:295–305. 22. Clarke AR, Gledhill S, Hooper ML, Bird CC, Wyllie AH. p53 dependence of early apoptotic and proliferative responses within the mouse intestinal epithelium following gamma-irradiation. Oncogene. 1994;9:1767–73. 23. Potten CS, Booth C. The role of radiation-induced and spontaneous apoptosis in the homeostasis of the gastrointestinal epithelium: a brief review. Comp Biochem Physiol B Biochem Mol Biol. 1997;118:473–8. 24. Dublineau I, Grison S, Grandcolas L, et al. Effects of chronic 137Cs ingestion on barrier properties of jejunal epithelium in rats. J Toxicol Environ Health Part A. 2007;70:810–9. 25. Guzman-Stein G, Bonsack M, Liberty J, Delaney JP. Abdominal radiation causes bacterial translocation. J Surg Res. 1989;46:104–7. 26. Baker DG, Krochak RJ. The response of the microvascular system to radiation: a review. Cancer Investig. 1989;7:287–94. 27. Hopewell JW, Calvo W, Jaenke R, Reinhold HS, Robbins ME, Whitehouse EM. Microvasculature and radiation damage. Recent Results Cancer Res Fortschr Krebsforschung Progres Rech Cancer. 1993;130:1–16. 28. Paris F, Fuks Z, Kang A, et al. Endothelial apoptosis as the primary lesion initiating intestinal radiation damage in mice. Science. 2001;293:293–7. 29. Wang J, Zheng H, Ou X, Fink LM, Hauer-Jensen M. Deficiency of microvascular thrombomodulin and up-regulation of proteaseactivated receptor-1 in irradiated rat intestine: possible link between endothelial dysfunction and chronic radiation fibrosis. Am J Pathol. 2002;160:2063–72. 30. Verheij M, Dewit LG, Boomgaard MN, Brinkman HJ, van Mourik JA. Ionizing radiation enhances platelet adhesion to the extracellular matrix of human endothelial cells by an increase in the release of von Willebrand factor. Radiat Res. 1994;137:202–7. 31. Dunn MM, Drab EA, Rubin DB. Effects of irradiation on endothelial cell-polymorphonuclear leukocyte interactions. J Appl Physiol. 1986;60:1932–7. 32. Wang J, Boerma M, Fu Q, Hauer-Jensen M. Significance of endothelial dysfunction in the pathogenesis of early and delayed radiation enteropathy. World J Gastroenterol. 2007;13:3047–55. 33. Denham JW, Hauer-Jensen M. The radiotherapeutic injury–a complex ‘wound’. Radiother Oncol J Eur Soc Ther Radiol Oncol. 2002;63:129–45. 34. Lebrun F, Francois A, Vergnet M, Lebaron-Jacobs L, Gourmelon P, Griffiths NM. Ionizing radiation stimulates muscarinic regulation of rat intestinal mucosal function. Am J Physiol. 1998;275:G1333–40. 35. Classen J, Belka C, Paulsen F, Budach W, Hoffmann W, Bamberg M. Radiation-induced gastrointestinal toxicity. Pathophysiology,

Curr Gastroenterol Rep (2014) 16:383

36.

37. 38.

39.

40.

41.

42. 43. 44.

45. 46.

47.

48. 49. 50.

51.

52.

53.

54.

55.

56.

57.

approaches to treatment and prophylaxis. Strahlenther Onkol Organ Deutsch Rontgengesellschaft. 1998;174 Suppl 3:82–4. Wedlake L, Thomas K, McGough C, Andreyev HJ. Small bowel bacterial overgrowth and lactose intolerance during radical pelvic radiotherapy: An observational study. Eur J Cancer. 2008;44: 2212–7. Sandle GI. Salt and water absorption in the human colon: a modern appraisal. Gut. 1998;43:294–9. Konishi T, Watanabe T, Nagawa H. Treatment of local ischaemia: another promising approach for gastrointestinal complications of pelvic radiotherapy. Gut. 2006;55:1209. Yarnold J, Brotons MC. Pathogenetic mechanisms in radiation fibrosis. Radiother Oncol J Eur Soc Ther Radiol Oncol. 2010;97: 149–61. Gervaz P, Morel P, Vozenin-Brotons MC. Molecular aspects of intestinal radiation-induced fibrosis. Curr Mol Med. 2009;9: 273–80. Haydont V, Vozenin-Brotons MC. Maintenance of radiationinduced intestinal fibrosis: cellular and molecular features. World J Gastroenterol. 2007;13:2675–83. Thompson JS, Saxena SK, Sharp JG. Regulation of intestinal regeneration: new insights. Microsc Res Tech. 2000;51:129–37. Kennedy GD, Heise CP. Radiation colitis and proctitis. Clin Colon Rectal Surg. 2007;20:64–72. Coia LR, Myerson RJ, Tepper JE. Late effects of radiation therapy on the gastrointestinal tract. Int J Radiat Oncol Biol Phys. 1995;31: 1213–36. Otterson MF. Effects of radiation upon gastrointestinal motility. World J Gastroenterol. 2007;13:2684–92. Overgaard J, Matsui M. Effect of radiation on glucose absorption in the mouse jejunum in vivo. Radiother Oncol J Eur Soc Ther Radiol Oncol. 1990;18:71–7. Packey CD, Ciorba MA. Microbial influences on the small intestinal response to radiation injury. Curr Opin Gastroenterol. 2010;26: 88–94. Ciorba MA, Stenson WF. Probiotic therapy in radiation-induced intestinal injury and repair. Ann N Y Acad Sci. 2009;1165:190–4. Crawford PA, Gordon JI. Microbial regulation of intestinal radiosensitivity. Proc Natl Acad Sci U S A. 2005;102:13254–9. Kuhn R, Lohler J, Rennick D, Rajewsky K, Muller W. Interleukin10-deficient mice develop chronic enterocolitis. Cell. 1993;75: 263–74. Willett CG, Ooi CJ, Zietman AL, et al. Acute and late toxicity of patients with inflammatory bowel disease undergoing irradiation for abdominal and pelvic neoplasms. Int J Radiat Oncol Biol Phys. 2000;46:995–8. Manichanh C, Varela E, Martinez C, et al. The gut microbiota predispose to the pathophysiology of acute postradiotherapy diarrhea. Am J Gastroenterol. 2008;103:1754–61. Algin O, Turkbey B, Ozmen E, Algin E. Magnetic resonance enterography findings of chronic radiation enteritis. Cancer Imaging Off Publ Int Cancer Imaging Soc. 2011;11:189–94. Cronin CG, Lohan DG, Browne AM, Alhajeri AN, Roche C, Murphy JM. MR enterography in the evaluation of small bowel dilation. Clin Radiol. 2009;64:1026–34. Khalid U, Norman AR, Andreyev HJ. Elevated C-reactive protein levels are not a feature of uncomplicated radiation-induced bowel injury. Eur J Cancer Care. 2007;16:346–50. Hille A, Schmidt-Giese E, Hermann RM, et al. A prospective study of faecal calprotectin and lactoferrin in the monitoring of acute radiation proctitis in prostate cancer treatment. Scand J Gastroenterol. 2008;43:52–8. Wedlake L, McGough C, Hackett C, et al. Can biological markers act as non-invasive, sensitive indicators of radiation-induced effects in the gastrointestinal mucosa? Aliment Pharmacol Ther. 2008;27: 980–7.

Page 7 of 9, 383 58. Lutgens L, Lambin P. Biomarkers for radiation-induced small bowel epithelial damage: an emerging role for plasma Citrulline. World J Gastroenterol. 2007;13:3033–42. 59. Lutgens LC, Blijlevens NM, Deutz NE, Donnelly JP, Lambin P, de Pauw BE. Monitoring myeloablative therapy-induced small bowel toxicity by serum citrulline concentration: a comparison with sugar permeability tests. Cancer. 2005;103:191–9. 60. Lutgens LC, Deutz NE, Gueulette J, et al. Citrulline: a physiologic marker enabling quantitation and monitoring of epithelial radiationinduced small bowel damage. Int J Radiat Oncol Biol Phys. 2003;57:1067–74. 61. Kopelman Y, Groissman G, Fireman Z. Radiation enteritis diagnosed by capsule endoscopy. Gastrointest Endosc. 2007;66:599. discussion. 62. Sidhu R, Sanders DS, Morris AJ, McAlindon ME. Guidelines on small bowel enteroscopy and capsule endoscopy in adults. Gut. 2008;57:125–36. 63. Kim HM, Kim YJ, Kim HJ, Park SW, Bang S, Song SY. A pilot study of capsule endoscopy for the diagnosis of radiation enteritis. Hepato-gastroenterology. 2011;58:459–64. 64. Caunedo-Alvarez A, Romero-Vazquez J, Herrerias-Gutierrez JM. Patency and Agile capsules. World J Gastroenterol. 2008;14: 5269–73. 65. Spada C, Riccioni ME, Costamagna G. The new, dissolving patency capsule: a safe and effective tool to avoid the complication of retained video capsules. J Clin Gastroenterol. 2008;42:761–2. 66. Ludgate SM, Merrick MV. The pathogenesis of post-irradiation chronic diarrhoea: measurement of SeHCAT and B12 absorption for differential diagnosis determines treatment. Clin Radiol. 1985;36:275–8. 67. Danielsson A, Nyhlin H, Persson H, Stendahl U, Stenling R, Suhr O. Chronic diarrhoea after radiotherapy for gynaecological cancer: occurrence and aetiology. Gut. 1991;32:1180–7. 68. Yeoh EK, Horowitz M, Russo A, Muecke T, Robb T, Chatterton BE. Gastrointestinal function in chronic radiation enteritis–effects of loperamide-N-oxide. Gut. 1993;34:476–82. 69. Rossel P, Sortsoe Jensen H, Qvist P, Arveschoug A. Prognosis of adult-onset idiopathic bile acid malabsorption. Scand J Gastroenterol. 1999;34:587–90. 70. Gasbarrini A, Lauritano EC, Gabrielli M, et al. Small intestinal bacterial overgrowth: diagnosis and treatment. Dig Dis. 2007;25: 237–40. 71. Beer WH, Fan A, Halsted CH. Clinical and nutritional implications of radiation enteritis. Am J Clin Nutr. 1985;41:85–91. 72. Green N, Iba G, Smith WR. Measures to minimize small intestine injury in the irradiated pelvis. Cancer. 1975;35:1633–40. 73. Gunderson LL, Russell AH, Llewellyn HJ, Doppke KP, Tepper JE. Treatment planning for colorectal cancer: radiation and surgical techniques and value of small-bowel films. Int J Radiat Oncol Biol Phys. 1985;11:1379–93. 74. Herbert SH, Solin LJ, Hoffman JP, et al. Volumetric analysis of small bowel displacement from radiation portals with the use of a pelvic tissue expander. Int J Radiat Oncol Biol Phys. 1993;25:885–93. 75. Shanahan TG, Mehta MP, Bertelrud KL, et al. Minimization of small bowel volume within treatment fields utilizing customized “belly boards”. Int J Radiat Oncol Biol Phys. 1990;19:469–76. 76. Russ JE, Smoron GL, Gagnon JD. Omental transposition flap in colorectal carcinoma: adjunctive use in prevention and treatment of radiation complications. Int J Radiat Oncol Biol Phys. 1984;10:55–62. 77. Lechner P, Cesnik H. Abdominopelvic omentopexy: preparatory procedure for radiotherapy in rectal cancer. Dis Colon Rectum. 1992;35:1157–60. 78. Devereux DF, Kavanah MT, Feldman MI, et al. Small bowel exclusion from the pelvis by a polyglycolic acid mesh sling. J Surg Oncol. 1984;26:107–12.

383, Page 8 of 9 79. Rodier JF, Janser JC, Rodier D, et al. Prevention of radiation enteritis by an absorbable polyglycolic acid mesh sling. A 60-case multicentric study. Cancer. 1991;68:2545–9. 80. Beitler A, Rodriguez-Bigas MA, Weber TK, Lee RJ, Cuenca R, Petrelli NJ. Complications of absorbable pelvic mesh slings following surgery for rectal carcinoma. Dis Colon Rectum. 1997;40:1336–41. 81. Hoffman JP, Sigurdson ER, Eisenberg BL. Use of saline-filled tissue expanders to protect the small bowel from radiation. Oncology. 1998;12:51–4. discussion 4, 60, 2, passim. 82. Sugarbaker PH. Intrapelvic prosthesis to prevent injury of the small intestine with high dosage pelvic irradiation. Surg Gynecol Obstet. 1983;157:269–71. 83. Freund H, Gunderson L, Krause R, Fischer JE. Prevention of radiation enteritis after abdominoperineal resection and radiotherapy. Surg Gynecol Obstet. 1979;149:206–8. 84. Jia D, Koonce NA, Griffin RJ, Jackson C, Corry PM. Prevention and mitigation of acute death of mice after abdominal irradiation by the antioxidant N-acetyl-cysteine (NAC). Radiat Res. 2010;173: 579–89. 85. Tannehill SP, Mehta MP, Larson M, et al. Effect of amifostine on toxicities associated with sequential chemotherapy and radiation therapy for unresectable non-small-cell lung cancer: results of a phase II trial. J Clin Oncol Off J Am Soc Clin Oncol. 1997;15: 2850–7. 86. Athanassiou H, Antonadou D, Coliarakis N, et al. Protective effect of amifostine during fractionated radiotherapy in patients with pelvic carcinomas: results of a randomized trial. Int J Radiat Oncol Biol Phys. 2003;56:1154–60. 87. Akpolat M, Gulle K, Topcu-Tarladacalisir Y, et al. Protection by Lcarnitine against radiation-induced ileal mucosal injury in the rat: pattern of oxidative stress, apoptosis and cytokines. Int J Radiat Biol. 2013;89:732–40. 88. Sekhon S. Chronic radiation enteritis: women’s food tolerances after radiation treatment for gynecologic cancer. J Am Diet Assoc. 2000;100:941–3. 89. McGough C, Baldwin C, Frost G, Andreyev HJ. Role of nutritional intervention in patients treated with radiotherapy for pelvic malignancy. Br J Cancer. 2004;90:2278–87. 90. Arlow FL, Dekovich AA, Priest RJ, Beher WT. Bile acids in radiation-induced diarrhea. South Med J. 1987;80:1259–61. 91. Walters JR, Pattni SS. Managing bile acid diarrhoea. Ther Adv Gastroenterol. 2010;3:349–57. 92. Jenkins C, Carmody TJ, Rush AJ. Depression in radiation oncology patients: a preliminary evaluation. J Affect Disord. 1998;50: 17–21. 93. Abayomi J, Kirwan J, Hackett A, Bagnall G. A study to investigate women’s experiences of radiation enteritis following radiotherapy for cervical cancer. J Hum Nutr Diet Off J Br Diet Assoc. 2005;18: 353–63. 94. Bories C, Messing B, Lacourt J, Bernier JJ. Evaluation of the use of parenteral nutrition in chronic and severe radiation enterocolitis. Gastroenterol Clin Biol. 1987;11:142–7. 95. Thompson JS, Weseman R, Rochling F, et al. Radiation therapy increases the risk of hepatobiliary complications in short bowel syndrome. Nutr Clin Pract Off Publ Am Soc Parenter Enter Nutr. 2011;26:474–8. 96. Dietz DW, Remzi FH, Fazio VW. Strictureplasty for obstructing small-bowel lesions in diffuse radiation enteritis–successful outcome in five patients. Dis Colon Rectum. 2001;44:1772–7. 97. Regimbeau JM, Panis Y, Gouzi JL, Fagniez PL, French University Association for Surgical R. Operative and long term results after surgery for chronic radiation enteritis. Am J Surg. 2001;182: 237–42. 98. Galland RB, Spencer J. Surgical management of radiation enteritis. Surgery. 1986;99:133–9.

Curr Gastroenterol Rep (2014) 16:383 99. Swan RW, Fowler Jr WC, Boronow RC. Surgical management of radiation injury to the small intestine. Surg Gynecol Obstet. 1976;142:325–7. 100. Amiot A, Joly F, Lefevre JH, et al. Long-term outcome after extensive intestinal resection for chronic radiation enteritis. Dig Liver Dis Off J Ital Soc Gastroenterol Ital Assoc Study Liver. 2013;45:110–4. 101. Onodera H, Nagayama S, Mori A, Fujimoto A, Tachibana T, Yonenaga Y. Reappraisal of surgical treatment for radiation enteritis. World J Surg. 2005;29:459–63. 102. Spyropoulos BG, Misiakos EP, Fotiadis C, Stoidis CN. Antioxidant properties of probiotics and their protective effects in the pathogenesis of radiation-induced enteritis and colitis. Dig Dis Sci. 2011;56: 285–94. 103. Han W, Mercenier A, Ait-Belgnaoui A, et al. Improvement of an experimental colitis in rats by lactic acid bacteria producing superoxide dismutase. Inflamm Bowel Dis. 2006;12:1044–52. 104. Carroll IM, Andrus JM, Bruno-Barcena JM, Klaenhammer TR, Hassan HM, Threadgill DS. Anti-inflammatory properties of Lactobacillus gasseri expressing manganese superoxide dismutase using the interleukin 10-deficient mouse model of colitis. Am Jo Physiol Gastrointest Liver Physiol. 2007;293:G729–38. 105. Kullisaar T, Zilmer M, Mikelsaar M, et al. Two antioxidative lactobacilli strains as promising probiotics. Int J Food Microbiol. 2002;72:215–24. 106. Chang SK, Hassan HM. Characterization of superoxide dismutase in Streptococcus thermophilus. Appl Environ Microbiol. 1997;63: 3732–5. 107. Molla M, Gironella M, Salas A, et al. Protective effect of superoxide dismutase in radiation-induced intestinal inflammation. Int J Radiat Oncol Biol Phys. 2005;61:1159–66. 108. Loguercio C, D’Argenio G, Delle Cave M, et al. Glutathione supplementation improves oxidative damage in experimental colitis. Dig Liver Dis Off J Ital Soc Gastroenterol Ital Assoc Study Liver. 2003;35:635–41. 109. Sengul N, Aslim B, Ucar G, et al. Effects of exopolysaccharideproducing probiotic strains on experimental colitis in rats. Dis Colon Rectum. 2006;49:250–8. 110. Delia P, Sansotta G, Donato V, et al. Use of probiotics for prevention of radiation-induced diarrhea. World J Gastroenterol. 2007;13:912–5. 111. Urbancsek H, Kazar T, Mezes I, Neumann K. Results of a doubleblind, randomized study to evaluate the efficacy and safety of Antibiophilus in patients with radiation-induced diarrhoea. Eur J Gastroenterol Hepatol. 2001;13:391–6. 112. Floch MH, Walker WA, Guandalini S, et al. Recommendations for probiotic use–2008. J Clin Gastroenterol. 2008;42 Suppl 2:S104–8. 113. Hille A, Christiansen H, Pradier O, et al. Effect of pentoxifylline and tocopherol on radiation proctitis/enteritis. Strahlenther Onkol Organ Deutsch Rontgengesellschaft. 2005;181:606–14. 114. Gothard L, Cornes P, Brooker S, et al. Phase II study of vitamin E and pentoxifylline in patients with late side effects of pelvic radiotherapy. Radiother Oncol J Eur Soc Ther Radiol Oncol. 2005;75: 334–41. 115. Delanian S, Lefaix JL. Current management for late normal tissue injury: radiation-induced fibrosis and necrosis. Semin Radiat Oncol. 2007;17:99–107. 116. Hamama S, Gilbert-Sirieix M, Vozenin MC, Delanian S. Radiationinduced enteropathy: molecular basis of pentoxifylline-vitamin E anti-fibrotic effect involved TGF-beta1 cascade inhibition. Radiother Oncol J Eur Soc Ther Radiol Oncol. 2012;105:305–12. 117. Kilic D, Egehan I, Ozenirler S, Dursun A. Double-blinded, randomized, placebo-controlled study to evaluate the effectiveness of sulphasalazine in preventing acute gastrointestinal complications due to radiotherapy. Radiother Oncol J Eur Soc Ther Radiol Oncol. 2000;57:125–9.

Curr Gastroenterol Rep (2014) 16:383 118. Loiudice TA, Lang JA. Treatment of radiation enteritis: a comparison study. Am J Gastroenterol. 1983;78:481–7. 119. Marshall GT, Thirlby RC, Bredfeldt JE, Hampson NB. Treatment of gastrointestinal radiation injury with hyperbaric oxygen. Undersea Hyperb Med J Undersea Hyperb Med Soc Inc. 2007;34:35–42. 120. Olgac V, Erbil Y, Barbaros U, et al. The efficacy of octreotide in pancreatic and intestinal changes: radiation-induced enteritis in animals. Dig Dis Sci. 2006;51:227–32. 121. Rocourt DV, Mehta VB, Besner GE. Heparin-binding EGF-like growth factor decreases inflammatory cytokine expression after intestinal ischemia/reperfusion injury. J Surg Res. 2007;139: 269–73. 122. Kuhn MA, Xia G, Mehta VB, Glenn S, Michalsky MP, Besner GE. Heparin-binding EGF-like growth factor (HB-EGF) decreases oxygen free radical production in vitro and in vivo. Antioxid Redox Signal. 2002;4:639–46. 123. Matthews MA, Watkins D, Darbyshire A, Carson WE, Besner GE. Heparin-binding EGF-like growth factor (HB-EGF) protects the intestines from radiation therapy-induced intestinal injury. J Pediatr Surg. 2013;48:1316–22. 124. Williams JP, Hernady E, Johnston CJ, et al. Effect of administration of lovastatin on the development of late pulmonary effects after whole-lung irradiation in a murine model. Radiat Res. 2004;161: 560–7. 125. Haydont V, Bourgier C, Pocard M, et al. Pravastatin Inhibits the Rho/CCN2/extracellular matrix cascade in human fibrosis explants and improves radiation-induced intestinal fibrosis in rats. Clin Cancer Res Off J Am Assoc Cancer Res. 2007;13:5331–40. 126. Yamashita M, Otsuka F, Mukai T, et al. Simvastatin antagonizes tumor necrosis factor-alpha inhibition of bone morphogenetic proteins-2-induced osteoblast differentiation by regulating Smad signaling and Ras/Rho-mitogen-activated protein kinase pathway. J Endocrinol. 2008;196:601–13. 127. Keskek M, Gocmen E, Kilic M, et al. Increased expression of cyclooxygenase-2 (COX-2) in radiation-induced small bowel injury in rats. J Surg Res. 2006;135:76–84. 128. Wang J, Zheng H, Ou X, et al. Hirudin ameliorates intestinal radiation toxicity in the rat: support for thrombin inhibition as strategy to minimize side-effects after radiation therapy and as

Page 9 of 9, 383

129.

130.

131.

132.

133.

134.

135.

136.

137.

138.

countermeasure against radiation exposure. J Thromb Haemost. 2004;2:2027–35. Wang J, Albertson CM, Zheng H, Fink LM, Herbert JM, HauerJensen M. Short-term inhibition of ADP-induced platelet aggregation by clopidogrel ameliorates radiation-induced toxicity in rat small intestine. Thromb Haemost. 2002;87:122–8. Wang J, Zheng H, Qiu X, Kulkarni A, Fink LM, Hauer-Jensen M. Modulation of the intestinal response to ionizing radiation by anticoagulant and non-anticoagulant heparins. Thromb Haemost. 2005;94:1054–9. De Cristofaro R, De Candia E, Landolfi R. Effect of high- and lowmolecular-weight heparins on thrombin-thrombomodulin interaction and protein C activation. Circulation. 1998;98:1297–301. Mohri M, Gonda Y, Oka M, et al. The antithrombotic effects of recombinant human soluble thrombomodulin (rhsTM) on tissue factor-induced disseminated intravascular coagulation in crabeating monkeys (Macaca fascicularis). Blood Coagul Fibrinolysis Int J Haemost Thromb. 1997;8:274–83. Solis MM, Vitti M, Cook J, et al. Recombinant soluble human thrombomodulin: a randomized, blinded assessment of prevention of venous thrombosis and effects on hemostatic parameters in a rat model. Thromb Res. 1994;73:385–94. Bernard GR, Vincent JL, Laterre PF, et al. Efficacy and safety of recombinant human activated protein C for severe sepsis. New Engl J Med. 2001;344:699–709. Andrade-Gordon P, Maryanoff BE, Derian CK, et al. Design, synthesis, and biological characterization of a peptide-mimetic antagonist for a tethered-ligand receptor. Proc Natl Acad Sci U S A. 1999;96:12257–62. Nguyen NP, Antoine JE, Dutta S, Karlsson U, Sallah S. Current concepts in radiation enteritis and implications for future clinical trials. Cancer. 2002;95:1151–63. Rose DM, Hochwald SN, Harrison LE, Burt M. Selective glutathione repletion with oral oxothiazolidine carboxylate (OTZ) in the radiated tumor-bearing rat. J Surg Res. 1996;62:224–8. Graca BM, Freire PA, Brito JB, Ilharco JM, Carvalheiro VM, Caseiro-Alves F. Gastroenterologic and radiologic approach to obscure gastrointestinal bleeding: how, why, and when? Radiogr Rev Publ Radiol Soc N Am Inc. 2010;30:235–52.

Radiation enteritis.

Radiation enteritis continues to be a major health concern in recipients of radiation therapy. The incidence of radiation enteritis is expected to con...
352KB Sizes 0 Downloads 3 Views