Stem cell transplantation Gastrointestinal graft-versus-host disease: from biomarkers to pathophysiology

J. Ferrara ABSTRACT

University of Michigan Hospitals and Acute graft-versus-host disease (GVHD) of the (GI) affects up to 60% of Health Centers, MI, USA patients receiving allogeneic hematopoietic cell transplantation (HCT). It causes nausea, vomiting, anorexia, secretory diarrhea and, in severe cases, abdominal pain and/or hemorrhage. Acute GVHD is often clinically indistinguishable from other causes of GI dysfunction such as conditioning regimen Correspondence: toxicity, infection or medications. Endoscopic biopsy is often used to confirm the diagnosis, but histo- James Ferrara logical severity on biopsy does not consistently correlate with clinical outcome. Clinical stage II or E-mail: [email protected] greater (more than 1 L of diarrhea per day) is associated with reduced survival, but daily stool volume can vary considerably. Lower GI GVHD responds poorly to treatment compared to other target organs, and treatment with high-dose systemic steroid therapy carries significant risks, especially infectious Hematology Education: complications in profoundly immunosuppressed patients. the education program for the annual congress of the European Hematology Association Learning goals At the conclusion of this activity, participants should: 2013;7:347-350 - know the three parameters that define risk for death at the onset of gastrointestinal graft-versus- host disease; - know the functional importance of Paneth cells in the gastrointestinal tract; - know the relationship between Reg3 and the gastrointestinal microbiome. α

versus Acute graft- -host disease (GVHD) of The research effort that identified REG3α the gastrointestinal tract (GI) affects up to 60% used a proteomics approach that analyzed of patients receiving allogeneic hematopoietic pooled plasma samples from 10 patients with cell transplantation (HCT).1,2 It causes nausea, biopsy-proven GI GVHD and 10 patients vomiting, anorexia, secretory diarrhea and, in without GVHD taken at similar times after severe cases, abdominal pain and/or hemor- HCT.12,13 We identified and quantified 562 pro- rhage.3 Acute GVHD is often clinically indis- teins, of which 74 were increased at least 2- tinguishable from other causes of GI dysfunc- fold in patients with GVHD. Of the 5 tion such as conditioning regimen toxicity, preferentially expressed in the GI tract, only infection or medications. Endoscopic biopsy is one had commercially available antibodies often used to confirm the diagnosis,4 but histo- suitable for quantification of plasma concen- logical severity on biopsy does not consistent- trations by ELISA. This process identified ly correlate with clinical outcome.4-6 Clinical REG3α as the lead candidate for a biomarker stage II or greater (more than 1 L of diarrhea of GI GVHD. per day) is associated with reduced survival,1,2 We validated REG3α as a biomarker by but daily stool volume can vary considerably. measuring plasma concentration in samples Lower GI GVHD responds poorly to treatment from 850 allogeneic HCT recipients from the compared to other target organs, 2 and treat- University of Michigan, USA. Plasma REG3α ment with high-dose systemic steroid therapy concentrations were 3 times higher in patients carries significant risks, especially infectious at the onset of GI GVHD than in all other complications in profoundly immunosup- patients, including those with non-GVHD pressed patients.7,8 enteritis. Serum REG3α concentrations were The standard treatment of acute GVHD is also higher in GI GVHD in an independent higher dose systemic steroids, which has not validation set of 143 HCT patients from changed in 40 years. One reason for this lack Regensburg, Germany, and Kyushu, Japan, of progress is the lack of validated biomarkers although the absolute values were lower. This for acute GVHD. We have recently identified difference may be due to a center effect that and validated laboratory regenerating islet- depends on several factors, including varia- derived 3-alpha (REG3α), a C-type tions in transplant conditioning regimens and secreted by Paneth cells,9,10 as a non-invasive, supportive care. For example, all patients in reliable blood biomarker specific for GVHD Regensburg and Kyushu received oral antibi- of the GI tract with diagnostic and prognostic otics as GVHD prophylaxis, whereas utility.11 Michigan patients did not, and thus increased

Hematology Education: the education program for the annual congress of the European Hematology Association | 2013; 7(1) | 347 | 18th Congress of the European Hematology Association

GI flora might account for greater REG3α secretion. number or pathological grade correlated better with clini- We next analyzed REG3α concentrations according to cal GVHD severity. Proportional odds logistical regres- diagnosis and type of GI symptom in all 1000 patients. In sion analysis demonstrated that Paneth cell numbers α patients with diarrhea caused by GVHD, REG3 concen- retained a strong correlation with GVHDP severity after trations at the onset of GVHD were 5 times higher than in accounting for pathological grade ( =0.008), but patho- α patients with diarrhea from other causes. REG3 concen- logical grade did not correlate with GVHDP severity after trations at the onset of symptoms continued to distinguish Paneth cell numbers were accounted for ( =0.27). Thus between GVHD and non-GVHD etiologies in patients Paneth cell number in the duodenum correlates better with with both small and large volumes of diarrhea. clinical GI GVHD severity than histological grade. The clinical utility of any biomarker increases if it pro- Patients with a complete response (CR) to therapy also vides prognostic information regarding the future status of had significantly more Paneth cells than patients with a a disease and/or patient, e.g. the likelihood of response to partial response (PR); patients with no response (NR) had treatment. We, therefore, evaluated the prognostic signifi- the fewest Paneth cells corresponding to a highly signifi- α cance of REG3 plasma levels in more than 160 patients cantP difference among the three treatment responses taken at the time of diagnosis of lower GI GVHD. Four ( <0.0001) (Figure 2). The number of Paneth cells also weeks is a common time point at which to evaluate stronglydata not correlatedshown with maximum GI GVHD severity response.14 REG3α concentrations were 3-fold lower at ( ). We found that patients whose duodenal biopsies con- the timeversus of GVHD diagnosis in patients who did not respond those in patients who experienced a com- tained a mean of 4 or fewer Paneth cells at the onset of vs. GVHD were more than twice as likely to die from GVHD- plete or partial responseP to therapy (635±132 ng/mL 240±61 ng/mL; <0.001). Patients responding to therapy related causes as comparedvs. P to patients with 5 or more α Paneth cells (55% 23%; <0.0001). This large differ- still exhibited REG3 concentrations more thanP 3 times that of non-GVHD controls (77±22 ng/mL; <0.001). ence in GVHD-related mortality translated into a signifi- Because the response to treatment at four weeks strongly cant difference in overall vs.survival atP six months from the correlates with non-relapse mortality (NRM), we hypoth- onset of GI GVHD (16% 42%, =0.01). esized that the REG3α concentration at GVHD diagnosis The current GI GVHD pathological grading system has would also correlate with NRM. We, therefore, divided the major limitations. First, the correlation between severity patients into two equal groups based upon the median of colonic changes and clinical GVHD severity is poor REG3α concentration. NRM was twice as high in patients and so many pathologists do not report pathological grade with high REG3α concentrations, and this difference in order to avoid confusion by clinicians. Second, the remained significant after adjusting for known risk factors grading system has not been standardized. Third, there is of donor type, degree of HLA match, conditioning intensi- no definition as to how to resolve the variable severity within biopsy sections from the same patient. These find- ty, age,vs. and baseline disease severity:P 59% (95%CI: 4%- 69%) 34% (95%CI: 2%-46%); <0.001). ings demonstrate that a straightforward quantification of Data regarding the clinical stage, histological grade, Paneth cell number can help establish the diagnosis of GI REG3α concentration, and level of GVHD at onset were GVHD and has prognostic importance. Recent research supports the correlation between Paneth available in 140vs patients. The plasma concentration of REG3α (above . below the median), the clinical severity cells and GI GVHD, and our observations provide insights vs into physiological mechanisms and suggest Paneth cells of GVHD (stage 1vs . stage 2-4), and the histological α severity (stage 1-3 . stage 4) at the time of GVHD diag- secrete α-defensins and REG3 , which are antimicrobial nosis independently predicted both lack of response to peptides. These peptides function to regulate the microbio- GVHD therapy and 1-year NRM after adjustment for the ta of the intestine through selective activity against non- commensal bacteria, while generally sparing commensal aforementioned risk factors. When lack of response to therapy and NRM were modeled simultaneously on all four parameters, all the three parameters remained statisti- cally significant. The inclusion of all three characteristics that remained statistically significant on simultaneous modeling demonstrated that patients with increasing num- bers of risk factors present at onset had increasing risk for NRM. Paneth cells are major producers of REG3α. Given the importance of these biomarkers, we evaluated 118 patients who had duodenal biopsies obtained at the time of GVHD- related diarrhea, and 15 patients who had minimal or no GI symptoms but had small duodenal biopsies available. We quantitated the number of Paneth cells per high pow- ered field (HPF) and found a very strong statistical corre- lation between the number of Paneth cells per HPF and the clinicalP severity of GI GVHD at the time of biopsy ( <0.0001) (Figure 1). Contrary decreasedto our hypothesis, how- ever, the number of Paneth cells as the severity of GVHD increased. Likewise, small intestine pathologi- caldata grade not shown also correlated with clinical GVHD severity Figure 1. Paneth cell count by clinical GI GVHD onset stage. ( ). We next determined whether Paneth cell

| 348 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2013; 7(1) Stockholm, Sweden, June 13-16, 2013

P bacteria. A loss of intestinal microbiota diversity, charac- ( <0.001). As shown in Figure 2, Paneth cells were read- terized by expansion of Lactobacillus species and contrac- ily identified by immuno-histochemical (IHC) staining of ture of the commensal bacteria flora, has been observed in lysozyme. On Day +14, GVHD caused an 80% decrease both mice and patients who developed GVHD.15 In a sep- in Paneth cell numbers. Similarly, GVHD caused more arate study, mice with GVHD exhibited decreased num- than an over 95% decrease in REG3α . Thus, the bers of Paneth cells, similar to our observations in clinical number of Paneth cells decreased dramatically in both GVHD.16 Other immune-mediated inflammatory condi- clinical and experimental GVHD. Yet, despite this tions, such as inflammatory bowel disease and reduced decrease, the levels in the plasma increased. Paneth cell numbers characterize and correlate with One possible explanation for this paradox could be that changes in the intestinal microbiota.17,18 Interestingly, the correlation of mucosal denudation with high REG3α α plasma concentration of REG3 dataand notPaneth shown cell numbers concentrations suggests that microscopic breaches in the do not correlate with each other ( ), suggest- mucosal epithelial barrier caused by severe GVHD permit ing that these two parameters measure different biological REG3α to traverse into the systemic circulation. The tight processes, and that Paneth cells may not be the primary proximity of Paneth cells with ISCs concentrates their drivers of the increased appearance of REG3α in the plas- secretory contents in that vicinity, so that mucosal barrier ma. Paneth cells are not normally present in the distant disruption caused by stem cell dropout may preferentially colon, although they are found in the proximal colon close allow Paneth cell secretions, including REG3α, to enter to the ileal-cecal junction. This focus on changes in the into the bloodstream. REG3α is a large molecule that is small intestine has precedent in other diseases, where concentrated in the mucus covering the apical surface of small intestinal pathology can account for the diarrhea the cells so that when the cell dies, highly concentrated observed in viral gastroenteritis,19 celiac disease,20 and REG3α translocates into the circulation system (Figure 3; some cases of bacterial overgrowth.21 Nonetheless, REG3a is represented by the red circle). Plasma levels of although Paneth cell quantification eliminates much of the REG3α may, therefore, serve as a surrogate marker for the subjectivity and variability within the current GI GVHD cumulative area of these breaches to GI mucosal barrier pathological grading system, incorporation of this meas- integrity, a parameter impossible to measure by individual ure into GI GVHD management will require acquisition of tissue biopsies and current endoscopic technology. Such duodenal biopsies. The additional expense and risk associ- an estimate of total damage to the mucosal barrier may ated with either upper GI endoscopy or colonoscopy that also help explain the prognostic value of REG3α with includes the terminal ileum may be offset by improved respect to therapy responsiveness and NRM. risk stratification early in the GI GVHD course, a time The importance of REG3α as an anti-microbial peptide when interventions are most likely to be effective. refocuses our attention on the potential role of the micro- The surprising inverse relationship between Paneth cell biota in the pathogenesis of GI GVHD. The importance of number and REG3α concentrations led to a further exam- the GI microbial flora to systematic GVHD was noted ination of the role of Paneth cells and REG3α in a well- almost forty years ago when germ-free mice developed defined animal model of acute GVHD. B6D2F1 mice significantly less GVHD.22,23 Clinical trials confirmed received 12 Gy total body irradiation on Day 1 and on Day these findings and led to the use of oral non-absorbable 0 were injected intravenously (iv) with 5 million BM cells as part of GVHD prophylaxis.24,25 As men- and 2.5 million B6 spleen cells. Syngeneic B6 BMT recip- tioned above, the crypts are the primary sites of GVHD ients served as non-GVHD controls. Mice were killed on damage in the GI tract, and intestinal stem cells (ISCs) Day +14 and samples of the ileum were scored for GVHD, appear to be the principal cellular targets because their as for human samples. GVHD scores were 2.3±0.4 in the damage amplifies systemic GVHD, and their protection allogeneic BMT group and 1.0±0.1 in the syngeneic group through wnt signals reduces alloreactive cascade of

Figure 2. GVHD damage to GI mucosal immunity. Figure 3. REG3 translocation into the bloodstream during GVHD. α

Hematology Education: the education program for the annual congress of the European Hematology Association | 2013; 7(1) | 349 | 18th Congress of the European Hematology Association

26 2010;2(13):13ra2. GVHD. ISCs reside in the crypts juxtaposedin to vitro Paneth cells, and Paneth cells are key to ISC function ,27,28 14. Levine JE, Logan B, Wu J, Alousi AM, Ho V, Bolanos-Meade 29 J, et al. Graft-versus-host disease treatment: predictors of sur- giving rise to their description as “guardians of ISCs”. vival. Biol Blood Marrow Transplant. 2010;16(12):1693-9. Paneth cells secrete antimicrobial peptides such as alpha 15. Jenq RR, Ubeda C, Taur Y, Menezes CC, Khanin R, Dudakov defensins and regenerative3 alpha (REG3α) that sterilize JA, et al. Regulation of intestinal by microbiota the mucus covering the luminal surface of enterocytes, following allogeneic bone marrow transplantation. J Exp Med. 30 2012;209(5):903-11. creating a protective barrier. 16. Eriguchi Y, Takashima S, Oka H, Shimoji S, Nakamura K, Uryu H, et al. Graft-versus-host disease disrupts intestinal microbial ecology by inhibiting Paneth cell production of α- References defensins. Blood. 2012;120(1):223-31. 17. Gersemann M Wehkamp J, Stange EF. Innate immune dys- function in inflammatory bowel disease. J Intern Med. 1. Martin PJ, McDonald GB, Sanders JE, Anasetti C, Appelbaum 2012;271(5):421-8. FR, Deeg HJ, et al. Increasingly frequent diagnosis of acute 18. Bevins CL Salzman NH. Paneth cells, antimicrobial peptides gastrointestinal graft-versus-host disease after allogeneic and maintenance of intestinal homeostasis. Nat Rev hematopoietic cell transplantation. Biol Blood Marrow Microbiol. 2011;9(5):356-68. Transpl. 2004;10(5):320-7. 19. Baldi F Bianco MA, Nardone G, Pilotto A, Zamparo E. Focus 2. MacMillan ML, Weisdorf DJ, Wagner JE, DeFor TE, Burns on acute diarrhoeal disease. World J Gastroenterol LJ, Ramsay NK, et al. Response of 443 patients to steroids as 2009;15(27):3341-8. primary therapy for acute graft-versus-host disease: compari- 20. Bao F Green PH, Bhagat G. An update on celiac disease son of grading systems. Biol Blood Marrow Transpl. histopathology and the road ahead. Arch Pathol Lab Med 2002;8(7):387-94. 2012;136(7):735-45. 3. Przepiorka D, Weisdorf D, Martin P, Klingemann HG, Beatty 21. Pyleris E Giamarellos-Bourboulis EJ, Tzivras D, Koussoulas P, Hows J, et al. 1994 Consensus Conference on Acute GVHD V, Barbatzas C, Pimentel M. The prevalence of overgrowth by Grading. Bone Marrow Transpl. 1995;15(6):825-8. aerobic bacteria in the small intestine by small bowel culture: 4. Shulman HM, Kleiner D, Lee SJ, Morton T, Pavletic SZ, relationship with irritable bowel syndrome. Dig Dis Sci. Farmer E, et al. Histopathologic diagnosis of chronic graft- 2012;57(5):1321-9. versus-host disease: National Institutes of Health Consensus 22. Jones JM, Wison R, Bealmear PM. Mortality and gross pathol- Development Project on Criteria for Clinical Trials in Chronic ogy of secondary disease in germfree mouse radiation Graft-versus-Host Disease: II. Pathology Working Group chimeras. RadiatRes. 1971;45:577. Report. Biol Blood Marrow Transpl. 2006;12(1):31-47. 23. van Bekkum DW, Roodenburg J, Heidt PJ, van der Waaij D. 5. Mowat A, Socie G. Intestinal Graft-vs.-Host Disease. In: Mitigation of secondary disease of allogeneic mouse radiation Ferrara JLM, Cooke KR, Deeg HJ (eds.) Graft-vs-Host chimeras by modification of the intestinal microflora. J Natl Disease. 3rd ed. New York: Marcel Dekker; 2004. p. 279-327. Cancer Inst. 1974;52(2):401-4. 6. Ertault-Daneshpouy M, Leboeuf C, Lemann M, Bouhidel F, 24. Storb R, Prentice RL, Buckner CD, Clift RA, Appelbaum F, Ades L, Gluckman E, et al. Pericapillary hemorrhage as crite- rion of severe human digestive graft-versus-host disease. Deeg J, et al. Graft-versus-host disease and survival in patients Blood. 2004;103(12):4681-4. with aplastic anemia treated by marrow grafts from HLA- 7. Weisdorf D, Haake R, Blazar B, Miller W, McGlave P, identical siblings. Beneficial effect of a protective environ- Ramsay N, et al. Treatment of moderate/severe acute graft- ment. N Engl J Med. 1983;308(6):302-7. versus-host disease after allogeneic bone marrow transplanta- 25. Beelen DW, Haralambie E, Brandt H, Linzenmeier G, Muller tion: an analysis of clinical risk features and outcome. Blood. KD, Quabeck K, et al. Evidence that sustained growth sup- 1990;75(4):1024-30. pression of intestinal anaerobic bacteria reduces the risk of 8. Deeg HJ. How I treat refractory acute GVHD. Blood. acute graft-versus-host disease after sibling marrow transplan- 2007;109(10):4119-26. tation. Blood. 1992;80(10):2668-76. 9. Cash HL, Whitham CV, Behrendt CL, Hooper LV. Symbiotic 26. Takashima S, Kadowaki M, Aoyama K, Koyama M, Oshima bacteria direct expression of an intestinal bactericidal lectin. T, Tomizuka K, et al. The Wnt agonist R-spondin1 regulates Science. 2006;313(5790):1126-30. systemic graft-versus-host disease by protecting intestinal 10. Gironella M, Iovanna JL, Sans M, Gil F, Penalva M, Closa D, stem cells. J Exp Med. 2011;208(2):285-94. et al. Anti-inflammatory effects of pancreatitis associated pro- 27. Sato T, Vries RG, Snippert HJ, van de Wetering M, Barker N, tein in inflammatory bowel disease. Gut. 2005;54(9):1244-53. Stange DE, et al. Single Lgr5 stem cells build crypt-villus 11. Ferrara JL, Harris AC, Greenson JK, Braun TM, Holler E, structures in vitro without a mesenchymal niche. Nature. Teshima T, et al. Regenerating islet-derived 3 alpha is a bio- 2009;459(7244):262-5. marker of gastrointestinal graft-versus-host disease. Blood. 28. Sato T, van Es JH, Snippert HJ, Stange DE, Vries RG, van den 2011;118(25):6702-8. Born M, et al. Paneth cells constitute the niche for Lgr5 stem 12. Faca V, Pitteri SJ, Newcomb L, Glukhova V, Phanstiel D, cells in intestinal crypts. Nature. 2011;469(7330):415-8. Krasnoselsky A, et al. Contribution of protein fractionation to 29. Ganz T. Defensins: antimicrobial peptides of innate immunity. depth of analysis of the serum and plasma proteomes. J Nat Rev Immunol. 2003;3(9):710-20. Proteome Res. 2007;6(9):3558-65. 30. Vaishnava S, Yamamoto M, Severson KM, Ruhn KA, Yu X, 13. Paczesny S, Braun TM, Levine JE, Hogan J, Crawford J, Koren O, et al. The antibacterial lectin RegIIIgamma promotes Coffing B, et al. Elafin is a biomarker of graft-versus-host dis- the spatial segregation of microbiota and host in the intestine. ease of the skin. Science Translational Medicine. Science. 2011;334(6053):255-8.

| 350 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2013; 7(1)