Double blind placebo-controlled trial for the prevention of ulcerative colitis relapses by β-fructan prebiotics: efficacy and metabolomic analysis =================================================================================================================================================== * Rosica Valcheva * Heather Armstrong * Ognjen Kovic * Michael Bording-Jorgensen * Simona Veniamin * María Elisa Pérez-Muñoz * Natasha Haskey * Melissa Silva * Farhad Peerani * Karen Wong * Dina H. Kao * Sander Veldhuyzen Van Zanten * Karen I. Kroeker * Deanna L. Gibson * Eytan Wine * Michael Gänzle * Jens Walter * Levinus A. Dieleman ## Abstract **Background** Ulcerative colitis (UC) is associated with altered intestinal microbiome (‘dysbiosis’), most significantly, reduced strict butyrate-producing anaerobes and increased facultative anaerobes. Inulin-type prebiotics appear to reduce and prevent colitis in preclinical studies and small clinical trials. However, these results need to be validated in randomized controlled clinical trial (RCT) studies. **Aims** The aim of this RCT study was to assess the efficacy of β-fructans (oligofructose and inulin) in preventing relapses in UC patients in clinical remission as well as identify potential mechanisms of activity. **Methods** Adult UC patients in clinical remission (total Mayo score ≤ 2) were randomized to supplement their diet with 15g/d of either β-fructans (oligofructose and inulin; Synergy1/Prebiotin) or placebo (maltodextrin) for 6 months. Partial Mayo scores, medications, adverse events and intervention compliance were monitored monthly. Fecal specimens were collected throughout the study to measure fecal calprotectin (FCP), along with stool metabolites. **Results** Eighty-nine UC patients in clinical remission were randomized to β-fructans (n=43) or placebo (n=46). Of those, 11 participants never started, 2 were withdrawn, and 76 were included in the study population (β-fructans n=35; placebo n=41). Although there was no difference in symptomatic clinical flare (β-fructans n=11 flare; placebo n=10 flare, P=0.60), patients randomized to oligofructose and inulin showed only a 3-fold median FCP increase versus 17-fold in the placebo group (P=0.038). Interestingly, antibiotics and serotonin reuptake inhibitors significantly increased the relative risk (RR) of flare (RR 3.321, 95% Cl 2.005 to 5.344, P < 0.0001). β-Fructan intake significantly increased anti-inflammatory fecal metabolites (arabinose, L-arabitol, 5-oxo-D-proline). **Conclusions** Although oligofructose and inulin did not prevent symptomatic relapses in UC patients, their oral administration significantly reduced the severity of biochemical relapse compared to placebo which was further associated with increased anti-inflammatory metabolites. Key Words * IBD * UC * dietary fibers * β-fructans * oligofructose * inulin * intestinal microbiota * gastroenterology * gut health ## Introduction Inflammatory bowel diseases (IBD), including Crohn disease (CD) and ulcerative colitis (UC), are common and severely debilitating chronic intestinal disorders affecting 1 in 150 people in developed nations and incidence is rising globally.1-4 The etiology of IBD remains poorly understood; however, associated risk factors include genetic susceptibility, urban lifestyle, dietary factors, heightened hygiene, and gut microbes.3, 5-10 Dysbiosis, or altered microbiome, is associated with IBD11-13 and can be impacted by diet.14-17 In particular, non-digestible carbohydrates (fiber and resistant starch) pass through the small intestine undigested; instead, some are fermented by gut microbes in the large bowel, representing a mechanistic link between diet and microbes.18-21 Fiber fermentation by microbes provides intestinal host cells with carbon and energy. Fermentation of dietary fibers produces gas, lactate, and short chain fatty acids (SCFAs)21 such as butyrate, acetate, and propionate in the colon.22, 23 SCFAs are absorbed and have beneficial physiological effects on shaping the gut microenvironment,23 immune response,24 and energy metabolism.25-29 The non-digestible inulin-type β-fructans are classified as prebiotics as they have been shown to beneficially alter activity of gut microbiota.30 Their clinical potential for use in IBD has been supported through studies demonstrating their ability to reduce intestinal inflammation in rodent colitis models,31 induce clinical response in mild UC patients associated with increased butyrate production,32 reduced fecal calprotectin (FCP) in UC patients,33 and decrease pro-inflammatory cytokines TNF-α and IL-1α mRNA expression in their colonic mucosa.34 Controversial results are reported in mild-moderate CD where inulin-type β-fructans were associated with induction of anti-inflammatory cytokines in CD mucosa, but with no reported clinical benefit.35 The efficacy of prebiotics in managing inflammation in IBD however, is not as well documented and it remains disputed whether their activity relates to the stimulation of specific members of the intestinal microbiota, or to metabolic alterations such as an increased production of SCFAs.30, 36 A reoccurring issue with such studies is that the duration of treatment may be too short to assess clinical and endoscopic improvement, and often, the colonic microbiota were not analyzed. The present randomized controlled trial (RCT) study aimed to examine safety, tolerability and efficacy, and to define mechanisms underlying the activity of inulin-type β-fructans for use in prevention of UC relapse in patients in clinical remission and who had a documented relapse in the preceding 18 months. Remission adult UC patients were randomized to supplement their diet with 15 g/d of either prebiotics (β-fructans) or placebo (maltodextrin) for 6 months. Fecal and serum specimens were collected throughout the study and outcome measurements included clinical parameters, FCP, and metabolites, which were associated with disease activity as defined by FCP change from baseline to month 6, or at relapse. Consumption of inulin-type β-fructans did not prevent clinical relapse in UC patients who were in clinical remission, however it significantly reduced risk of relapse, defined by increased FCP (subclinical or biochemical colonic inflammation). ## Methods ### Study Settings and Design This study was a single-center, double-blind, randomized, parallel, placebo controlled clinical trial (RCT) comparing the effectiveness of β-fructans (Synergy-1/Prebiotin; combination of oligofructose and inulin) versus maltodextrin (as placebo) to prevent clinical relapse among patients with inactive UC. The study, conducted at University of Alberta (Edmonton, Canada), occurred between 4 July 2016 and 1 December 2020. All research procedures performed in this trial were in strict accordance with a predefined protocol and adherence to international GCP guidelines and Declaration of Helsinki. The research protocol was approved by Health Research Ethics Board (Study ID Pro00041938) at the University of Alberta and Natural Health Directorate at Health Canada. The study is publicly accessible at the U.S. National Institute of Health database ([clinicaltrials.gov](http://clinicaltrials.gov) identification number [NCT02865707](http://medrxiv.org/lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT02865707&atom=%2Fmedrxiv%2Fearly%2F2022%2F01%2F18%2F2022.01.16.22269376.atom)). ### Patients, inclusion and exclusion criteria Patients aged 18-65 years, with clinical and endoscopic confirmed remission UC, Mayo Clinical Score ≤2 on the 12-point Mayo scale at baseline,37 endoscopic score 0 or 1, and with no documented clinical relapse within 18 months before the study, were eligible to participate. Inclusion criteria were treatment on stable doses of 5-aminosalicylic acid (5-ASA) for at least 2 weeks, azathioprine and/or biologics (infliximab, vedolizumab) for at least 2 months prior to screening, or no medication for at least 1 week prior to the start of the trial, and negative tests for Clostridoides difficile toxin, stool pathogens, and pregnancy. Exclusion criteria included use of oral or steroids 4 weeks prior to the screening, topical 5-ASA and steroids within 1 week, use of methotrexate or 6-mercaptopurine, use of antibiotics within 2 months, and use of anti-diarrheal agents within 3 days of the screening visit. Patients with significant chronic disorders (severe cardiac disease, renal failure, severe pulmonary disease or severe psychiatric disorder), any active infection, or pregnancy were also excluded. Patients matching the inclusion and exclusion criteria were approached and only those who provided written informed consent were recruited into the study. ### Study Intervention and Procedures All study products were supplied by BENEO GmbH (Germany) in sachets. The study intervention consisted of two arms: 1) β-fructans (Prebiotin/Synergy1®, oligofructose and inulin in ratio 1:1 manufactured by BENEO GmbH, Germany, and kindly provided by Jackson GI Medical LLC, Harrisburg, PA, USA) 15 g/day; and 2) Placebo (Agenamalt 22.222 maltodextrin DE19 manufactured by AGRANA Beteiligungs-AG, Austria) 15 g/day. Maltodextrin was chosen as placebo since it is thought to be fully metabolized in the small intestines and would have no effect on the colonic gut microbiota fermentation.38, 39 Participants were advised to dissolve the powder products in 200 ml warm liquid (water or other beverage) and drink it during meal. To improve tolerability, participants began with an adjustment period of 2 weeks consuming half of total daily product’s amount (7.5 g/day). A summary of study procedures is shown in **Figure 1**. At screening, consented patient underwent a sigmoidoscopic and physical examinations to confirm eligibility and total Mayo clinical score was calculated. If patients qualified, then biopsies were collected at 20-25 cm from the anal verge. In addition, demographics [including age, sex, height and weight to calculate body mass index (BMI)], full medical history, UC disease location, list of current medications and supplements were recorded. The same procedures were performed at 6 months, or at relapse. Patients were randomized in 1:1 fashion to one of the treatment arms using random block sizes of 2 and 4, stratified based on sex (males versus females). The randomization sequence was generated by University of Alberta Epidemiology Coordinating and Research (EPICORE) Centre and embedded into REDCap (Research Electronic Data Capture)40 electronic case report form (eCRF) system. Participants were required to maintain a stable dietary pattern, monitored by web-based Diet History Questionnaire II (DHQII)41 completed at baseline and 6-months/relapse as well as to remain on the same dosage and type of medication for UC maintenance (confirmed at month 1, 2, 3, 4, 5, 6 or at relapse). During the study period participants provided stool at baseline (month 0), month 1, month 3, and month 6 or disease relapse. Blood samples for haematology and chemistry were collected at baseline, month 3 and month 6 or at relapse. Incidence and severity of all adverse events were captured during the follow-up interviews every month. The adverse events were classified by the Investigator with regards to possible relationship to treatment (unrelated, unlikely, possible or probable) and according to severity (mild, moderate, severe). Compliance to study supplementation was defined as having used ≥ 85% of the provided sachets. It was assessed at month 3 and month 6, or at relapse. ![Figure 1:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/01/18/2022.01.16.22269376/F1.medium.gif) [Figure 1:](http://medrxiv.org/content/early/2022/01/18/2022.01.16.22269376/F1) Figure 1: Summary of study procedures and collected samples ### Outcome Measures The primary outcome measure was patient clinical relapse rate over 6 months of active supplemental intervention. Clinical relapse was based on partial Mayo score ≥ 3.42 Key secondary outcomes were 1) changes in FCP at month 6 or at relapse, versus baseline FCP; 2) changes in partial Mayo clinical score at month 6 or at relapse, versus baseline Mayo; 3) changes in endoscopic score at month 6 or at relapse, versus baseline endoscopic score; and 4) time to relapse. Treatment failure (disease relapse) was defined as an increase in UC disease activity measured as 1) a partial Mayo score ≥ 3; or 2) FCP concentration > 250 µg g-1.42 Treatment response was defined as maintained remission based on the FCP at month 6 versus month 0 (FCP <250 µg g-1 and/or FCP rise of less than 100 at endpoint). For participants who withdrew before the time of treatment assessment (month 6 or relapse) or were lost to follow-up missing data were imputed using last observation carried forward. Additional safety outcome measures included number of patients experiencing a serious adverse event (SAE) during treatment period as well as stool culture testing positive for intestinal pathogens. Safety assessment also included measures of C-reactive protein (CRP), blood haematology (hemoglobin, MCV, platelets, WBC, albumin), iron deficiency [iron, total iron binding capacity (TIBC), ferritin iron saturation index], serum liver and kidney markers (serum creatinine, aspartate aminotransferase (AST), alanine transaminase (ALT), gamma-glutamyl transferase (GGT), alkaline phosphatase (ALP), bilirubin) as well as 25-hydroxy vitamin D at baseline versus month 3 and endpoint (month 6 or relapse). Tolerability outcome measures were defined based on total treatment emergent adverse events (TEAE) versus such TEAE leading to withdrawal. ### Sample Size Determination Data from our pilot study in UC patients with active disease supplemented with β-fructans (Synergy1®) to stable dose 5-ASA treatment for 9 weeks demonstrated a 77% response in 15 g/day dose versus 33% in a lower dose of 7.5 g/day.32 Based on this study outcome we have chosen a dose of 15 g/day to be tested in the current study with an estimated 30% difference in the incidence of UC patients experiencing clinical relapse by 6 months. A total of 84 patients, 42 in each arm, were required to detect a difference of 30% with a power of 80% using a two-sided p=0.05 level test. With an anticipated dropout rate of 20%, the overall sample size for this trial was proposed be 100 patients. ### Metabolomics Untargeted metabolites were isolated from patient stool samples as directed by the Calgary Metabolomics Research Facility, University of Calgary. On ice, 100-200mg of stool were measured into 1.5mL Eppendorf tubes and weights were recorded. Five volumes of pre-chilled methanol was added to each sample, then homogenized at max speed in the MPBio Fastprep24 bead beater for 1 min. Samples were incubated on ice for 30min then centrifuged at max speed, 4c, for 10min. Supernatants were processed for µM concentration of metabolites by Calgary Metabolomics Research Facility staff. ### Quantification and Statistical Analysis #### Statistical Analysis Groups were compared using paired Wilcoxon *t*-test (two-tailed) analysis, ANOVA, or Kendall, depending on the relevant question, using GraphPad Prism. A p-value of <0.05 was considered significant in all cases and all error deviations are described by ± SEM. ## Results ### Recruitment and Participant Flow We screened 113 potential participants, of whom 34 failed to meet the eligibility criteria. **Figure 2** outlines the patient flow in the recruitment, intervention allocation and reasons for exclusion. Of the 89 patients who successfully passed screening, 43 were randomly assigned to the β-fructans arm and 46 to the placebo arm. One participant developed allergic reaction to β-fructans, manifested by skin rash, shortly after starting the intervention (24-48h), and was immediately withdrawn. Two participants, one in each group, had to be withdrawn from the study as they became ineligible (one had recurrent *C. difficile* infection, and one had protocol violation related to medication and failure to randomization). Finally, 10 participants never started the study procedures and were lost to follow-up. Thus, the safety study population included 76 subjects with 35 in the β-fructans arm and 41 participants in the placebo arm who participated in at least one follow-up study visits. The demographics and baseline characteristics of the safety population are shown in **Table 1** and were similar across the treatment groups. Overall, 57% of the participants were women and mean age at entry was 45.0 ± 13.4 years. The median body mass index (BMI, kg m-2) was 27.0 (IQR 24.5-30.0) suggesting a population mostly in the overweight category. Baseline median FCP concentrations were 113.8 µg g-1 (IQR 48.9 - 471.1) with about one third of the participants in each arm having elevated concentrations > 250 µg g-1 (β-fructans n=9, 26%; placebo n=13, 31%) though exhibiting no clinical symptoms. A history of pan-colitis accounted for 60% of all UC. At baseline 33% of the participants were taking biologic medications, 41% were only on 5-ASA and 16% were on no medication related to UC. View this table: [Table 1.](http://medrxiv.org/content/early/2022/01/18/2022.01.16.22269376/T1) Table 1. Baseline demographics of the Safety study population ![Figure 2:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/01/18/2022.01.16.22269376/F2.medium.gif) [Figure 2:](http://medrxiv.org/content/early/2022/01/18/2022.01.16.22269376/F2) Figure 2: Patient flow in the recruitment, intervention allocation and reason for exclusion. Of the 89 patients who successfully passed screening, 43 were randomly assigned to receive β-fructans intervention and 46 to the placebo arm. 13 patients either never started the trial or were lost to follow-up therefore, the intention-to-treat study population included 76 subjects with 35 in the β-fructans arm and 41 in the placebo arm. Further patients withdrew their consent during the study, therefore in total 65 participants completed the study including 29 in the β-fructans arm and 36 in the placebo arm. ### Adverse Events and Safety Parameters All symptoms occurring during the study were documented and rated for severity and potential relationship with the study products. **Table 2** summarizes all treatment emergent adverse events (TEAE) described by the study population during the course of 6 months intervention. Significantly more participants in the β-fructans group reported the onset of new symptoms (n=25, 71%, p=0.006) in comparison to such TEAE in the placebo population (n=16, 39%). Inulin type β-fructans intake was associated with mild or moderate flatulence and bloating reported by 51% of the study population (n=18) and thus was considered as a treatment related adverse event. Five participants elected withdrawal due to this TEAE. FCP analysis determined that this TEAE was not associated with increased subclinical inflammation [baseline FCP median 76.1 µg g-1 (IQR 45.4 - 589.3) versus early withdrawal FCP median 42.9 µg g-1 (IQR 21.8 - 92.3), p = 0.625] (**Supplementary Figure 1**). For the remaining patients experiencing flatulence and bloating, this symptom resolved within 2-8 weeks. Other gastrointestinal symptoms, such as pyrosis, diarrhea or constipation were insignificant and equally reported by both treatment groups, although resulted in one participant withdrawal in the placebo group. Besides gastrointestinal-associated TEAEs, 12 patients, 6 in each treatment group, reported acute infections (respiratory, tooth abscess, STI) and 9 were treated with antibiotics (β-fructans n = 4; placebo n = 5). Finally, most haematologic and serum parameters remained in the normal ranges and did not significantly change across groups from baseline (**Table 3**). CRP levels remained low and BMI index did not alter after 6 months prebiotic intervention. ![Supplementary Figure 1:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/01/18/2022.01.16.22269376/F5.medium.gif) [Supplementary Figure 1:](http://medrxiv.org/content/early/2022/01/18/2022.01.16.22269376/F5) Supplementary Figure 1: Fecal calprotectin measured at baseline and withdrawal in participants who withdrew from the study early. View this table: [Table 2.](http://medrxiv.org/content/early/2022/01/18/2022.01.16.22269376/T2) Table 2. Summary ofTreatment Emergent Adverse Events Recorded in the Safety Population View this table: [Table 3.](http://medrxiv.org/content/early/2022/01/18/2022.01.16.22269376/T3) Table 3. Changes in blood haematology, serum liver and kidney markers in UC patients in clinical remission consuming inulin-type β-fructans for 6 months. ### Primary and Secondary Outcomes The primary outcome of the study was maintenance of clinical remission during 6 months intervention. Treatment failure (relapse) was described as an increase in clinical symptoms (partial Mayo score ≥ 3; symptomatic relapse) and/or by changes in the biochemical marker FCP (endpoint FCP > 250 µg g-1 or FCP rise of more than 100 µg g-1 in patients with high baseline FCP; FCP relapse). Linear regression analysis showed strong correlation between symptom changes (partial Mayo score) and FCP changes (F-value = 70.39, r2 = 0.53, p < 0.0001). During the 6-months intervention 11 individuals in β-fructans group (31%) and 10 in the placebo group (24%) experienced symptomatic relapse (chi-square 1.23, p = 0.26; **Figure 3A**). Similarly, there was no significant difference in the proportion of patients who showed biochemical relapse (β-fructans n = 10, 29%; placebo n = 16, 39%; chi-square 0.094, p = 0.76). In addition, 4 patients (β-fructans n = 2; placebo n = 2) experienced symptomatic relapse during the 6-months post-intervention observational period. Log-rank test comparing the relapse events over the 12 months period (6 months intervention followed by 6 months observation) showed no significant difference in the time to relapse between β-fructans and placebo (p = 0.92; **Figure 3B**). Based on this outcome we concluded that β-fructans intake was ineffective in improving the clinical relapse rates or time to clinical relapse. Consequently, the trial was prematurely terminated due to lack of efficacy and further recruitment was halted. ![Figure 3:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/01/18/2022.01.16.22269376/F3.medium.gif) [Figure 3:](http://medrxiv.org/content/early/2022/01/18/2022.01.16.22269376/F3) Figure 3: Clinical outcomes as measured in participants at baseline and endpoint. We then assessed the secondary outcome measures, notably changes in clinical parameters partial Mayo score, colitis endoscopic score, and FCP. At baseline both treatment groups had similar partial Mayo score (β-fructans 0.0±0.0; placebo 0.0±0.2, p > 0.99), endoscopic scores (β-fructans 0.5±0.5; placebo 0.5±0.5, p > 0.99), and FCP (β-fructans median 113.3, IQR 48.7-333.9; placebo median 129.1, IQR 47.8-539.5, p = 0.63). Two-way ANOVA test showed that both β-fructans and placebo groups were characterized with a significant increase in the UC-related symptoms (partial Mayo score) versus the baseline measures (p=0.002; endpoint β-fructans 0.9±1.8; placebo 0.9±1.9; **Figure 3C**) and insignificant increase in the endoscopic scores (endpoint β-fructans 0.8±1.0; placebo 0.7±1.0; **Figure 3D**), however there was no difference between the two treatments (p>0.99). In contrast, assessment of FCP changes showed a slight but not significant reduction in FCP levels in β-fructans group (endpoint β-fructans median 73.3, IQR 22.4-1072.0, p=0.35). Placebo group demonstrated a significant increase in the FCP levels at treatment endpoint (endpoint placebo median 285.5, IQR 49.1-1122.0, p = 0.036; **Figure 3E**). Finally, there was no statistical difference in FCP absolute levels (p = 0.19), nor in FCP fold change (p = 0.10), between groups at the intervention endpoint. However, data showed that while those clinically relapsing patients randomized to β-fructans had a 3-fold median increase in their FCP from baseline (IQR 2.3-8.6), relapsing patients randomized to the placebo had a 17-fold median increase (IQR 4.6-47.0; p=0.038; **Figure 3F**); a significant difference between groups. ### Consumption of β-fructans induced significant changes in metabolites in participant stool To understand difference between patients in relapse vs remission, we examined metabolite abundance changes over time via untargeted metabolomics of stool collected from β-fructans remission (n=15), β-fructans relapse (n=10), placebo remission (n=15), and placebo relapse (n=15) participants (**Figure 4**). At month 0, participants randomized to the β-fructans arm who remained in remission during the study, had significantly higher fecal arabinose, 3-hydroxy-3-methylglutarate, 3-hydroxybutanoic acid, α-D-glucose, D-alanine, acetoacetate, ferulate, and D-gulonic acid γ-lactone, and significantly less leucine and L-glutamic acid than those in the β-fructan arm who relapsed (**Figure 4A**). In contrast, participants randomized to the placebo arm who remained in remission had significantly higher arabinose (like β-fructans arm) and N-acetyl-L-aspartic acid, compared to those in the placebo arm who relapsed. ![Figure 4:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/01/18/2022.01.16.22269376/F4.medium.gif) [Figure 4:](http://medrxiv.org/content/early/2022/01/18/2022.01.16.22269376/F4) Figure 4: Metabolites were examined in patient stool samples isolated from synergy remission (n=8), synergy relapse (n=3), placebo remission (n=10), and placebo relapse (n=4) taken at **(A)** month 0, **(B)** month 6, and **(C)** month 6 versus month 0. **p<0.01, *p<0.05. At month 6, participants randomized to the β-fructans arm who remained in remission had significantly higher 3-hydroxy-3-methylglutarate, 3-hydroxybutanoic acid, α-D-glucose, and L-arabitol, along with significantly less 4-methyl-2-oxo-pentanoic acid and L-serine than those in the β-fructans arm who relapsed (**Figure 4B**). In contrast, participants randomized to the placebo arm who remained in remission had significantly higher arabinose, N-acetyl-DL-serine, 2”“-deoxyguanosine, D-gulonic acid γ-lactone, thymidine, D-glucuronolactone, guanine, L-arginine, formyl-L-methionyl peptide, corticosterone, cortisone, and cortisol along with decreased salicylate compared to those in the placebo arm who relapsed. Comparing the fold-change in metabolites from month 6 versus month 0, participants randomized to the β-fructans arm who remained in remission had significantly greater increased fold-change in N-acetyl-DL-serine, L-arabitol, 5-oxo-D-proline, 3-[4-hydroxyphenyl]pyruvate, and omega-hydroxydodecanoic acid, along with significantly decreased 2-quinolinecarboxylin acid, than those in the β-fructans arm who relapsed (**Figure 4C**). In contrast, participants randomized to the placebo arm who remained in remission had significantly higher 2”“-deoxyguanosine, mannitol, allantoin, and guanine compared to those in the placebo arm who relapsed. ## Discussion An altered intestinal microbiome is well linked to IBD43-45 and mounting evidence supports the use of dietary interventions as a successful approach to re-establish microbiota in mild to moderately active IBD. Dietary fibres, in particular, are a key energy source for gut microbiota and fructan fibres have previously been shown to reduce colitis in animal models of colitis,31, 46 and in small clinical studies.32-34 Unfortunately, studies related to the clinical efficacy of fructan fibres remain limited, and those that have been published suffer from short treatment duration and small participant numbers. This RCT shows that consumption of β-fructans for 6 months by UC patients in remission was ineffective at improving clinical relapse rate or time to relapse however, β-fructans were able to reduce the overall severity of relapse by preventing subclinical inflammation or significantly reducing the amount of FCP increase during symptomatic relapse; UC patients randomized to β-fructans who relapsed had a 3-fold median increase in their FCP from baseline compared to patients randomized to placebo who relapsed (17-fold median increase; p = 0.038). A number of metabolites were significantly altered in response to β-fructan consumption. Arabinose exerts anti-inflammatory effects in mouse models of colitis47 and was found to be higher at baseline in both the β-fructan and placebo arm participants who remained in remission at month 6, suggesting that high fecal arabinose may be a predictor of remission in UC patients. Unique to overall response to β-fructan consumption, those patients who remained in remission also had significantly higher L-arabitol than patients who relapsed; arabitol is produced from arabinose, although it has not previously been well defined, it can indicate overgrowth of select microbes in the gut.48 Increased 5-oxo-D-proline was also identified in the β-fructan remission group, compared to relapse, and has been previously shown to be significantly higher in IBD patients, linked to coping mechanisms of oxidative stress and inflammation.49 3-[4-hydroxyphenyl]pyruvate, increased in those who remained in remission following consumption of β-fructans, has also previously been shown to be upregulated in IBD, although its potential role remains unknown.50 Lastly, hydroxydodecanoic acid, increased in those who remained in remission following consumption of β-fructans, has been shown to have anti-inflammatory effects in limited studies.51 Overall, the specific pathways associated with remission following consumption of β-fructans have previously been identified for their significant anti-inflammatory effects, demonstrating potential mechanistic effects that will need to be investigated in future biochemical studies. In conclusion, this RCT study suggest that intake of 15 g/day β-fructans over a 6-month period could induce significant reduction in FCP and subclinical remission. These findings suggest that high-dose inulin-type fructan fibre supplement improves subclinical colitis demonstrating their promising potential as an adjunct treatment to aid in maintaining remission in UC. Further well-powered RCT with these prebiotics are warranted to better define personalized response to β-fructans and identify biomarkers predicting which patients are most likely to benefit from these interventions. ## Data Availability All data produced in the present work are contained in the manuscript ## AUTHOR CONTRIBUTIONS LD, RV, and HA conceived, developed, and coordinated the project. RV, OK, and MS acquired the patient consent. RV, HA, OK, MBJ, SV, MS, FP, KW, DK, SVZ, KK, MG, and LD collected and/or processed stool, mucosal washings and biopsy tissues. RV, SV, HA, NH, MEPM, and MBJ performed and/or analyzed experiments. RV and HA were responsible for the figure preparation and statistical analyses. JW, DG, and EW provided additional supervision and oversight. HA and RV drafted the manuscript. All authors contributed to manuscript edits and approved the final version of the manuscript. ## DECLARATION OF INTERESTS The authors have no competing interests to declare. ## GRANT SUPPORT The funders had no role in study design, collection, or interpretation of data. The following grants were used to fund the project: Weston Family Foundation Microbiome Initiative (LD). Weston Family Foundation Microbiome Initiative grant (EW, HA, RV, LD). Canadian Institute of Health Research grants (LD, EW). HA was supported by a Canadian Institute of Health Research postdoctoral fellowship. HA and MBJ were supported by Mitacs Elevate fellowships in partner with the Weston Family Foundation. ## ACKNOWLEDGEMENTS We graciously thank the patients for agreeing to participate and provide samples, as well as the IBD research coordinators, nurses, and endoscopy staff at the Zeidler Ledcor IBD Clinic, University of Alberta Hospital. We would like to acknowledge The Applied Genomic Core at the University of Alberta along with IMPACTT Microbiome and the Calgary Metabolomics Research Facility at the University of Calgary for their help in experiment preparations and technical procedures. We also thank CEGIIR for providing material support. LD was supported by CIHR and Weston Family Foundation grants and also sponsored by Jackson GI; along with Weston Family Foundation funding awarded to EW, HA, LD, and RV. The funders had no role in study design, collection, or interpretation of data. * Received January 16, 2022. * Revision received January 16, 2022. * Accepted January 18, 2022. * © 2022, Posted by Cold Spring Harbor Laboratory The copyright holder for this pre-print is the author. All rights reserved. The material may not be redistributed, re-used or adapted without the author's permission. ## References 1. 1.Collaborators GBDIBD. The global, regional, and national burden of inflammatory bowel disease in 195 countries and territories, 1990-2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet Gastroenterol Hepatol 2020;5:17–30. 2. 2.Kaplan GG. The global burden of IBD: from 2015 to 2025. Nat Rev Gastroenterol Hepatol 2015;12:720–7. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nrgastro.2015.150&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26323879&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F01%2F18%2F2022.01.16.22269376.atom) 3. 3.Abu Freha N, Schwartz D, Elkrinawi J, et al. Inflammatory bowel disease among Bedouin Arabs in southern Israel: urbanization and increasing prevalence rates. Eur J Gastroenterol Hepatol 2015;27:230–4. 4. 4.Coward S, Clement F, Benchimol EI, et al. Past and Future Burden of Inflammatory Bowel Diseases Based on Modeling of Population-Based Data. Gastroenterology 2019;156:1345–1353 e4. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F01%2F18%2F2022.01.16.22269376.atom) 5. 5.Ng SC. Emerging leadership lecture: Inflammatory bowel disease in Asia: emergence of a “Western” disease. J Gastroenterol Hepatol 2015;30:440–5. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/jgh.12859&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25469874&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F01%2F18%2F2022.01.16.22269376.atom) 6. 6.Gasparetto M, Guariso G. Highlights in IBD Epidemiology and Its Natural History in the Paediatric Age. Gastroenterol Res Pract 2013;2013:829040. 7. 7.Benchimol EI, Mack DR, Guttmann A, et al. Inflammatory bowel disease in immigrants to Canada and their children: a population-based cohort study. Am J Gastroenterol 2015;110:553–63. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ajg.2015.52&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25756238&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F01%2F18%2F2022.01.16.22269376.atom) 8. 8.Wine E. Should we be treating the bugs instead of cytokines and T cells? Dig Dis 2014;32:403–9. 9. 9.Aujnarain A, Mack DR, Benchimol EI. The role of the environment in the development of pediatric inflammatory bowel disease. Curr Gastroenterol Rep 2013;15:326. 10. 10.Levine A, Wine E. Effects of enteral nutrition on Crohn’s disease: clues to the impact of diet on disease pathogenesis. Inflamm Bowel Dis 2013;19:1322–9. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1097/MIB.0b013e3182802acc&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23399738&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F01%2F18%2F2022.01.16.22269376.atom) 11. 11.Carding S, Verbeke K, Vipond DT, et al. Dysbiosis of the gut microbiota in disease. Microb Ecol Health Dis 2015;26:26191. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3402/mehd.v26.26191&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25651997&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F01%2F18%2F2022.01.16.22269376.atom) 12. 12.Casen C, Vebo HC, Sekelja M, et al. Deviations in human gut microbiota: a novel diagnostic test for determining dysbiosis in patients with IBS or IBD. Aliment Pharmacol Ther 2015;42:71–83. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/apt.13236&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25973666&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F01%2F18%2F2022.01.16.22269376.atom) 13. 13.Imhann F, Vich Vila A, Bonder MJ, et al. Interplay of host genetics and gut microbiota underlying the onset and clinical presentation of inflammatory bowel disease. Gut 2018;67:108–119. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiZ3V0am5sIjtzOjU6InJlc2lkIjtzOjg6IjY3LzEvMTA4IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMDEvMTgvMjAyMi4wMS4xNi4yMjI2OTM3Ni5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 14. 14.Singh RK, Chang HW, Yan D, et al. Influence of diet on the gut microbiome and implications for human health. J Transl Med 2017;15:73. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/s12967-017-1175-y&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28388917&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F01%2F18%2F2022.01.16.22269376.atom) 15. 15.Eid N, Enani S, Walton G, et al. The impact of date palm fruits and their component polyphenols, on gut microbial ecology, bacterial metabolites and colon cancer cell proliferation. J Nutr Sci 2014;3:e46. 16. 16.Francavilla R, Calasso M, Calace L, et al. Effect of lactose on gut microbiota and metabolome of infants with cow’s milk allergy. Pediatr Allergy Immunol 2012;23:420–7. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/j.1399-3038.2012.01286.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22435727&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F01%2F18%2F2022.01.16.22269376.atom) 17. 17.Suez J, Korem T, Zeevi D, et al. Artificial sweeteners induce glucose intolerance by altering the gut microbiota. Nature 2014;514:181–6. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature13793&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25231862&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F01%2F18%2F2022.01.16.22269376.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000342663100032&link_type=ISI) 18. 18.Pistollato F, Sumalla Cano S, Elio I, et al. Role of gut microbiota and nutrients in amyloid formation and pathogenesis of Alzheimer disease. Nutr Rev 2016;74:624–34. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/nutrit/nuw023&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27634977&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F01%2F18%2F2022.01.16.22269376.atom) 19. 19.Lozupone CA, Stombaugh JI, Gordon JI, et al. Diversity, stability and resilience of the human gut microbiota. Nature 2012;489:220–30. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature11550&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22972295&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F01%2F18%2F2022.01.16.22269376.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000308635900030&link_type=ISI) 20. 20.Sonnenburg ED, Sonnenburg JL. Starving our microbial self: the deleterious consequences of a diet deficient in microbiota-accessible carbohydrates. Cell Metab 2014;20:779–786. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cmet.2014.07.003&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25156449&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F01%2F18%2F2022.01.16.22269376.atom) 21. 21.Armstrong H, Mander, I., Zhang, Z., Armstrong, D., Wine, E. Not all fibres are born equal; variable response to dietary fibre subtypes in IBD. Frontiers in Pediatrics 2021;8. 22. 22.Cummings JH, Pomare EW, Branch WJ, et al. Short chain fatty acids in human large intestine, portal, hepatic and venous blood. Gut 1987;28:1221–7. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiZ3V0am5sIjtzOjU6InJlc2lkIjtzOjEwOiIyOC8xMC8xMjIxIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMDEvMTgvMjAyMi4wMS4xNi4yMjI2OTM3Ni5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 23. 23.Rios-Covian D, Ruas-Madiedo P, Margolles A, et al. Intestinal Short Chain Fatty Acids and their Link with Diet and Human Health. Front Microbiol 2016;7:185. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3389/fmicb.2016.00185&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F01%2F18%2F2022.01.16.22269376.atom) 24. 24.Smith PM, Howitt MR, Panikov N, et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 2013;341:569–73. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEyOiIzNDEvNjE0NS81NjkiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMi8wMS8xOC8yMDIyLjAxLjE2LjIyMjY5Mzc2LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 25. 25.den Besten G, van Eunen K, Groen AK, et al. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J Lipid Res 2013;54:2325–40. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamxyIjtzOjU6InJlc2lkIjtzOjk6IjU0LzkvMjMyNSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIyLzAxLzE4LzIwMjIuMDEuMTYuMjIyNjkzNzYuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 26. 26.Chambers ES, Preston T, Frost G, et al. Role of Gut Microbiota-Generated Short-Chain Fatty Acids in Metabolic and Cardiovascular Health. Curr Nutr Rep 2018;7:198–206. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s13668-018-0248-8&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F01%2F18%2F2022.01.16.22269376.atom) 27. 27.Parada Venegas D, De la Fuente MK, Landskron G, et al. Short Chain Fatty Acids (SCFAs)-Mediated Gut Epithelial and Immune Regulation and Its Relevance for Inflammatory Bowel Diseases. Front Immunol 2019;10:277. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3389/FIMMU.2019.00277/BIBTEX&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F01%2F18%2F2022.01.16.22269376.atom) 28. 28.Zhao Y, Chen F, Wu W, et al. GPR43 mediates microbiota metabolite SCFA regulation of antimicrobial peptide expression in intestinal epithelial cells via activation of mTOR and STAT3. Mucosal Immunol 2018;11:752–762. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/mi.2017.118&link_type=DOI) 29. 29.Macia L, Tan J, Vieira AT, et al. Metabolite-sensing receptors GPR43 and GPR109A facilitate dietary fibre-induced gut homeostasis through regulation of the inflammasome. Nat Commun 2015;6:6734. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ncomms7734&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25828455&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F01%2F18%2F2022.01.16.22269376.atom) 30. 30.Flint HJ, Scott KP, Duncan SH, et al. Microbial degradation of complex carbohydrates in the gut. Gut Microbes 2012;3:289–306. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.4161/gmic.19897&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22572875&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F01%2F18%2F2022.01.16.22269376.atom) 31. 31.Koleva PT, Valcheva RS, Sun X, et al. Inulin and fructo-oligosaccharides have divergent effects on colitis and commensal microbiota in HLA-B27 transgenic rats. Br J Nutr 2012;108:1633–43. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1017/S0007114511007203&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22243836&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F01%2F18%2F2022.01.16.22269376.atom) 32. 32.Valcheva R, Koleva P, Martinez I, et al. Inulin-type fructans improve active ulcerative colitis associated with microbiota changes and increased short-chain fatty acids levels. Gut Microbes 2019;10:334–357. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1080/19490976.2018.1526583&link_type=DOI) 33. 33.Casellas F, Borruel N, Torrejon A, et al. Oral oligofructose-enriched inulin supplementation in acute ulcerative colitis is well tolerated and associated with lowered faecal calprotectin. Aliment Pharmacol Ther 2007;25:1061–7. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/j.1365-2036.2007.03288.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17439507&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F01%2F18%2F2022.01.16.22269376.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000245602300007&link_type=ISI) 34. 34.Furrie E, Macfarlane S, Kennedy A, et al. Synbiotic therapy (Bifidobacterium longum/Synergy 1) initiates resolution of inflammation in patients with active ulcerative colitis: a randomised controlled pilot trial. Gut 2005;54:242–9. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiZ3V0am5sIjtzOjU6InJlc2lkIjtzOjg6IjU0LzIvMjQyIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMDEvMTgvMjAyMi4wMS4xNi4yMjI2OTM3Ni5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 35. 35.Benjamin JL, Hedin CR, Koutsoumpas A, et al. Randomised, double-blind, placebo-controlled trial of fructo-oligosaccharides in active Crohn’s disease. Gut 2011;60:923–9. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiZ3V0am5sIjtzOjU6InJlc2lkIjtzOjg6IjYwLzcvOTIzIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMDEvMTgvMjAyMi4wMS4xNi4yMjI2OTM3Ni5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 36. 36.Bindels LB, Neyrinck AM, Salazar N, et al. Non Digestible Oligosaccharides Modulate the Gut Microbiota to Control the Development of Leukemia and Associated Cachexia in Mice. PLoS One 2015;10:e0131009. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26098097&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F01%2F18%2F2022.01.16.22269376.atom) 37. 37.Schroeder KW, Tremaine WJ, Ilstrup DM. Coated oral 5-aminosalicylic acid therapy for mildly to moderately active ulcerative colitis. A randomized study. N Engl J Med 1987;317:1625–9. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJM198712243172603&link_type=DOI) 38. 38.Hofman DL, van Buul VJ, Brouns FJ. Nutrition, Health, and Regulatory Aspects of Digestible Maltodextrins. Crit Rev Food Sci Nutr 2016;56:2091–100. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F01%2F18%2F2022.01.16.22269376.atom) 39. 39.Zhang X, Leemhuis H, van der Maarel M. Digestion kinetics of low, intermediate and highly branched maltodextrins produced from gelatinized starches with various microbial glycogen branching enzymes. Carbohydr Polym 2020;247:116729. 40. 40.Harris PA, Taylor R, Thielke R, et al. Research electronic data capture (REDCap)--a metadata-driven methodology and workflow process for providing translational research informatics support. J Biomed Inform 2009;42:377–81. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jbi.2008.08.010&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18929686&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F01%2F18%2F2022.01.16.22269376.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000264958800018&link_type=ISI) 41. 41.National institute of Health EaRP, Institute. Canadian Diet History Questionnaire (CDHQ-II). [https://canadiandhqii.com/survey_resources.html](https://canadiandhqii.com/survey_resources.html), 2014. 42. 42.D’Haens G, Ferrante M, Vermeire S, et al. Fecal calprotectin is a surrogate marker for endoscopic lesions in inflammatory bowel disease. Inflamm Bowel Dis 2012;18:2218–24. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/ibd.22917&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22344983&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F01%2F18%2F2022.01.16.22269376.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000311242400003&link_type=ISI) 43. 43.Morgan XC, Tickle TL, Sokol H, et al. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol 2012;13:R79. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/gb-2012-13-9-r79&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23013615&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F01%2F18%2F2022.01.16.22269376.atom) 44. 44.Erickson AR, Cantarel BL, Lamendella R, et al. Integrated metagenomics/metaproteomics reveals human host-microbiota signatures of Crohn’s disease. PLoS One 2012;7:e49138. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pone.0049138&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23209564&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F01%2F18%2F2022.01.16.22269376.atom) 45. 45.Sokol H, Pigneur B, Watterlot L, et al. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc Natl Acad Sci U S A 2008;105:16731–6. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMjoiMTA1LzQzLzE2NzMxIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMDEvMTgvMjAyMi4wMS4xNi4yMjI2OTM3Ni5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 46. 46.Hoentjen F, Welling GW, Harmsen HJ, et al. Reduction of colitis by prebiotics in HLA-B27 transgenic rats is associated with microflora changes and immunomodulation. Inflamm Bowel Dis 2005;11:977–85. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1097/01.MIB.0000183421.02316.d5&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16239843&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F01%2F18%2F2022.01.16.22269376.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000233211100004&link_type=ISI) 47. 47.Li Y, Pan H, Liu JX, et al. l-Arabinose Inhibits Colitis by Modulating Gut Microbiota in Mice. J Agric Food Chem 2019;67:13299–13306. 48. 48.Lehtonen L, Anttila VJ, Ruutu T, et al. Diagnosis of disseminated candidiasis by measurement of urine D-arabinitol/L-arabinitol ratio. J Clin Microbiol 1996;34:2175–9. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNtIjtzOjU6InJlc2lkIjtzOjk6IjM0LzkvMjE3NSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIyLzAxLzE4LzIwMjIuMDEuMTYuMjIyNjkzNzYuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 49. 49.Martin FP, Su MM, Xie GX, et al. Urinary metabolic insights into host-gut microbial interactions in healthy and IBD children. World J Gastroenterol 2017;23:3643–3654. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3748/wjg.v23.i20.3643&link_type=DOI) 50. 50.Martin FP, Ezri J, Cominetti O, et al. Urinary Metabolic Phenotyping Reveals Differences in the Metabolic Status of Healthy and Inflammatory Bowel Disease (IBD) Children in Relation to Growth and Disease Activity. Int J Mol Sci 2016;17. 51. 51.Chen YF, Wang K, Zhang YZ, et al. In Vitro Anti-Inflammatory Effects of Three Fatty Acids from Royal Jelly. Mediators Inflamm 2016;2016:3583684.