586598

research-article2015

NCPXXX10.1177/0884533615586598Nutrition in Clinical PracticeParian et al

CAM Corner

Nutraceutical Supplements for Inflammatory Bowel Disease Alyssa M. Parian, MD1; Berkeley N. Limketkai, MD1,2; Neha D. Shah, MPH, RD, CNSC3; and Gerard E. Mullin, MD, CNSP1

Inflammatory bowel diseases (IBDs), specifically Crohn’s disease (CD) and ulcerative colitis (UC), are relapsing and remitting inflammatory diseases of the intestines. The etiology remains unknown; however, aberrations in genetics, imbalances in the gut microbiome, dietary and lifestyle factors such as cigarette smoking, medications, and environmental triggers are all believed to play a role in disease pathogenesis. A diet high in fiber and omega-3 fatty acids is protective for the development of IBD, whereas a diet high in refined sugars, complex carbohydrates, and omega-6 fatty acids (ie, red meat) increases the risk of acquiring IBD.1-5 The gut microbiome of patients with IBD is characterized by a reduced biodiversity compared with that of disease-free healthy controls.6,7 An aberrant mucosal immune system and irregular mucosal epithelium with increased permeability may permit the translocation of bacterial-derived toxins with resulting gut inflammation.8 This review aims to summarize the data behind complementary and nutrition therapies that directly influence the key steps in the pathogenesis of IBD and thus should be considered in the treatment of IBD.

Prebiotics Prebiotics are nondigestible fibrous products that selectively promote the growth or activity of commensal microorganisms that improve the well-being of the host.9 Prebiotics are carbohydrates that are not typically digestible by the human gastrointestinal tract; therefore, they are selectively fermented by commensal bacteria (ie, Bifidobacteria) and provide nutrients for their metabolism. There are multiple different prebiotics including inulin, fructo-oligosaccharides, galacto-oligosaccharides, soybean oligosaccharides, and complex polysaccharides. Fermentation of prebiotics produces short-chain fatty acids including butyrate, which has anti-inflammatory properties, promotes colonic epithelial integrity and healing, reduces neoplasia, and more.10-14 Inulin and oligofructoses promote the growth of lactobacilli and bifidobacteria, which may reduce inflammation. Prebiotics are not as well studied as probiotics for their influence in the natural history of IBD, with only small noncontrolled studies, typically including only patients with UC. Inulin was found to lower fecal calprotectin level, a marker of colonic

Nutrition in Clinical Practice Volume XX Number X Month 201X 1­–8 © 2015 American Society for Parenteral and Enteral Nutrition DOI: 10.1177/0884533615586598 ncp.sagepub.com hosted at online.sagepub.com

inflammation in patients with UC,15 and germinated barley foodstuff decreased UC clinical activity scores16 and maintained remission.17 Plantago ovata improved UC clinical symptoms and increased the health-promoting short-chain fatty acid butyrate in the stool, although the adverse effects of Plantago ovata, including constipation and flatulence, limit its use.18,19

Probiotics Probiotics are defined by the World Health Organization as “live micro-organisms which, when administered in adequate amounts, confer a health benefit on the host.”20 Probiotics aim to alter the microflora of the intestines to a more favorable subset of organisms that may have anti-inflammatory and gut healing properties. An increase in commensal bacteria and decrease in proinflammatory organisms such as Escherichia coli, Enterobacter aerogenes, Klebsiella pneumonia, Streptococcus viridans, Bacteriodes fragilis, Bacteriodes uniformis, and Clostridium ramosum can promote the production of shortchain fatty acids (SCFAs) such as butyrate. SCFAs lower the pH of the colon and therefore prohibit the growth of pathogenic organisms. In addition, probiotics may decrease intestinal permeability and improve the barrier function of the intestines.21 Probiotics also down regulate proinflammatory mediators and induce T regulatory cells to dampen damaging autoimmune responses.22-25 VSL#3 (Sigma-Tau Pharmaceuticals, Gaithersburg, MD) contains 3 genera of bacteria: Lactobacilli (L casei, L plantarum, L acidophilus, L delbrueckii), Bifidobacteria (B longum, B breve, B infantis), and Streptococcus (S salivarius). Multiple trials have found it to have an excellent safety profile. A From 1Division of Gastroenterology & Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland; 2Division of Gastroenterology & Hepatology, Stanford University School of Medicine, Palo Alto, California; and 3Digestive Health Center, Stanford Health Care, Palo Alto, California. Financial disclosure: None declared. Corresponding Author: Alyssa M. Parian, MD, Johns Hopkins Hospital, 4940 Eastern Avenue, A-Building, Suite 502, Baltimore, MD 21224, USA. Email: [email protected]

Downloaded from ncp.sagepub.com at DALHOUSIE UNIV on June 19, 2015

2

Nutrition in Clinical Practice XX(X)

randomized, placebo-controlled trial found that UC patients with pouchitis treated with VSL#3 had an 85% decrease in the rate of relapse.26 A 30% decreased rate of pouchitis was found when using VSL#3 as the primary prevention of pouchitis in patients with UC.27 Current guidelines for the management of pouchitis suggest that VSL#3 be used for prevention of recurrent pouchitis in patients with UC, but there are not enough data to recommend it for the treatment of acute pouchitis.28 Multiple small studies have found positive results for the use of VSL#3 in the induction and maintenance of remission of UC.29-34 A meta-analysis by Jonkers et al35 showed that higher doses of VSL#3 (3600 billion bacteria daily) induced remission in active UC 70% more frequently than placebo.29 Studies of VSL#3 in patients with CD are more limited and less appealing.36-39 A prospective, placebo-controlled trial found that VSL#3 started within 30 days of ileal resection and re-anastomosis trended toward a decreased endoscopic recurrence (9.3% vs 15.7%, P = .19). In addition, compared with placebo, those who received VSL#3 had a decrease in proinflammatory mucosal cytokine expression. The timing seemed to make a difference, as those who were started on the probiotic > 90 days after surgery did not have any less severe recurrence or alteration in cytokine production than placebo.40 E coli Nissle 1917 has been found in several studies to induce and maintain remission in patients with UC and is equally as effective as mesalamine therapy.41-44 Bifidobacteriafermented milk (BFM) containing Bifidobacterium breve, B bifidum, and Lactobacillus acidophilus is effective in inducing and maintaining remission of UC, preventing flares, and increasing the stool concentration of butyrate and short-chain fatty acids.45,46 BIO-THREE probiotic, containing 3 genera of “good” bacteria, was found to induce remission in 45% of patients with UC with mild to moderate distal colitis and increase the levels of Bifidobacteria in the stool and had no significant adverse events.47 Lactobacillus rhamnosus GG (LGG) did not improve active pouchitis symptoms or inflammation in patients with UC despite a proven change in the microbiome.48 LGG has also been studied in active and postoperative CD with negative results.49-51 Another Lactobacillus strain, johnsonii, has been studied in the prevention of postoperative recurrence in CD without a difference clinically or endoscopically compared with placebo.52,53 A small study found that adding Saccharomyces boulardii to standard medical therapy was helpful in maintaining remission in CD.54 However, a recent meta-analysis showed that there was no benefit of Saccharomyces for the treatment of CD.55 A pilot study in patients with active UC found positive results using Saccharomyces boulardii in combination with mesalamine therapy.56 Two meta-analyses found some benefit in probiotics in inducing remission in UC, especially when combined with conventional therapy.57,58 The meta-analyses also demonstrated a benefit in maintaining remission in UC.57,58 Several Cochrane reviews and meta-analyses in patients with CD did

not find a significant benefit of probiotics in decreasing relapse rates,38 preventing postoperative recurrences,36 or maintaining remission.37,39 The small size of the studies, the heterogeneous methodology, and the varied measured outcomes limit these studies. Larger trials are needed to better evaluate probiotics in the treatment of IBD.

Synbiotics Synbiotics are combinations of prebiotics and probiotics that are felt to exert a synergistic effect on the gastrointestinal tract. Two studies of different synbiotics in active CD found promising results with improved clinical symptoms, a decrease in tumor necrosis factor (TNF)-α, and an increase in fecal Bifidobacteria.59,60 In a trial of 30 patients with CD treated with Synbiotic 2000 (combination of 4 probiotics and 4 prebiotics), there was no effect on postoperative recurrence.61 Bifidobacterium longum/Synergy 1 was given to 18 patients with active UC for 1 month and they were found to have improvement in endoscopic inflammation and significantly decreased levels of inflammatory cytokines including TNF-α and interleukin (IL) 1-α. A study of 41 patients with mild to moderate UC randomized to synbiotics (Bifidobacterium breve strain and galactooligosaccharide) or placebo showed more improvement in endoscopic score and lower levels of the myeloperoxidase inflammatory marker.62 A larger study of 120 patients with UC randomized to probiotic, prebiotic, or synbiotic therapy similarly found synbiotics to have greater improvements in symptom scores and C-reactive protein levels.63

Fish Oil Essential fatty acids (EFAs) consist of omega-3 and omega-6 polyunsaturated fatty acids (PUFAs). EFAs are not endogenously produced and can be acquired only through dietary intake. Omega-3 EFAs have been shown to have anti-inflammatory properties, whereas omega-6 EFAs are believed to have proinflammatory effects. Fish oil is a valuable source of omega-3 and has been shown to affect the gut immune system by suppressing T cell signaling, inhibiting proinflammatory cytokine synthesis, reducing inflammatory cell recruitment, decreasing the key proinflammatory transcription factor NF-kB, and enhancing epithelial barrier function. There have been 6 individual studies showing benefit from fish oil for the treatment of chemical-induced colitis in animal models.64-69

Induction There are no randomized trials that specifically investigated the use of oral n-3 PUFA for induction of remission in active CD. Several small initial studies showed positive results in the treatment of active UC with fish oil,70-73 including 3

Downloaded from ncp.sagepub.com at DALHOUSIE UNIV on June 19, 2015

Parian et al

3

randomized, placebo-controlled studies showing significant results.70,71,74 One of the randomized, placebo-controlled trials with 18 patients found an improvement in clinical, endoscopic, and histologic scores.74 A larger randomized trial of 121 patients with active UC compared a nutritionally balanced oral supplement enriched with fish oil fructo-oligosaccharides, gum arabic, vitamin E, vitamin C, and selenium with a carbohydrate-based placebo formula for 6 months. No difference in clinical, endoscopic, and histologic scores was seen, but there was a more rapid reduction in prednisone dose in those who received the nutrition supplement.75 A small crossover trial that compared fish oil 5.4 grams per day with sulfasalazine 2 grams per day in 10 patients with mild to moderate active UC found significantly increased inflammatory markers (ie, C-reactive protein, erythrocyte sedimentation rate, and platelets) in the fish oil arm.76 Despite lower sigmoidoscopy scores, there was no difference in histologic scores. The study investigators therefore concluded that sulfasalazine was superior to fish oil in the treatment of mild to moderate active UC.

Maintenance The Epanova Program in Crohn’s Study 1 (EPIC-1) and 2 (EPIC-2) were 2 randomized controlled trials that recruited patients with quiescent CD from 98 centers in the United States, Canada, Europe, and Israel.77 The participants were randomized to commercial omega-3 fatty acid capsules (50%60% EPA, 15%-25% docosahexaenoic acid) or placebo. Relapse rates were similar in both treatment arms. A recent meta-analysis by the Cochrane Collaboration evaluated the efficacy of n-3 PUFA for the maintenance of remission in CD.78 The pooled analysis included 6 studies with 1039 participants and showed a marginal benefit of n-3 PUFA compared with placebo (relative risk [RR] = 0.77; 95% confidence interval [CI], 0.61-0.98). However, there was unexplained heterogeneity (I2 = 58%) and 4 studies were graded as having high or unknown risk of bias.79-82 The remaining 2 studies considered to have no significant heterogeneity and low risk of bias were the EPIC-1 and EPIC-2 studies. The protective effect of n-3 PUFA was no longer statistical significant (RR = 0.88; 95% CI, 0.74-1.05) with these 2 combined studies. The efficacy of fish oil seems to be improved when combined with mesalamine therapy80 and when used for Crohn’s colitis rather than small bowel enteritis.79 A systematic review is difficult to perform on this topic due to variation in fish oil formulations, differences in disease location, and severity. Fish oil may have its greatest effects in patients with IBD with proven essential fatty acid deficiency, and once levels are replete, there may be no further benefit.83 For the maintenance of remission in UC, the 3 randomized trials that compared fish oil with placebo in 138 patients did not detect a difference in 1- to 2-year relapse rate.84-86

Curcumin Curcumin is a naturally occurring substance from the plant Curcuma longa and has been used successfully in Ayurvedic medicine for hundreds of years. Curcumin has natural antiinflammatory properties acting on the arachidonic acid cascade and inhibiting NF-kB as well as immunosuppressive effects by inhibiting IL-2 synthesis and TNF-α.87,88 Multiple animal studies found that curcumin was able to treat as well as prevent chemically induced colitis.89-91 In a human pilot study, 10 patients (5 CD, 5 UC) were treated with curcumin and 90% improved symptomatically and serologically. Four of the 5 patients with UC were able to decrease or stop concurrent colitis medication.92 A larger, randomized, double-blind, placebocontrolled trial of 89 patients with quiescent UC on baseline mesalamine therapy found that the addition of curcumin 1 gram orally twice daily resulted in symptomatic and endoscopic improvement as well as a significantly lower relapse rate.93 Curcumin has been shown to have an excellent safety profile and is well tolerated.94,95 Due to its chemopreventive activity against colon cancer,96 curcumin may be an ideal adjunctive therapy for colitis patients due to their increased risk of colorectal cancer. A Cochrane review was performed to evaluate the efficacy and safety of curcumin for maintenance of remission in patients with UC.97 Only 1 study by Hanai et al,93 mentioned above, met the inclusion criteria, and the Cochrane analysis concluded that curcumin may be a safe and effective adjunctive therapy when coupled with mesalamine for maintenance of remission, but further studies are needed.

Boswellia Boswellia serrata is another frequently used Ayurvedic herb to treat inflammatory conditions by also acting on the arachidonic acid cascade and inhibiting leukotrienes, leading to a decrease in proinflammatory cytokines.98 A study by Gupta et al99 found that B serrata (350 mg 3 times a day) was as effective as sulfasalazine therapy in reducing symptoms in patients with UC. A randomized, double-blind controlled trial by Gerhardt et al100 showed a Boswellia extract (H15) to be equivalent to mesalamine in the treatment of CD. A double-blind randomized controlled trial of 108 patients confirmed the safety and patient tolerability of B serrata.101

Other Herbal Therapies Aloe vera is well-known in traditional medicine to promote gut epithelial healing and has been shown to be an antioxidant with possible anti-inflammatory properties and found to decrease reactive oxygen metabolite production in a dose-dependent manner in in vitro studies.102 The studies on aloe vera are limited to mostly case reports showing improved colitis with oral therapy.102-109 One placebo-controlled trial in patients with UC

Downloaded from ncp.sagepub.com at DALHOUSIE UNIV on June 19, 2015

4

Nutrition in Clinical Practice XX(X)

did find aloe vera efficacious in achieving clinical and microscopic improvement and with minimal side effects.108 Wheat grass juice (triticum aestivum) is also believed to have antioxidant properties110 and has been used for the treatment of thalassemias, intestinal diseases, and IBD. Pigenin, the major component of wheat grass, blocks production of proinflammatory cytokines IL-1β, IL-8, and TNF through inactivation of NF-κB.111 A randomized, placebo-controlled trial in patients with active UC found that 100 mL of wheat grass juice was associated with decreased disease activity and rectal bleeding.112 An herbal treatment consisting of myrrh, dry extract of chamomile flowers, and coffee charcoal has been shown to have anti-inflammatory and antidiarrheal properties. A study of this herbal preparation found it noninferior to low-dose mesalamine treatment in the maintenance of remission of UC.113

Vitamin D Vitamin D regulates cell growth and is an important cofactor for the immune system. Deficiencies in vitamin D have been linked to several inflammatory conditions.114-116 Along with calcium, vitamin D is essential for bone health, which is prudent in patients with IBD who have many risk factors for poor bone health, including chronic inflammation and steroid use. Vitamin D supplementation has been shown to restore immunoreactivity and decrease monocyte production of IL-6 and TNF-α.117-120 Endogenous production of vitamin D in the skin during exposure to UVB sun rays is the major source. Fatty fishes, cod liver oil, beef liver, egg yolks, and fortified milk contain high levels of vitamin D.121 Levels of vitamin D < 20 ng/mL are considered deficient, whereas levels between 20 and 30 ng/mL are considered insufficient.121 Patients with IBD are frequently deficient in vitamin D due to sun avoidance in those on thiopurines, small bowel inflammation affecting absorption, ileal resection affecting bile salt resorption, increased excretion in the stool, and higher requirements.121-125 Vitamin D deficiency may be a risk factor for the development of IBD supported by higher rates of IBD in northern latitudes and an increased number of flares during winter (low sunlight) months.126-128 In the Nurses’ Health Study, a lower risk of CD was seen in participants with higher vitamin D levels. In addition, those taking vitamin D supplements had a lower risk of the development of UC.129 Ananthakrishnan et al130 found in a prospective study that patients with CD who normalized their vitamin D level through supplementation had a lower rate of hospitalization and surgery. Smaller studies in patients with CD taking vitamin D supplementations have shown a trend toward a decrease in relapse rates and a lower disease activity score.131,132

Conclusion Several nutraceuticals show promising results for the treatment of inflammatory bowel disease with minimal to no side effects

Table 1.  Therapeutic Modalities in the Treatment of Inflammatory Bowel Diseases. Therapy Probiotics Fish oil Curcumin Boswellia Butyrate enemas Aminosalicylates Corticosteroids Azathioprine Infliximab

Level of Evidencea

Level of Risk

A B B B B A A B A

Low Low NA Low NA Moderate High High High

NA, not available. a Level of evidence is based on Strength of Recommendation Taxonomy (SORT). Grade A is based on consistent, good-quality, patient-oriented evidence such as systematic reviews or meta-analyses, or high-quality patient-oriented randomized controlled trial. Grade B is based on inconsistent or limited-quality patient-oriented evidence. Grade C is based on consensus, usual practice, opinion, or disease-oriented evidence.

(Table 1). Prebiotics and probiotics appear to alter the gut microbiome by increasing biodiversity and increasing the production of butyrate, an anti-inflammatory and antineoplastic agent of the gut. Fish oil acts as a direct anti-inflammatory agent as well as an immune regulator. Curcumin and Boswellia both are anti-inflammatory agents that act on a variety of receptors, and vitamin D affects cell growth and immune system regulation. As per President Obama, we are at the dawn of the age of personalized medicine and the U.S. federal government will be further investing to bring this to fruition. Thus, in the future we may be able to predict response to these natural therapies based on gut microbiome analyses, serological markers, and more. Continued trials are needed to determine the subset of patients with IBD who would respond best to each nutraceutical and how to best combine natural medicines with conventional pharmaceuticals.

References 1. Tragnone A, Valpiani D, Miglio F, et al. Dietary habits as risk factors for inflammatory bowel disease. Eur J Gastroenterol Hepatol. 1995;7(1):4751. 2. Persson PG, Ahlbom A, Hellers G. Diet and inflammatory bowel disease: a case-control study. Epidemiology. 1992;3(1):47-52. 3. Sakamoto N, Kono S, Wakai K, et al. Dietary risk factors for inflammatory bowel disease: a multicenter case-control study in Japan. Inflamm Bowel Dis. 2005;11(2):154-163. 4. Amre DK, D’Souza S, Morgan K, et al. Imbalances in dietary consumption of fatty acids, vegetables, and fruits are associated with risk for Crohn’s disease in children. Am J Gastroenterol. 2007;102(9):2016-2025. 5. Ananthakrishnan AN, Khalili H, Konijeti GG, et al. A prospective study of long-term intake of dietary fiber and risk of Crohn’s disease and ulcerative colitis. Gastroenterology. 2013;145(5):970-977. 6. Porter CK, Tribble DR, Aliaga PA, Halvorson HA, Riddle MS. Infectious gastroenteritis and risk of developing inflammatory bowel disease. Gastroenterology. 2008;135(3):781-786. 7. Gradel KO, Nielsen HL, Schønheyder HC, Ejlertsen T, Kristensen B, Nielsen H. Increased short- and long-term risk of inflammatory bowel disease

Downloaded from ncp.sagepub.com at DALHOUSIE UNIV on June 19, 2015

Parian et al

5

after salmonella or campylobacter gastroenteritis. Gastroenterology. 2009; 137(2):495-501. 8. Hollander D. Crohn’s disease—a permeability disorder of the tight junction? Gut. 1988;29(12):1621-1624. 9. Roberfroid M. Prebiotics: the concept revisited. J Nutr. 2007;137(3, suppl 2):830S-837S. 10. Wollowski I, Rechkemmer G, Pool-Zobel BL. Protective role of probiotics and prebiotics in colon cancer. Am J Clin Nutr. 2001;73(2, suppl): 451S-455S. 11. Butzner JD, Parmar R, Bell CJ, Dalal V. Butyrate enema therapy stimulates mucosal repair in experimental colitis in the rat. Gut. 1996;38(4): 568-573. 12. Scheppach W, Bartram P, Richter A, et al. Effect of short-chain fatty acids on the human colonic mucosa in vitro. JPEN J Parenter Enteral Nutr. 1992;16(1):43-48. 13. Segain JP, Raingeard de la Blétière D, Bourreille A, et al. Butyrate inhibits inflammatory responses through NFkappaB inhibition: implications for Crohn’s disease. Gut. 2000;47(3):397-403. 14. Menzel T, Lührs H, Zirlik S, et al. Butyrate inhibits leukocyte adhesion to endothelial cells via modulation of VCAM-1. Inflamm Bowel Dis. 2004;10(2):122-128. 15. Casellas F, Borruel N, Torrejón 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(9):1061-1067. 16. Kanauchi O, Mitsuyama K, Homma T, et al. Treatment of ulcerative colitis patients by long-term administration of germinated barley foodstuff: multi-center open trial. Int J Mol Med. 2003;12(5):701-704. 17. Hanai H, Kanauchi O, Mitsuyama K, et al. Germinated barley foodstuff prolongs remission in patients with ulcerative colitis. Int J Mol Med. 2004;13(5):643-647. 18. Fernández-Bañares F, Hinojosa J, Sánchez-Lombraña JL, et al. Randomized clinical trial of Plantago ovata seeds (dietary fiber) as compared with mesalamine in maintaining remission in ulcerative colitis. Spanish Group for the Study of Crohn’s Disease and Ulcerative Colitis (GETECCU). Am J Gastroenterol. 1999;94(2):427-433. 19. Hallert C, Kaldma M, Petersson BG. Ispaghula husk may relieve gastrointestinal symptoms in ulcerative colitis in remission. Scand J Gastroenterol. 1991;26(7):747-750. 20. Hill C, Guarner F, Reid G, et al. Expert consensus document. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat Rev Gastroenterol Hepatol. 2014;11(8):506-514. 21. Garcia Vilela E, De Lourdes De Abreu Ferrari M, Oswaldo Da Gama Torres H, et al. Influence of Saccharomyces boulardii on the intestinal permeability of patients with Crohn’s disease in remission. Scand J Gastroenterol. 2008;43(7):842-848. 22. Stephani J, Radulovic K, Niess JH. Gut microbiota, probiotics and inflammatory bowel disease. Arch Immunol Ther Exp (Warsz). 2011;59(3): 161-177. 23. Prisciandaro L, Geier M, Butler R, Cummins A, Howarth G. Probiotics and their derivatives as treatments for inflammatory bowel disease. Inflamm Bowel Dis. 2009;15(12):1906-1914. 24. O’Mahony C, Scully P, O’Mahony D, et al. Commensal-induced regulatory T cells mediate protection against pathogen-stimulated NF-kappaB activation. PLoS Pathog. 2008;4(8):e1000112. 25. Matsumoto S, Watanabe N, Imaoka A, Okabe Y. Preventive effects of Bifidobacterium- and Lactobacillus-fermented milk on the development of inflammatory bowel disease in senescence-accelerated mouse P1/Yit strain mice. Digestion. 2001;64(2):92-99. 26. Gionchetti P, Rizzello F, Venturi A, et al. Oral bacteriotherapy as maintenance treatment in patients with chronic pouchitis: a double-blind, placebo-controlled trial. Gastroenterology. 2000;119(2): 305-309.

27. Gionchetti P, Rizzello F, Helwig U, et al. Prophylaxis of pouchitis onset with probiotic therapy: a double-blind, placebo-controlled trial. Gastroenterology. 2003;124(5):1202-1209. 28. Pardi DS, D’Haens G, Shen B, Campbell S, Gionchetti P. Clinical guidelines for the management of pouchitis. Inflamm Bowel Dis. 2009;15(9):1424-1431. 29. Bibiloni R, Fedorak RN, Tannock GW, et al. VSL#3 probiotic-mixture induces remission in patients with active ulcerative colitis. Am J Gastroenterol. 2005;100(7):1539-1546. 30. Huynh HQ, deBruyn J, Guan L, et al. Probiotic preparation VSL#3 induces remission in children with mild to moderate acute ulcerative colitis: a pilot study. Inflamm Bowel Dis. 2009;15(5):760-768. 31. Miele E, Pascarella F, Giannetti E, Quaglietta L, Baldassano RN, Staiano A. Effect of a probiotic preparation (VSL#3) on induction and maintenance of remission in children with ulcerative colitis. Am J Gastroenterol. 2009;104(2):437-443. 32. Sood A, Midha V, Makharia GK, et al. The probiotic preparation, VSL#3 induces remission in patients with mild-to-moderately active ulcerative colitis. Clin Gastroenterol Hepatol. 2009;7(11):1202-1209. 33. Tursi A, Brandimarte G, Giorgetti GM, Forti G, Modeo ME, Gigliobianco A. Low-dose balsalazide plus a high-potency probiotic preparation is more effective than balsalazide alone or mesalazine in the treatment of acute mild-to-moderate ulcerative colitis. Med Sci Monit. 2004;10(11): PI126- PI131. 34. Tursi A, Brandimarte G, Papa A, et al. Treatment of relapsing mildto-moderate ulcerative colitis with the probiotic VSL#3 as adjunctive to a standard pharmaceutical treatment: a double-blind, randomized, placebo-controlled study. Am J Gastroenterol. 2010;105(10): 2218-2227. 35. Jonkers D, Penders J, Masclee A, Pierik M. Probiotics in the management of inflammatory bowel disease: a systematic review of intervention studies in adult patients. Drugs. 2012;72(6):803-823. 36. Doherty G, Bennett G, Patil S, Cheifetz A, Moss AC. Interventions for prevention of post-operative recurrence of Crohn’s disease. Cochrane Database Syst Rev. 2009;(4):CD006873. 37. Rahimi R, Nikfar S, Rahimi F, et al. A meta-analysis on the efficacy of probiotics for maintenance of remission and prevention of clinical and endoscopic relapse in Crohn’s disease. Dig Dis Sci. 2008;53(9):25242531. 38. Rolfe VE, Fortun PJ, Hawkey CJ, Bath-Hextall F. Probiotics for maintenance of remission in Crohn’s disease. Cochrane Database Syst Rev. 2006;(4):CD004826. 39. Shen J, Ran HZ, Yin MH, Zhou TX, Xiao DS. Meta-analysis: the effect and adverse events of Lactobacilli versus placebo in maintenance therapy for Crohn disease. Intern Med J. 2009;39(2):103-109. 40. Fedorak RN, Feagan BG, Hotte N, et al. The probiotic VSL#3 has antiinflammatory effects and could reduce endoscopic recurrence after surgery for Crohn’s disease. Clin Gastroenterol Hepatol. 2015;13(5): 928-935. 41. Rembacken BJ, Snelling AM, Hawkey PM, Chalmers DM, Axon AT. Non-pathogenic Escherichia coli versus mesalazine for the treatment of ulcerative colitis: a randomised trial. Lancet. 1999;354(9179): 635-639. 42. Kruis W, Schütz E, Fric P, Fixa B, Judmaier G, Stolte M. Double-blind comparison of an oral Escherichia coli preparation and mesalazine in maintaining remission of ulcerative colitis. Aliment Pharmacol Ther. 1997;11(5):853-858. 43. Kruis W, Fric P, Pokrotnieks J, et al. Maintaining remission of ulcerative colitis with the probiotic Escherichia coli Nissle 1917 is as effective as with standard mesalazine. Gut. 2004;53(11):1617-1623. 44. Matthes H, Krummenerl T, Giensch M, Wolff C, Schulze J. Clinical trial: probiotic treatment of acute distal ulcerative colitis with rectally administered Escherichia coli Nissle 1917 (EcN). BMC Complement Altern Med. 2010;10:13.

Downloaded from ncp.sagepub.com at DALHOUSIE UNIV on June 19, 2015

6

Nutrition in Clinical Practice XX(X)

45. Kato K, Mizuno S, Umesaki Y, et al. Randomized placebo-controlled trial assessing the effect of bifidobacteria-fermented milk on active ulcerative colitis. Aliment Pharmacol Ther. 2004;20(10):1133-1141. 46. Ishikawa H, Akedo I, Umesaki Y, Tanaka R, Imaoka A, Otani T. Randomized controlled trial of the effect of bifidobacteria-fermented milk on ulcerative colitis. J Am Coll Nutr. 2003;22(1):56-63. 47. Tsuda Y, Yoshimatsu Y, Aoki H, et al. Clinical effectiveness of probiotics therapy (BIO-THREE) in patients with ulcerative colitis refractory to conventional therapy. Scand J Gastroenterol. 2007;42(11):1306-1311. 48. Kuisma J, Mentula S, Jarvinen H, Kahri A, Saxelin M, Farkkila M. Effect of Lactobacillus rhamnosus GG on ileal pouch inflammation and microbial flora. Aliment Pharmacol Ther. 2003;17(4):509-515. 49. Bousvaros A, Guandalini S, Baldassano RN, et al. A randomized, double-blind trial of Lactobacillus GG versus placebo in addition to standard maintenance therapy for children with Crohn’s disease. Inflamm Bowel Dis. 2005;11(9):833-839. 50. Prantera C, Scribano ML, Falasco G, Andreoli A, Luzi C. Ineffectiveness of probiotics in preventing recurrence after curative resection for Crohn’s disease: a randomised controlled trial with Lactobacillus GG. Gut. 2002;51(3):405-409. 51. Schultz M, Timmer A, Herfarth HH, Sartor RB, Vanderhoof JA, Rath HC. Lactobacillus GG in inducing and maintaining remission of Crohn’s disease. BMC Gastroenterol. 2004;4:5. 52. Van Gossum A, Dewit O, Louis E, et al. Multicenter randomized-controlled clinical trial of probiotics (Lactobacillus johnsonii, LA1) on early endoscopic recurrence of Crohn’s disease after ileo-caecal resection. Inflamm Bowel Dis. 2007;13(2):135-142. 53. Marteau P, Lémann M, Seksik P, et al. Ineffectiveness of Lactobacillus johnsonii LA1 for prophylaxis of postoperative recurrence in Crohn’s disease: a randomised, double blind, placebo controlled GETAID trial. Gut. 2006;55(6):842-847. 54. Guslandi M, Mezzi G, Sorghi M, Testoni PA. Saccharomyces boulardii in maintenance treatment of Crohn’s disease. Dig Dis Sci. 2000;45(7): 1462-1464. 55. McFarland LV. Systematic review and meta-analysis of Saccharomyces boulardii in adult patients. World J Gastroenterol. 2010;16(18):22022222. 56. Guslandi M, Giollo P, Testoni PA. A pilot trial of Saccharomyces boulardii in ulcerative colitis. Eur J Gastroenterol Hepatol. 2003;15(6): 697-698. 57. Sang LX, Chang B, Zhang WL, Wu XM, Li XH, Jiang M. Remission induction and maintenance effect of probiotics on ulcerative colitis: a meta-analysis. World J Gastroenterol. 2010;16(15):1908-1915. 58. Zigra PI, Maipa VE, Alamanos YP. Probiotics and remission of ulcerative colitis: a systematic review. Neth J Med. 2007;65(11):411-418. 59. Fujimori S, Tatsuguchi A, Gudis K, et al. High dose probiotic and prebiotic cotherapy for remission induction of active Crohn’s disease. J Gastroenterol Hepatol. 2007;22(8):1199-1204. 60. Steed H, Macfarlane GT, Blackett KL, et al. Clinical trial: the microbiological and immunological effects of synbiotic consumption—a randomized double-blind placebo-controlled study in active Crohn’s disease. Aliment Pharmacol Ther. 2010;32(7):872-883. 61. Chermesh I, Tamir A, Reshef R, et al. Failure of Synbiotic 2000 to prevent postoperative recurrence of Crohn’s disease. Dig Dis Sci. 2007;52(2):385-389. 62. Ishikawa H, Matsumoto S, Ohashi Y, et al. Beneficial effects of probiotic bifidobacterium and galacto-oligosaccharide in patients with ulcerative colitis: a randomized controlled study. Digestion. 2011;84(2):128-133. 63. Fujimori S, Gudis K, Mitsui K, et al. A randomized controlled trial on the efficacy of synbiotic versus probiotic or prebiotic treatment to improve the quality of life in patients with ulcerative colitis. Nutrition. 2009;25(5): 520-525. 64. Whiting CV, Bland PW, Tarlton JF. Dietary n-3 polyunsaturated fatty acids reduce disease and colonic proinflammatory cytokines in a mouse model of colitis. Inflamm Bowel Dis. 2005;11(4):340-349.

65. Camuesco D, Gálvez J, Nieto A, et al. Dietary olive oil supplemented with fish oil, rich in EPA and DHA (n-3) polyunsaturated fatty acids, attenuates colonic inflammation in rats with DSS-induced colitis. J Nutr. 2005;135(4):687-694. 66. Hegazi RA, Saad RS, Mady H, Matarese LE, O’Keefe S, Kandil HM. Dietary fatty acids modulate chronic colitis, colitis-associated colon neoplasia and COX-2 expression in IL-10 knockout mice. Nutrition. 2006;22(3):275-282. 67. Arita M, Yoshida M, Hong S, et al. Resolvin E1, an endogenous lipid mediator derived from omega-3 eicosapentaenoic acid, protects against 2,4,6-trinitrobenzene sulfonic acid-induced colitis. Proc Natl Acad Sci U S A. 2005;102(21):7671-7676. 68. Hudert CA, Weylandt KH, Lu Y, et al. Transgenic mice rich in endogenous omega-3 fatty acids are protected from colitis. Proc Natl Acad Sci U S A. 2006;103(30):11276-11281. 69. Meister D, Ghosh S. Effect of fish oil enriched enteral diet on inflammatory bowel disease tissues in organ culture: differential effects on ulcerative colitis and Crohn’s disease. World J Gastroenterol. 2005;11(47): 7466-7472. 70. Aslan A, Triadafilopoulos G. Fish oil fatty acid supplementation in active ulcerative colitis: a double-blind, placebo-controlled, crossover study. Am J Gastroenterol. 1992;87(4):432-437. 71. Stenson WF, Cort D, Rodgers J, et al. Dietary supplementation with fish oil in ulcerative colitis. Ann Intern Med. 1992;116(8):609-614. 72. Barbosa DS, Cecchini R, El Kadri MZ, Rodríguez MA, Burini RC, Dichi I. Decreased oxidative stress in patients with ulcerative colitis supplemented with fish oil omega-3 fatty acids. Nutrition. 2003;19(10):837-842. 73. Lorenz R, Weber PC, Szimnau P, Heldwein W, Strasser T, Loeschke K. Supplementation with n-3 fatty acids from fish oil in chronic inflammatory bowel disease—a randomized, placebo-controlled, double-blind cross-over trial. J Intern Med Suppl. 1989;731:225-232. 74. Almallah YZ, Richardson S, O’Hanrahan T, et al. Distal procto-colitis, natural cytotoxicity, and essential fatty acids. Am J Gastroenterol. 1998;93(5):804-809. 75. Seidner DL, Lashner BA, Brzezinski A, et al. An oral supplement enriched with fish oil, soluble fiber, and antioxidants for corticosteroid sparing in ulcerative colitis: a randomized, controlled trial. Clin Gastroenterol Hepatol. 2005;3(4):358-369. 76. Dichi I, Frenhane P, Dichi JB, et al. Comparison of omega-3 fatty acids and sulfasalazine in ulcerative colitis. Nutrition. 2000;16(2):87-90. 77. Feagan BG, Sandborn WJ, Mittmann U, et al. Omega-3 free fatty acids for the maintenance of remission in Crohn disease: the EPIC Randomized Controlled Trials. JAMA. 2008;299(14):1690-1697. 78. Lev-Tzion R, Griffiths AM, Leder O, Turner D. Omega 3 fatty acids (fish oil) for maintenance of remission in Crohn’s disease. Cochrane Database Syst Rev. 2014;2:CD006320. 79. Lorenz-Meyer H, Bauer P, Nicolay C, et al. Omega-3 fatty acids and low carbohydrate diet for maintenance of remission in Crohn’s disease. A randomized controlled multicenter trial. Study Group Members (German Crohn’s Disease Study Group). Scand J Gastroenterol. 1996;31(8): 778-785. 80. Romano C, Cucchiara S, Barabino A, Annese V, Sferlazzas C. Usefulness of omega-3 fatty acid supplementation in addition to mesalazine in maintaining remission in pediatric Crohn’s disease: a doubleblind, randomized, placebo-controlled study. World J Gastroenterol. 2005;11(45):7118-7121. 81. Belluzzi A, Brignola C, Campieri M, Pera A, Boschi S, Miglioli M. Effect of an enteric-coated fish-oil preparation on relapses in Crohn’s disease. N Engl J Med. 1996;334(24):1557-1560. 82. Belluzzi A, Brignola C, Campieri M, et al. A new enteric coated preparation of omega-3 fatty acids for preventing post-surgical recurrence in Crohn’s disease. Gastroenterology. 1997;112(4):A494. 83. Siguel EN, Lerman RH. Prevalence of essential fatty acid deficiency in patients with chronic gastrointestinal disorders. Metabolism. 1996;45(1):12-23.

Downloaded from ncp.sagepub.com at DALHOUSIE UNIV on June 19, 2015

Parian et al

7

84. Hawthorne AB, Daneshmend TK, Hawkey CJ, et al. Treatment of ulcerative colitis with fish oil supplementation: a prospective 12 month randomised controlled trial. Gut. 1992;33(7):922-928. 85. Loeschke K, Ueberschaer B, Pietsch A, et al. n-3 fatty acids only delay early relapse of ulcerative colitis in remission. Dig Dis Sci. 1996;41(10): 2087-2094. 86. Mantzaris GJ, Archavlis E, Zografos C, Petraki K, Spiliades C, Triantafyllou G. A prospective, randomized, placebo-controlled study of fish oil in ulcerative colitis. Hellenic J Gastroenterol. 1996;9(2): 138-141. 87. Jobin C, Bradham CA, Russo MP, et al. Curcumin blocks cytokinemediated NF-kappa B activation and proinflammatory gene expression by inhibiting inhibitory factor I-kappa B kinase activity. J Immunol. 1999;163(6):3474-3483. 88. Duvoix A, Blasius R, Delhalle S, et al. Chemopreventive and therapeutic effects of curcumin. Cancer Lett. 2005;223(2):181-190. 89. Jian YT, Mai GF, Wang JD, Zhang YL, Luo RC, Fang YX. Preventive and therapeutic effects of NF-kappaB inhibitor curcumin in rats colitis induced by trinitrobenzene sulfonic acid. World J Gastroenterol. 2005;11(12):1747-1752. 90. Deguchi Y, Andoh A, Inatomi O, et al. Curcumin prevents the development of dextran sulfate sodium (DSS)-induced experimental colitis. Dig Dis Sci. 2007;52(11):2993-2998. 91. Sugimoto K, Hanai H, Tozawa K, et al. Curcumin prevents and ameliorates trinitrobenzene sulfonic acid-induced colitis in mice. Gastroenterology. 2002;123(6):1912-1922. 92. Holt PR, Katz S, Kirshoff R. Curcumin therapy in inflammatory bowel disease: a pilot study. Dig Dis Sci. 2005;50(11):2191-2193. 93. Hanai H, Iida T, Takeuchi K, et al. Curcumin maintenance therapy for ulcerative colitis: randomized, multicenter, double-blind, placebo-controlled trial. Clin Gastroenterol Hepatol. 2006;4(12):1502-1506. 94. Bengmark S. Curcumin, an atoxic antioxidant and natural NFkappaB, cyclooxygenase-2, lipooxygenase, and inducible nitric oxide synthase inhibitor: a shield against acute and chronic diseases. JPEN J Parenter Enteral Nutr. 2006;30(1):45-51. 95. Cheng AL, Hsu CH, Lin JK, et al. Phase I clinical trial of curcumin, a chemopreventive agent, in patients with high-risk or pre-malignant lesions. Anticancer Res. 2001;21(4B):2895-2900. 96. Perkins S, Verschoyle RD, Hill K, et al. Chemopreventive efficacy and pharmacokinetics of curcumin in the min/+ mouse, a model of familial adenomatous polyposis. Cancer Epidemiol Biomarkers Prev. 2002;11(6):535-540. 97. Kumar S, Ahuja V, Sankar MJ, Kumar A, Moss AC. Curcumin for maintenance of remission in ulcerative colitis. Cochrane Database Syst Rev. 2012;10:CD008424. 98. Gayathri B, Manjula N, Vinaykumar KS, Lakshmi BS, Balakrishnan A. Pure compound from Boswellia serrata extract exhibits anti-inflammatory property in human PBMCs and mouse macrophages through inhibition of TNFalpha, IL-1beta, NO and MAP kinases. Int Immunopharmacol. 2007;7(4):473-482. 99. Gupta I, Parihar A, Malhotra P, et al. Effects of gum resin of Boswellia serrata in patients with chronic colitis. Planta Med. 2001;67(5): 391-395. 100. Gerhardt H, Seifert F, Buvari P, Vogelsang H, Repges R. Therapy of active Crohn disease with Boswellia serrata extract H 15 [in German]. Z Gastroenterol. 2001;39(1):11-17. 101. Holtmeier W, Zeuzem S, Preiss J, et al. Randomized, placebo-controlled, double-blind trial of Boswellia serrata in maintaining remission of Crohn’s disease: good safety profile but lack of efficacy. Inflamm Bowel Dis. 2011;17(2):573-582. 102. Langmead L, Makins RJ, Rampton DS. Anti-inflammatory effects of aloe vera gel in human colorectal mucosa in vitro. Aliment Pharmacol Ther. 2004;19(5):521-527. 103. Aloe vera helps ulcerative colitis. Health News. 2004;10(6):2.

104. Carty E, Ballinger A, Azooz O, et al. Does topical aloe vera reduce the severity of trinitrobenzene sulphonic acid (TNBS) colitis in rats? Gastroenterology. 1998;114(4):A948. 105. Korkina L, Suprun M, Petrova A, Mikhal’chik E, Luci A, De Luca C. The protective and healing effects of a natural antioxidant formulation based on ubiquinol and aloe vera against dextran sulfate-induced ulcerative colitis in rats. Biofactors. 2003;18(1-4):255-264. 106. Langmead L, Ballinger A, Carty E, et al. Aloe vera enemas ameliorate trinitrobenzene sulphonic acid (TNBS) colitis in rats. Gastroenterology. 1999;116(4):A802. 107. Langmead L, Feakins RM, Goldthorpe S, et al. Randomised, doubleblind, placebo-controlled trial of oral aloe vera gel for mild-moderately active ulcerative colitis. Gastroenterology. 2004;126(4):A463. 108. Langmead L, Feakins RM, Goldthorpe S, et al. Randomized, doubleblind, placebo-controlled trial of oral aloe vera gel for active ulcerative colitis. Aliment Pharmacol Ther. 2004;19(7):739-747. 109. Langmead L, Feakins RM, Goldthorpe S, et al. Randomised, double blind, placebo controlled trial of oral aloe vera gel for mild-moderately active ulcerative colitis. Gut. 2004;53:A30. 110. Kulkarni SD, Tilak JC, Acharya R, Rajurkar NS, Devasagayam TP, Reddy AV. Evaluation of the antioxidant activity of wheatgrass (Triticum aestivum L.) as a function of growth under different conditions. Phytother Res. 2006;20(3):218-227. 111. Nicholas C, Batra S, Vargo MA, et al. Apigenin blocks lipopolysaccharide-induced lethality in vivo and proinflammatory cytokines expression by inactivating NF-kappaB through the suppression of p65 phosphorylation. J Immunol. 2007;179(10):7121-7127. 112. Ben-Arye E, Goldin E, Wengrower D, Stamper A, Kohn R, Berry E. Wheat grass juice in the treatment of active distal ulcerative colitis: a randomized double-blind placebo-controlled trial. Scand J Gastroenterol. 2002;37(4):444-449. 113. Langhorst J, Varnhagen I, Schneider SB, et al. Randomised clinical trial: a herbal preparation of myrrh, chamomile and coffee charcoal compared with mesalazine in maintaining remission in ulcerative colitis—a double-blind, double-dummy study. Aliment Pharmacol Ther. 2013;38(5): 490-500. 114. Lemire JM, Adams JS, Kermani-Arab V, Bakke AC, Sakai R, Jordan SC. 1,25-Dihydroxyvitamin D3 suppresses human T helper/inducer lymphocyte activity in vitro. J Immunol. 1985;134(5):3032-3035. 115. Cantorna MT, Mahon BD. D-hormone and the immune system. J Rheumatol Suppl. 2005;76:11-20. 116. Cantorna MT, Zhu Y, Froicu M, Wittke A. Vitamin D status, 1,25-dihydroxyvitamin D3, and the immune system. Am J Clin Nutr. 2004;80(6 suppl):1717S-1720S. 117. Toss G, Symreng T. Delayed hypersensitivity response and vitamin D deficiency. Int J Vitam Nutr Res. 1983;53(1):27-31. 118. Wu S, Sun J. Vitamin D, vitamin D receptor, and macroautophagy in inflammation and infection. Discov Med. 2011;11(59):325-335. 119. Yuk JM, Shin DM, Lee HM, et al. Vitamin D3 induces autophagy in human monocytes/macrophages via cathelicidin. Cell Host Microbe. 2009;6(3):231-243. 120. Zhang Y, Leung DY, Richers BN, et al. Vitamin D inhibits monocyte/ macrophage proinflammatory cytokine production by targeting MAPK phosphatase-1. J Immunol. 2012;188(5):2127-2135. 121. Holick MF. Vitamin D deficiency. N Engl J Med. 2007;357(3):266-281. 122. Leichtmann GA, Bengoa JM, Bolt MJ, Sitrin MD. Intestinal absorption of cholecalciferol and 25-hydroxycholecalciferol in patients with both Crohn’s disease and intestinal resection. Am J Clin Nutr. 1991;54(3):548552. 123. Rosen CJ. Clinical practice. Vitamin D insufficiency. N Engl J Med. 2011;364(3):248-254. 124. Sentongo TA, Semaeo EJ, Stettler N, Piccoli DA, Stallings VA, Zemel BS. Vitamin D status in children, adolescents, and young adults with Crohn disease. Am J Clin Nutr. 2002;76(5):1077-1081.

Downloaded from ncp.sagepub.com at DALHOUSIE UNIV on June 19, 2015

8

Nutrition in Clinical Practice XX(X)

125. Tajika M, Matsuura A, Nakamura T, et al. Risk factors for vitamin D deficiency in patients with Crohn’s disease. J Gastroenterol. 2004;39(6): 527-533. 126. Lim WC, Hanauer SB, Li YC. Mechanisms of disease: vitamin D and inflammatory bowel disease. Nat Clin Pract Gastroenterol Hepatol. 2005;2(7):308-315. 127. Harries AD, Brown R, Heatley RV, Williams LA, Woodhead S, Rhodes J. Vitamin D status in Crohn’s disease: association with nutrition and disease activity. Gut. 1985;26(11):1197-1203. 128. Vogelsang H, Ferenci P, Woloszczuk W, et al. Bone disease in vitamin D-deficient patients with Crohn’s disease. Dig Dis Sci. 1989;34(7): 1094-1099.

129. Ananthakrishnan AN, Khalili H, Higuchi LM, et al. Higher predicted vitamin D status is associated with reduced risk of Crohn’s disease. Gastroenterology. 2012;142(3):482-489. 130. Ananthakrishnan AN, Cagan A, Gainer VS, et al. Normalization of plasma 25-hydroxy vitamin D is associated with reduced risk of surgery in Crohn’s disease. Inflamm Bowel Dis. 2013;19(9):1921-1927. 131. Jørgensen SP, Agnholt J, Glerup H, et al. Clinical trial: vitamin D3 treatment in Crohn’s disease—a randomized double-blind placebo-controlled study. Aliment Pharmacol Ther. 2010;32(3):377-383. 132. Yang L, Weaver V, Smith JP, Bingaman S, Hartman TJ, Cantorna MT. Therapeutic effect of vitamin D supplementation in a pilot study of Crohn’s patients. Clin Transl Gastroenterol. 2013;4:e33.

Downloaded from ncp.sagepub.com at DALHOUSIE UNIV on June 19, 2015

Nutraceutical Supplements for Inflammatory Bowel Disease.

Nutraceutical Supplements for Inflammatory Bowel Disease. - PDF Download Free
321KB Sizes 0 Downloads 12 Views