702
Views
1
CrossRef citations to date
0
Altmetric
REVIEW

Personalized Dietary Regimens for Inflammatory Bowel Disease: Current Knowledge and Future Perspectives

ORCID Icon, , , &
Pages 15-27 | Received 23 Aug 2022, Accepted 06 Jan 2023, Published online: 12 Jan 2023

References

  • Molodecky NA, Soon IS, Rabi DM, et al. Increasing incidence and prevalence of the inflammatory bowel diseases with time, based on systematic review. Gastroenterology. 2012;142(1):46–54.e42. doi:10.1053/j.gastro.2011.10.001
  • Glade MJ, Meguid MM. A glance at. dietary emulsifiers, the human intestinal mucus and microbiome, and dietary fiber. Nutrition. 2016;32(5):609–614. doi:10.1016/j.nut.2015.12.036
  • Yang Y, Chung owyang GW, Wu GD. Summary of the Asia America Assembly of IBD (AAA IBD 2016): east meets west–the rising incidence of IBD in Asia as a paradigm for environmental effects on the pathogenesis of immune-mediated disease. Gastroenterology. 2016;151(6):e1–e5. doi:10.1016/j.physbeh.2017.03.040
  • Levine A, Sigall Boneh R, Wine E. Evolving role of diet in the pathogenesis and treatment of inflammatory bowel diseases. Gut. 2018;67(9):1726–1738. doi:10.1136/gutjnl-2017-315866
  • Hou JK, Abraham B, El-Serag H. Dietary intake and risk of developing inflammatory bowel disease: a systematic review of the literature. Am J Gastroenterol. 2011;106(4):563–573. doi:10.1038/ajg.2011.44
  • Wood JA, Halmos EP, Taylor KM, Gibson PR. The role of epidemiological evidence from prospective population studies in shaping dietary approaches to therapy in Crohn’s disease. Mol Nutr Food Res. 2021;65(5):2000294. doi:10.1002/mnfr.202000294
  • Monteiro CA, Moubarac JC, Cannon G, Ng SW, Popkin B. Ultra-processed products are becoming dominant in the global food system. Obes Rev. 2013;14(S2):21–28. doi:10.1111/obr.12107
  • Narula N, Wong ECL, Dehghan M, et al. Association of ultra-processed food intake with risk of inflammatory bowel disease: prospective cohort study. BMJ. 2021;374. doi:10.1136/bmj.n1554
  • Lo C-H, Khandpur N, Rossato SL, et al. Ultra-processed foods and risk of Crohn’s disease and ulcerative colitis: a prospective cohort study. Clin Gastroenterol Hepatol. 2021. doi:10.1016/j.cgh.2021.08.031
  • Halmos EP, Mack A, Gibson PR. Review article: emulsifiers in the food supply and implications for gastrointestinal disease. Aliment Pharmacol Ther. 2019;49(1):41–50. doi:10.1111/apt.15045
  • Dimitrijevic D, Shaw AJ, Florence A. Effects of some non-ionic surfactants on transepithelial permeability in Caco-2 cells. J Pharm Pharmacol. 2000;52(2):157–162. doi:10.1211/0022357001773805
  • Chassaing B, Koren O, Goodrich JK, et al. Dietary emulsifiers impact the mouse gut microbiota promoting colitis and metabolic syndrome. Nature. 2015;519(7541):92–96. doi:10.1038/nature14232
  • Watt J, Marcus R. Carrageenan-induced ulceration of the large intestine in the Guinea pig. Gut. 1971;12(2):164–171. doi:10.1136/gut.12.2.164
  • Bhattacharyya S, Dudeja PK, Tobacman JK. Tumor necrosis factor α-induced inflammation is increased but apoptosis is inhibited by common food additive carrageenan. J Biol Chem. 2010;285(50):39511–39522. doi:10.1074/jbc.M110.159681
  • Chassaing B, Compher C, Bonhomme B, et al. Randomized controlled-feeding study of dietary emulsifier carboxymethyl cellulose reveals detrimental impacts on the gut microbiota and metabolome. Gastroenterology. 2021. doi:10.1053/j.gastro.2021.11.006
  • Roberts CL, Keita ÅV, Duncan SH, et al. Translocation of Crohn’s disease Escherichia coli across M-cells: contrasting effects of soluble plant fibres and emulsifiers. Gut. 2010;59(10):1331–1339. doi:10.1136/gut.2009.195370
  • Wedlake L, Slack N, Andreyev HJN, Whelan K. Fiber in the treatment and maintenance of inflammatory bowel disease: a systematic review of randomized controlled trials. Inflamm Bowel Dis. 2014;20(3):576–586. doi:10.1097/01.MIB.0000437984.92565.31
  • Ordovas JM, Ferguson LR, Tai ES, Mathers JC. Personalised nutrition and health. BMJ. 2018;361(k2173):1–7. doi:10.1136/bmj.k2173
  • Celis-Morales C, Livingstone KM, Marsaux CFM, et al. Effect of personalized nutrition on health-related behaviour change: evidence from the Food4Me European randomized controlled trial. Int J Epidemiol. 2017;46(2):578–588. doi:10.1093/ije/dyw186
  • Ferguson LR, De Caterina R, Görman U, et al. Guide and position of the international society of nutrigenetics/nutrigenomics on personalised nutrition: part 1 - fields of precision nutrition. J Nutrigenet Nutrigenomics. 2016;9(1):12–27. doi:10.1159/000445350
  • Wyatt P, Berry SE, Finlayson G, et al. Postprandial glycaemic dips predict appetite and energy intake in healthy individuals. Nat Metab. 2021;3(4):523–529. doi:10.1038/s42255-021-00383-x
  • Moore SE, McMullan M, McEvoy CT, McKinley MC, Woodside JV. The effectiveness of peer-supported interventions for encouraging dietary behaviour change in adults: a systematic review. Public Health Nutr. 2019;22. doi:10.1017/S1368980018003294
  • Cederholm T, Jensen GL, Correia MI, et al. GLIM criteria for the diagnosis of malnutrition – a consensus report from the global clinical nutrition community. Clin Nutr. 2019;38(1):1–9. doi:10.1016/j.clnu.2018.08.002
  • Filippi J, Al-Jaouni R, Wiroth JB, Hébuterne X, Schneider SM. Nutritional deficiencies in patients with Crohn’s disease in remission. Inflamm Bowel Dis. 2006;12(3):185–191. doi:10.1097/01.MIB.0000206541.15963.c3
  • Kondrup J, Rasmussen H, Hamberg O, Stanga Z. Nutritional risk screening (NRS 2002): a new method based on an analysis of controlled clinical trials. Clin Nutr. 2003;22:321–336. doi:10.1016/S0261-5614(02)00214-5
  • BAPEN. The “MUST” explanatory booklet: a guide to the “Malnutrition Universal Screening Tool” (‘MUST’) for adults; 2011. Available from: https://www.health.gov.il/download/ng/N500-19.pdf. Accessed January 9, 2023.
  • Day AS, Yao CK, Costello SP, Andrews JM, Bryant RV. Food avoidance, restrictive eating behaviour and association with quality of life in adults with inflammatory bowel disease: a systematic scoping review. Appetite. 2021;167:105650. doi:10.1016/j.appet.2021.105650
  • Czuber-Dochan W, Morgan M, Hughes LD, Lomer MCE, Lindsay JO, Whelan K. Perceptions and psychosocial impact of food, nutrition, eating and drinking in people with inflammatory bowel disease: a qualitative investigation of food-related quality of life. J Hum Nutr Diet. 2020;33(1):115–127. doi:10.1111/jhn.12668
  • Dunleavy K, Ungaro R, Manning L, Novak J, Gold S, Colombel J. Vitamin C deficiency in inflammatory bowel disease: the forgotten micronutrient. J Crohns Colitis. 2020;14(Supplement_1):S209–S209. doi:10.1093/ecco-jcc/jjz203.272
  • Bischoff SC, Escher J, Hébuterne X, et al. ESPEN practical guideline: clinical Nutrition in inflammatory bowel disease. Clin Nutr. 2020;39(3):632–653. doi:10.1016/j.clnu.2019.11.002
  • Torres J, Bonovas S, Doherty G, et al. ECCO guidelines on therapeutics in Crohn’s disease: medical treatment. J Crohns Colitis. 2020;14(1):4–22. doi:10.1093/ecco-jcc/jjz180
  • Raine T, Spinelli A, Bonovas S, et al. ECCO guidelines on therapeutics in ulcerative colitis: surgical treatment. J Crohns Colitis. 2022;16(2):179–189. doi:10.1093/ecco-jcc/jjab177
  • Chiu E, Oleynick C, Raman M, Bielawska B. Optimizing inpatient nutrition care of adult patients with inflammatory bowel disease in the 21st century. Nutrients. 2021;13(5):1581. doi:10.3390/nu13051581
  • Danese S, Solitano V, Jairath V, Biroulet LP. The future of drug development for inflammatory bowel disease: the need to ACT (advanced combination treatment). Gut. 2022;71:2380–2387. doi:10.1136/gutjnl-2022-327025
  • Wellens J, Vermeire S, Sabino J. Let food be thy medicine — its role in Crohn’s disease. Nutrients. 2021;13(832):1–16. doi:10.3390/nu13030832
  • Dziechciarz P, Horvath A, Shamir R, Szajewska H. Meta-analysis: enteral nutrition in active Crohn’s disease in children. Aliment Pharmacol Ther. 2007;26(6):795–806. doi:10.1111/j.1365-2036.2007.03431.x
  • Gunasekeera V, Mendall MA, Chan D, Kumar D. Treatment of Crohn’s disease with an IgG4-guided exclusion diet: a randomized controlled trial. Dig Dis Sci. 2016;61(4):1148–1157. doi:10.1007/s10620-015-3987-z
  • Komperød MJ, Sommer C, Mellin-Olsen T, Iversen PO, Røseth AG, Valeur J. Persistent symptoms in patients with Crohn’s disease in remission: an exploratory study on the role of diet. Scand J Gastroenterol. 2018;53(5):573–578. doi:10.1080/00365521.2017.1397736
  • Levine A, Wine E, Assa A, et al. Crohn’s disease exclusion diet plus partial enteral nutrition induces sustained remission in a randomized controlled trial. Gastroenterology. 2019;157(2):440–450.e8. doi:10.1053/j.gastro.2019.04.021
  • Svolos V, Hansen R, Nichols B, et al. Treatment of active Crohn’s disease with an ordinary food-based diet that replicates exclusive enteral nutrition. Gastroenterology. 2019;156(5):1354–1367.e6. doi:10.1053/j.gastro.2018.12.002
  • Olendzki BC, Silverstein TD, Persuitte GM, Ma Y, Baldwin KR, Cave D. An anti-inflammatory diet as treatment for inflammatory bowel disease: discovery service for Endeavour College of Natural Health Library. Nutr J. 2014;13(5):1–7. doi:10.1186/1475-2891-13-5
  • Konijeti GG, Kim N, Lewis JD, et al. Efficacy of the autoimmune protocol diet for inflammatory bowel disease. Inflamm Bowel Dis. 2017;23(11):2054–2060. doi:10.1097/MIB.0000000000001221
  • Whelan K, Martin LD, Staudacher HM, Lomer MCE. The low FODMAP diet in the management of irritable bowel syndrome: an evidence-based review of FODMAP restriction, reintroduction and personalisation in clinical practice. J Hum Nutr Diet. 2018;31(2):239–255. doi:10.1111/jhn.12530
  • Sabino J, Lewis JD, Colombel JF. Treating inflammatory bowel disease with diet: a taste test. Gastroenterology. 2019;157(2):295–297. doi:10.1053/j.gastro.2019.06.027
  • Noor NM, Verstockt B, Parkes M, Lee JC. Personalised medicine in Crohn’s disease. lancet Gastroenterol Hepatol. 2020;5(1):80–92. doi:10.1016/S2468-1253(19)30340-1
  • Schwärzler J, Mayr L, Vich Vila A, et al. PUFA-induced metabolic enteritis as a fuel for Crohn’s disease. Gastroenterology. 2022;162(6):1690–1704. doi:10.1053/j.gastro.2022.01.004
  • Sabino J. Understanding the role of PUFAs in Crohn’s disease. Gastroenterology. 2022;162(6):1590–1591. doi:10.1053/j.gastro.2022.02.048
  • Triggs CM, Munday K, Hu R, et al. Dietary factors in chronic inflammation: food tolerances and intolerances of a New Zealand Caucasian Crohn’s disease population. Mutat Res. 2010;690(1–2):123–138. doi:10.1016/j.mrfmmm.2010.01.020
  • Cohen AB, Lee D, Long MD, et al. Dietary patterns and self-reported associations of diet with symptoms of inflammatory bowel disease. Dig Dis Sci. 2013;58(5):1322–1328. doi:10.1007/s10620-012-2373-3.Dietary
  • Pittet V, Vaucher C, Maillard MH, et al. Information needs and concerns of patients with inflammatory bowel disease: what can we learn from participants in a bilingual clinical cohort? PLoS One. 2016;11(3):1–17. doi:10.1371/journal.pone.0150620
  • Armstrong H, Mander I, Zhang Z, Armstrong D, Wine E. Not all fibers are born equal; variable response to dietary fiber subtypes in IBD. Front Pediatr. 2021;8:1–15. doi:10.3389/fped.2020.620189
  • Beukema M, Faas MM, de Vos P. The effects of different dietary fiber pectin structures on the gastrointestinal immune barrier: impact via gut microbiota and direct effects on immune cells. Exp Mol Med. 2020;52(9):1364–1376. doi:10.1038/s12276-020-0449-2
  • Cox SR, Lindsay JO, Fromentin S, et al. Effects of low FODMAP diet on symptoms, fecal microbiome, and markers of inflammation in patients with quiescent inflammatory bowel disease in a randomized trial. Gastroenterology. 2020;158(1):176–188.e7. doi:10.1053/j.gastro.2019.09.024
  • Halmos EP, Christophersen CT, Bird AR, Shepherd SJ, Muir JG, Gibson PR. Consistent prebiotic effect on gut microbiota with altered fodmap intake in patients with crohn s disease: a randomised, controlled cross-over trial of well-defined diets. Clin Transl Gastroenterol. 2016;7(4):e164–10. doi:10.1038/ctg.2016.22
  • Loughman A, Staudacher HM. Treating the individual with diet: is gut microbiome testing the answer? Lancet Gastroenterol Hepatol. 2020;5(5):437. doi:10.1016/S2468-1253(20)30023-6
  • Gardner CD, Trepanowski JF, Gobbo LCD, et al. Effect of low-fat VS low-carbohydrate diet on 12-month weight loss in overweight adults and the association with genotype pattern or insulin secretion the DIETFITS randomized clinical trial. JAMA. 2018;319(7):667–679. doi:10.1001/jama.2018.0245
  • Zeevi D, Korem T, Zmora N, et al. Personalized nutrition by prediction of glycemic responses. Cell. 2015;163(5):1079–1094. doi:10.1016/j.cell.2015.11.001
  • Mirkov MU, Verstockt B, Cleynen I. Genetics of inflammatory bowel disease: beyond NOD2. Lancet Gastroenterol Hepatol. 2017;2(3):224–234. doi:10.1016/S2468-1253(16)30111-X
  • Petermann I, Triggs CM, Huebner C, et al. Mushroom intolerance: a novel diet gene interaction in Crohn’s disease. Br J Nutr. 2009;102(4):506–508. doi:10.1017/S0007114509276446
  • Takahashi Y, Masuda H, Ishii Y, Nishida Y, Kobayashi M, Asai S. Decreased expression of thioredoxin interacting protein mRNA in inflamed colonic mucosa in patients with ulcerative colitis. Oncol Rep. 2007;18(3):531–535. doi:10.3892/or.18.3.531
  • Laing BB, Lim AG, Ferguson LR. A personalised dietary approach-A way forward to manage nutrient deficiency, effects of the western diet, and food intolerances in inflammatory bowel disease. Nutrients. 2019;11(7):1–28. doi:10.3390/nu11071532
  • Wild CP. Complementing the genome with an “exposome”: the outstanding challenge of environmental exposure measurement in molecular epidemiology. Cancer Epidemiol Biomarkers Prev. 2005;14(8):1847–1850. doi:10.1158/1055-9965.EPI-05-0456
  • Sudhakar P, Alsoud D, Wellens J, Verstockt S. Tailoring multi-omics to inflammatory bowel diseases: all for one and one for all. JCC. 2022;16:1306–1320. doi:10.1016/S2468-1253(21)00065-0
  • Zeng Z, Mukherjee A, Zhang H. From genetics to epigenetics, roles of epigenetics in inflammatory bowel disease. Front Genet. 2019;10:1–17. doi:10.3389/fgene.2019.01017
  • Hardy TM, Tollefsbol TO. Epigenetic diet: impact on the epigenome and cancer. Epigenomics. 2011;3(4):503–518. doi:10.2217/epi.11.71
  • Biesiekierski JR, Jalanka J, Staudacher HM. Can gut microbiota composition predict response to dietary treatments? Nutrients. 2019;11(5):1–15. doi:10.3390/nu11051134
  • Bennet SMP, Böhn L, Störsrud S, et al. Multivariate modelling of faecal bacterial profiles of patients with IBS predicts responsiveness to a diet low in FODMAPs. Gut. 2018;67(5):872–881. doi:10.1136/gutjnl-2016-313128
  • Qi Q, Li J, Yu B, et al. Host and gut microbial tryptophan metabolism and type 2 diabetes: an integrative analysis of host genetics, diet, gut microbiome and circulating metabolites in cohort studies. Gut. 2021;71:1095–1105. doi:10.1136/gutjnl-2021-324053
  • Tebani A, Bekri S. Paving the way to precision nutrition through metabolomics. Front Nutr. 2019;6:1–10. doi:10.3389/fnut.2019.00041
  • Koeth RA, Wang Z, Levison BS, et al. Intestinal microbiota metabolism of l-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med. 2013;19(5):576–585. doi:10.1038/nm.3145
  • Tang WHW, Wang Z, Levison BS, et al. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N Engl J Med. 2013;368(17):1575–1584. doi:10.1056/nejmoa1109400
  • Aldubayan MA, Pigsborg K, Gormsen SMO, et al. A double-blinded, randomized, parallel intervention to evaluate biomarker-based nutrition plans for weight loss: the PREVENTOMICS study. Clin Nutr. 2022;41:101890. doi:10.1016/j.clnu.2022.06.032
  • Kim AH, Roberts C, Feagan BG, et al. Developing a standard set of patient-centred outcomes for inflammatory bowel disease-an international, cross-disciplinary consensus. J Crohns Colitis. 2018;12(4):408–418. doi:10.1093/ecco-jcc/jjx161
  • Laville M, Segrestin B, Alligier M, et al. Evidence-based practice within nutrition: what are the barriers for improving the evidence and how can they be dealt with? Trials. 2017;18(1). doi:10.1186/s13063-017-2160-8
  • Staudacher HM, Irving PM, Lomer MCE, Whelan K. The challenges of control groups, placebos and blinding in clinical trials of dietary interventions. Proc Nutr Soc. 2017;76(3):203–212. doi:10.1017/S0029665117000350
  • Dron L, Taljaard M, Cheung YB, et al. The role and challenges of cluster randomised trials for global health. Lancet Glob Health. 2021;9(5):e701–e710. doi:10.1016/S2214-109X(20)30541-6
  • Kaplan HC, Opipari-Arrigan L, Yang J, et al. Personalized research on diet in ulcerative colitis and crohn’s disease: a series of N-of-1 diet trials. Am J Gastroenterol. 2022;117(6):902–917. doi:10.14309/ajg.0000000000001800
  • Dimbleby H. The national food strategy: the plan. Environ Rural Aff. 2021;290. doi:10.2307/j.ctt4cgqfz.3