Impact of fibre and red/processed meat intake on treatment outcomes among patients with chronic inflammatory diseases initiating biological therapy: A prospective cohort study

Silja H Overgaard, Signe B Sørensen, Heidi L Munk, Anders B Nexøe, Henning Glerup, Rikke H Henriksen, Tanja Guldmann, Natalia Pedersen, Sanaz Saboori, Lone Hvid, Jens F Dahlerup, Christian L Hvas, Mohamad Jawhara, Karina W Andersen, Andreas K Pedersen, Ole H Nielsen, Fredrik Bergenheim, Jacob B Brodersen, Berit L Heitmann, Thorhallur I Halldorsson, Uffe Holmskov, Anette Bygum, Robin Christensen, Jens Kjeldsen, Torkell Ellingsen, Vibeke Andersen, Silja H Overgaard, Signe B Sørensen, Heidi L Munk, Anders B Nexøe, Henning Glerup, Rikke H Henriksen, Tanja Guldmann, Natalia Pedersen, Sanaz Saboori, Lone Hvid, Jens F Dahlerup, Christian L Hvas, Mohamad Jawhara, Karina W Andersen, Andreas K Pedersen, Ole H Nielsen, Fredrik Bergenheim, Jacob B Brodersen, Berit L Heitmann, Thorhallur I Halldorsson, Uffe Holmskov, Anette Bygum, Robin Christensen, Jens Kjeldsen, Torkell Ellingsen, Vibeke Andersen

Abstract

Background: Biologic disease-modifying drugs have revolutionised the treatment of a number of chronic inflammatory diseases (CID). However, up to 60% of the patients do not have a sufficient response to treatment and there is a need for optimization of treatment strategies.

Objective: To investigate if the treatment outcome of biological therapy is associated with the habitual dietary intake of fibre and red/processed meat in patients with a CID.

Methods: In this multicentre prospective cohort study, we consecutively enrolled 233 adult patients with a diagnosis of Crohn's Disease, Ulcerative Colitis, Rheumatoid Arthritis (RA), Axial Spondyloarthritis, Psoriatic Arthritis and Psoriasis, for whom biologic therapy was planned, over a 3 year period. Patients with completed baseline food frequency questionnaires were stratified into a high fibre/low red and processed meat exposed group (HFLM) and an unexposed group (low fibre/high red and processed meat intake = LFHM). The primary outcome was the proportion of patients with a clinical response to biologic therapy after 14-16 weeks of treatment.

Results: Of the 193 patients included in our primary analysis, 114 (59%) had a clinical response to biologic therapy. In the HFLM group (N = 64), 41 (64%) patients responded to treatment compared to 73 (56%) in the LFHM group (N = 129), but the difference was not statistically significant (OR: 1.48, 0.72-3.05). For RA patients however, HFLM diet was associated with a more likely clinical response (82% vs. 35%; OR: 9.84, 1.35-71.56).

Conclusion: Habitual HFLM intake did not affect the clinical response to biological treatment across CIDs. HFLM diet in RA patients might be associated with better odds for responding to biological treatment, but this would need confirmation in a randomised trial.

Trial registration: (clinicaltrials.gov), identifier [NCT03173144].

Keywords: biologic therapy; chronic inflammatory disease; diet; fibre; inflammatory bowel disease; processed meat; red meat; rheumatoid arthritis.

Conflict of interest statement

Author CH has received speaker fee from Takeda Pharma and Tillotts Pharma (unrelated to the present work). The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2022 Overgaard, Sørensen, Munk, Nexøe, Glerup, Henriksen, Guldmann, Pedersen, Saboori, Hvid, Dahlerup, Hvas, Jawhara, Andersen, Pedersen, Nielsen, Bergenheim, Brodersen, Heitmann, Halldorsson, Holmskov, Bygum, Christensen, Kjeldsen, Ellingsen and Andersen.

Figures

Figure 1
Figure 1
Flow chart of the enrolment of participants. Number of Chronic Inflammatory Disease patients who were screened, included in the study and included in the analysis. Patients screened for eligibility were not explicitly recorded in the study. For CD, UC, and PsO we estimated a number based on the mean inclusion rate from two clinics that kept a pre-screening log. For RA, axSpA, and PsA, the number is not an estimate but the total number of patients that initiated biologic treatment in the clinic during enrolment. IBD, Inflammatory bowel disease (Crohn's disease and Ulcerative colitis); CD, Crohn's disease; UC, Ulcerative colitis; RA, Rheumatoid arthritis; axSpA, Axial spondyloarthritis; PsA, Psoriatic arthritis; PsO, Psoriasis; FFQ, Food Frequency Questionnaire; HFLM, high fibre/low meat; LFHM, low fibre/high meat; ITT, intention-to-treat; SF-12 PCS + MCS, short form health survey the physical and mental component summary, The short health scale includes four health dimensions (symptom burden, functional status, disease-related burden and general wellbeing); CRP, C-reactive protein.
Figure 2
Figure 2
Meta-analysis (random effects model) of the included Chronic Inflammatory Diseases (CIDs) on the “As Observed” population comparing clinical response to biologics in patients with a high intake of fibre and low intake of red/processed meat (HFLM) vs. patients with a low intake of fibre and high intake of red/processed meat (LFHM). The horizontal lines represent the odds ratio (OR) ± 95% confidence interval. Event = clinical response according to the specified criteria for each CID, i.e., the number shows how many out of the total number of participants in the group, that have had a clinical response. The “As Observed” populations means patients with complete data for clinical response (i.e., 17 patients are excluded).
Figure 3
Figure 3
Receiver operating characteristics curve analysis for the predictive value of the ratio of fibre and red/processed meat intake (A), fibre intake (B) and red/processed meat intake (C) on clinical response. The ROC curves were constructed by calculating the sensitivity and specificity for prediction of clinical response for (A) the ratio of fibre to red/processed meat intake, (B) fibre intake, and (C) red/processed meat intake. Thereafter the sensitivity was plotted against 1-specificity. ROC, receiver operating characteristics.

References

    1. Li P, Zheng Y, Chen X. Drugs for autoimmune inflammatory diseases: from small molecule compounds to anti-TNF biologics. Front Pharmacol. (2017) 8:460. 10.3389/fphar.2017.00460
    1. Sieper J, Poddubnyy D. Axial spondyloarthritis. Lancet. (2017) 390:73–84. 10.1016/S0140-6736(16)31591-4
    1. Ng SC, Shi HY, Hamidi N, Underwood FE, Tang W, Benchimol EI, et al. . Worldwide incidence and prevalence of inflammatory bowel disease in the 21st century: a systematic review of population-based studies. Lancet. (2018) 390:2769–78. 10.1016/S0140-6736(17)32448-0
    1. Ogdie A, Weiss P. The epidemiology of psoriatic arthritis. Rheum Dis Clin North Am. (2015) 41:545–68. 10.1016/j.rdc.2015.07.001
    1. Parisi R, Iskandar IYK, Kontopantelis E, Augustin M, Griffiths CEM, Ashcroft DM. National, regional, and worldwide epidemiology of psoriasis: systematic analysis and modelling study. BMJ. (2020) 369:m1590. 10.1136/bmj.m1590
    1. Alamanos Y, Drosos AA. Epidemiology of adult rheumatoid arthritis. Autoimmun Rev. (2005) 4:130–6. 10.1016/j.autrev.2004.09.002
    1. Cross M, Smith E, Hoy D, Carmona L, Wolfe F, Vos T, et al. . The global burden of rheumatoid arthritis: estimates from the global burden of disease 2010 study. Ann Rheum Dis. (2014) 73:1316–22. 10.1136/annrheumdis-2013-204627
    1. Knowles SR, Graff LA, Wilding H, Hewitt C, Keefer L, Mikocka-Walus A. Quality of life in inflammatory bowel disease: a systematic review and meta-analyses—part I. Inflamm Bowel Dis. (2018) 24:742–51. 10.1093/ibd/izx100
    1. Ritchlin CT, Colbert RA, Gladman DD. Psoriatic arthritis. N Engl J Med. (2017) 376:957–70. 10.1056/NEJMra1505557
    1. Stern RS, Nijsten T, Feldman SR, Margolis DJ, Rolstad T. Psoriasis is common, carries a substantial burden even when not extensive, and is associated with widespread treatment dissatisfaction. J Investig Dermatol Symp Proc. (2004) 9:136–9. 10.1046/j.1087-0024.2003.09102.x
    1. Aletaha D, Smolen JS. Diagnosis and management of rheumatoid arthritis: a review. JAMA. (2018) 320:1360–72. 10.1001/jama.2018.13103
    1. Digby-Bell JL, Atreya R, Monteleone G, Powell N. Interrogating host immunity to predict treatment response in inflammatory bowel disease. Nat Rev Gastroenterol Hepatol. (2020) 17:9–20. 10.1038/s41575-019-0228-5
    1. Nielsen OH, Ainsworth MA. Tumour necrosis factor inhibitors for inflammatory bowel disease. N Engl J Med. (2013) 369:754–62. 10.1056/NEJMct1209614
    1. Zaiss MM, Joyce Wu HJ, Mauro D, Schett G, Ciccia F. The gut-joint axis in rheumatoid arthritis. Nat Rev Rheumatol. (2021) 17:224–37. 10.1038/s41584-021-00585-3
    1. Neurath MF. Host-microbiota interactions in inflammatory bowel disease. Nat Rev Gastroenterol Hepatol. (2020) 17:76–7. 10.1038/s41575-019-0248-1
    1. Sikora M, Stec A, Chrabaszcz M, Knot A, Waskiel-Burnat A, Rakowska A, et al. . Gut microbiome in psoriasis: an updated review. Pathogens. (2020) 9:463–77. 10.3390/pathogens9060463
    1. Kanda N, Hoashi T, Saeki H. Nutrition and psoriasis. Int J Mol Sci. (2020) 21:5405–24. 10.3390/ijms21155405
    1. Campos-Perez W, Martinez-Lopez E. Effects of short chain fatty acids on metabolic and inflammatory processes in human health. Biochim Biophys Acta Mol Cell Biol Lipids. (2021) 1866:158900. 10.1016/j.bbalip.2021.158900
    1. Couto MR, Gonçalves P, Magro F, Martel F. Microbiota-derived butyrate regulates intestinal inflammation: focus on inflammatory bowel disease. Pharmacol Res. (2020) 159:104947. 10.1016/j.phrs.2020.104947
    1. Desai MS, Seekatz AM, Koropatkin NM, Kamada N, Hickey CA, Wolter M, et al. . A dietary fibre-deprived gut microbiota degrades the colonic mucus barrier and enhances pathogen susceptibility. Cell. (2016) 167:1339–1353.e21. 10.1016/j.cell.2016.10.043
    1. Yao CK, Muir JG, Gibson PR. Review article: insights into colonic protein fermentation, its modulation and potential health implications. Aliment Pharmacol Ther. (2016) 43:181–96. 10.1111/apt.13456
    1. Christensen R, Heitmann BL, Andersen KW, Nielsen OH, Sorensen SB, Jawhara M, et al. . Impact of red and processed meat and fibre intake on treatment outcomes among patients with chronic inflammatory diseases: protocol for a prospective cohort study of prognostic factors and personalised medicine. BMJ Open. (2018) 8:e018166. 10.1136/bmjopen-2017-018166
    1. Vandenbroucke JP, von Elm E, Altman DG, Gøtzsche PC, Mulrow CD, Pocock SJ, et al. . Strengthening the reporting of observational studies in epidemiology (STROBE): explanation and elaboration. Ann Intern Med. (2007) 147:W163–94. 10.7326/0003-4819-147-8-200710160-00010-w1
    1. Harris PA, Taylor R, Thielke R, Payne J, Gonzalez N, Conde JG. 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. 10.1016/j.jbi.2008.08.010
    1. Harris PA, Taylor R, Minor BL, Elliott V, Fernandez M, O'Neal L, et al. . The REDCap consortium: building an international community of software platform partners. J Biomed Inform. (2019) 95:103208. 10.1016/j.jbi.2019.103208
    1. Eriksen L, Grønbæk M, Helge JW, Tolstrup JS, Curtis T. The Danish health examination survey 2007–2008 (DANHES 2007–2008). Scand J Public Health. (2011) 39:203–11. 10.1177/1403494810393557
    1. Køster-Rasmussen R, Siersma V, Halldorsson TI, de Fine Olivarius N, Henriksen JE, Heitmann BL. Missing portion sizes in FFQ—alternatives to use of standard portions. Public Health Nutr. (2015) 18:1914–21. 10.1017/S1368980014002389
    1. Felson DT, Anderson JJ, Boers M, Bombardier C, Furst D, Goldsmith C, et al. . American college of rheumatology. Preliminary definition of improvement in rheumatoid arthritis. Arthritis Rheum. (1995) 38:727–35. 10.1002/art.1780380602
    1. Sieper J, Rudwaleit M, Baraliakos X, Brandt J, Braun J, Burgos-Vargas R, et al. . The assessment of spondyloarthritis international Society (ASAS) handbook: a guide to assess spondyloarthritis. Ann Rheum Dis. (2009) 68 Suppl 2:ii1–44. 10.1136/ard.2008.104018
    1. Austin PC. Balance diagnostics for comparing the distribution of baseline covariates between treatment groups in propensity-score matched samples. Stat Med. (2009) 28:3083–107. 10.1002/sim.3697
    1. Berglund P, Heeringa S. Multiple Imputation of Missing Data Using SAS. Cary, NC: SAS Institute Inc; (2014).
    1. Rubin DB. Multiple Imputation for Nonresponse in Surveys. Wiley Series in Probability and Mathematical Statistics. Applied Probability and Statistics. New York, NY: Wiley; (1987). p. 253. 10.1002/9780470316696
    1. Cao J, Zhang S. Multiple comparison procedures. JAMA. (2014) 312:543–4. 10.1001/jama.2014.9440
    1. Altman DG, Bland JM. Interaction revisited: the difference between two estimates. BMJ. (2003) 326:219. 10.1136/bmj.326.7382.219
    1. Christensen R, Bours MJL, Nielsen SM. Effect modifiers and statistical tests for interaction in randomised trials. J Clin Epidemiol. (2021) 134:174–7. 10.1016/j.jclinepi.2021.03.009
    1. Nguyen DL, Palmer LB, Nguyen ET, McClave SA, Martindale RG, Bechtold ML. Specialised enteral nutrition therapy in Crohn's disease patients on maintenance infliximab therapy: a meta-analysis. Therap Adv Gastroenterol. (2015) 8:168–75. 10.1177/1756283X15578607
    1. Chiba M, Tsuji T, Nakane K, Komatsu M. High amount of dietary fibre not harmful but favourable for Crohn's disease. Perm J. (2015) 19:58–61. 10.7812/TPP/14-124
    1. García-Montero C, Fraile-Martínez O, Gómez-Lahoz AM, Pekarek L, Castellanos AJ, Noguerales-Fraguas F, et al. . Nutritional components in western diet vs. mediterranean diet at the gut microbiota-immune system interplay. Implications for health and disease. Nutrients. (2021) 13:699–48. 10.3390/nu13020699
    1. Sofi F, Cesari F, Abbate R, Gensini GF, Casini A. Adherence to mediterranean diet and health status: meta-analysis. BMJ. (2008) 337:a1344. 10.1136/bmj.a1344
    1. Guerreiro CS, Calado Â, Sousa J, Fonseca JE. Diet, microbiota, and gut permeability-the unknown triad in rheumatoid arthritis. Front Med. (2018) 5:349. 10.3389/fmed.2018.00349
    1. Stephen AM, Champ MM, Cloran SJ, Fleith M, van Lieshout L, Mejborn H, et al. . Dietary fibre in Europe: current state of knowledge on definitions, sources, recommendations, intakes and relationships to health. Nutr Res Rev. (2017) 30:149–90. 10.1017/S095442241700004X
    1. Pedersen AN, Christensen T, Matthiessen J, Knudsen VK, Rosenlund-Sørensen M, Biltoft-Jensen A, et al. . Danskernes kostvaner 2011–2013. DTU National Food Institute; (2015). Available online at:
    1. Willett W, Rockström J, Loken B, Springmann M, Lang T, Vermeulen S, et al. . Food in the anthropocene: the EAT-lancet commission on healthy diets from sustainable food systems. Lancet. (2019) 393:447–92. 10.1016/S0140-6736(18)31788-4
    1. Cocking C, Walton J, Kehoe L, Cashman KD, Flynn A. The role of meat in the European diet: current state of knowledge on dietary recommendations, intakes and contribution to energy and nutrient intakes and status. Nutr Res Rev. (2020) 33:181–9. 10.1017/S0954422419000295
    1. McGonagle D, McDermott MF. A proposed classification of the immunological diseases. PLoS Med. (2006) 3:e297. 10.1371/journal.pmed.0030297
    1. Szekanecz Z, McInnes IB, Schett G, Szamosi S, Benko S, Szucs G. Autoinflammation and autoimmunity across rheumatic and musculoskeletal diseases. Nat Rev Rheumatol. (2021) 17:585–95. 10.1038/s41584-021-00652-9
    1. Sarkar MK, Tsoi LC, Gudjonsson JE. Psoriasis: a mixed autoimmune and autoinflammatory disease. Curr Opin Immunol. (2017) 49:1–8. 10.1016/j.coi.2017.07.007
    1. McGonagle D, Watad A, Savic S. Mechanistic immunological based classification of rheumatoid arthritis. Autoimmun Rev. (2018) 17:1115–23. 10.1016/j.autrev.2018.06.001
    1. Mease PJ, Karki C, Liu M, Li Y, Gershenson B, Feng H, et al. . Discontinuation and switching patterns of tumour necrosis factor inhibitors (TNFis) in TNFi-naive and TNFi-experienced patients with psoriatic arthritis: an observational study from the US-based corrona registry. RMD Open. (2019) 5:e000880. 10.1136/rmdopen-2018-000880
    1. Pope J, Thorne JC, Haraoui BP, Psaradellis E, Sampalis J. Do patients with active RA have differences in disease activity and perceptions if anti-TNF naive vs. anti-TNF experienced? Baseline results of the optimization of adalimumab trial. Med Sci Monit. (2012) 18:Pi17–20. 10.12659/MSM.883250
    1. Gomez-Reino JJ, Carmona L. Switching TNF antagonists in patients with chronic arthritis: an observational study of 488 patients over a 4-year period. Arthritis Res Ther. (2006) 8:R29. 10.1186/ar1881
    1. Vandenbroucke JP, von Elm E, Altman DG, Gøtzsche PC, Mulrow CD, Pocock SJ, et al. . Strengthening the reporting of observational studies in epidemiology (STROBE): explanation and elaboration. Int J Surg. (2014) 12:1500–24. 10.1016/j.ijsu.2014.07.014
    1. Cox SR, Clarke H, O'Keeffe M, Dubois P, Irving PM, Lindsay JO, et al. . Nutrient, fibre, and FODMAP intakes and food-related quality of life in patients with inflammatory bowel disease, and their relationship with gastrointestinal symptoms of differing aetiologies. J Crohns Colitis. (2021) 15:2041–53. 10.1093/ecco-jcc/jjab116
    1. Principi M, Losurdo G, Iannone A, Contaldo A, Deflorio V, Ranaldo N, et al. . Differences in dietary habits between patients with inflammatory bowel disease in clinical remission and a healthy population. Ann Gastroenterol. (2018) 31:469–73. 10.20524/aog.2018.0273
    1. Bolte LA, Vich Vila A, Imhann F, Collij V, Gacesa R, Peters V, et al. . Long-term dietary patterns are associated with pro-inflammatory and anti-inflammatory features of the gut microbiome. Gut. (2021) 70:1287–98. 10.1136/gutjnl-2020-322670

Source: PubMed

3
Se inscrever