Improvements to postprandial glucose control in subjects with type 2 diabetes: a multicenter, double blind, randomized placebo-controlled trial of a novel probiotic formulation

Fanny Perraudeau, Paul McMurdie, James Bullard, Andrew Cheng, Colleen Cutcliffe, Achal Deo, John Eid, Jessica Gines, Mohan Iyer, Nicholas Justice, Wesley T Loo, Madeleine Nemchek, Marcus Schicklberger, Michael Souza, Brendon Stoneburner, Surabhi Tyagi, Orville Kolterman, Fanny Perraudeau, Paul McMurdie, James Bullard, Andrew Cheng, Colleen Cutcliffe, Achal Deo, John Eid, Jessica Gines, Mohan Iyer, Nicholas Justice, Wesley T Loo, Madeleine Nemchek, Marcus Schicklberger, Michael Souza, Brendon Stoneburner, Surabhi Tyagi, Orville Kolterman

Abstract

Introduction: A growing body of evidence suggests that specific, naturally occurring gut bacteria are under-represented in the intestinal tracts of subjects with type 2 diabetes (T2D) and that their functions, like gut barrier stability and butyrate production, are important to glucose and insulin homeostasis. The objective of this study was to test the hypothesis that enteral exposure to microbes with these proposed functions can safely improve clinical measures of glycemic control and thereby play a role in the overall dietary management of diabetes.

Research design and methods: We evaluated whether a probiotic comprised of these anaerobic bacteria would enhance dietary management by (1) manufacturing two novel probiotic formulations containing three (WBF-010) or five (WBF-011) distinct strains in a Current Good Manufacturing Practice (cGMP) facility, (2) establishing consistent live-cell concentrations, (3) confirming safety at target concentrations dispensed in both animal and human studies and (4) conducting a 12-week parallel, double-blind, placebo-controlled, proof-of-concept study in which subjects previously diagnosed with T2D (n=76) were randomly assigned to a two times a day regimen of placebo, WBF-010 or WBF-011.

Results: No safety or tolerability issues were observed. Compared with the placebo group, subjects administered WBF-011 (which contains inulin, Akkermansia muciniphila, Clostridium beijerinckii, Clostridium butyricum, Bifidobacterium infantis and Anaerobutyricum hallii) significantly improved in the primary outcome, glucose total area under the curve (AUC): -36.1 mg/dL/180 min, p=0.0500 and also improved in secondary outcomes, glycated hemoglobin (A1c): -0.6, glucose incremental-AUC: -28.6 mg/dL/180 min.

Conclusions: To our knowledge, this is the first randomized controlled trial to administer four of the five strains to human subjects with T2D. This proof-of-concept study (clinical trial number NCT03893422) shows that the intervention was safe and well tolerated and that supplementation with WBF-011 improves postprandial glucose control. The limited sample size and intersubject variability justifies future studies designed to confirm and expand on these observations.

Keywords: diabetes mellitus, Type 2; fatty acids; glycated hemoglobin A.

Conflict of interest statement

Competing interests: All authors are employees and stock/stock option shareholders of Pendulum Therapeutics, Inc (formerly known as ‘Whole Biome Inc.’). OK owns stock in GlySens, Inc, has stock options in ViaCyte, Inc, and is a consultant to NuSirt BioPharma, Circius, and NanoPrecision Medical.

© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Summary of trial. (A) CONSORT flow diagram of subject enrollment, allocation, and follow-up counts. A total of 76 subjects were randomized at one of six US study sites and allocated by block to one of three experimental arms. Withdrawal from study participation was similar across arms, whereas those on placebo were more likely to discontinue the intervention. (B) Ingredients present in respective formulation capsules. Specific per-strain viability is provided in online supplementary table S1. A description of study formulation production is provided in online supplementary text S4. (C) Graphical summary of glucose control measurement outcomes. Primary endpoint was total glucose AUC0-180 min, while incremental glucose AUC0-180 min and A1c were key secondary measures. Additional detail is provided in table 2 and online supplementary figure S1. AUC, area under the curve.
Figure 2
Figure 2
Rates of detection of probiotic strains via qPCR at each of four collection timepoints. Each point indicates the fraction of subjects in a study arm that had at least two replicates with positive detection. Light gray box indicates the y-axis range of proportions observed in samples from subjects randomized to the placebo group. Point shape distinguishes separate primer-pairs for the same strain. Primer sequences are defined in online supplementary table S1. All stool homogenate samples were measured in at least quadruplicate reactions for each primer-pair.
Figure 3
Figure 3
Summary of subject-wise changes in stool SCFAs. Each point in each panel represents a different subject in the study. (A) Change in the stool butyrate fraction of total SCFA (acetate, propionate, butyrate), shown as the per-subject Log10-ratio of week-12 to baseline. (B) Changes in the millimolar concentration in stool. Changes are represented as baseline subtracted from week-12 (median difference for technical replicate pairs). Panels separate the values for each SCFA. Further detail is provided in online supplementary text S4. SCFA, short-chain fatty acid.

References

    1. Centers for Disease Control and Prevention C for DC and P National diabetes statistics report 2017.
    1. American Diabetes Association Economic costs of diabetes in the U.S. in 2017. Diabetes Care 2018;41:917–28. 10.2337/dci18-0007
    1. Bullard KM, Cowie CC, Lessem SE, et al. . Prevalence of Diagnosed Diabetes in Adults by Diabetes Type - United States, 2016. MMWR Morb Mortal Wkly Rep 2018;67:359–61. 10.15585/mmwr.mm6712a2
    1. Dean L, McEntyre J. The Genetic Landscape of Diabetes - NCBI Bookshelf 2004.
    1. Qi L, Cornelis MC, Zhang C, et al. . Genetic predisposition, Western dietary pattern, and the risk of type 2 diabetes in men. Am J Clin Nutr 2009;89:1453–8. 10.3945/ajcn.2008.27249
    1. Trowell HC. Dietary-fiber hypothesis of the etiology of diabetes mellitus. Diabetes 1975;24:762–5. 10.2337/diab.24.8.762
    1. Sanna S, van Zuydam NR, Mahajan A, et al. . Causal relationships among the gut microbiome, short-chain fatty acids and metabolic diseases. Nat Genet 2019;51:600–5. 10.1038/s41588-019-0350-x
    1. Hartstra AV, Bouter KEC, Bäckhed F, et al. . Insights into the role of the microbiome in obesity and type 2 diabetes. Diabetes Care 2015;38:159–65. 10.2337/dc14-0769
    1. Cani PD, de Vos WM. Next-Generation Beneficial Microbes: The Case of Akkermansia muciniphila. Front Microbiol 2017;8:8. 10.3389/fmicb.2017.01765
    1. Canfora EE, Meex RCR, Venema K, et al. . Gut microbial metabolites in obesity, NAFLD and T2DM. Nat Rev Endocrinol 2019;15:261–73. 10.1038/s41574-019-0156-z
    1. Lazar V, Ditu L-M, Pircalabioru GG, et al. . Gut microbiota, host organism, and diet trialogue in diabetes and obesity. Front Nutr 2019;6:21. 10.3389/fnut.2019.00021
    1. Vrieze A, Van Nood E, Holleman F, et al. . Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology 2012;143:913–6. 10.1053/j.gastro.2012.06.031
    1. Merenstein D, El-Nachef N, Lynch SV. Fecal microbial therapy: promises and pitfalls. J Pediatr Gastroenterol Nutr 2014;59:157–61. 10.1097/MPG.0000000000000415
    1. DeFilipp Z, Bloom PP, Torres Soto M, et al. . Drug-Resistant E. coli Bacteremia Transmitted by Fecal Microbiota Transplant. N Engl J Med 2019;381:2043–50. 10.1056/NEJMoa1910437
    1. Sun Z, Sun X, Li J, et al. . Using probiotics for type 2 diabetes mellitus intervention: advances, questions, and potential. Crit Rev Food Sci Nutr 2020;60:1–14. 10.1080/10408398.2018.1547268
    1. McFarland LV, Evans CT, Goldstein EJC. Strain-Specificity and Disease-Specificity of probiotic efficacy: a systematic review and meta-analysis. Front Med 2018;5:124. 10.3389/fmed.2018.00124
    1. Samah S, Ramasamy K, Lim SM, et al. . Probiotics for the management of type 2 diabetes mellitus: a systematic review and meta-analysis. Diabetes Res Clin Pract 2016;118:172–82. 10.1016/j.diabres.2016.06.014
    1. Shetty SA, Zuffa S, Bui TPN, et al. . Reclassification of Eubacterium hallii as Anaerobutyricum hallii gen. nov., comb. nov., and description of Anaerobutyricum soehngenii sp. nov., a butyrate and propionate-producing bacterium from infant faeces. Int J Syst Evol Microbiol 2018;68:3741–6. 10.1099/ijsem.0.003041
    1. Derrien M, Vaughan EE, Plugge CM, et al. . Akkermansia muciniphila gen. nov., sp. nov., a human intestinal mucin-degrading bacterium. Int J Syst Evol Microbiol 2004;54:1469–76. 10.1099/ijs.0.02873-0
    1. Everard A, Belzer C, Geurts L, et al. . Cross-Talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc Natl Acad Sci U S A 2013;110:9066–71. 10.1073/pnas.1219451110
    1. Belzer C, de Vos WM. Microbes inside--from diversity to function: the case of Akkermansia. Isme J 2012;6:1449–58. 10.1038/ismej.2012.6
    1. Allin KH, Tremaroli V, Caesar R, et al. . Aberrant intestinal microbiota in individuals with prediabetes. Diabetologia 2018;61:810–20. 10.1007/s00125-018-4550-1
    1. Dao MC, Everard A, Aron-Wisnewsky J, et al. . Akkermansia muciniphila and improved metabolic health during a dietary intervention in obesity: relationship with gut microbiome richness and ecology. Gut 2016;65:426–36. 10.1136/gutjnl-2014-308778
    1. Yassour M, Lim MY, Yun HS, et al. . Sub-clinical detection of gut microbial biomarkers of obesity and type 2 diabetes. Genome Med 2016;8:17. 10.1186/s13073-016-0271-6
    1. Shin N-R, Lee J-C, Lee H-Y, et al. . An increase in the Akkermansia spp. population induced by metformin treatment improves glucose homeostasis in diet-induced obese mice. Gut 2014;63:727–35. 10.1136/gutjnl-2012-303839
    1. Plovier H, Everard A, Druart C, et al. . A purified membrane protein from Akkermansia muciniphila or the pasteurized bacterium improves metabolism in obese and diabetic mice. Nat Med 2017;23:107–13. 10.1038/nm.4236
    1. Martens EC, Chiang HC, Gordon JI. Mucosal glycan foraging enhances fitness and transmission of a saccharolytic human gut bacterial symbiont. Cell Host Microbe 2008;4:447–57. 10.1016/j.chom.2008.09.007
    1. Tailford LE, Crost EH, Kavanaugh D, et al. . Mucin glycan foraging in the human gut microbiome. Front Genet 2015;6:81. 10.3389/fgene.2015.00081
    1. Depommier C, Everard A, Druart C, et al. . Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: a proof-of-concept exploratory study. Nat Med 2019;25:1096–103. 10.1038/s41591-019-0495-2
    1. Kolodziejczyk AA, Zheng D, Elinav E. Diet-microbiota interactions and personalized nutrition. Nat Rev Microbiol 2019;17:742–53. 10.1038/s41579-019-0256-8
    1. Koh A, De Vadder F, Kovatcheva-Datchary P, et al. . From dietary fiber to host physiology: short-chain fatty acids as key bacterial metabolites. Cell 2016;165:1332–45. 10.1016/j.cell.2016.05.041
    1. Vital M, Karch A, Pieper DH. Colonic butyrate-producing communities in humans: an overview using omics data. mSystems 2017;2:e00130-17. 10.1128/mSystems.00130-17
    1. Belenguer A, Duncan SH, Calder AG, et al. . Two routes of metabolic cross-feeding between Bifidobacterium adolescentis and butyrate-producing anaerobes from the human gut. Appl Environ Microbiol 2006;72:3593–9. 10.1128/AEM.72.5.3593-3599.2006
    1. Morrison DJ, Mackay WG, Edwards CA, et al. . Butyrate production from oligofructose fermentation by the human faecal flora: what is the contribution of extracellular acetate and lactate? Br J Nutr 2006;96:570–7.
    1. Gilijamse PW, Hartstra AV, Levin E, et al. . Treatment with Anaerobutyricum soehngenii: a pilot study of safety and dose-response effects on glucose metabolism in human subjects with metabolic syndrome. NPJ Biofilms Microbiomes 2020;6:16. 10.1038/s41522-020-0127-0
    1. Geerlings SY, Kostopoulos I, de Vos WM, et al. . Akkermansia muciniphila in the Human Gastrointestinal Tract: When, Where, and How? Microorganisms 2018;6:75. 10.3390/microorganisms6030075
    1. Bunesova V, Lacroix C, Schwab C. Mucin Cross-Feeding of infant bifidobacteria and Eubacterium hallii. Microb Ecol 2018;75:228–38. 10.1007/s00248-017-1037-4
    1. Belzer C, Chia LW, Aalvink S, et al. . Microbial Metabolic Networks at the Mucus Layer Lead to Diet-Independent Butyrate and Vitamin B12 Production by Intestinal Symbionts. mBio 2017;8:e00770-17. 10.1128/mBio.00770-17
    1. Ling Z, Liu X, Cheng Y, et al. . Clostridium butyricum combined with Bifidobacterium infantis probiotic mixture restores fecal microbiota and attenuates systemic inflammation in mice with antibiotic-associated diarrhea. Biomed Res Int 2015;2015:1–9. 10.1155/2015/582048
    1. McDonald TJ, Warren R. Diagnostic confusion? repeat HbA1c for the diagnosis of diabetes. Diabetes Care 2014;37:e135–6. 10.2337/dc14-0055
    1. O'Toole PW, Marchesi JR, Hill C. Next-Generation probiotics: the spectrum from probiotics to live biotherapeutics. Nat Microbiol 2017;2:17057. 10.1038/nmicrobiol.2017.57
    1. Saarela MH. Safety aspects of next generation probiotics. Curr Opin Food Sci 2019;30:8–13. 10.1016/j.cofs.2018.09.001
    1. David LA, Maurice CF, Carmody RN, et al. . Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014;505:559–63. 10.1038/nature12820
    1. Singh RK, Chang H-W, Yan D, et al. . Influence of diet on the gut microbiome and implications for human health. J Transl Med 2017;15:73. 10.1186/s12967-017-1175-y
    1. Hornung BVH, Zwittink RD, Kuijper EJ. Issues and current standards of controls in microbiome research. FEMS Microbiol Ecol 2019;95:fiz045. 10.1093/femsec/fiz045
    1. Donaldson GP, Lee SM, Mazmanian SK. Gut biogeography of the bacterial microbiota. Nat Rev Microbiol 2016;14:20–32. 10.1038/nrmicro3552
    1. Primec M, Mičetić-Turk D, Langerholc T. Analysis of short-chain fatty acids in human feces: a scoping review. Anal Biochem 2017;526:9–21. 10.1016/j.ab.2017.03.007

Source: PubMed

3
Suscribir