The Commensal Microbe Veillonella as a Marker for Response to an FGF19 Analog in NASH

Rohit Loomba, Lei Ling, Duy M Dinh, Alex M DePaoli, Hsiao D Lieu, Stephen A Harrison, Arun J Sanyal, Rohit Loomba, Lei Ling, Duy M Dinh, Alex M DePaoli, Hsiao D Lieu, Stephen A Harrison, Arun J Sanyal

Abstract

Background and aims: The composition of the human gut microbiota is linked to health and disease, and knowledge of the impact of therapeutics on the microbiota is essential to decipher their biological roles and to gain new mechanistic insights. Here we report the effect of aldafermin, an analog of the gut hormone FGF19, versus placebo on the gut microbiota in a prospective, phase 2 study in patients with NASH.

Approach and results: A total of 176 patients with biopsy-confirmed nonalcoholic steatohepatitis (NASH) (nonalcoholic fatty liver disease activity score ≥ 4), fibrosis (F1-F3 by NASH Clinical Research Network criteria), and elevated liver fat content (≥ 8% by magnetic resonance imaging-proton density fat fraction) received 0.3 mg (n = 23), 1 mg (n = 49), 3 mg (n = 49), and 6 mg (n = 28) aldafermin or placebo (n = 27) for 12 weeks. Stool samples were collected on day 1 and week 12 and profiled using 16S ribosomal RNA gene sequencing; 122 patients had paired stool microbiome profiles at both day 1 and week 12. Overall, the state of the gut microbial community was distinctly stable in patients treated with aldafermin, with all major phyla and genera unaltered during therapy. Patients treated with aldafermin showed a significant, dose-dependent enrichment in the rare genus Veillonella, a commensal microbe known to have lactate-degrading and performance-enhancing properties, which correlated with changes in serum bile acid profile.

Conclusions: Veillonella may be a bile acid-sensitive bacteria whose enrichment is enabled by aldafermin-mediated suppression of bile acid synthesis and, in particular, decreases in toxic bile acids. This study provides an integrated analysis of gut microbiome, serum bile acid metabolome, imaging, and histological measurements in clinical trials testing aldafermin for NASH. Our results provide a better understanding of the intricacies of microbiome-host interactions (clinicaltrials.gov trial No. NCT02443116).

© 2020 by the American Association for the Study of Liver Diseases.

Figures

Fig. 1
Fig. 1
Stable gut microbiome with aldafermin therapy in patients with NASH. (A) Study design. Cohort 1 was a placebo‐controlled, double‐blind study comparing aldafermin 3 mg and 6 mg versus placebo for 12 weeks; cohort 2 was a dose‐expansion study evaluating aldafermin 0.3 mg, 1 mg, and 3 mg for 12 weeks; and cohort 3 further expanded the assessment of aldafermin 1 mg in additional patients for 12 weeks. Overall, a total of 176 patients with NASH received 0.3 mg (n = 23), 1 mg (n = 49), 3 mg (n = 49), or 6 mg (n = 28) aldafermin or placebo (n = 27) for 12 weeks in this phase 2 trial of aldafermin. The 16S rRNA sequencing of fecal samples collected at baseline (day 1) and week 12 (end of treatment) was performed. Patients had liver biopsy at baseline and underwent MRI and laboratory tests at baseline and week 12. (B) Gut microbial richness and evenness measured by alpha diversity (P values by Mann‐Whitney U test with FDR corrections using Benjamini‐Hochberg method). (C) Gut microbial beta biodiversity. Beta diversity was evaluated using UniFrac‐based analysis. In the principal coordinates analysis, no clustering was observed at the PC1 versus PC2 plot, indicating stable phylogenetic composition of the samples. All patients with paired stool samples at both day 1 and week 12 were included in the analysis: placebo (n = 18), aldafermin 0.3 mg (n = 17), 1 mg (n = 30), 3 mg (n = 36), and 6 mg (n = 21). Abbreviations: D1, day 1; W12, week 12.
Fig. 2
Fig. 2
Phylogenetic abundance at the phylum level. (A) Relative abundance of top phyla by visits in patients treated with placebo or aldafermin. Taxonomic compositions of the most prevalent phyla are displayed, with ranks ordered from left to right by their decreasing abundance. Only the top 10 phylotypes are shown for clarity (P > 0.05 for all comparisons by Kruskal‐Wallis test with Benjamini‐Hochberg FDR‐adjusted multiple test correction). (B) Relative abundance of top phyla by treatment groups (P > 0.05 for all comparisons by Mann‐Whitney U test with Benjamini‐Hochberg FDR‐adjusted multiple test correction). The boxes represent the interquartile range (IQR), from the first and third quartiles, and the inside line represents the median. The whiskers denote the lowest and highest values within 1.5 IQR from the first and third quartiles. The circles represent outliers beyond the whiskers. All patients with paired stool samples at both day 1 and week 12 were included in the analysis: placebo (n = 18), aldafermin 0.3 mg (n = 17), 1 mg (n = 30), 3 mg (n = 36), and 6 mg (n = 21).
Fig. 3
Fig. 3
Phylogenetic abundance at the genus level. (A) Relative abundance of top genera by visits in patients treated with placebo or aldafermin. The phylotypes with median relative abundances greater than 0.01% of total abundance in either the placebo or aldafermin groups were included for analysis. Only the 10 most abundant genera in each group are shown for clarity (P > 0.05 for all comparisons by Kruskal‐Wallis test with Benjamini‐Hochberg FDR‐adjusted multiple test correction). (B) Relative abundance of top genera by treatment groups (P > 0.05 for all comparisons by Mann‐Whitney U test with Benjamini‐Hochberg FDR‐adjusted multiple test correction). The boxes represent the IQR, from the first and third quartiles, and the inside line represents the median. The whiskers denote the lowest and highest values within 1.5 IQR from the first and third quartiles. The circles represent outliers beyond the whiskers. All patients with paired stool samples at both day 1 and week 12 were included in the analysis: placebo (n = 18), aldafermin 0.3 mg (n = 17), 1 mg (n = 30), 3 mg (n = 36), and 6 mg (n = 21).
Fig. 4
Fig. 4
Aldafermin enriches the rare genus Veillonella. (A) Box and whisker plot of relative abundance of Veillonella. Stool samples from patients treated with placebo, aldafermin 0.3 mg, aldafermin 1 mg, aldafermin 3 mg, and aldafermin 6 mg were assessed by 16S rRNA gene sequencing. Dose‐dependent increases in Veillonella abundance were observed in the aldafermin groups, but not in the placebo group (***P < 0.001, **P < 0.01 vs. placebo by Kruskal‐Wallis test with Benjamini‐Hochberg FDR‐adjusted multiple test correction). (B) Proportion of subjects with fecal samples that were positive for Veillonella at week 12. Presence of Veillonella in stool samples was considered positive (**P < 0.01, *P < 0.05 vs. placebo by Fisher’s exact test). (C) Aldafermin did not affect other microbes that are of oral origin or typically associated with Veillonella, nor did it affect microbes that ferment other substrates or have an ethanol‐producing property. Shown are box and whisker plots of relative abundance in placebo and aldafermin groups. All patients with paired stool samples at both day 1 and week 12 were included in the analysis: placebo (n = 18), aldafermin 0.3 mg (n = 17), 1 mg (n = 30), 3 mg (n = 36), and 6 mg (n = 21). Abbreviations: D1, day 1; W12, week 12.
Fig. 5
Fig. 5
Enrichment of Veillonella inversely correlates with toxic, hydrophobic bile acids. (A) Box and whisker plots of serum bile acids from placebo and aldafermin‐treated patients by GC‐MS. Shown are concentrations of GCA, GCDCA, GDCA, and DCA at baseline (day 1) and end‐of‐treatment (week 12) (***P < 0.001, **P < 0.01, *P < 0.05 vs. placebo): placebo (n = 27), aldafermin 0.3 mg (n = 23), 1 mg (n = 49), 3 mg (n = 49), and 6 mg (n = 28). (B) Correlation between Veillonella OTU reads and concentrations of GCA, GCDCA, GDCA, and DCA at week 12. Spearman’s correlation coefficients were calculated; rho and P values are shown on the graphs.
Fig. 6
Fig. 6
Correlation between gut microbiome and LFC measured by MRI‐PDFF. (A) Patients were grouped into the top quartile (Q4) versus the bottom quartile (Q1) according to baseline LFC. Differences in alpha diversity were observed between Q1 and Q4 subjects using the observed OTUs but not the Shannon index. (B) Weighted UniFrac analysis showed no separation between low liver fat (Q1) and high liver fat (Q4) populations at baseline, whereas unweighted UniFrac showed a trend of clustering. (C) No significant difference at the phylum and genus level between low liver fat (Q1) and high liver fat (Q4) populations. (D) Relative abundance of top phyla in subjects with ≥ 70% reduction in LFC (i.e., super‐responders). At the phylum level, Euryarchaeota significantly differed between aldafermin‐treated subjects who had ≥ 70% reduction in LFC and the placebo group (*P < 0.05 by Mann‐Whitney U test with Benjamini‐Hochberg FDR‐adjusted multiple test correction). (E) Relative abundance of top genera in subjects with ≥ 70% reduction in LFC (i.e., super‐responders). Genera are displayed with ranks ordered from left to right by their decreasing abundance (left panel) or significance (right panel). Only the top 10 phylotypes are shown for clarity. At the genus level, although the top 10 genera are similar between groups, the minor genus Haemophilus is more abundant in subjects with ≥ 70% reduction in LFC than placebo‐treated subjects (*P < 0.05 by Mann‐Whitney U test with Benjamini‐Hochberg FDR‐adjusted multiple test correction). The boxes represent the IQR, from the first and third quartiles, and the inside line represents the median. The whiskers denote the lowest and highest values within 1.5 IQR from the first and third quartiles. The circles represent outliers beyond the whiskers. All patients with paired stool samples at both day 1 and week 12 were included in the analysis: placebo (n = 18), aldafermin 0.3 mg (n = 17), 1 mg (n = 30), 3 mg (n = 36), and 6 mg (n = 21).

References

    1. Backhed F, Ley RE, Sonnenburg JL, Peterson DA, Gordon JI. Host‐bacterial mutualism in the human intestine. Science 2005;307:1915‐1920.
    1. Sonnenburg JL, Backhed F. Diet‐microbiota interactions as moderators of human metabolism. Nature 2016;535:56‐64.
    1. Honda K, Littman DR. The microbiota in adaptive immune homeostasis and disease. Nature 2016;535:75‐84.
    1. Thaiss CA, Zmora N, Levy M, Elinav E. The microbiome and innate immunity. Nature 2016;535:65‐74.
    1. Younossi ZM, Tampi R, Priyadarshini M, Nader F, Younossi IM, Racila A. Burden of illness and economic model for patients with nonalcoholic steatohepatitis in the United States. Hepatology 2019;69:564‐572.
    1. Sharpton SR, Ajmera V, Loomba R. Emerging role of the gut microbiome in nonalcoholic fatty liver disease: from composition to function. Clin Gastroenterol Hepatol 2019;17:296‐306.
    1. Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A, Ley RE, et al. A core gut microbiome in obese and lean twins. Nature 2009;457:480‐484.
    1. Caussy C, Loomba R. Gut microbiome, microbial metabolites and the development of NAFLD. Nat Rev Gastroenterol Hepatol 2018;15:719‐720.
    1. Loomba R, Seguritan V, Li W, Long T, Klitgord N, Bhatt A, et al. Gut microbiome‐based metagenomic signature for non‐invasive detection of advanced fibrosis in human nonalcoholic fatty liver disease. Cell Metab 2017;25:1054‐1062.e1055.
    1. Hoyles L, Fernandez‐Real JM, Federici M, Serino M, Abbott J, Charpentier J, et al. Molecular phenomics and metagenomics of hepatic steatosis in non‐diabetic obese women. Nat Med 2018;24:1070‐1080.
    1. Boursier J, Mueller O, Barret M, Machado M, Fizanne L, Araujo‐Perez F, et al. The severity of nonalcoholic fatty liver disease is associated with gut dysbiosis and shift in the metabolic function of the gut microbiota. Hepatology 2016;63:764‐775.
    1. Raman M, Ahmed I, Gillevet PM, Probert CS, Ratcliffe NM, Smith S, et al. Fecal microbiome and volatile organic compound metabolome in obese humans with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol 2013;11:868‐875.e861‐863.
    1. Wong VW, Tse CH, Lam TT, Wong GL, Chim AM, Chu WC, et al. Molecular characterization of the fecal microbiota in patients with nonalcoholic steatohepatitis—a longitudinal study. PLoS One 2013;8:e62885.
    1. Wang B, Jiang X, Cao M, Ge J, Bao Q, Tang L, et al. Altered fecal microbiota correlates with liver biochemistry in nonobese patients with non‐alcoholic fatty liver disease. Sci Rep 2016;6:32002.
    1. Inagaki T, Moschetta A, Lee YK, Peng L, Zhao G, Downes M, et al. Regulation of antibacterial defense in the small intestine by the nuclear bile acid receptor. Proc Natl Acad Sci U S A 2006;103:3920‐3925.
    1. Parks DJ, Blanchard SG, Bledsoe RK, Chandra G, Consler TG, Kliewer SA, et al. Bile acids: natural ligands for an orphan nuclear receptor. Science 1999;284:1365‐1368.
    1. Kliewer SA, Mangelsdorf DJ. Bile acids as hormones: the FXR‐FGF15/19 pathway. Dig Dis 2015;33:327‐331.
    1. Degirolamo C, Sabba C, Moschetta A. Therapeutic potential of the endocrine fibroblast growth factors FGF19, FGF21 and FGF23. Nat Rev Drug Discov 2016;15:51‐69.
    1. Zhou M, Learned RM, Rossi SJ, DePaoli AM, Tian H, Ling L. Engineered FGF19 eliminates bile acid toxicity and lipotoxicity leading to resolution of steatohepatitis and fibrosis in mice. Hepatol Commun 2017;1:1024‐1042.
    1. DePaoli AM, Zhou M, Kaplan DD, Hunt SC, Adams TD, Learned RM, et al. FGF19 analog as a surgical factor mimetic that contributes to metabolic effects beyond glucose homeostasis. Diabetes 2019;68:1315‐1328.
    1. Harrison SA, Rinella ME, Abdelmalek MF, Trotter JF, Paredes AH, Arnold HL, et al. NGM282 for treatment of non‐alcoholic steatohepatitis: a multicentre, randomised, double‐blind, placebo‐controlled, phase 2 trial. Lancet 2018;391:1174‐1185.
    1. Harrison SA, Rossi SJ, Paredes AH, Trotter JF, Bashir MR, Guy CD, et al. NGM282 improves liver fibrosis and histology in 12 weeks in patients with nonalcoholic steatohepatitis. Hepatology 2020;71:1198‐1212.
    1. Rinella ME, Trotter JF, Abdelmalek MF, Paredes AH, Connelly MA, Jaros MJ, et al. Rosuvastatin improves the FGF19 analogue NGM282‐associated lipid changes in patients with non‐alcoholic steatohepatitis. J Hepatol 2019;70:735‐744.
    1. Zhu L, Baker SS, Gill C, Liu W, Alkhouri R, Baker RD, et al. Characterization of gut microbiomes in nonalcoholic steatohepatitis (NASH) patients: a connection between endogenous alcohol and NASH. Hepatology 2013;57:601‐609.
    1. Yoshimoto S, Loo TM, Atarashi K, Kanda H, Sato S, Oyadomari S, et al. Obesity‐induced gut microbial metabolite promotes liver cancer through senescence secretome. Nature 2013;499:97‐101.
    1. Scheiman J, Luber JM, Chavkin TA, MacDonald T, Tung A, Pham LD, et al. Meta‐omics analysis of elite athletes identifies a performance‐enhancing microbe that functions via lactate metabolism. Nat Med 2019;25:1104‐1109.
    1. Sorribas M, Jakob MO, Yilmaz B, Li H, Stutz D, Noser Y, et al. FXR modulates the gut‐vascular barrier by regulating the entry sites for bacterial translocation in experimental cirrhosis. J Hepatol 2019;71:1126‐1140.
    1. Qin N, Yang F, Li A, Prifti E, Chen Y, Shao L, et al. Alterations of the human gut microbiome in liver cirrhosis. Nature 2014;513:59‐64.
    1. Chen Y, Ji F, Guo J, Shi D, Fang D, Li L. Dysbiosis of small intestinal microbiota in liver cirrhosis and its association with etiology. Sci Rep 2016;6:34055.
    1. Oh TG, Kim SM, Caussy C, Fu T, Guo J, Bassirian S, Singh S, et al. A Universal Gut‐Microbiome‐Derived Signature Predicts Cirrhosis. Cell Metab 2020;32:901.
    1. Schwartz CC, Almond HR, Vlahcevic ZR, Swell L. Bile acid metabolism in cirrhosis. V. Determination of biliary lipid secretion rates in patients with advanced cirrhosis. Gastroenterology 1979;77:1177‐1182.
    1. Merritt ME, Donaldson JR. Effect of bile salts on the DNA and membrane integrity of enteric bacteria. J Med Microbiol 2009;58:1533‐1541.
    1. Wang Y, Gao X, Zhang X, Xiao Y, Huang J, Yu D, Li X, et al. Gut microbiota dysbiosis is associated with altered bile acid metabolism in infantile cholestasis. mSystems 2019;4:e00463.
    1. Drolz A, Horvatits T, Rutter K, Landahl F, Roedl K, Meersseman P, et al. Lactate improves prediction of short‐term mortality in critically Ill patients with cirrhosis: a multinational study. Hepatology 2019;69:258‐269.
    1. Gaci N, Borrel G, Tottey W, O'Toole PW, Brugere JF. Archaea and the human gut: new beginning of an old story. World J Gastroenterol 2014;20:16062‐16078.
    1. Ghoshal U, Shukla R, Srivastava D, Ghoshal UC. Irritable bowel syndrome, particularly the constipation‐predominant form, involves an increase in methanobrevibacter smithii, which is associated with higher methane production. Gut Liv 2016;10:932‐938.
    1. Oduyebo I, Camilleri M, Nelson AD, Khemani D, Nord SL, Busciglio I, et al. Effects of NGM282, an FGF19 variant, on colonic transit and bowel function in functional constipation: a randomized phase 2 trial. Am J Gastroenterol 2018;113:725‐734.
    1. Caussy C, Tripathi A, Humphrey G, Bassirian S, Singh S, Faulkner C, et al. A gut microbiome signature for cirrhosis due to nonalcoholic fatty liver disease. Nat Commun 2019;10:1406.
    1. Caussy C, Hsu C, Lo MT, Liu A, Bettencourt R, Ajmera VH, et al. Link between gut‐microbiome derived metabolite and shared gene‐effects with hepatic steatosis and fibrosis in NAFLD. Hepatology 2018;68:918‐932.
    1. Tripathi A, Debelius J, Brenner DA, Karin M, Loomba R, Schnabl B, et al. The gut‐liver axis and the intersection with the microbiome. Nat Rev Gastroenterol Hepatol 2018;15:397‐411.
    1. Castera L, Friedrich‐Rust M, Loomba R. Noninvasive assessment of liver disease in patients with nonalcoholic fatty liver disease. Gastroenterology 2019;156:1264‐1281.e1264.
    1. Demir M, Lang S, Martin A, Farowski F, Wisplinghoff H, Vehreschild M, et al. Phenotyping non‐alcoholic fatty liver disease by the gut microbiota: Ready for prime time? J Gastroenterol Hepatol 2020. Apr 8. 10.1111/jgh.15071. [Epub ahead of print]
    1. Falony G, Joossens M, Vieira‐Silva S, Wang J, Darzi Y, Faust K, et al. Population‐level analysis of gut microbiome variation. Science 2016;352:560‐564.

Source: PubMed

3
Abonneren