Abstract
The landscape of clinical microbiome research has dramatically evolved over the past decade. By leveraging in vivo and in vitro experimentation, multiomic approaches and computational biology, we have uncovered mechanisms of action and microbial metrics of association and identified effective ways to modify the microbiome in many diseases and treatment modalities. This Review explores recent advances in the clinical application of microbiome research over the past 5 years, while acknowledging existing barriers and highlighting opportunities. We focus on the translation of microbiome research into clinical practice, spearheaded by Food and Drug Administration (FDA)-approved microbiome therapies for recurrent Clostridioides difficile infections and the emerging fields of microbiome-based diagnostics and therapeutics. We highlight key examples of studies demonstrating how microbiome mechanisms, metrics and modifiers can advance clinical practice. We also discuss forward-looking perspectives on key challenges and opportunities toward integrating microbiome data into routine clinical practice, precision medicine and personalized healthcare and nutrition.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$259.00 per year
only $21.58 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout


Similar content being viewed by others
References
Chang, D. et al. Gut Microbiome Wellness Index 2 enhances health status prediction from gut microbiome taxonomic profiles. Nat. Commun. 15, 7447 (2024).
Hickman, B. et al. Gut microbiota wellbeing index predicts overall health in a cohort of 1000 infants. Nat. Commun. 15, 8323 (2024).
Schmartz, G. P. et al. Decoding the diagnostic and therapeutic potential of microbiota using pan-body pan-disease microbiomics. Nat. Commun. 15, 8261 (2024).
Weingarden, A. et al. Dynamic changes in short- and long-term bacterial composition following fecal microbiota transplantation for recurrent Clostridium difficile infection. Microbiome 3, 10 (2015).
Ross, F. C. et al. The interplay between diet and the gut microbiome: implications for health and disease. Nat. Rev. Microbiol. 22, 671–686 (2024).
Shanahan, F., Ghosh, T. S. & O’Toole, P. W. The healthy microbiome—what is the definition of a healthy gut microbiome? Gastroenterology 160, 483–494 (2021).
Mensah, G. E., Maseng, M. G., Allard, S. & Gilbert, J. A. in Precision Nutrition (eds Heber, D. et al.) Ch. 6 (Elsevier, 2024).
Quinn-Bohmann, N. et al. Microbial community-scale metabolic modelling predicts personalized short-chain fatty acid production profiles in the human gut. Nat. Microbiol. 9, 1700–1712 (2024).
Armstrong, G. et al. Swapping metagenomics preprocessing pipeline components offers speed and sensitivity increases. mSystems 7, e0137821 (2022).
Piperni, E. et al. Intestinal Blastocystis is linked to healthier diets and more favorable cardiometabolic outcomes in 56,989 individuals from 32 countries. Cell 187, 4554–4570 (2024).
Bermingham, K. M. et al. Effects of a personalized nutrition program on cardiometabolic health: a randomized controlled trial. Nat. Med. 30, 1888–1897 (2024).
Zeevi, D. et al. Personalized nutrition by prediction of glycemic responses. Cell 163, 1079–1094 (2015).
Wastyk, H. C. et al. Gut-microbiota-targeted diets modulate human immune status. Cell 184, 4137–4153 (2021).
Gilbert, J. A. & Lynch, S. V. Community ecology as a framework for human microbiome research. Nat. Med. 25, 884–889 (2019).
Sala, A. et al. A new phenotype in Candida–epithelial cell interaction distinguishes colonization- versus vulvovaginal candidiasis-associated strains. mBio 14, e0010723 (2023).
Abt, M. C. et al. Commensal bacteria calibrate the activation threshold of innate antiviral immunity. Immunity 37, 158–170 (2012).
Steed, A. L. et al. The microbial metabolite desaminotyrosine protects from influenza through type I interferon. Science 357, 498–502 (2017).
Erttmann, S. F. et al. The gut microbiota prime systemic antiviral immunity via the cGAS–STING–IFN-I axis. Immunity 55, 847–861 (2022).
Vasquez Ayala, A. et al. Commensal bacteria promote type I interferon signaling to maintain immune tolerance in mice. J. Exp. Med. 221, e20230063 (2024).
Glick, V. J. et al. Vaginal lactobacilli produce anti-inflammatory β-carboline compounds. Cell Host Microbe 32, 1897–1909 (2024).
Kwon, M. S. & Lee, H. K. Host and microbiome interplay shapes the vaginal microenvironment. Front. Immunol. 13, 919728 (2022).
Pramanick, R. et al. Vaginal microbiota of asymptomatic bacterial vaginosis and vulvovaginal candidiasis: are they different from normal microbiota? Microb. Pathog. 134, 103599 (2019).
McCauley, K. E. et al. Heritable vaginal bacteria influence immune tolerance and relate to early-life markers of allergic sensitization in infancy. Cell Rep. Med. 3, 100713 (2022).
Lebeer, S. et al. A citizen-science-enabled catalogue of the vaginal microbiome and associated factors. Nat. Microbiol. 8, 2183–2195 (2023).
Spencer, B. L. et al. Heterogeneity of the group B streptococcal type VII secretion system and influence on colonization of the female genital tract. Mol. Microbiol. 120, 258–275 (2023).
Heath, P. T. & Jardine, L. A. Neonatal infections: group B streptococcus. BMJ Clin. Evid. 2014, 0323 (2014).
Ni, J. et al. Early antibiotic exposure and development of asthma and allergic rhinitis in childhood. BMC Pediatr. 19, 225 (2019).
Chelimo, C., Camargo, C. A. Jr., Morton, S. M. B. & Grant, C. C. Association of repeated antibiotic exposure up to age 4 years with body mass at age 4.5 years. JAMA Netw. Open 3, e1917577 (2020).
Reid, G., Beuerman, D., Heinemann, C. & Bruce, A. W. Probiotic Lactobacillus dose required to restore and maintain a normal vaginal flora. FEMS Immunol. Med. Microbiol. 32, 37–41 (2001).
Lev-Sagie, A. et al. Vaginal microbiome transplantation in women with intractable bacterial vaginosis. Nat. Med. 25, 1500–1504 (2019).
Wrønding, T. et al. Antibiotic-free vaginal microbiota transplant with donor engraftment, dysbiosis resolution and live birth after recurrent pregnancy loss: a proof of concept case study. EClinicalMedicine 61, 102070 (2023).
Petricevic, L. et al. Effect of vaginal probiotics containing Lactobacillus casei rhamnosus (Lcr regenerans) on vaginal dysbiotic microbiota and pregnancy outcome, prospective, randomized study. Sci. Rep. 13, 7129 (2023).
Ali, A., Jørgensen, J. S. & Lamont, R. F. The contribution of bacteriophages to the aetiology and treatment of the bacterial vaginosis syndrome. Fac. Rev. 11, 8 (2022).
Landlinger, C. et al. Engineered phage endolysin eliminates Gardnerella biofilm without damaging beneficial bacteria in bacterial vaginosis ex vivo. Pathogens 10, 54 (2021).
Al-Anany, A. M. et al. Phage therapy in the management of urinary tract infections: a comprehensive systematic review. Phage 4, 112–127 (2023).
Bogaert, D. et al. Mother-to-infant microbiota transmission and infant microbiota development across multiple body sites. Cell Host Microbe 31, 447–460 (2023).
Jašarević, E. et al. The composition of human vaginal microbiota transferred at birth affects offspring health in a mouse model. Nat. Commun. 12, 6289 (2021).
Li, H.-T., Zhou, Y.-B. & Liu, J.-M. The impact of cesarean section on offspring overweight and obesity: a systematic review and meta-analysis. Int. J. Obes. 37, 893–899 (2013).
Kuhle, S., Tong, O. S. & Woolcott, C. G. Association between caesarean section and childhood obesity: a systematic review and meta‐analysis. Obes. Rev. 16, 295–303 (2015).
Yuan, C. et al. Association between cesarean birth and risk of obesity in offspring in childhood, adolescence, and early adulthood. JAMA Pediatr. 170, e162385 (2016).
Dominguez-Bello, M. G. et al. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc. Natl Acad. Sci. USA 107, 11971–11975 (2010).
Mueller, N. T. et al. Maternal bacterial engraftment in multiple body sites of cesarean section born neonates after vaginal seeding—a randomized controlled trial. mBio 14, e0049123 (2023).
Korpela, K. et al. Maternal fecal microbiota transplantation in cesarean-born infants rapidly restores normal gut microbial development: a proof-of-concept study. Cell 183, 324–334 (2020).
Dubois, L. et al. Paternal and induced gut microbiota seeding complement mother-to-infant transmission. Cell Host Microbe 32, 1011–1024 (2024).
Song, S. J. et al. Naturalization of the microbiota developmental trajectory of cesarean-born neonates after vaginal seeding. Med 2, 951–964 (2021).
Bode, L. et al. It’s alive: microbes and cells in human milk and their potential benefits to mother and infant. Adv. Nutr. 5, 571–573 (2014).
Xu, D. et al. Complement in breast milk modifies offspring gut microbiota to promote infant health. Cell 187, 750–763 (2024).
Johnson-Hence, C. B. et al. Stability and heterogeneity in the antimicrobiota reactivity of human milk-derived immunoglobulin A. J. Exp. Med. 220, e20220839 (2023).
Christian, P. et al. The need to study human milk as a biological system. Am. J. Clin. Nutr. 113, 1063–1072 (2021).
Shenhav, L. et al. Microbial colonization programs are structured by breastfeeding and guide healthy respiratory development. Cell 187, 5431–5452 (2024).
Henrick, B. M. et al. Bifidobacteria-mediated immune system imprinting early in life. Cell 184, 3884–3898 (2021).
Barratt, M. J. et al. Bifidobacterium infantis treatment promotes weight gain in Bangladeshi infants with severe acute malnutrition. Sci. Transl. Med. 14, eabk1107 (2022).
Nguyen, M. et al. Impact of probiotic B. infantis EVC001 feeding in premature infants on the gut microbiome, nosocomially acquired antibiotic resistance, and enteric inflammation. Front. Pediatr. 9, 618009 (2021).
van den Akker, C. H. P. et al. Reevaluating the FDA’s warning against the use of probiotics in preterm neonates: a societal statement by ESPGHAN and EFCNI. J. Pediatr. Gastroenterol. Nutr. 78, 1403–1408 (2024).
Wang, Y. et al. Probiotics, prebiotics, lactoferrin, and combination products for prevention of mortality and morbidity in preterm infants: a systematic review and network meta-analysis. JAMA Pediatr. 177, 1158–1167 (2023).
Gilbert, J. A. et al. Current understanding of the human microbiome. Nat. Med. 24, 392–400 (2018).
Gilbert, J. A. et al. Microbiome-wide association studies link dynamic microbial consortia to disease. Nature 535, 94–103 (2016).
Mullish, B. H. & Allegretti, J. R. The contribution of bile acid metabolism to the pathogenesis of Clostridioides difficile infection. Therap. Adv. Gastroenterol. 14, 17562848211017724 (2021).
Sinha, S. R. et al. Dysbiosis-induced secondary bile acid deficiency promotes intestinal inflammation. Cell Host Microbe 27, 659–670 (2020).
Feuerstadt, P. et al. SER-109, an oral microbiome therapy for recurrent Clostridioides difficile infection. N. Engl. J. Med. 386, 220–229 (2022).
Ben-Yacov, O. et al. Gut microbiome modulates the effects of a personalised postprandial-targeting (PPT) diet on cardiometabolic markers: a diet intervention in pre-diabetes. Gut 72, 1486–1496 (2023).
Culp, E. J., Nelson, N. T., Verdegaal, A. A. & Goodman, A. L. Microbial transformation of dietary xenobiotics shapes gut microbiome composition. Cell 187, 6327–6345 (2024).
She, J. et al. Statins aggravate insulin resistance through reduced blood glucagon-like peptide-1 levels in a microbiota-dependent manner. Cell Metab. 36, 408–421 (2024).
Wilmanski, T. et al. Heterogeneity in statin responses explained by variation in the human gut microbiome. Med 3, 388–405 (2022).
Lee, K. A. et al. Cross-cohort gut microbiome associations with immune checkpoint inhibitor response in advanced melanoma. Nat. Med. 28, 535–544 (2022).
Bolte, L. A. et al. Association of a Mediterranean diet with outcomes for patients treated with immune checkpoint blockade for advanced melanoma. JAMA Oncol. 9, 705–709 (2023).
Björk, J. R. et al. Longitudinal gut microbiome changes in immune checkpoint blockade-treated advanced melanoma. Nat. Med. 30, 785–796 (2024).
Abdelsalam, N. A., Hegazy, S. M. & Aziz, R. K. The curious case of Prevotella copri. Gut Microbes 15, 2249152 (2023).
Baldelli, V., Scaldaferri, F., Putignani, L. & Del Chierico, F. The role of Enterobacteriaceae in gut microbiota dysbiosis in inflammatory bowel diseases. Microorganisms 9, 697 (2021).
Mills, R. H. et al. Multi-omics analyses of the ulcerative colitis gut microbiome link Bacteroides vulgatus proteases with disease severity. Nat. Microbiol. 7, 262–276 (2022).
Federici, S. et al. Targeted suppression of human IBD-associated gut microbiota commensals by phage consortia for treatment of intestinal inflammation. Cell 185, 2879–2898 (2022).
Grubb, D. S. et al. PHAGE-2 Study: supplemental bacteriophages extend Bifidobacterium animalis subsp. lactis BL04 benefits on gut health and microbiota in healthy adults. Nutrients 12, 2474 (2020).
Li, X., Zhang, S., Guo, G., Han, J. & Yu, J. Gut microbiome in modulating immune checkpoint inhibitors. EBioMedicine 82, 104163 (2022).
Spencer, C. N. et al. Dietary fiber and probiotics influence the gut microbiome and melanoma immunotherapy response. Science 374, 1632–1640 (2021).
Davar, D. et al. Fecal microbiota transplant overcomes resistance to anti-PD-1 therapy in melanoma patients. Science 371, 595–602 (2021).
Baruch, E. N. et al. Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients. Science 371, 602–609 (2021).
Stein-Thoeringer, C. K. et al. A non-antibiotic-disrupted gut microbiome is associated with clinical responses to CD19-CAR-T cell cancer immunotherapy. Nat. Med. 29, 906–916 (2023).
Zhang, Y., Wang, Y., Zhang, B., Li, P. & Zhao, Y. Methods and biomarkers for early detection, prediction, and diagnosis of colorectal cancer. Biomed. Pharmacother. 163, 114786 (2023).
Fan, Y. & Pedersen, O. Gut microbiota in human metabolic health and disease. Nat. Rev. Microbiol. 19, 55–71 (2021).
Wu, C. et al. Obesity-enriched gut microbe degrades myo-inositol and promotes lipid absorption. Cell Host Microbe 32, 1301–1314 (2024).
DiNicolantonio, J. J. & O’Keefe, J. H. Myo-inositol for insulin resistance, metabolic syndrome, polycystic ovary syndrome and gestational diabetes. Open Heart 9, e001989 (2022).
Diener, C. et al. Baseline gut metagenomic functional gene signature associated with variable weight loss responses following a healthy lifestyle intervention in humans. mSystems 6, e0096421 (2021).
Corbin, L. J. et al. The metabolomic signature of weight loss and remission in the Diabetes Remission Clinical Trial (DiRECT). Diabetologia 67, 74–87 (2024).
Zhang, T. et al. Free fatty acid receptor 4 modulates dietary sugar preference via the gut microbiota. Nat. Microbiol. 10, 348–361 (2025).
Tan, S. et al. Interaction between the gut microbiota and colonic enteroendocrine cells regulates host metabolism. Nat. Metab. 6, 1076–1091 (2024).
Christensen, L. et al. Prevotella abundance and salivary amylase gene copy number predict fat loss in response to wholegrain diets. Front. Nutr. 9, 947349 (2022).
Christensen, L., Roager, H. M., Astrup, A. & Hjorth, M. F. Microbial enterotypes in personalized nutrition and obesity management. Am. J. Clin. Nutr. 108, 645–651 (2018).
Christensen, L. et al. Prevotella abundance predicts weight loss success in healthy, overweight adults consuming a whole-grain diet ad libitum: a post hoc analysis of a 6-wk randomized controlled trial. J. Nutr. 149, 2174–2181 (2019).
Mei, Z. et al. Strain-specific gut microbial signatures in type 2 diabetes identified in a cross-cohort analysis of 8,117 metagenomes. Nat. Med. 30, 2265–2276 (2024).
Goodrich, J. K. et al. Human genetics shape the gut microbiome. Cell 159, 789–799 (2014).
Ruaud, A. et al. Syntrophy via interspecies H2 transfer between Christensenella and Methanobrevibacter underlies their global cooccurrence in the human gut. mBio 11, e03235-19 (2020).
Waters, J. L. & Ley, R. E. The human gut bacteria Christensenellaceae are widespread, heritable, and associated with health. BMC Biol. 17, 83 (2019).
Akbuğa-Schön, T. et al. The keystone gut species Christensenella minuta boosts gut microbial biomass and voluntary physical activity in mice. mBio 15, e0283623 (2024).
Mazier, W. et al. A new strain of Christensenella minuta as a potential biotherapy for obesity and associated metabolic diseases. Cells 10, 823 (2021).
Ignatyeva, O. et al. Christensenella minuta, a new candidate next-generation probiotic: current evidence and future trajectories. Front. Microbiol. 14, 1241259 (2023).
Relizani, K. et al. Selection of a novel strain of Christensenella minuta as a future biotherapy for Crohn’s disease. Sci. Rep. 12, 6017 (2022).
Kropp, C. et al. Christensenella minuta protects and restores intestinal barrier in a colitis mouse model by regulating inflammation. npj Biofilms Microbiomes 10, 88 (2024).
Zahavi, L. et al. Bacterial SNPs in the human gut microbiome associate with host BMI. Nat. Med. 29, 2785–2792 (2023).
Kurt, Ö., Doğruman Al, F. & Tanyüksel, M. Eradication of Blastocystis in humans: really necessary for all? Parasitol. Int. 65, 797–801 (2016).
Li, H. et al. Resistant starch intake facilitates weight loss in humans by reshaping the gut microbiota. Nat. Metab. 6, 578–597 (2024).
Van Hul, M. & Cani, P. D. The gut microbiota in obesity and weight management: microbes as friends or foe? Nat. Rev. Endocrinol. 19, 258–271 (2023).
Maifeld, A. et al. Fasting alters the gut microbiome reducing blood pressure and body weight in metabolic syndrome patients. Nat. Commun. 12, 1970 (2021).
Koutoukidis, D. A. et al. The association of weight loss with changes in the gut microbiota diversity, composition, and intestinal permeability: a systematic review and meta-analysis. Gut Microbes 14, 2020068 (2022).
Wolter, M. et al. Diet-driven differential response of Akkermansia muciniphila modulates pathogen susceptibility. Mol. Syst. Biol. 20, 596–625 (2024).
Zhang, Z. et al. Impact of fecal microbiota transplantation on obesity and metabolic syndrome—a systematic review. Nutrients 11, 2291 (2019).
Thaiss, C. A. et al. Persistent microbiome alterations modulate the rate of post-dieting weight regain. Nature 540, 544–551 (2016).
Tsai, C.-Y. et al. Gut microbial signatures for glycemic responses of GLP-1 receptor agonists in type 2 diabetic patients: a pilot study. Front. Endocrinol. 12, 814770 (2021).
Chen, R. Y. et al. A microbiota-directed food intervention for undernourished children. N. Engl. J. Med. 384, 1517–1528 (2021).
Chang, H.-W. et al. Prevotella copri and microbiota members mediate the beneficial effects of a therapeutic food for malnutrition. Nat. Microbiol. 9, 922–937 (2024).
Cheng, J., Venkatesh, S., Ke, K., Barratt, M. J. & Gordon, J. I. A human gut Faecalibacterium prausnitzii fatty acid amide hydrolase. Science 386, eado6828 (2024).
Ecklu-Mensah, G. et al. Gut microbiota and fecal short chain fatty acids differ with adiposity and country of origin: the METS-microbiome study. Nat. Commun. 14, 5160 (2023).
Suez, J. et al. Artificial sweeteners induce glucose intolerance by altering the gut microbiota. Nature 514, 181–186 (2014).
Suez, J. et al. Personalized microbiome-driven effects of non-nutritive sweeteners on human glucose tolerance. Cell 185, 3307–3328 (2022).
Rein, M. et al. Effects of personalized diets by prediction of glycemic responses on glycemic control and metabolic health in newly diagnosed T2DM: a randomized dietary intervention pilot trial. BMC Med. 20, 56 (2022).
Perraudeau, F. et al. Improvements to postprandial glucose control in subjects with type 2 diabetes: a multicenter, double blind, randomized placebo-controlled trial of a novel probiotic formulation. BMJ Open Diabetes Res. Care 8, e001319 (2020).
Wang, C.-H. et al. Adjuvant probiotics of Lactobacillus salivarius subsp. salicinius AP-32, L. johnsonii MH-68, and Bifidobacterium animalis subsp. lactis CP-9 attenuate glycemic levels and inflammatory cytokines in patients with type 1 diabetes mellitus. Front. Endocrinol. 13, 754401 (2022).
Kumar, S. et al. A high potency multi‐strain probiotic improves glycemic control in children with new‐onset type 1 diabetes mellitus: a randomized, double‐blind, and placebo‐controlled pilot study. Pediatr. Diabetes 22, 1014–1022 (2021).
Savilahti, E. et al. Probiotic intervention in infancy is not associated with development of beta cell autoimmunity and type 1 diabetes. Diabetologia 61, 2668–2670 (2018).
Moravejolahkami, A. R., Shakibaei, M., Fairley, A. M. & Sharma, M. Probiotics, prebiotics, and synbiotics in type 1 diabetes mellitus: a systematic review and meta-analysis of clinical trials. Diabetes Metab. Res. Rev. 40, e3655 (2024).
Groele, L. et al. Lack of effect of Lactobacillus rhamnosus GG and Bifidobacterium lactis Bb12 on beta-cell function in children with newly diagnosed type 1 diabetes: a randomised controlled trial. BMJ Open Diabetes Res. Care 9, e001523 (2021).
Shabani-Mirzaee, H. et al. The effect of oral probiotics on glycated haemoglobin levels in children with type 1 diabetes mellitus — a randomized clinical trial. Pediatr. Endocrinol. Diabetes Metab. 29, 128–133 (2023).
Ali, Z., Ma, H., Ayim, I. & Wali, A. Efficacy of new beverage made of dates vinegar and garlic juice in improving serum lipid profile parameters and inflammatory biomarkers of mildly hyperlipidemic adults: a double-blinded, randomized, placebo-controlled study. J. Food Biochem. 42, e12545 (2018).
Johnston, C. S., Kim, C. M. & Buller, A. J. Vinegar improves insulin sensitivity to a high-carbohydrate meal in subjects with insulin resistance or type 2 diabetes. Diabetes Care 27, 281–282 (2004).
Ali, Z., Ma, H., Wali, A., Ayim, I. & Sharif, M. N. Daily date vinegar consumption improves hyperlipidemia, β-carotenoid and inflammatory biomarkers in mildly hypercholesterolemic adults. J. Herb. Med. 17–18, 100265 (2019).
Armstrong, H. K. et al. Unfermented β-fructan fibers fuel inflammation in select inflammatory bowel disease patients. Gastroenterology 164, 228–240 (2023).
Arifuzzaman, M. et al. Dietary fiber is a critical determinant of pathologic ILC2 responses and intestinal inflammation. J. Exp. Med. 221, e20232148 (2024).
Girard, C., Tromas, N., Amyot, M. & Shapiro, B. J. Gut microbiome of the Canadian Arctic Inuit. mSphere 2, e00297-16 (2017).
Campbell, R., Hauptmann, A., Campbell, K., Fox, S. & Marco, M. L. Better understanding of food and human microbiomes through collaborative research on Inuit fermented foods. Microbiome Res. Rep. 1, 5 (2022).
Jones, S. An ethical way forward for Indigenous microbiome research. Nature https://doi.org/10.1038/d41586-024-02792-w (2024).
Jensen, N. et al. Dietary fiber monosaccharide content alters gut microbiome composition and fermentation. Appl. Environ. Microbiol. 90, e00964-24 (2024).
Berry, S. E. et al. Human postprandial responses to food and potential for precision nutrition. Nat. Med. 26, 964–973 (2020).
Abed, J. et al. Fap2 mediates Fusobacterium nucleatum colorectal adenocarcinoma enrichment by binding to tumor-expressed Gal-GalNAc. Cell Host Microbe 20, 215–225 (2016).
Yang, Y. et al. Fusobacterium nucleatum increases proliferation of colorectal cancer cells and tumor development in mice by activating Toll-like receptor 4 signaling to nuclear factor-κB, and up-regulating expression of microRNA-21. Gastroenterology 152, 851–866 (2017).
Ternes, D. et al. The gut microbial metabolite formate exacerbates colorectal cancer progression. Nat. Metab. 4, 458–475 (2022).
Komiya, Y. et al. Patients with colorectal cancer have identical strains of Fusobacterium nucleatum in their colorectal cancer and oral cavity. Gut 68, 1335–1337 (2019).
Zepeda-Rivera, M. et al. A distinct Fusobacterium nucleatum clade dominates the colorectal cancer niche. Nature 628, 424–432 (2024).
Kerns, K. A. et al. Localized microbially induced inflammation influences distant healthy tissues in the human oral cavity. Proc. Natl Acad. Sci. USA 120, e2306020120 (2023).
Grodner, B. et al. Spatial mapping of mobile genetic elements and their bacterial hosts in complex microbiomes. Nat. Microbiol. 9, 2262–2277 (2024).
Surma, S. et al. Periodontitis, blood pressure, and the risk and control of arterial hypertension: epidemiological, clinical, and pathophysiological aspects—review of the literature and clinical trials. Curr. Hypertens. Rep. 23, 27 (2021).
Huang, S. et al. Predictive modeling of gingivitis severity and susceptibility via oral microbiota. ISME J. 8, 1768–1780 (2014).
O’Dwyer, D. N. et al. Commensal oral microbiota, disease severity, and mortality in fibrotic lung disease. Am. J. Respir. Crit. Care Med. 209, 1101–1110 (2024).
Jemimah, S., Chabib, C. M. M., Hadjileontiadis, L. & AlShehhi, A. Gut microbiome dysbiosis in Alzheimer’s disease and mild cognitive impairment: a systematic review and meta-analysis. PLoS ONE 18, e0285346 (2023).
Naughten, S. et al. The re-emerging role of linoleic acid in paediatric asthma. Eur. Respir. Rev. 32, 230063 (2023).
Stewart, C. J. Homing in on 12,13-diHOME in asthma. Nat. Microbiol. 4, 1774–1775 (2019).
Lin, D. L. et al. 12,13-diHOME promotes inflammatory macrophages and epigenetically modifies their capacity to respond to microbes and allergens. J. Immunol. Res. 2024, 2506586 (2024).
Levan, S. R. et al. Elevated faecal 12,13-diHOME concentration in neonates at high risk for asthma is produced by gut bacteria and impedes immune tolerance. Nat. Microbiol. 4, 1851–1861 (2019).
Fujimura, K. E. et al. Neonatal gut microbiota associates with childhood multisensitized atopy and T cell differentiation. Nat. Med. 22, 1187–1191 (2016).
Yan, Z. et al. Multi-omics analyses of airway host–microbe interactions in chronic obstructive pulmonary disease identify potential therapeutic interventions. Nat. Microbiol. 7, 1361–1375 (2022).
Mitropoulou, G. et al. Phage therapy for pulmonary infections: lessons from clinical experiences and key considerations. Eur. Respir. Rev. 31, 220121 (2022).
Nakatsuji, T., Cheng, J. Y. & Gallo, R. L. Mechanisms for control of skin immune function by the microbiome. Curr. Opin. Immunol. 72, 324–330 (2021).
Sanford, J. A., O’Neill, A. M., Zouboulis, C. C. & Gallo, R. L. Short-chain fatty acids from Cutibacterium acnes activate both a canonical and epigenetic inflammatory response in human sebocytes. J. Immunol. 202, 1767–1776 (2019).
Lai, Y. et al. Commensal bacteria regulate Toll-like receptor 3-dependent inflammation after skin injury. Nat. Med. 15, 1377–1382 (2009).
Nakatsuji, T. et al. Development of a human skin commensal microbe for bacteriotherapy of atopic dermatitis and use in a phase 1 randomized clinical trial. Nat. Med. 27, 700–709 (2021).
Myles, I. A. et al. Therapeutic responses to Roseomonas mucosa in atopic dermatitis may involve lipid-mediated TNF-related epithelial repair. Sci. Transl. Med. 12, eaaz8631 (2020).
Guéniche, A. et al. Improvement of atopic dermatitis skin symptoms by Vitreoscilla filiformis bacterial extract. Eur. J. Dermatol. 16, 380–384 (2006).
Shen, Z. et al. A genome catalog of the early-life human skin microbiome. Genome Biol. 24, 252 (2023).
Farias Amorim, C. et al. Multiomic profiling of cutaneous leishmaniasis infections reveals microbiota-driven mechanisms underlying disease severity. Sci. Transl. Med. 15, eadh1469 (2023).
Oh, J., Robison, J. & Kuchel, G. A. Frailty-associated dysbiosis of human microbiotas in older adults in nursing homes. Nat. Aging 2, 876–877 (2022).
Larson, P. J. et al. Associations of the skin, oral and gut microbiome with aging, frailty and infection risk reservoirs in older adults. Nat. Aging 2, 941–955 (2022).
Sulakvelidze, A., Alavidze, Z. & Morris, J. G. Jr. Bacteriophage therapy. Antimicrob. Agents Chemother. 45, 649–659 (2001).
Pirnay, J.-P. et al. Personalized bacteriophage therapy outcomes for 100 consecutive cases: a multicentre, multinational, retrospective observational study. Nat. Microbiol. 9, 1434–1453 (2024).
He, Y. et al. Regional variation limits applications of healthy gut microbiome reference ranges and disease models. Nat. Med. 24, 1532–1535 (2018).
Olsson, L. M. et al. Dynamics of the normal gut microbiota: a longitudinal one-year population study in Sweden. Cell Host Microbe 30, 726–739 (2022).
Zaramela, L. S., Tjuanta, M., Moyne, O., Neal, M. & Zengler, K. SynDNA — a synthetic DNA spike-in method for absolute quantification of shotgun metagenomic sequencing. mSystems 7, e0044722 (2022).
Uyttebroek, S. et al. Safety and efficacy of phage therapy in difficult-to-treat infections: a systematic review. Lancet Infect. Dis. 22, e208–e220 (2022).
Zmora, N. et al. Personalized gut mucosal colonization resistance to empiric probiotics is associated with unique host and microbiome features. Cell 174, 1388–1405 (2018).
Suez, J. et al. Post-antibiotic gut mucosal microbiome reconstitution is impaired by probiotics and improved by autologous FMT. Cell 174, 1406–1423 (2018).
Montassier, E. et al. Probiotics impact the antibiotic resistance gene reservoir along the human GI tract in a person-specific and antibiotic-dependent manner. Nat. Microbiol. 6, 1043–1054 (2021).
Shalon, D. et al. Profiling the human intestinal environment under physiological conditions. Nature 617, 581–591 (2023).
Schmidt, F. et al. Noninvasive assessment of gut function using transcriptional recording sentinel cells. Science 376, eabm6038 (2022).
Crossette, E. et al. Metagenomic quantification of genes with internal standards. mBio 12, e03173-20 (2021).
Chiu, C. Y. & Miller, S. A. Clinical metagenomics. Nat. Rev. Genet. 20, 341–355 (2019).
Wilson, M. R. et al. Clinical metagenomic sequencing for diagnosis of meningitis and encephalitis. N. Engl. J. Med. 380, 2327–2340 (2019).
Gu, W. et al. Rapid pathogen detection by metagenomic next-generation sequencing of infected body fluids. Nat. Med. 27, 115–124 (2021).
Miller, S. et al. Laboratory validation of a clinical metagenomic sequencing assay for pathogen detection in cerebrospinal fluid. Genome Res. 29, 831–842 (2019).
Blauwkamp, T. A. et al. Analytical and clinical validation of a microbial cell-free DNA sequencing test for infectious disease. Nat. Microbiol. 4, 663–674 (2019).
Tan, J. K. et al. Laboratory validation of a clinical metagenomic next-generation sequencing assay for respiratory virus detection and discovery. Nat. Commun. 15, 9016 (2024).
Benoit, P. et al. Seven-year performance of a clinical metagenomic next-generation sequencing test for diagnosis of central nervous system infections. Nat. Med. 30, 3522–3533 (2024).
Langelier, C. et al. Integrating host response and unbiased microbe detection for lower respiratory tract infection diagnosis in critically ill adults. Proc. Natl Acad. Sci. USA 115, E12353–E12362 (2018).
Ma, P. et al. Bacterial droplet-based single-cell RNA-seq reveals antibiotic-associated heterogeneous cellular states. Cell 186, 877–891 (2023).
Kuchina, A. et al. Microbial single-cell RNA sequencing by split-pool barcoding. Science 371, eaba5257 (2021).
Lötstedt, B., Stražar, M., Xavier, R., Regev, A. & Vickovic, S. Spatial host–microbiome sequencing reveals niches in the mouse gut. Nat. Biotechnol. 42, 1394–1403 (2023).
McNulty, R. et al. Probe-based bacterial single-cell RNA sequencing predicts toxin regulation. Nat. Microbiol. 8, 934–945 (2023).
Valdés-Mas, R. et al. Metagenome-informed metaproteomics of the human gut microbiome, host, and dietary exposome uncovers signatures of health and inflammatory bowel disease. Cell 188, 1062–1083 (2025).
Duncan, S. H., Conti, E., Ricci, L. & Walker, A. W. Links between diet, intestinal anaerobes, microbial metabolites and health. Biomedicines 11, 1338 (2023).
Beresford-Jones, B. S. et al. The Mouse Gastrointestinal Bacteria Catalogue enables translation between the mouse and human gut microbiotas via functional mapping. Cell Host Microbe 30, 124–138 (2022).
Zaramela, L. S. et al. The sum is greater than the parts: exploiting microbial communities to achieve complex functions. Curr. Opin. Biotechnol. 67, 149–157 (2021).
Bolyen, E. et al. Reproducible, interactive, scalable and extensible microbiome data science using QIIME 2. Nat. Biotechnol. 37, 852–857 (2019).
Louie, T. et al. VE303, a defined bacterial consortium, for prevention of recurrent Clostridioides difficile infection. JAMA 329, 1356–1366 (2023).
Ponce, D. M. et al. Safety and efficacy results from a randomized, double-blind, placebo-controlled cohort 2 of a phase 1b study of an investigational live biotherapeutic, SER-155, in adults undergoing allo-HCT. Transplant. Cell. Ther. 31, S86 (2025).
Abbott, M. & Ustoyev, Y. Cancer and the immune system: the history and background of immunotherapy. Semin. Oncol. Nurs. 35, 150923 (2019).
Karikó, K., Buckstein, M., Ni, H. & Weissman, D. Suppression of RNA recognition by Toll-like receptors: the impact of nucleoside modification and the evolutionary origin of RNA. Immunity 23, 165–175 (2005).
Khanna, S. et al. A novel microbiome therapeutic increases gut microbial diversity and prevents recurrent Clostridium difficile infection. J. Infect. Dis. 214, 173–181 (2016).
McGovern, B. H. et al. SER-109, an investigational microbiome drug to reduce recurrence after Clostridioides difficile infection: lessons learned from a phase 2 trial. Clin. Infect. Dis. 72, 2132–2140 (2021).
Sims, M. D. et al. Safety and tolerability of SER-109 as an investigational microbiome therapeutic in adults with recurrent Clostridioides difficile infection: a phase 3, open-label, single-arm trial. JAMA Netw. Open 6, e2255758 (2023).
Straub, T. J. et al. Impact of a purified microbiome therapeutic on abundance of antimicrobial resistance genes in patients with recurrent Clostridioides difficile infection. Clin. Infect. Dis. 78, 833–841 (2024).
Khanna, S. et al. SER-109: an oral investigational microbiome therapeutic for patients with recurrent Clostridioides difficile infection (rCDI). Antibiotics 11, 1234 (2022).
Jain, N., Umar, T. P., Fahner, A.-F. & Gibietis, V. Advancing therapeutics for recurrent Clostridioides difficile infections: an overview of Vowst’s FDA approval and implications. Gut Microbes 15, 2232137 (2023).
Food and Drug Administration. Early Clinical Trials with Live Biotherapeutic Products: Chemistry, Manufacturing, and Control Information. FDA www.fda.gov/media/82945/download (2016).
Food and Drug Administration. Enforcement Policy Regarding Investigational New Drug Requirements for Use of Fecal Microbiota for Transplantation to Treat Clostridioides difficile Infection Not Responsive to Standard Therapies. FDA www.fda.gov/media/86440/download (2022).
van Nood, E. et al. Duodenal infusion of donor feces for recurrent Clostridium difficile. N. Engl. J. Med. 368, 407–415 (2013).
Green, J. E. et al. Efficacy and safety of fecal microbiota transplantation for the treatment of diseases other than Clostridium difficile infection: a systematic review and meta-analysis. Gut Microbes 12, 1–25 (2020).
Food and Drug Administration. Fecal Microbiota Products. FDA www.fda.gov/vaccines-blood-biologics/fecal-microbiota-products (2024).
Canada’s Drug Agency. Fecal microbiota therapy in Canada: an environmental scan. CDA FMT www.cda-amc.ca/fecal-microbiota-therapy-canada-environmental-scan#:~:text=FMT%20is%20approved%20in%20Canada,clinical%20criteria%20to%20receive%20treatment (2024).
Australian Department of Health and Aged Care. Faecal Microbiota Transplant Products Regulation. Understand How We Regulate Faecal Microbiota Transplant (FMT) Products www.tga.gov.au/products/biologicals-blood-and-tissues-and-advanced-therapies/biologicals/faecal-microbiota-transplant-products-regulation (2023).
Lee, C. et al. Safety of fecal microbiota, live-jslm (REBYOTA™) in individuals with recurrent Clostridioides difficile infection: data from five prospective clinical trials. Therap. Adv. Gastroenterol. 16, 17562848231174276 (2023).
Microbiome Therapeutics Innovation Group. Navigating regulatory and analytical challenges in live biotherapeutic product development and manufacturing. Front. Microbiomes https://doi.org/10.3389/frmbi.2024.1441290 (2024).
Author information
Authors and Affiliations
Corresponding author
Ethics declarations
Competing interests
J.A.G. is associated with Holobiome, BiomeSense, Bened Life and Sun Genomics. C.Y.C. is associated with Delve Bio and receives research support from Abbott Laboratories. P.S. is associated with Holobiome. E.E. and E.S. are associated with DayTwo. M.R.H. is associated with Seres Therapeutics. T.D.S. is associated with ZOE. S.V.L. is associated with Siolta Therapeutics. M.J.B. is associated with Seed Health, ProDermIQ and Elysium Health. M.B.A. is associated with Tiny Health. R.K. is associated with BiomeSense, GenCirq, DayTwo, Cybele and Biota.
Peer review
Peer review information
Nature Medicine thanks the anonymous reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Karen O’Leary, in collaboration with the Nature Medicine team.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Gilbert, J.A., Azad, M.B., Bäckhed, F. et al. Clinical translation of microbiome research. Nat Med 31, 1099–1113 (2025). https://doi.org/10.1038/s41591-025-03615-9
Received:
Accepted:
Published:
Issue date:
DOI: https://doi.org/10.1038/s41591-025-03615-9
This article is cited by
-
Multifaceted effects of the microbiome in pancreatic cancer: from association to modulation
Nature Reviews Gastroenterology & Hepatology (2025)
-
Distinguishing the causative, correlative and bidirectional roles of the gut microbiota in mental health
Nature Mental Health (2025)
-
Call for papers on the clinical microbiome
Nature Microbiology (2025)
-
Nutritional Status and Fecal Microbiota in School Children from the Galapagos and the Andean Region
Microbial Ecology (2025)
-
Microbial proteases as emerging anti-inflammatory therapeutics: a comprehensive review
Archives of Microbiology (2025)