Next Article in Journal
In Vitro Synergy Evaluation of Trimethoprim/Sulfamethoxazole Combined with Levofloxacin and Ceftazidime Against Stenotrophomonas maltophilia: A Comparative Study Using Checkerboard and Gradient Diffusion Methods
Previous Article in Journal
Pancreatic Stone Protein: A Multifaceted Biomarker—A Comprehensive Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Microbiome-Targeted Therapies in Gastrointestinal Diseases: Clinical Evidence and Emerging Innovations

by
Enoch Chi Ngai Lim
1 and
Chi Eung Danforn Lim
1,2,3,*
1
Translational Research Department, Specialist Medical Services Group, Earlwood, NSW 2206, Australia
2
School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia
3
NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia
*
Author to whom correspondence should be addressed.
Acta Microbiol. Hell. 2025, 70(3), 36; https://doi.org/10.3390/amh70030036
Submission received: 9 August 2025 / Revised: 30 August 2025 / Accepted: 10 September 2025 / Published: 13 September 2025

Abstract

Microbiome-targeted therapies are redefining gastroenterology by delivering precision interventions that align with the body’s natural microbial ecosystem. This narrative review evaluates evidence for established approaches, probiotics, prebiotics, fecal microbiota transplantation (FMT), and postbiotics, and examines emerging innovations such as engineered probiotics, bacteriophage therapy, and metabolite-based interventions. Cure rates for recurrent Clostridium difficile infection in randomized trials range from 67% to 94%, depending on route and donor protocol, while multi-strain probiotics provide moderate benefits in inflammatory bowel disease. New modalities, including engineered bacteria and defined bacterial consortia, have progressed to Phase 3 trials, with several granted FDA breakthrough therapy designation. Approvals of Rebyota and Vowst mark a pivotal milestone, creating validated regulatory pathways for microbiome therapeutics. Despite progress, challenges remain in protocol standardisation, patient selection, cost-effectiveness, and clinical integration. Over 200 active trials and growing pharmaceutical investment signal a robust pipeline, with applications expanding to oncology, metabolic disorders, and immune modulation. Continued progress depends on validated biomarkers and personalized strategies guided by microbiome profiling. International regulatory harmonization will also be required to ensure safe and equitable adoption. The field is shifting toward working with, rather than against, the body’s microbial ecosystem, offering substantial potential for personalized gastrointestinal disease management.

1. Introduction

The human gastrointestinal tract harbours trillions of microorganisms that profoundly influence health and disease through complex interactions with host physiology, metabolism, and immune function [1]. Disruptions in this delicate microbial ecosystem, known as dysbiosis, contribute to a range of gastrointestinal disorders, from functional conditions to inflammatory diseases and infections [1]. Traditional therapeutic approaches often fail to address underlying microbial imbalances, leading to symptom recurrence and treatment resistance. Microbiome-targeted therapies offer a paradigm shift toward precision interventions that restore microbial balance and enhance therapeutic outcomes [2]. These approaches range from established interventions, such as probiotics and fecal microbiota transplantation, to cutting-edge innovations, including engineered bacteria and metabolite-based therapies [2]. Recent U.S. Food and Drug Administration (FDA) approvals of the first microbiome-derived pharmaceuticals mark a pivotal transition from experimental treatments to validated clinical interventions.
This review aims to provide a comprehensive analysis of current clinical evidence for microbiome-targeted therapies across major gastrointestinal diseases, examine emerging therapeutic innovations, and evaluate practical considerations for clinical implementation. We focus on clinical outcomes data, comparative effectiveness studies, and real-world implementation experiences while highlighting future directions in this rapidly advancing field.

2. Methods

This narrative review was conducted as a narrative synthesis of recent clinical and translational evidence. Structured literature searches were carried out across PubMed/MEDLINE, Cochrane, ClinicalTrials.gov, EMBASE, Web of Science, and Scopus for the period January 2015 to August 2025. The following combined search terms were used: “microbiome”, “gut microbiota”, “probiotics”, “prebiotics”, “synbiotics”, “postbiotics”, “fecal microbiota transplantation”, “FMT”, “engineered probiotics”, “bacteriophage”, “metabolite therapy”, “gastrointestinal disease”, “inflammatory bowel disease”, “irritable bowel syndrome”, “Clostridioides difficile”, “non-alcoholic fatty liver disease”, “NAFLD”, “NASH”, “Helicobacter pylori”.
Additional disease-specific terms were added to refine searches. Reference lists of relevant systematic reviews and meta-analyses were hand-searched to identify further eligible studies. Priority was given to randomized controlled trials, systematic reviews, meta-analyses, regulatory documents (FDA, EMA), and guideline statements from major gastroenterology societies.
Inclusion criteria were: peer-reviewed English-language publications, clinical trials and reviews reporting patient outcomes, and regulatory or guideline documents addressing microbiome-targeted therapies. Exclusion criteria were: case reports, single-patient studies, preclinical-only studies, conference abstracts, and non-English publications.
Evidence was synthesized qualitatively, with emphasis on clinical relevance, magnitude of effect, and translational implications. Where available, pooled estimates from high-quality meta-analyses and regulatory submissions were prioritized to reflect evidence strength.

3. Established Therapeutic Approaches

3.1. Fecal Microbiota Transplantation Leads to Clinical Efficacy

Fecal microbiota transplantation represents the most clinically validated microbiome intervention, with robust evidence across multiple conditions. The most compelling results emerge from C. difficile infection treatment, where FMT demonstrates superior clinical outcomes compared to standard antibiotic therapies [1,2]. Meta-analyses demonstrate that fecal microbiota transplantation (FMT) is highly effective for the treatment of recurrent and refractory Clostridium difficile infection. In a systematic review and meta-analysis, Quraishi et al. [3] reported markedly higher cure rates with FMT compared to conventional antibiotic therapy. Similarly, Fischer et al. [4] found FMT to be both safe and efficacious for patients with inflammatory bowel disease who developed recurrent or refractory C. difficile infection. Cure rates for recurrent Clostridium difficile infection (CDI) in randomized trials range from 67% to 94%, depending on delivery route and donor protocol [1,3]. Most studies define cure as absence of recurrence within 8–12 weeks. Meta-analyses report an absolute risk reduction of about 30% compared with vancomycin, yielding a number needed to treat between 3 and 5 [2,3].
Recent randomised controlled trials have provided further insight into microbiome-based therapies. In the ECOSPOR III trial, Feuerstadt et al. [5] demonstrated that the investigational oral microbiome therapeutic SER-109 significantly reduced the risk of C. difficile recurrence compared with placebo. Clinical guidelines from the American College of Gastroenterology summarise these and other high-quality studies, underscoring the role of FMT in current practice [6]. Safety profiles remain favourable across studies. Wang et al. [7] reported serious adverse events in fewer than 11% of patients receiving FMT, with rates similar to those observed in other gastrointestinal interventions. The most commonly reported side effects include transient abdominal discomfort, diarrhoea, and bloating. Following documented cases of pathogen transmission during FMT, including transmission of drug-resistant Escherichia coli described by DeFilipp et al. [8], enhanced donor screening protocols have been implemented to further mitigate risk.

3.2. Probiotics and Condition-Specific Benefits

Probiotics demonstrate variable efficacy across gastrointestinal conditions, with the strongest signals in specific clinical scenarios. In irritable bowel syndrome, a systematic review and meta-analysis of 53 randomised controlled trials involving 5545 patients found that selected probiotics improve global symptoms and abdominal pain. However, conclusions were limited by heterogeneity and variable certainty [9]. Efficacy is strain- and disease-specific. For IBS, Lactobacillus plantarum 299v and Bifidobacterium infantis 35624 have the most consistent supportive evidence, while results for other single-strain and multi-strain products are mixed [10].
For inflammatory bowel disease, meta-analysis indicates that some probiotics, particularly multi-strain formulations such as VSL#3, can be beneficial in ulcerative colitis, although effect sizes and certainty vary, and optimal dosing is not established by the included studies [11]. For maintenance of remission in ulcerative colitis, a Cochrane review concluded that overall effectiveness remains uncertain due to low to very low-certainty evidence [12]. In pouchitis after ileal pouch–anal anastomosis, the evidence base for probiotics and other microbiome-targeted options is limited and uncertain, and available reviews do not provide a robust ranking that places multi-strain probiotics among the top options for preventing clinical relapse [13]. Substantial heterogeneity exists across trials due to differences in probiotic strain, dose, treatment duration, and outcome measures. These factors limit pooled estimates and explain divergent conclusions. Null findings occur frequently and should be considered alongside positive results to avoid overstating efficacy.

3.3. Prebiotics and Synbiotics Expand Therapeutic Options

Prebiotic interventions, defined as substrates selectively utilised by host microorganisms conferring a health benefit, have shown benefits in a variety of conditions, including functional gastrointestinal disorders. Evidence supports their role in modulating specific microbial metabolic pathways, though clinical outcomes vary across studies [14]. Synbiotic formulations, which combine probiotics with prebiotic substrates, are designed to enhance probiotic survival and colonisation in the gut. Limited emerging evidence suggests they may offer advantages over probiotic use alone. However, current research is constrained by heterogeneous study designs and the absence of standardised protocols and outcome measures [15].

4. Disease-Specific Clinical Applications

4.1. Inflammatory Bowel Disease Treatment Advances

Inflammatory bowel disease is a priority area for microbiome interventions, but current guideline recommendations are conservative. The AGA advises that probiotics for ulcerative colitis or Crohn’s disease should be used only within clinical trials, with a conditional (very low-certainty) suggestion limited to a specific 8-strain formulation in pouchitis [16]. A meta-analysis of Crohn’s disease found no significant benefit of probiotics in maintaining remission, underscoring that their applications in this setting remain investigational [17]. Randomised trials of FMT in ulcerative colitis demonstrate that multidonor, intensive-dosing regimens can induce remission in a subset of patients. In Paramsothy et al., 2017, steroid-free clinical remission with endoscopic improvement at 8 weeks was achieved in about 27% of patients receiving FMT versus ~8% receiving placebo [18]. Multidonor, intensive regimens have induced remission in about 25–30% of patients [18]. However, reproducibility is limited and optimal protocols remain undefined. Protocol heterogeneity continues to limit conclusions on optimal regimens. Beyond IBD, FMT has also been explored in other gastrointestinal disorders, such as slow transit constipation, where a small pilot study suggested potential symptomatic benefits [19]. These findings illustrate the broader therapeutic interest in microbiome-directed approaches while highlighting the need for standardised protocols and validated outcome measures before routine clinical use in IBD [16,17,18,19]. Despite some positive findings in ulcerative colitis and pouchitis, the evidence for probiotics in IBD remains of low certainty, with small sample sizes, heterogeneous trial designs, and limited reproducibility reducing confidence in their clinical utility.

4.2. Clostridium Difficile Infection Management

C. difficile infection is the flagship indication for microbiome therapeutics, as reflected by regulatory approvals and clinical guideline recommendations. For primary prevention, a Cochrane review found that probiotics reduce the risk of antibiotic-associated C. difficile-associated diarrhoea in adults and children, though evidence on prevention of recurrence was insufficient [20]. FDA-approved microbiome-based products show consistent efficacy in preventing recurrence: Vowst reduced 8-week recurrence to 12.4% compared with 39.8% for placebo [21], and Rebyota achieved treatment success at 8 weeks in 70.6% of recipients versus 57.5% with placebo [22]. Current IDSA/SHEA treatment guidelines recommend FMT for multiply recurrent CDI after failure of appropriate antibiotic regimens, with fidaxomicin and vancomycin as standard antimicrobial options [23].

4.3. Irritable Bowel Syndrome Symptom Management

IBS is a heterogeneous condition, and microbiome interventions show variable but sometimes meaningful benefits. Recent randomised controlled trials, summarised in a systematic review, found that certain probiotic regimens improve IBS symptoms compared with a placebo, with multi-strain formulations tending to perform better than single-strain products. However, effect sizes and study designs varied [24]. Efficacy is strain-specific. An evidence-based international guide identified particular preparations with supportive data for IBS outcomes, including specific Bifidobacterium strains such as B. infantis 35624 for global symptoms and abdominal pain; this review did not assess Saccharomyces cerevisiae I-3856 [25].
Regarding treatment duration, trials reviewed in [24] more often demonstrated significant improvements with interventions lasting at least eight weeks, suggesting that benefits may require sustained administration rather than very short courses [26]. Quantitative pooled estimates vary across analyses. An earlier systematic review reported a relative risk of persistent global symptoms of approximately 0.71 for probiotics versus placebo, corresponding to a number needed to treat of approximately four; however, heterogeneity was substantial, and optimal strains or doses could not be identified [26]. Safety profiles for probiotics remain favourable. Large-scale meta-analyses in other gastrointestinal contexts, such as antibiotic-associated diarrhea, have found no significant increase in adverse events compared with placebo, supporting their overall tolerability [27]. Despite this safety profile, evidence of clinical efficacy in IBS remains low certainty because of small sample sizes, heterogeneous trial designs, and limited reproducibility.

4.4. Metabolic Liver Disease Interventions

Non-alcoholic fatty liver disease (NAFLD) is emerging as a target for microbiome-based interventions, with growing but still limited clinical evidence. Reviews of clinical studies report improvements in liver enzymes, inflammatory markers, and imaging-based assessments of hepatic steatosis and stiffness in some probiotic and FMT trials. However, robust meta-analytic confirmation is lacking [28]. Mechanistic insights from microbiome data have identified NAFLD-associated microbial signatures and predicted metabolic pathway changes, including altered profiles of gut-derived metabolites [29]. Probiotics containing butyrate-producing species have demonstrated beneficial effects on hepatic steatosis and liver enzyme levels in randomised controlled trials, with anti-inflammatory effects, such as reductions in high-sensitivity C-reactive protein. Proposed mechanisms include enhanced short-chain fatty acid production, although this has not always been directly measured [30]. Animal models consistently demonstrate therapeutic benefit, but human evidence remains limited to small randomized or pilot trials [31]. Translation to clinical practice is therefore still preliminary.

4.5. Helicobacter Pylori Eradication Enhancement

H. pylori eradication therapy can benefit from probiotic adjuncts, with multiple meta-analyses, some focused on paediatric populations, showing modest increases in eradication rates and reductions in antibiotic-associated adverse effects, particularly diarrhoea [32,33,34,35]. In children, Lactobacillus-supplemented triple therapy has been associated with improved outcomes compared to standard therapy alone [32]. A broader paediatric meta-analysis likewise supports the use of probiotics as an add-on during eradication regimens [33]. Across trials synthesised in these reviews, Lactobacillus and Saccharomyces boulardii are the most consistently studied adjuncts; both are associated with higher eradication rates and fewer overall side effects compared with antibiotics alone [34,35]. Strain-specific superiority (e.g., Bifidobacterium vs. others) cannot be concluded from these citations, and effect sizes vary by study design and population [32,33,34,35]. In practice, the improved tolerability observed with probiotic co-therapy may help support adherence and reduce antibiotic-associated complications, even as absolute gains in eradication remain modest and strain- and regimen-dependent [34,35].
A summary of the evidence for microbiome-targeted therapies across major gastrointestinal conditions, including their clinical outcomes, evidence strength, and limitations, is presented in Table 1 and Table 2.

5. Emerging Therapeutic Innovations

5.1. Engineered Probiotics Enter Clinical Development

Next-generation probiotics built with synthetic biology represent an advanced frontier in microbiome therapeutics. Engineered organisms have been developed with programmable therapeutic functions, improved stability, and targeted delivery features [36,37]. Current research includes the design of sense-and-respond systems in probiotics to detect pathogen-associated signals and release antimicrobial agents, as outlined in conceptual frameworks for synthetic microbes as drug delivery platforms [38]. Applications in metabolic diseases include engineered strains capable of producing therapeutic enzymes, such as phenylalanine ammonia lyase, for phenylketonuria [39].
SYNB1618, based on E. coli Nissle 1917 and engineered with phenylalanine-degrading pathways, demonstrated safety, tolerability, and pharmacodynamic activity in early-phase human trials [40]. Safety innovation strategies described for engineered probiotics include antibiotic resistance-free selection systems, genetic kill switches, and other containment approaches [41]. CRISPR-Cas9 technology further enables precise, sequence-specific genetic targeting of bacteria, supporting the development of highly controlled therapeutic functions [42]. Published human trials report only early safety and pharmacodynamic outcomes. No Phase 3 efficacy data are yet available. Regulatory approval will depend on robust biosafety safeguards, including genetic containment strategies and mechanisms to minimize unintended ecological effects.

5.2. Bacteriophage Therapy Development

Bacteriophage-based approaches offer targeted antimicrobial options against gastrointestinal pathogens, including Clostridioides difficile. Endolysins derived from C. difficile phages have demonstrated activity in vitro and in infection models, supporting their potential as precision adjuncts to existing therapies [43,44]. Technical strategies under investigation include combining multiple phages to broaden host-range and reduce the emergence of resistance, as demonstrated by phage cocktails that curtailed C. difficile growth in vitro and limited proliferation in vivo models [45]. The CD27L endolysin, cloned from a C. difficile bacteriophage, exhibits lytic activity against C. difficile with selectivity that helps spare commensal bacteria in experimental settings [44]. Clinical development remains early-stage. Human studies have primarily evaluated the safety and tolerability of orally administered phage preparations, which appear acceptable in healthy adults; formulation and dosing considerations (including survival through the gastrointestinal tract) are active areas of work [46,47]. Overall, preclinical findings, including infection-model data with C. difficile endolysins, support continued development, but robust clinical efficacy data are still limited [43,44,45,46,47]. Human efficacy data remain absent, with formulation stability and resistance risk as major hurdles. For bacteriophage therapies, effective resistance monitoring and stability controls will be critical to meet regulatory standards and ensure safe clinical translation.

5.3. Metabolite-Based Interventions Advance

Microbial metabolite-based strategies aim to influence specific host—microbe biochemical pathways. Short-chain fatty acids (SCFAs) such as butyrate, propionate, and acetate have well-described roles in gut health, metabolic regulation, and immune modulation, with proposed applications in gastrointestinal and metabolic diseases [48,49]. Bile acid metabolism is another promising target, as microbially modified bile acids can act as farnesoid X receptor (FXR) agonists, influencing metabolic homeostasis and liver function [50]. Tryptophan-derived indole metabolites activate the aryl hydrocarbon receptor, enhancing mucosal immunity and supporting intestinal barrier integrity. Reduced levels of microbiota-derived metabolites, including secondary bile acids and tryptophan catabolites, have been associated with loss of colonization resistance, intestinal inflammation, and increased risk of disorders such as inflammatory bowel disease and metabolic dysfunction [51,52,53]. Postbiotic approaches are being explored for their safety, stability, and defined composition advantages compared with live microorganisms. Akkermansia muciniphila is among the candidates investigated for metabolic and gut barrier benefits [54]. The ISAPP consensus defines postbiotics broadly and summarises evidence for beneficial effects on gut barrier function and immune modulation, while noting that clinical outcomes depend on the specific preparation and context [55]. Defined metabolite supplements differ from complex postbiotic mixtures, which face regulatory uncertainty due to variable composition.
A summary figure (Figure 1) maps the developmental stage, mechanisms of action, and key translational hurdles for engineered probiotics, bacteriophage therapies, and metabolite-based interventions, highlighting the current opportunities and barriers to clinical adoption.

6. Regulatory Landscape and Clinical Implementation

6.1. FDA Approvals Establish Therapeutic Validation

The November 2022 approval of Rebyota and the April 2023 approval of Vowst marked watershed moments for microbiome therapeutics, representing the first FDA-approved microbiota-based products for gastrointestinal indications and establishing recognised regulatory pathways [56,57]. Rebyota is a donor-derived fecal microbiota preparation administered as a single-dose rectal enema. Clinical trials have demonstrated superiority over placebo for prevention of recurrent Clostridium difficile infection, with published data from pivotal studies supporting its approval [58]. Vowst contains purified live Firmicutes spores sourced from qualified donors, delivered as oral capsules taken over three consecutive days, and achieved 12.4% recurrence versus 39.8% with placebo at 8 weeks in its pivotal study [59].
Vowst received Breakthrough Therapy, Priority Review, and Orphan Drug designations, reflecting its significant clinical advantages over existing treatments. Both products are regulated under the FDA’s framework for biologics, with the Live Biotherapeutic Products guidance from the Center for Biologics Evaluation and Research providing relevant chemistry, manufacturing, and control expectations [60]. Additional donor screening requirements, including enhanced pathogen testing such as for COVID-19, have been implemented following safety alerts [61]. The manufacturing of these products must comply with current Good Manufacturing Practice standards, with comprehensive quality control systems in place [62].

6.2. Cost-Effectiveness Challenges Implementation

High acquisition costs remain a barrier to broad adoption despite clinical efficacy. The Institute for Clinical and Economic Review (ICER) lists estimated wholesale acquisition costs of approximately $17,500 per treatment course for Vowst and $9500 for Rebyota, compared with substantially lower procedural costs for conventional fecal microbiota transplantation [63]. Insurance coverage varies by plan and region. UnitedHealthcare policy documents confirm coverage of fecal microbiota transplantation, including product-based preparations, starting at first recurrence for eligible beneficiaries [64]. Manufacturer-reported patient support programs offer co-pay assistance of up to $5000–$10,000 annually for eligible insured patients [65,66]. Patient assistance programs provide a no-cost product to qualifying uninsured patients. Specialty pharmacy networks manage product distribution and offer benefits verification and reimbursement support, which may assist in navigating prior authorisation processes [65,66]. Costs should also be considered in the context of existing gastrointestinal therapies. Traditional FMT in procedural settings may cost under USD 1500, far less than Rebyota or Vowst. Their relative value depends on long-term durability of benefit and payer support. Conventional treatments such as vancomycin for recurrent CDI or immunosuppressants for IBD remain far less costly, whereas biologics carry high but reimbursed prices. Framing microbiome therapies against these benchmarks highlights both their therapeutic promise and economic challenge.

6.3. Implementation Barriers Require Systematic Solutions

Clinical implementation faces several practical considerations, including storage requirements, administration procedures, and provider training. Rebyota must be stored in a dedicated freezer at −20 °C or colder and used within seven days after thawing if kept refrigerated [67]. Vowst, as an oral capsule formulation, follows standard oral medication handling procedures without requiring special cold storage needs, with stability features designed for routine pharmacy use [68]. Successful integration into care pathways requires trained clinical staff familiar with preparation and administration protocols. Manufacturer and professional society resources provide clinical education, as well as guidance on benefits verification and reimbursement processes to support adoption [67,68,69]. Ongoing safety monitoring is conducted through FDA post-market surveillance systems and professional registries such as the AGA FMT and gut microbial therapies registry [69,70]. These systems track reported adverse events and contribute to the long-term safety profile of microbiota-based therapeutics. Ethical considerations include obtaining informed consent for donor-derived products and providing transparent disclosure of associated risks. Access inequities are significant in low- and middle-income countries, where donor screening and cold-chain infrastructure are limited. Addressing these disparities will be essential for equitable adoption.

7. Comparative Effectiveness and Precision Medicine

To summarise, the relative efficacy, certainty of evidence, and clinical features of major microbiome-based therapies are summarised in Table 3.

7.1. Patient Stratification Advances Personalized Approaches

Microbiome profiling offers a path toward precision medicine by using baseline composition and diversity data to inform treatment selection. Evidence shows that certain baseline microbiome characteristics, including diversity measures, are associated with improved response to interventions, supporting a personalized approach to microbiome-targeted therapy [71]. In clinical practice, this could involve a workflow where patients provide stool samples for sequencing, profiles are compared with known response-associated patterns, and clinicians select the most suitable intervention accordingly. Follow-up profiling could track compositional changes to guide therapy adjustments. Predictive biomarkers are not yet validated and vary across populations. Combining these with AI-based models raises significant ethical and data privacy concerns, necessitating safeguards and transparency prior to clinical integration. While conceptual frameworks exist, detailed stepwise protocols and EHR integration remain proposals rather than routine practice [71].
Host genetic variation also influences treatment outcomes. For example, PNPLA3 gene polymorphisms are associated with varying metabolic phenotypes in NAFLD, which may interact with microbiota-targeted strategies [72]. Evidence from functional gastrointestinal disorders also supports this approach, as microbiota-targeted therapies such as rifaximin have demonstrated clinical benefit in IBS [73]. Biomarker research is progressing toward the development of predictive and monitoring tools. In IBD, baseline bacterial composition and shifts in microbial metabolite profiles have been associated with therapeutic response, suggesting potential for integrating microbiome and metabolite data into treatment algorithms [74]. Population-specific considerations include tailoring approaches for paediatric patients, who may have different safety profiles; ensuring close monitoring in immunocompromised individuals; and recognising that geographic and lifestyle factors influence baseline microbiome composition [75]. The baseline microbiome composition varies across populations due to diet, lifestyle, and environmental exposures. These differences influence treatment response and limit the generalizability of trial results, highlighting the importance of geographic diversity in clinical research.

7.2. Long-Term Outcomes Support Sustained Benefits

Durability data show sustained benefit over weeks to a few months. In a randomised trial, frozen and fresh FMT were non-inferior for recurrent C. difficile infection, with clinical resolution assessed over up to 13 weeks [76]. Longer-term follow-up comes mainly from cohort protocols rather than national registries; for example, a severe/complicated CDI protocol combining FMT with selected vancomycin reported high success but is not a registry and does not provide 10-year outcomes [77]. Risk–benefit considerations remain favourable in appropriately selected patients, but rare serious events can occur. A report of fatal aspiration pneumonia during FMT administration highlights the need for strict procedural safeguards; overall safety depends on careful screening and technique, and benefits include meaningful symptom improvement in recurrent CDI settings [78].

8. Future Directions and Clinical Implications

8.1. Technological Convergence Drives Innovation

The convergence of artificial intelligence, synthetic biology, and precision medicine is accelerating the development of microbiome-focused therapeutics. Machine-learning approaches can analyse high-dimensional clinical and -omics data to support precision treatment strategies, aligning with broader AI-enabled decision support in medicine [79]. The manufacturing of defined microbial consortia is moving toward greater standardisation and quality control, and next-generation sequencing is routinely used in research settings for the longitudinal monitoring of therapeutic microbes and host responses [80]. In oncology, multiple studies demonstrated that the gut microbiome is associated with response to anti-PD-1 immunotherapy in melanoma, highlighting the potential for microbiome-informed strategies alongside existing treatments [81,82]. While these findings motivate the use of combination approaches, prospective interventional trials are still needed to determine whether adding microbiome-directed therapies improves efficacy or affects toxicity profiles [81,82].

8.2. Regulatory Evolution Supports Innovation

International regulatory frameworks from both the FDA and the EMA offer models that can inform the development of novel microbiome therapeutics. While the EMA’s reflection paper on advanced therapy medicinal product (ATMP) classification [83] and the FDA’s gene therapy guidance for rare diseases [84] are not specific to microbiome products, they illustrate approaches to defining product categories, establishing manufacturing standards, and setting safety monitoring expectations for novel biologics. Clear product classification, such as the FDA’s Live Biotherapeutic Product designation, can facilitate consistent regulatory review processes. Expedited pathways, including the FDA’s Breakthrough Therapy designation [85] and the EMA’s PRIME (Priority Medicines) program [86], provide enhanced scientific and regulatory support for promising interventions. These frameworks employ a risk-based approach to strike a balance between innovation and patient safety, potentially offering adaptable models for microbiome-based products.

8.3. Market Expansion Drives Investment

The global microbiome therapeutics market is projected to grow from approximately USD 300 million in 2021 to USD 3.2 billion by 2032, driven by increasing clinical validation, regulatory approvals, and commercial uptake [87]. Strategic collaborations between major pharmaceutical companies and microbiome-focused biotechnology firms are accelerating product development by combining clinical, manufacturing, and market expertise [88]. Public market activity continues to expand. For example, Microba Life Sciences’ listing on the Australian Securities Exchange (ASX) underscored investor confidence, with the company citing an estimated global sector value of USD 4.89 billion [89]. In the private sector, microbiome-focused companies have attracted more than USD 1.6 billion in venture capital funding over a two-year period, reflecting sustained market interest despite ongoing clinical and regulatory hurdles [90]. The evolution of the patent landscape strongly shapes competitive dynamics. In the pharmaceutical industry, patenting and enforcement activities frequently increase following major product approvals as companies work to secure and extend market exclusivity [91]. Concurrently, intellectual property strategies such as tiered licensing, patent pools, and collaborative agreements can help balance the need for innovation incentives with broader access goals, particularly in global health contexts [92]. A consolidated summary of technological, regulatory, and market trends shaping the future of microbiome-targeted therapies is presented in Table 4.

9. Clinical Practice Recommendations

9.1. Evidence-Based Implementation Guidelines

Clinical practice integration should follow evidence hierarchies. Fecal microbiota transplantation (FMT) is recommended for recurrent Clostridium difficile infection after appropriate antibiotic regimens, supported by systematic review and meta-analysis evidence [93,94]. Selected probiotic preparations have supportive evidence in certain gastrointestinal indications, but benefits are strain- and condition-specific rather than universal [95]. Patient selection should consider clinical severity, comorbidities, and contraindications. Cohort and trial data underlying FMT commonly exclude severely immunocompromised patients and highlight procedural risks, underscoring the need for careful screening and monitoring in higher-risk groups [94]. Treatment protocols should standardise dosing, route, and follow-up according to published trial methods. For probiotics, effectiveness is strain-specific; while some multi-strain combinations show benefit in particular settings, consistent superiority over single-strain products is not established across conditions [95]. Additional considerations apply to vulnerable populations. In pediatric patients, responses to microbiome-targeted therapies may differ from adults, and long-term safety data remain limited. In immunocompromised individuals, careful donor screening and close monitoring are essential, as the risk of infection remains a concern.

9.2. Safety Monitoring Frameworks

Comprehensive safety monitoring for microbiome therapies should include pre-treatment screening, active surveillance during therapy, and long-term follow-up, reflecting practices used in published FMT cohorts and calls for standardised protocols [96,97,98]. Screening protocols and pathogen-detection methods are critical components of risk control in gut-microbiota-modulating interventions, given the potential for unintended microbial effects noted in broader microbiome literature [96]. Adverse-event tracking should capture both immediate and delayed complications using standardised documentation, with sustained follow-up to characterise durability and late events. Long-term observational data after colonoscopic FMT illustrate the feasibility and value of such surveillance [97]. Professional guidance urging standardisation of FMT procedures underscores the role of provider education in appropriate patient selection and complication recognition [98]. Risk mitigation should emphasise awareness of contraindications, clear response protocols for complications, and multidisciplinary input, alongside quality-assurance/standardisation efforts to ensure consistent implementation across centres, as advocated in proposals to formalise FMT practice [98].

10. Conclusions

Microbiome-targeted therapies have progressed from experimental use to validated treatments, with FDA-approved products demonstrating clear clinical benefits. The strongest evidence supports FMT for recurrent C. difficile infection, while strain-specific probiotics show benefits in selected gastrointestinal conditions. Emerging approaches—such as engineered bacteria, bacteriophage therapy, and metabolite-based interventions—signal the next wave of precision microbiome medicine. Implementation requires evidence-based patient selection, standardised protocols, and robust safety monitoring, while cost-effectiveness and operational barriers remain challenges. Key research priorities include standardizing FMT protocols, validating predictive biomarkers for therapy selection, and establishing long-term safety registries. Future work should also evaluate cost-effectiveness and ensure equitable access across health systems.

Author Contributions

Conceptualization, E.C.N.L. and C.E.D.L.; methodology, E.C.N.L.; software, E.C.N.L.; validation, E.C.N.L. and C.E.D.L.; formal analysis, E.C.N.L. and C.E.D.L.; investigation, E.C.N.L.; resources, E.C.N.L. and C.E.D.L.; data curation, E.C.N.L.; writing—original draft preparation, E.C.N.L.; writing—review and editing, E.C.N.L. and C.E.D.L.; visualization, C.E.D.L.; supervision, C.E.D.L.; project administration, E.C.N.L.; funding acquisition, C.E.D.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analysed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

None of the authors declared financial and non-financial relationships, activities, or conflicts of interest regarding this manuscript.

Abbreviations

The following abbreviations are used in this manuscript:
AACAntibiotic-Associated Colitis
ACGAmerican College of Gastroenterology
ACSAmerican Chemical Society
AEAdverse Event
AGAAmerican Gastroenterological Association
AIArtificial Intelligence
ARRAbsolute Risk Reduction
ASXAustralian Securities Exchange
ATMPAdvanced Therapy Medicinal Product
CACalifornia
CBERCenter for Biologics Evaluation and Research
CDCrohn’s Disease
CDIClostridium difficile Infection
COVID-19Coronavirus Disease 2019
CRISPRClustered Regularly Interspaced Short Palindromic Repeats
DOIDigital Object Identifier
EHRElectronic Health Record
EMAEuropean Medicines Agency
EMBASEExcerpta Medica Database
FDAFood and Drug Administration
FMTFecal Microbiota Transplantation
FXRFarnesoid X Receptor
GIGastrointestinal
GMPGood Manufacturing Practice
HBVHepatitis B Virus
IBDInflammatory Bowel Disease
IBSIrritable Bowel Syndrome
ICERIncremental Cost-Effectiveness Ratio
IDSAInfectious Diseases Society of America
IIIPhase III Clinical Trial
IPOInitial Public Offering
ISAPPInternational Scientific Association for Probiotics and Prebiotics
JAMAJournal of the American Medical Association
JBJournal of Bacteriology
LBPLive Biotherapeutic Product
MAMeta-Analysis
MCGMicrogram
MDDoctor of Medicine
MIBMicrobiome
NAFLDNon-Alcoholic Fatty Liver Disease
NASHNon-Alcoholic Steatohepatitis
NNTNumber Needed to Treat
PD-1Programmed Death-1
PHAGEBacteriophage
PMIDPubMed Identifier
PRIMEPriority Medicines
RCTRandomized Controlled Trial
RNARibonucleic Acid
SER-109Microbiome Therapeutic Product SER-109
SHEASociety for Healthcare Epidemiology of America
SSIEMSociety for the Study of Inborn Errors of Metabolism
UCUlcerative Colitis
USUnited States
USDUnited States Dollar
VCVenture Capital
VOWSTMicrobiome Therapeutic Product Vowst
VSLProbiotic Formulation VSL#3

References

  1. Allegretti, J.R.; Kearney, S.; Li, N.; Bogart, E.; Bullock, K.; Gerber, G.K.; Bry, L.; Clish, C.B.; Alm, E.; Korzenik, J.R. Recurrent C. difficile infection associates with distinct bile acid and microbiome profiles. Aliment. Pharmacol. Ther. 2016, 43, 1142–1153. [Google Scholar] [CrossRef]
  2. Cammarota, G.; Ianiro, G.; Tilg, H.; Rajilić-Stojanović, M.; Kump, P.; Satokari, R.; Sokol, H.; Arkkila, P.; Pintus, C.; Hart, A.; et al. European consensus conference on faecal microbiota transplantation in clinical practice. Gut 2017, 66, 569–580. [Google Scholar] [CrossRef] [PubMed]
  3. Quraishi, M.N.; Widlak, M.; Bhala, N.; Moore, D.; Price, M.; Sharma, N.; Iqbal, T.H. Systematic review with meta-analysis: The efficacy of faecal microbiota transplantation for the treatment of recurrent and refractory Clostridium difficile infection. Aliment. Pharmacol. Ther. 2017, 46, 479–493. [Google Scholar] [CrossRef] [PubMed]
  4. Fischer, M.; Kao, D.; Kelly, C.; Kuchipudi, A.; Jafri, S.-M.; Blumenkehl, M.; Rex, D.; Mellow, M.; Kaur, N.; Sokol, H.; et al. Fecal microbiota transplantation is safe and efficacious for recurrent or refractory Clostridium difficile infection in patients with inflammatory bowel disease. Inflamm. Bowel Dis. 2016, 22, 2402–2409. [Google Scholar] [CrossRef] [PubMed]
  5. Feuerstadt, P.; Louie, T.J.; Lashner, B.; Wang, E.E.; Diao, L.; Bryant, J.A.; Sims, M.; Kraft, C.S.; Cohen, S.H.; Berenson, C.S.; et al. SER-109, an oral microbiome therapy for recurrent Clostridioides difficile infection. N. Engl. J. Med. 2022, 386, 220–229. [Google Scholar] [CrossRef]
  6. Kelly, C.R.; Fischer, M.; Allegretti, J.R.; LaPlante, K.; Stewart, D.B.; Limketkai, B.N.; Stollman, N.H. ACG clinical guidelines: Prevention, diagnosis, and treatment of Clostridioides difficile infections. Am. J. Gastroenterol. 2021, 116, 1124–1147. [Google Scholar] [CrossRef]
  7. Wang, S.; Xu, M.; Wang, W.; Cao, X.; Piao, M.; Khan, S.; Yan, F.; Cao, H.; Wang, B.; Grivennikov, S. Systematic review: Adverse events of fecal microbiota transplantation. PLoS ONE 2016, 11, e0161174. [Google Scholar] [CrossRef]
  8. DeFilipp, Z.; Bloom, P.P.; Torres Soto, M.; Mansour, M.K.; Sater, M.R.A.; Huntley, M.H.; Turbett, S.; Chung, R.T.; Chen, Y.-B.; Hohmann, E.L. Drug-resistant E. coli bacteremia transmitted by fecal microbiota transplant. N. Engl. J. Med. 2019, 381, 2043–2050. [Google Scholar] [CrossRef]
  9. Ford, A.C.; Harris, L.A.; Lacy, B.E.; Quigley, E.M.M.; Moayyedi, P. Systematic review with meta-analysis: The efficacy of prebiotics, probiotics, synbiotics and antibiotics in irritable bowel syndrome. Aliment. Pharmacol. Ther. 2018, 48, 1044–1060. [Google Scholar] [CrossRef]
  10. Sniffen, J.C.; McFarland, L.V.; Evans, C.T.; Goldstein, E.J.C.; Lobo, L.A. Choosing an appropriate probiotic product for your patient: An evidence-based practical guide. PLoS ONE 2018, 13, e0209205. [Google Scholar] [CrossRef]
  11. Derwa, Y.; Gracie, D.J.; Hamlin, P.J.; Ford, A.C. Systematic review with meta-analysis: The efficacy of probiotics in inflammatory bowel disease. Aliment. Pharmacol. Ther. 2017, 46, 389–400. [Google Scholar] [CrossRef]
  12. Iheozor-Ejiofor, Z.; Kaur, L.; Gordon, M.; Baines, P.A.; Sinopoulou, V.; Akobeng, A.K.; Cochrane IBD Group. Probiotics for maintenance of remission in ulcerative colitis. Cochrane Database Syst. Rev. 2020, 3, CD007443. [Google Scholar] [CrossRef] [PubMed]
  13. Singh, S.; Stroud, A.M.; Holubar, S.D.; Sandborn, W.J.; Pardi, D.S.; Cochrane IBD Group. Treatment and prevention of pouchitis after ileal pouch-anal anastomosis for chronic ulcerative colitis. Cochrane Database Syst. Rev. 2015, 11, CD001176. [Google Scholar] [CrossRef] [PubMed]
  14. Gibson, G.R.; Hutkins, R.; Sanders, M.E.; Prescott, S.L.; Reimer, R.A.; Salminen, S.J.; Scott, K.; Stanton, C.; Swanson, K.S.; Cani, P.D.; et al. Expert consensus document: The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 491–502. [Google Scholar] [CrossRef] [PubMed]
  15. Swanson, K.S.; Gibson, G.R.; Hutkins, R.; Reimer, R.A.; Reid, G.; Verbeke, K.; Scott, K.P.; Holscher, H.D.; Azad, M.B.; Delzenne, N.M.; et al. The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of synbiotics. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 687–701. [Google Scholar] [CrossRef]
  16. Su, G.L.; Ko, C.W.; Bercik, P.; Falck-Ytter, Y.; Sultan, S.; Weizman, A.V.; Morgan, R.L. AGA clinical practice guidelines on the role of probiotics in the management of gastrointestinal disorders. Gastroenterology 2020, 159, 697–705. [Google Scholar] [CrossRef]
  17. Rahimi, R.; Nikfar, S.; Rahimi, F.; Elahi, B.; Derakhshani, S.; Vafaie, M.; Abdollahi, M. A meta-analysis on the efficacy of probiotics for maintenance of remission and prevention of clinical and endoscopic relapse in Crohn’s disease. Dig. Dis. Sci. 2008, 53, 2524–2531. [Google Scholar] [CrossRef]
  18. Paramsothy, S.; Kamm, M.A.; Kaakoush, N.O.; Walsh, A.J.; van den Bogaerde, J.; Samuel, D.; Leong, R.W.L.; Connor, S.; Ng, W.; Paramsothy, R.; et al. Multidonor intensive faecal microbiota transplantation for active ulcerative colitis: A randomised placebo-controlled trial. Lancet 2017, 389, 1218–1228. [Google Scholar] [CrossRef]
  19. Tian, H.; Ding, C.; Gong, J.; Ge, X.; McFarland, L.V.; Gu, L.; Wei, Y.; Chen, Q.; Zhu, W.; Li, J.; et al. Treatment of slow transit constipation with fecal microbiota transplantation: A pilot study. J. Clin. Gastroenterol. 2016, 50, 865–870. [Google Scholar] [CrossRef]
  20. Goldenberg, J.Z.; Yap, C.; Lytvyn, L.; Lo, C.K.-F.; Beardsley, J.; Mertz, D.; Johnston, B.C.; Cochrane IBD Group. Probiotics for the prevention of Clostridium difficile-associated diarrhea in adults and children. Cochrane Database Syst. Rev. 2017, 12, CD006095. [Google Scholar] [CrossRef]
  21. Seres Therapeutics. VOWST Prescribing Information; Seres Therapeutics: Cambridge, MA, USA, 2023; Available online: https://www.vowst.com/prescribing-information (accessed on 7 August 2025).
  22. Ferring Pharmaceuticals. REBYOTA Monograph. 2025. Available online: https://www.ferring.ca/media/1385/rebyota-pm-control-no-285129-en-mar-5-2025.pdf (accessed on 7 August 2025).
  23. McDonald, L.C.; Gerding, D.N.; Johnson, S.; Bakken, J.S.; Carroll, K.C.; Coffin, S.E.; Dubberke, E.R.; Garey, K.W.; Gould, C.V.; Kelly, C.; et al. Clinical practice guidelines for Clostridium difficile infection in adults and children: 2017 update by the Infectious Diseases Society of America (IDSA) and Society for Healthcare Epidemiology of America (SHEA). Clin. Infect. Dis. 2018, 66, e1–e48. [Google Scholar] [CrossRef]
  24. Dale, H.F.; Rasmussen, S.H.; Asiller, Ö.Ö.; Lied, G.A. Probiotics in irritable bowel syndrome: An up-to-date systematic review. Nutrients 2019, 11, 2048. [Google Scholar] [CrossRef]
  25. Hungin, A.P.S.; Mulligan, C.; Pot, B.; Whorwell, P.; Agréus, L.; Fracasso, P.; Lionis, C.; Mendive, J.; de Foy, J.-M.P.; Rubin, G.; et al. Systematic review: Probiotics in the management of lower gastrointestinal symptoms in clinical practice—An evidence-based international guide. Aliment. Pharmacol. Ther. 2013, 38, 864–886. [Google Scholar] [CrossRef]
  26. Moayyedi, P.; Ford, A.C.; Talley, N.J.; Cremonini, F.; Foxx-Orenstein, A.E.; Brandt, L.J.; Quigley, E.M.M. The efficacy of probiotics in the treatment of irritable bowel syndrome: A systematic review. Gut 2010, 59, 325–332. [Google Scholar] [CrossRef] [PubMed]
  27. Newberry, S.J.; Hempel, S.; Maher, A.R.; Wang, Z.; Miles, J.N.V.; Shanman, R.; Johnsen, B.; Shekelle, P.G. Probiotics for the prevention and treatment of antibiotic-associated diarrhea: A systematic review and meta-analysis. JAMA 2012, 307, 1959–1969. [Google Scholar] [CrossRef] [PubMed]
  28. Paratore, M.; Santopaolo, F.; Cammarota, G.; Pompili, M.; Gasbarrini, A.; Ponziani, F.R. Fecal Microbiota Transplantation in Patients with HBV Infection or Other Chronic Liver Diseases: Update on Current Knowledge and Future Perspectives. J. Clin. Med. 2021, 10, 2605. [Google Scholar] [CrossRef] [PubMed]
  29. Nychas, E.; Marfil-Sánchez, A.; Chen, X.; Mirhakkak, M.; Li, H.; Jia, W.; Xu, A.; Nielsen, H.B.; Nieuwdorp, M.; Loomba, R.; et al. Discovery of robust and highly specific microbiome signatures of non-alcoholic fatty liver disease. Microbiome 2025, 13, 10. [Google Scholar] [CrossRef]
  30. Ahn, S.B.; Jun, D.W.; Kang, B.-K.; Lim, J.H.; Lim, S.; Chung, M.-J. Randomized, double-blind, placebo-controlled study of a multispecies probiotic mixture in nonalcoholic fatty liver disease. Sci. Rep. 2019, 9, 5688. [Google Scholar] [CrossRef]
  31. Zhou, D.; Pan, Q.; Shen, F.; Cao, H.-X.; Ding, W.-J.; Chen, Y.-W.; Fan, J.-G. Total fecal microbiota transplantation alleviates high-fat diet-induced steatohepatitis in mice via beneficial regulation of gut microbiota. Sci. Rep. 2017, 7, 1529. [Google Scholar] [CrossRef]
  32. Fang, H.-R.; Zhang, G.-Q.; Cheng, J.-Y.; Li, Z.-Y. Efficacy of Lactobacillus-supplemented triple therapy for Helicobacter pylori infection in children: A meta-analysis of randomized controlled trials. Eur. J. Pediatr. 2019, 178, 7–16. [Google Scholar] [CrossRef]
  33. Li, S.; Huang, X.-L.; Sui, J.-Z.; Chen, S.-Y.; Xie, Y.-T.; Deng, Y.; Wang, J.; Xie, L.; Li, T.-J.; He, Y.; et al. Meta-analysis of randomized controlled trials on the efficacy of probiotics in Helicobacter pylori eradication therapy in children. Eur. J. Pediatr. 2014, 173, 153–161. [Google Scholar] [CrossRef]
  34. Dang, Y.; Reinhardt, J.D.; Zhou, X.; Zhang, G.; Cardona, P.-J. The effect of probiotics supplementation on Helicobacter pylori eradication rates and side effects during eradication therapy: A meta-analysis. PLoS ONE 2014, 9, e111030. [Google Scholar] [CrossRef]
  35. Szajewska, H.; Horvath, A.; Piwowarczyk, A. Meta-analysis: The effects of Saccharomyces boulardii supplementation on Helicobacter pylori eradication rates and side effects during treatment. Aliment. Pharmacol. Ther. 2010, 32, 1069–1079. [Google Scholar] [CrossRef]
  36. Mimee, M.; Citorik, R.J.; Lu, T.K. Microbiome therapeutics—Advances and challenges. Adv. Drug Deliv. Rev. 2016, 105, 44–54. [Google Scholar] [CrossRef]
  37. Riglar, D.T.; Silver, P.A. Engineering bacteria for diagnostic and therapeutic applications. Nat. Rev. Microbiol. 2018, 16, 214–225. [Google Scholar] [CrossRef]
  38. Claesen, J.; Fischbach, M.A. Synthetic microbes as drug delivery systems. ACS Synth. Biol. 2015, 4, 358–364. [Google Scholar] [CrossRef] [PubMed]
  39. Isabella, V.M.; Ha, B.N.; Castillo, M.J.; Lubkowicz, D.J.; Rowe, S.E.; Millet, Y.A.; Anderson, C.L.; Li, N.; Fisher, A.B.; West, K.A.; et al. Development of a synthetic live bacterial therapeutic for the human metabolic disease phenylketonuria. Nat. Biotechnol. 2018, 36, 857–864. [Google Scholar] [CrossRef]
  40. Synlogic Inc. A Phase 1/2a Oral Placebo-controlled Study of SYNB1618 in Healthy Adult Volunteers and Subjects with Phenylketonuria, Society for the Study of Inborn Error of Metabolism (SSIEM) 2019, 4 September 2019. Available online: https://investor.synlogictx.com/static-files/e7c256fa-9b6a-47cb-9ef7-7d9cf44d7685 (accessed on 7 August 2025).
  41. Torres, L.; Krüger, A.; Csibra, E.; Gianni, E.; Pinheiro, V.B. Synthetic biology approaches to biological containment: Pre-emptively tackling potential risks. Essays Biochem. 2016, 60, 393–410. [Google Scholar] [CrossRef] [PubMed]
  42. Citorik, R.J.; Mimee, M.; Lu, T.K. Sequence-specific antimicrobials using efficiently delivered RNA-guided nucleases. Nat. Biotechnol. 2014, 32, 1141–1145. [Google Scholar] [CrossRef] [PubMed]
  43. Brady, T.S.; Fajardo, C.P.; Merrill, B.D.; Hilton, J.A.; Graves, K.A.; Eggett, D.L.; Hope, S. Efficacy of an endolysin against clostridioides difficile assessed in a galleria mellonella infection model. Antibiotics 2018, 7, 105. [Google Scholar] [CrossRef]
  44. Mayer, M.J.; Narbad, A.; Gasson, M.J. Molecular characterization of a Clostridium difficile bacteriophage and its cloned biologically active endolysin. J. Bacteriol. 2008, 190, 6734–6740. [Google Scholar] [CrossRef]
  45. Nale, J.Y.; Spencer, J.; Hargreaves, K.R.; Buckley, A.M.; Trzepiński, P.; Douce, G.R.; Clokie, M.R.J. Bacteriophage combinations significantly reduce Clostridium difficile growth in vitro and proliferation in vivo. Antimicrob. Agents Chemother. 2016, 60, 968–981. [Google Scholar] [CrossRef] [PubMed]
  46. Gindin, M.; Febvre, H.P.; Rao, S.; Wallace, T.C.; Weir, T.L. Bacteriophage for gastrointestinal health (PHAGE) study: Evaluating the safety and tolerability of supplemental bacteriophage consumption. J. Am. Coll. Nutr. 2019, 38, 68–75. [Google Scholar] [CrossRef] [PubMed]
  47. Chen, W.H.; Woolston, J.; Grant-Beurmann, S.; Robinson, C.K.; Bansal, G.; Nkeze, J.; Permala-Booth, J.; Fraser, C.M.; Tennant, S.M.; Shriver, M.C.; et al. Safety and Tolerability of ShigActive™, a Shigella spp. Targeting Bacteriophage Preparation, in a Phase 1 Randomized, Double-Blind, Controlled Clinical Trial. Antibiotics 2024, 13, 858. [Google Scholar] [CrossRef] [PubMed]
  48. Gonçalves, P.; Martel, F. Butyrate and colorectal cancer: The role of butyrate transport. Curr. Drug Metab. 2013, 14, 994–1008. [Google Scholar] [CrossRef]
  49. Koh, A.; De Vadder, F.; Kovatcheva-Datchary, P.; Bäckhed, F. From dietary fiber to host physiology: Short-chain fatty acids as key bacterial metabolites. Cell 2016, 165, 1332–1345. [Google Scholar] [CrossRef]
  50. Wahlström, A.; Sayin, S.I.; Marschall, H.-U.; Bäckhed, F. Intestinal crosstalk between bile acids and microbiota and its impact on host metabolism. Cell Metab. 2016, 24, 41–50. [Google Scholar] [CrossRef]
  51. Thanissery, R.; Winston, J.A.; Theriot, C.M. Inhibition of spore germination, growth, and toxin activity of clinically relevant C. difficile strains by gut microbiota derived secondary bile acids. Anaerobe 2017, 45, 86–100. [Google Scholar] [CrossRef]
  52. Zelante, T.; Iannitti, R.G.; Cunha, C.; De Luca, A.; Giovannini, G.; Pieraccini, G.; Zecchi, R.; D’Angelo, C.; Massi-Benedetti, C.; Fallarino, F.; et al. Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity 2013, 39, 372–385. [Google Scholar] [CrossRef]
  53. Roager, H.M.; Licht, T.R. Microbial tryptophan catabolites in health and disease. Nat. Commun. 2018, 9, 3294. [Google Scholar] [CrossRef]
  54. Cani, P.D.; de Vos, W.M. Next-generation beneficial microbes: The case of Akkermansia muciniphila. Front. Microbiol. 2017, 8, 1765. [Google Scholar] [CrossRef]
  55. Salminen, S.; Collado, M.C.; Endo, A.; Hill, C.; Lebeer, S.; Quigley, E.M.M.; Sanders, M.E.; Shamir, R.; Swann, J.R.; Szajewska, H.; et al. The International Scientific Association of Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of postbiotics. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 649–667. [Google Scholar] [CrossRef] [PubMed]
  56. US Food and Drug Administration. FDA Approves First Orally Administered Fecal Microbiota Product for the Prevention of Recurrence of Clostridioides Difficile Infection. 2023. Available online: https://www.fda.gov/news-events/press-announcements/fda-approves-first-orally-administered-fecal-microbiota-product-prevention-recurrence-clostridioides (accessed on 7 August 2025).
  57. Mullard, A. FDA approves second microbiome-based C. difficile therapy. Nat. Rev. Drug Discov. 2023, 22, 436. [Google Scholar] [CrossRef] [PubMed]
  58. Dubberke, E.R.; Lee, C.H.; Orenstein, R.; Khanna, S.; Hecht, G.; Gerding, D.N. Results from a randomized, placebo-controlled clinical trial of a RBX2660-A microbiota-based drug for the prevention of recurrent Clostridium difficile infection. Clin. Infect. Dis. 2018, 67, 1198–1204. [Google Scholar] [CrossRef] [PubMed]
  59. McGovern, B.H.; Ford, C.B.; Henn, M.R.; Pardi, D.S.; Khanna, S.; Hohmann, E.L.; O’bRien, E.J.; Desjardins, C.A.; Bernardo, P.; Wortman, J.R.; 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. 2021, 72, 2132–2140. [Google Scholar] [CrossRef]
  60. US Food and Drug Administration. Guidance for Industry: Early Clinical Trials with Live Biotherapeutic Products. 2016. Available online: https://www.fda.gov/files/vaccines,%20blood%20%26%20biologics/published/Early-Clinical-Trials-With-Live-Biotherapeutic-Products--Chemistry--Manufacturing--and-Control-Information--Guidance-for-Industry.pdf (accessed on 7 August 2025).
  61. US Food and Drug Administration. Information Pertaining to Additional Safety Protections Regarding Use of Fecal Microbiota for Transplantation. 2020. Available online: https://www.fda.gov/vaccines-blood-biologics/safety-availability-biologics/information-pertaining-additional-safety-protections-regarding-use-fecal-microbiota-transplantation (accessed on 7 August 2025).
  62. US Food and Drug Administration. Current Good Manufacturing Practice Regulations. 2025. Available online: https://www.fda.gov/drugs/pharmaceutical-quality-resources/current-good-manufacturing-practice-cgmp-regulations (accessed on 7 August 2025).
  63. Institute for Clinical and Economic Review (ICER). Microbiome-Based Therapies for Recurrent Clostridioides Difficile Infection: Effectiveness and Value. 2023. Available online: https://icer.org/assessment/value-assessment-framework-2023 (accessed on 7 August 2025).
  64. UnitedHealthcare. Medical Policy: Fecal Microbiota Transplantation. 2025. Available online: https://www.uhcprovider.com/content/dam/provider/docs/public/policies/umr/fecal-microbiota-transplantation-umr.pdf (accessed on 7 August 2025).
  65. Rebiotix Inc. REBYOTA Patient Resources. 2023. Available online: https://www.rebyota.com/rebyota-support-resources/ (accessed on 7 August 2025).
  66. Nestle HealthScience. VOWST Support Program. 2023. Available online: https://www.vowst.com/savings-and-support (accessed on 7 August 2025).
  67. Rebiotix Inc. REBYOTA Healthcare Provider Resources. 2022. Available online: https://www.rebyotahcp.com (accessed on 7 August 2025).
  68. Seres Therapeutics. VOWST Healthcare Provider Resources. 2023. Available online: https://www.vowsthcp.com (accessed on 7 August 2025).
  69. American Gastroenterological Association. FMT and Other Gut Microbial Therapies National Registry. 2023. Available online: https://gastro.org/research-and-awards/registries-and-studies/fecal-microbiota-transplantation-national-registry/ (accessed on 7 August 2025).
  70. US Food and Drug Administration. Postmarket Drug Safety Information for Patients and Providers; FDA: Silver Spring, MD, USA, 2023. Available online: https://www.fda.gov/drugs/drug-safety-and-availability (accessed on 7 August 2025).
  71. Kashyap, P.C.; Chia, N.; Nelson, H.; Segal, E.; Elinav, E. Microbiome at the frontier of personalized medicine. Mayo Clin. Proc. 2017, 92, 1855–1864. [Google Scholar] [CrossRef]
  72. Aron-Wisnewsky, J.; Vigliotti, C.; Witjes, J.; Le, P.; Holleboom, A.G.; Verheij, J.; Nieuwdorp, M.; Clément, K. Gut microbiota and human NAFLD: Disentangling microbial signatures from metabolic phenotypes. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 279–297. [Google Scholar] [CrossRef]
  73. Pimentel, M.; Lembo, A.; Chey, W.D.; Zakko, S.; Ringel, Y.; Yu, J.; Mareya, S.M.; Shaw, A.L.; Bortey, E.; Forbes, W.P. Rifaximin therapy for patients with irritable bowel syndrome without constipation. N. Engl. J. Med. 2011, 364, 22–32. [Google Scholar] [CrossRef]
  74. Lloyd-Price, J.; Arze, C.; Ananthakrishnan, A.N.; Schirmer, M.; Avila-Pacheco, J.; Poon, T.W.; Andrews, E.; Ajami, N.J.; Bonham, K.S.; Brislawn, C.J.; et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature 2019, 569, 655–662. [Google Scholar] [CrossRef]
  75. Francino, M.P. Antibiotics and the human gut microbiome: Dysbioses and accumulation of resistances. Front. Microbiol. 2016, 6, 1543. [Google Scholar] [CrossRef]
  76. Lee, C.H.; Steiner, T.; Petrof, E.O.; Smieja, M.; Roscoe, D.; Nematallah, A.; Weese, J.S.; Collins, S.; Moayyedi, P.; Crowther, M.; et al. Frozen vs fresh fecal microbiota transplantation and clinical resolution of diarrhea in patients with recurrent Clostridium difficile infection: A randomized clinical trial. JAMA 2016, 315, 142–149. [Google Scholar] [CrossRef] [PubMed]
  77. Fischer, M.; Sipe, B.W.; Rogers, N.A.; Cook, G.K.; Robb, B.W.; Vuppalanchi, R.; Rex, D.K. Faecal microbiota transplantation plus selected use of vancomycin for severe-complicated Clostridium difficile infection: Description of a protocol with high success rate. Aliment. Pharmacol. Ther. 2015, 42, 470–476. [Google Scholar] [CrossRef] [PubMed]
  78. Baxter, M.; Ahmad, T.; Colville, A.; Sheridan, R. Fatal aspiration pneumonia as a complication of fecal microbiota transplant. Clin. Infect. Dis. 2015, 61, 136–137. [Google Scholar] [CrossRef] [PubMed]
  79. Topol, E.J. High-performance medicine: The convergence of human and artificial intelligence. Nat. Med. 2019, 25, 44–56. [Google Scholar] [CrossRef]
  80. Zmora, N.; Soffer, E.; Elinav, E. Transforming medicine with the microbiome. Sci. Transl. Med. 2019, 11, eaaw1815. [Google Scholar] [CrossRef]
  81. Gopalakrishnan, V.; Spencer, C.N.; Nezi, L.; Reuben, A.; Andrews, M.C.; Karpinets, T.V.; Prieto, P.A.; Vicente, D.; Hoffman, K.; Wei, S.C.; et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 2018, 359, 97–103. [Google Scholar] [CrossRef]
  82. Matson, V.; Fessler, J.; Bao, R.; Chongsuwat, T.; Zha, Y.; Alegre, M.-L.; Luke, J.J.; Gajewski, T.F. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science 2018, 359, 104–108. [Google Scholar] [CrossRef]
  83. European Medicines Agency. Reflection Paper on Classification of Advanced Therapy Medicinal Products. 2012. Available online: https://www.ema.europa.eu/en/documents/regulatory-procedural-guideline/reflection-paper-classification-advanced-therapy-medicinal-products_en.pdf (accessed on 7 August 2025).
  84. US Food and Drug Administration. Human Gene Therapy for Rare Diseases: Guidance for Industry. 2020. Available online: https://www.fda.gov/regulatory-information/search-fda-guidance-documents/human-gene-therapy-rare-diseases (accessed on 7 August 2025).
  85. US Food and Drug Administration. Breakthrough Therapy. 2018. Available online: https://www.fda.gov/patients/fast-track-breakthrough-therapy-accelerated-approval-priority-review/breakthrough-therapy (accessed on 7 August 2025).
  86. European Medicines Agency. PRIME: Priority Medicines; EMA: Amsterdam, The Netherlands, 2023; Available online: https://www.ema.europa.eu/en/human-regulatory/research-development/prime-priority-medicines (accessed on 7 August 2025).
  87. Grand View Research. Microbiome Therapeutics Market Size, Share and Trends Analysis Report; Grand View Research: San Francisco, CA, USA, 2023; Available online: https://www.grandviewresearch.com/industry-analysis/microbiome-therapeutics-market (accessed on 7 August 2025).
  88. Ferring Pharmaceuticals. Ferring and PharmaBiome enter into a new microbiome R&D collaboration and exclusive licensing agreement. 2023. Available online: https://www.ferring.com/ferring-and-pharmabiome-enter-into-a-new-microbiome-rd-collaboration-and-exclusive-licensing-agreement/ (accessed on 8 August 2025).
  89. ASX. Microba Life Sciences Trusts Gut Instinct with IPO. Australian Securities Exchange. 2022. Available online: https://www.asx.com.au/blog/listed-at-asx/microba-life-sciences-trusts-gut-instinct-with-ipo (accessed on 8 August 2025).
  90. Securities.io. 5 Best Microbiome Companies (August 2025). 2025. Available online: https://www.securities.io/microbiome-companies/ (accessed on 8 August 2025).
  91. Feldman, R. May Your Drug Price Be Evergreen. J. Law Biosci. 2018, 5, 590–647. [Google Scholar] [CrossRef]
  92. Moon, S.; Jambert, E.; Childs, M.; von Schoen-Angerer, T. A Win–Win Solution? A Critical Analysis of Tiered Pricing to Improve Access to Medicines in Developing Countries. Glob. Health 2011, 7, 39. [Google Scholar] [CrossRef]
  93. Surawicz, C.M.; Brandt, L.J.; Binion, D.G.; Ananthakrishnan, A.N.; Curry, S.R.; Gilligan, P.H.; McFarland, L.V.; Mellow, M.; Zuckerbraun, B.S. Guidelines for diagnosis, treatment, and prevention of Clostridium difficile infections. Am. J. Gastroenterol. 2013, 108, 478–498. [Google Scholar] [CrossRef]
  94. Kassam, Z.; Lee, C.H.; Yuan, Y.; Hunt, R.H. Fecal microbiota transplantation for Clostridium difficile infection: Systematic review and meta-analysis. Am. J. Gastroenterol. 2013, 108, 500–508. [Google Scholar] [CrossRef]
  95. Wilkins, T.; Sequoia, J. Probiotics for gastrointestinal conditions: A summary of the evidence. Am. Fam. Physician 2017, 96, 170–178. [Google Scholar] [PubMed]
  96. Ianiro, G.; Tilg, H.; Gasbarrini, A. Antibiotics as deep modulators of gut microbiota: Between good, bad and ugly. Gut 2016, 65, 1906–1915. [Google Scholar] [CrossRef]
  97. Brandt, L.J.; Aroniadis, O.C.; Mellow, M.; Kanatzar, A.; Kelly, C.; Park, T.; Stollman, N.; Rohlke, F.; Surawicz, C. Long-term follow-up of colonoscopic fecal microbiota transplant for recurrent Clostridium difficile infection. Am. J. Gastroenterol. 2012, 107, 1079–1087. [Google Scholar] [CrossRef]
  98. Zhang, F.; Luo, W.; Shi, Y.; Fan, Z.; Ji, G. Should we standardize the 1700-year-old fecal microbiota transplantation? Am. J. Gastroenterol. 2012, 107, 1755. [Google Scholar] [CrossRef]
Figure 1. Developmental stage, mechanisms of action, and translational hurdles of emerging microbiome-targeted innovations.
Figure 1. Developmental stage, mechanisms of action, and translational hurdles of emerging microbiome-targeted innovations.
Amh 70 00036 g001
Table 1. Summary of Clinical Evidence for Microbiome-Targeted Therapies Across Gastrointestinal Conditions.
Table 1. Summary of Clinical Evidence for Microbiome-Targeted Therapies Across Gastrointestinal Conditions.
ConditionInterventionKey FindingsEvidence Strength (GRADE)Limitations/Notes
Inflammatory Bowel Disease (IBD)ProbioticsAGA: Use only in clinical trials for UC/CD; conditional (very low-certainty) suggestion for 8-strain mix in pouchitis [16]. No significant benefit in Crohn’s remission maintenance [17].Very low–lowBenefit limited to specific strains/formulations; remains investigational.
FMT (UC)Multidonor, intensive regimens induced steroid-free remission with endoscopic improvement in ~27% vs. ~8% placebo at 8 weeks [18].ModerateProtocol heterogeneity; optimal regimens unclear.
FMT (Other GI)Small pilot study in slow transit constipation showed potential benefit [19].Very lowSmall sample sizes; preliminary data only.
Clostridium difficile infection (CDI)Probiotics (Primary prevention)Cochrane review: reduced risk of antibiotic-associated CDI; insufficient data for recurrence prevention [20].ModeratePrevention effect mostly in primary setting.
FDA-approved microbiome productsVowst: recurrence 12.4% vs. 39.8% placebo [21]; Rebyota: success 70.6% vs. 57.5% placebo [22].HighSpecific to recurrent CDI; long-term durability still being studied.
FMTRecommended by IDSA/SHEA for multiply recurrent CDI after antibiotic failure [23].HighRequires donor screening; procedural infrastructure needed.
Irritable Bowel Syndrome (IBS)ProbioticsMulti-strain > single strain for symptom improvement; e.g., B. infantis 35624 effective for global symptoms/abdominal pain [24,25].Low–ModerateStrain-specific effects; optimal dose/duration unclear.
Treatment duration≥8 weeks associated with better outcomes [26]. Short courses less effective.
SafetyNo increased adverse events vs. placebo in large GI meta-analyses [27].HighWell-tolerated across trials.
Metabolic Liver Disease (NAFLD/NASH)ProbioticsSome RCTs show improvements in liver enzymes, steatosis, inflammatory markers [28,30].Low–ModerateMeta-analytic confirmation lacking.
FMTEarly pilot data suggest benefit; animal models support potential [29,31].Very lowHuman translation at early stage.
Helicobacter pyloriProbiotic adjuncts to eradication therapyModest increase in eradication rates; reduced antibiotic-associated diarrhoea [34,35].ModerateEffects strain- and regimen-dependent.
Strains studiedLactobacillus spp., S. boulardii most consistent; paediatric triple therapy with Lactobacillus improved outcomes [32,33]. No clear strain superiority established.
This table summarises the current clinical evidence for microbiome-targeted interventions across major gastrointestinal disease areas, highlighting intervention types, key findings, evidence strength, and limitations; Abbreviations: AGA—American Gastroenterological Association; UC—ulcerative colitis; CD—Crohn’s disease; CDI—Clostridium difficile infection; FMT—fecal microbiota transplantation; GI—gastrointestinal; IDSA/SHEA—Infectious Diseases Society of America/Society for Healthcare Epidemiology of America; NAFLD—non-alcoholic fatty liver disease; NASH—non-alcoholic steatohepatitis; RCT—randomised controlled trial.
Table 2. Summary of evidence characteristics for key microbiome-targeted therapies in gastrointestinal disease.
Table 2. Summary of evidence characteristics for key microbiome-targeted therapies in gastrointestinal disease.
ConditionInterventionSample Size/Follow-UpEvidence SourceEvidence Strength (GRADE)
Recurrent CDIFMT (various delivery routes)RCTs: 40–232 patients;
8–12 weeks follow-up [3]
RCTs
+ Meta-analysis
High
Vowst (SER-109)Phase 3 RCT: 182 patients;
8 weeks [5]
Regulatory trial
(FDA submission)
High
Rebyota (RBX2660)Phase 3 RCTs + pooled analysis: ~270 patients; 8 weeks [22]Regulatory trial
+ integrated analyses
High
Ulcerative colitisMultidonor intensive FMTRCT: 81 patients; 8 weeks [18]RCTModerate
Inflammatory Bowel DiseaseProbiotics (VSL#3/multi-strain)Trials 50–200 patients;
8–52 weeks [16,17]
RCTs
+ Cochrane reviews
Low–very low
Probiotics (various strains)Trials 80–400 patients;
4–12 weeks [9,24]
RCTs
+ Systematic reviews
Low–moderate
H. pylori
eradication
Adjunct probiotics (Bifidobacterium-based)RCTs ~200 patients;
4–8 weeks [30]
RCTsLow–moderate
Abbreviations: CDI—Clostridium difficile infection; FMT—fecal microbiota transplantation; RCT—randomised controlled trial; VSL#3—Proprietary 8-strain high-potency probiotic formulation (originally produced by VSL Pharmaceuticals; now marketed as Vivomixx® or Visbiome®).
Table 3. Comparative clinical outcomes across microbiome therapies.
Table 3. Comparative clinical outcomes across microbiome therapies.
TherapyConditionBest-Supported OutcomeEffect vs. ControlApprox NNTEvidence QualityKey Notes
FMTRecurrent CDIClinical cure/recurrence preventionSubstantially higher cure vs. antibiotics; high effectiveness in recurrent/refractory CDI Moderate (mixed RCT/obs.)Strong benefit across studies, but heterogeneity precludes a single pooled “success %” [3].
Multi-strain probioticsUC maintenanceMaintenance of remissionEvidence uncertain; no robust pooled benefit Low–very lowCochrane review could not confirm routine benefit for UC maintenance [12].
Vowst (SER-109)Recurrent CDIRecurrence at 8 weeks12.4% vs. 39.8% recurrence (absolute ↓ 27.4%)~4High (Phase 3 RCT)Oral capsules × 3 days; FDA-approved microbiota product [59].
Rebyota (RBX2660)Recurrent CDITreatment success at 8 weeks70.6% vs. 57.5% success (absolute ↑ 13.1%)~8High (integrated licensure analyses + RCTs)Single-dose rectal administration; FDA-approved [58].
Bifidobacterium-containing probiotics (adjunct)H. pylori eradicationEradication rate; adverse effectsModest increase in eradication and reduced AEs (strain/regimen dependent) Low–moderateBenefits are small and strain-specific; strongest data are mixed-probiotic or S. boulardii analyses [32,33,34,35].
VSL#3 (multi-strain)UC induction/maintenanceClinical remission/maintenanceSignals of benefit in UC in some analyses; heterogeneity limits precision Low–moderateEffect sizes vary by study; dosing/optimal use not firmly established [11].
This table summarises key outcomes from microbiome-based therapies where results are supported by cited clinical trials or systematic reviews. “Effect vs. control” shows the most relevant and reproducible figure from high-quality sources. Approximate numbers needed to treat (NNT) are calculated only where absolute risk reduction (ARR) can be derived from published percentages; otherwise, this field is left blank. Evidence quality reflects the certainty ratings in the cited systematic reviews or regulatory submissions, or is inferred from trial design strength (e.g., Phase 3 RCT = high). ↑ indicates an absolute increase in the proportion of patients achieving the primary outcome compared with control; ↓ indicates an absolute reduction in the proportion of patients experiencing the adverse outcome compared with control. Abbreviations: AE: adverse event; ARR: absolute risk reduction; CDI: Clostridium difficile infection; FDA: U.S. Food and Drug Administration; FMT: fecal microbiota transplantation; H. pylori: Helicobacter pylori; NNT: number needed to treat; obs.: observational studies; RCT: randomised controlled trial; UC: ulcerative colitis.
Table 4. Future Directions and Clinical Implications for Microbiome-Targeted Therapies.
Table 4. Future Directions and Clinical Implications for Microbiome-Targeted Therapies.
ThemeKey DevelopmentsClinical/Commercial ImplicationsEvidence or Market IndicatorsLimitations/Gaps
Technological
convergence
AI + -omics integration for precision treatment selection (↑) [79]Supports personalized microbiome therapy; aligns with AI-enabled decision-making in medicineAI models analyse high-dimensional clinical & microbiome data; early success in oncologyProspective validation needed; risk of overfitting in small datasets
Standardisation of defined microbial consortia manufacturing (↑) [80]Improves reproducibility, quality, and safety of therapeuticsGMP-aligned processes emerging; regulatory interest in consistencyHigh cost of compliance; technical complexity
Longitudinal monitoring via next-generation sequencing (↑) [80]Enables dynamic tracking of microbiome composition & host responseUsed in research; potential in clinical follow-upCost, data interpretation challenges
Gut microbiome linked to anti-PD-1 immunotherapy response in melanoma (↑) [81,82]Potential for microbiome-informed combination oncology regimensMultiple studies show associations; rationale for interventional trialsCausality unproven; effect on toxicity unclear
Regulatory evolutionEMA ATMP reflection paper & FDA gene therapy guidance [83,84]Provide frameworks for novel biologics, adaptable to microbiome productsClarify product categories, manufacturing, and safety expectationsNot microbiome-specific; interpretation may vary
FDA Live Biotherapeutic Product (LBP) designationStreamlines regulatory classification and reviewRecognised pathway for microbiome-based productsRequires detailed manufacturing and clinical data
Expedited pathways: FDA Breakthrough Therapy & EMA PRIME (↑) [85,86]Accelerate development of promising microbiome therapeuticsOffer enhanced scientific/regulatory supportReserved for high-impact products; stringent entry criteria
Market
expansion
Global market projected USD 300M (2021) → USD 3.2B (2032) (↑) [87]Demonstrates rapid sector growth potentialCAGR driven by approvals, validation, and adoptionProjections depend on regulatory success
Pharma–biotech strategic collaborations (↑) [88]Accelerate product development through shared expertiseExamples in manufacturing scale-up and trial executionRisk of dependency on single large partners
Public market example: Microba Life Sciences IPO on ASX (↑) [89]Signals investor confidence; sector valued at USD 4.89BIncreased visibility for microbiome companiesMarket volatility risk
Private market: >USD 1.6B VC funding in 2 years (↑) [90]Sustained investor interest despite hurdlesSupports pipeline diversificationFunding concentrated in select markets
Patent & IP strategies [91,92]Secure market exclusivity post-approval; promote access via tiered licensing/poolsShapes competitive landscape; encourages innovationTension between exclusivity & equitable access
Abbreviation: ↑ = growth or positive development/trend. AI: artificial intelligence; ATMP: advanced therapy medicinal product; EMA: European Medicines Agency; FDA: United States Food and Drug Administration; GMP: Good Manufacturing Practice; IPO: initial public offering; LBP: Live Biotherapeutic Product; PD-1: programmed cell death protein 1; PRIME: Priority Medicines; USD: United States dollars; VC: venture capital.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lim, E.C.N.; Lim, C.E.D. Microbiome-Targeted Therapies in Gastrointestinal Diseases: Clinical Evidence and Emerging Innovations. Acta Microbiol. Hell. 2025, 70, 36. https://doi.org/10.3390/amh70030036

AMA Style

Lim ECN, Lim CED. Microbiome-Targeted Therapies in Gastrointestinal Diseases: Clinical Evidence and Emerging Innovations. Acta Microbiologica Hellenica. 2025; 70(3):36. https://doi.org/10.3390/amh70030036

Chicago/Turabian Style

Lim, Enoch Chi Ngai, and Chi Eung Danforn Lim. 2025. "Microbiome-Targeted Therapies in Gastrointestinal Diseases: Clinical Evidence and Emerging Innovations" Acta Microbiologica Hellenica 70, no. 3: 36. https://doi.org/10.3390/amh70030036

APA Style

Lim, E. C. N., & Lim, C. E. D. (2025). Microbiome-Targeted Therapies in Gastrointestinal Diseases: Clinical Evidence and Emerging Innovations. Acta Microbiologica Hellenica, 70(3), 36. https://doi.org/10.3390/amh70030036

Article Metrics

Back to TopTop