Next Article in Journal
Bovine Ultra-Long CDR H3 Specific for Bovine Rotavirus Displays Potent Virus Neutralization and Therapeutic Effects in Infected Calves
Previous Article in Journal
MECP2 mRNA Profile in Brain Tissues from a Rett Syndrome Patient and Three Human Controls: Mutated Allele Preferential Transcription and In Situ RNA Mapping
Previous Article in Special Issue
Microbiota Metabolite Profiles and Dietary Intake in Older Individuals with Insomnia of Short vs. Normal Sleep Duration
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Effect of Microbiome-Derived Metabolites in Inflammation-Related Cancer Prevention and Treatment

by
Alice N. Mafe
1 and
Dietrich Büsselberg
2,*
1
Department of Biological Sciences, Faculty of Sciences, Taraba State University, Main Campus, Jalingo 660101, Taraba State, Nigeria
2
Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha Metropolitan Area, Al Rayyan P.O. Box 22104, Qatar
*
Author to whom correspondence should be addressed.
Biomolecules 2025, 15(5), 688; https://doi.org/10.3390/biom15050688
Submission received: 5 March 2025 / Revised: 29 April 2025 / Accepted: 6 May 2025 / Published: 8 May 2025
(This article belongs to the Special Issue Advances in Metabolites Produced by Microbiota)

Abstract

:
Chronic inflammation plays a crucial role in cancer development, yet the mechanisms linking the microbiome to inflammation-related carcinogenesis remain unclear. Emerging evidence suggests that microbiome-derived metabolites influence inflammatory pathways, presenting both challenges and opportunities for therapy. However, a deeper understanding of how these metabolites regulate inflammation and contribute to cancer prevention is still needed. This review explores recent advances in microbiome-derived metabolites and their roles in inflammation-related carcinogenesis. It highlights key molecular mechanisms, emerging therapies, and unresolved challenges. Synthesizing current research, including clinical trials and experimental models, bridges the gap between microbiome science and cancer therapy. Microbial metabolites such as short-chain fatty acids (SCFAs), polyamines, indoles, and bile acids play vital roles in regulating inflammation and suppressing cancer. Many metabolites exhibit potent anti-inflammatory and immunomodulatory effects, demonstrating therapeutic potential. Case studies show promising results, but challenges such as metabolite stability, bioavailability, and individual variability remain. Understanding microbiome–metabolite interactions offers novel strategies for cancer prevention and treatment. This review identifies knowledge gaps and proposes future research directions to harness microbiome-derived metabolites for innovative cancer therapies. Addressing these issues may pave the way for microbiome-targeted cancer interventions.

1. Introduction

Chronic inflammation is a well-established driver of cancer, contributing to tumor initiation, progression, and metastasis [1]. Recent evidence highlights the crucial role of the gut microbiome in modulating inflammation and tumorigenesis [2]. Among its many functions, the microbiome produces many metabolites that serve as critical signaling molecules, influencing immune responses, oxidative stress, and cellular pathways in cancer development [3]. These microbiome-derived metabolites have the potential to act as both pro- and anti-inflammatory agents, making them critical targets for cancer prevention and therapy [4]. Despite their growing recognition, the precise mechanisms through which these metabolites regulate inflammation and cancer risk remain incompletely understood [5]. Several gaps in knowledge and controversies hinder the full therapeutic utilization of microbiome-derived metabolites. The individual variability of the microbiome presents a challenge, as differences in microbial composition among individuals can significantly alter metabolite production and function [6]. Equally, the complex interplay in host–microbe interactions and the bioavailability of these metabolites in cancer therapy remains unclear [7]. While some metabolites exhibit promising anti-inflammatory and immunomodulatory effects, others may promote tumorigenesis under certain conditions, adding further complexity to their role in cancer biology [8]. Resolving these uncertainties is essential for developing effective microbiome-based interventions.
This review aims to synthesize recent discoveries, highlight emerging therapeutic strategies, and evaluate current obstacles in leveraging microbiome-derived metabolites for inflammation-related cancer prevention and treatment. This article provides insights into their clinical relevance and translational potential by critically assessing the molecular pathways and therapeutic innovations associated with these metabolites. Furthermore, this review explores the limitations of existing research, proposing novel interpretations and identifying core areas that require further investigation. The scope of this review encompasses the latest advancements in microbiome metabolite research, focusing on their role in modulating inflammation and their potential applications in cancer therapy. It critically evaluates preclinical and clinical findings, examines innovative therapeutic approaches, and discusses the broader implications of microbiome science in oncology. By tackling knowledge gaps and unresolved controversies, this study contributes to a more profound knowledge of microbiome–metabolite interactions and their significance in cancer prevention and treatment. This review comprehensively synthesizes microbiome–metabolite interactions in cancer, uncovering unexplored therapeutic potential and novel clinical applications. As research in this field progresses, realizing these interactions could pave the way for personalized microbiome-targeted strategies, offering innovative cancer prevention and treatment solutions.

2. Microbiome-Derived Metabolites and Their Role in Inflammation-Related Cancer

The gut microbiome produces a diverse range of metabolites that influence various physiological processes, including immune regulation, oxidative stress, and cellular signaling [9]. These microbial metabolites play a crucial role in modulating inflammation, which is an essential driver of cancer initiation and progression [10]. While some metabolites exhibit anti-inflammatory and tumor-suppressive effects, others contribute to carcinogenesis under specific conditions [11]. Perception of the functions of these significant microbial metabolites is essential for developing targeted strategies to mitigate inflammation-related cancer. Several studies have highlighted the impact of microbiome-derived metabolites on cancer progression (Table 1).

2.1. Major Microbial Metabolites and Their Functions

2.1.1. Short-Chain Fatty Acids (SCFAs)

SCFAs, including butyrate, propionate, and acetate, are produced through bacterial fermentation of dietary fibers in the colon [23]. These metabolites are central to immune modulation and maintaining intestinal homeostasis [24]. Butyrate, in particular, has been extensively studied for its ability to induce apoptosis in cancer cells, regulate immune responses, and enhance gut barrier integrity [25]. Propionate and acetate also contribute to anti-inflammatory processes by modulating cytokine production and immune cell function [26]. The tumor-suppressive properties of SCFAs highlight their potential as therapeutic agents in cancer prevention and treatment [27].

2.1.2. Polyamines

Polyamines, such as spermidine, are essential for cellular growth, differentiation, and immune regulation [28]. While polyamines are critical for normal physiological functions, dysregulated polyamine metabolism has been implicated in both inflammatory disorders and cancer [29]. Spermidine has shown promise in modulating inflammation by influencing autophagy, oxidative stress, and immune cell activity [30]. However, the dual role of polyamines in promoting both cell proliferation and apoptosis denotes the complexity of their involvement in cancer biology [31]. Further research is needed to harness the therapeutic potential of polyamine regulation in cancer treatment.

2.1.3. Indoles and Tryptophan Metabolites

Indoles and tryptophan-derived metabolites, primarily produced by gut bacteria, play a significant role in modulating immune responses and inflammation [32]. These metabolites activate the aryl hydrocarbon receptor (AHR), a principal regulator of immune homeostasis and anti-inflammatory signaling [33]. By influencing T cell differentiation and cytokine production, indole derivatives contribute to immune tolerance and the suppression of chronic inflammation [34]. Their ability to counteract inflammatory pathways makes them potential candidates for microbiome-targeted cancer therapies [35].

2.1.4. Bile Acid Derivatives

Secondary bile acids, produced through microbial metabolism of primary bile acids, exhibit diverse effects on immune regulation and tumorigenesis [36]. Some secondary bile acids, such as deoxycholic acid (DCA), have been linked to pro-inflammatory and tumor-promoting activities, while others, like lithocholic acid (LCA), exhibit immunosuppressive and anti-inflammatory properties [37]. The balance between these opposing effects is crucial in determining their role in cancer progression [38]. Discerning the interplay between bile acid metabolism and host immune responses is essential for designing microbiome-based interventions in cancer prevention [39]. The intricate relationship between microbiome-derived metabolites and inflammation-related cancer indicates the need for further investigation into their mechanisms of action [40]. By elucidating their molecular functions, researchers can develop innovative strategies to manipulate the microbiome for therapeutic benefits, offering new possibilities for cancer prevention and treatment [41].

2.1.5. Lipopolysaccharide (LPS)—The Pro-Inflammatory Metabolite

Lipopolysaccharide (LPS), a key component of the outer membrane of Gram-negative bacteria, functions as a potent microbial-associated molecular pattern (MAMP) [42]. Under healthy conditions, LPS is typically confined to the gut lumen at low concentrations, where it contributes to the maturation and modulation of the immune system [43]. However, disruptions in the gut microbiota, which is referred to as dysbiosis, can compromise intestinal barrier integrity, leading to increased translocation of LPS into the bloodstream [44], as dysbiosis has been shown to increase LPS in circulation and leads to increased chronic inflammation, leading to a tumor-favoring environment; furthermore, it even acts molecularly to promote cancer. This rise in systemic LPS levels is known to trigger chronic low-grade inflammation, primarily through the activation of toll-like receptor 4 (TLR4), which stimulates the production of inflammatory mediators such as TNF-α, IL-6, and IL-1β [45]. Such sustained inflammation plays a pivotal role in creating a tumor-supportive microenvironment by promoting genomic instability, neovascularization, and immune suppression [46]. In addition to its immunostimulatory effects, LPS has been directly implicated in cancer progression. Recent findings demonstrate that in human cholangiocarcinoma, LPS enhances cancer cell migration and invasion by activating the METTL3/PI3K/AKT signaling axis, thereby contributing to tumor aggressiveness and metastatic potential [22]. Collectively, these insights emphasize the dualistic nature of microbial metabolites, with LPS exemplifying how dysbiosis-associated products can exacerbate inflammation-related carcinogenesis. Building on the earlier discussion of key microbial metabolites such as short-chain fatty acids (SCFAs), polyamines, and indoles, it is also essential to highlight the unique contributions of beneficial microbes like Akkermansia muciniphila and Faecalibacterium prausnitzii. These species are gaining recognition for their significant roles in inflammation regulation and cancer prevention. They are increasingly considered next-generation probiotics due to their strong anti-inflammatory and immunoregulatory capabilities [47]. A. muciniphila is notable for its ability to degrade mucin, strengthen gut barrier function, and downregulate pro-inflammatory cytokines, thereby potentially inhibiting tumor initiation and progression [48]. In contrast, F. prausnitzii is a key butyrate producer and is consistently associated with reduced incidence of inflammatory bowel disease and colorectal cancer [49]. Compared with traditional probiotics such as Bifidobacteria and Lactobacilli, which are primarily known for promoting overall gut health and supporting immune function, A. muciniphila and F. prausnitzii offer more specialized effects by modulating host signaling pathways linked to inflammation and carcinogenesis. Their ability to influence pathways such as NF-κB, IL-10, and Treg cell responses highlights their potential as therapeutic agents in inflammation-associated cancers [50].

3. Molecular Mechanisms Linking Microbiome Metabolites to Cancer Prevention

Microbiome-derived metabolites exert profound effects on cancer prevention through multiple molecular pathways. These metabolites influence immune responses, epigenetic modifications, and systemic signaling networks, all of which play critical roles in tumor suppression [51]. Microbial metabolites can promote or inhibit cancer progression by modulating inflammation, metabolic reprogramming, and neuroimmune interactions [52]. Knowing these molecular mechanisms provides new opportunities for harnessing the microbiome as a therapeutic target in cancer prevention [53]. Figure 1 illustrates the protective role of microbiome metabolites in cancer prevention and therapy. As illustrated in Figure 2, the gut microbiome plays a dual role in cancer prevention and promotion depending on its composition.

3.1. Immunomodulation and Anti-Inflammatory Pathways

The interplay between microbiome-derived metabolites and immune signaling pathways plays a crucial role in modulating inflammation and promoting anticancer effects [54].

3.1.1. SCFAs and Regulatory T Cell Activation

Short-chain fatty acids (SCFAs), particularly butyrate, propionate, and acetate, are central players in immune regulation [55]. These microbial metabolites promote the differentiation and activation of regulatory T cells (Tregs) [56], which are essential for maintaining immune tolerance and suppressing chronic inflammation [57]. SCFAs enhance Treg function by modulating histone deacetylase (HDAC) activity and increasing the production of anti-inflammatory cytokines such as IL-10 [58]. SCFAs promote an immunosuppressive environment, helping counteract the pro-inflammatory conditions that drive cancer development. Thus, they are crucial mediators of cancer prevention.

3.1.2. Gut Microbiota and Tumor Microenvironment Modulation

The gut microbiota plays a fundamental role in shaping the tumor microenvironment by influencing immune cell recruitment, cytokine signaling, and metabolic interactions [59]. Microbiome-derived metabolites regulate the balance between pro- and anti-inflammatory factors within the tumor microenvironment, thereby affecting cancer progression [60]. For instance, certain bacterial species produce metabolites that activate immune surveillance mechanisms, enhancing the cytotoxic activity of natural killer (NK) cells and cytotoxic T lymphocytes [61]. Short-chain fatty acids (SCFAs), important microbial metabolites, influence gene regulation not by directly binding to DNA but by inhibiting histone deacetylases (HDACs), which leads to changes in chromatin structure and subsequently affects gene expression [62]. Conversely, dysbiosis or an imbalance in microbiome composition can lead to an inflammatory tumor-promoting environment, underscoring the importance of microbiome stability in cancer prevention [63].

3.2. Epigenetic and Metabolic Reprogramming

3.2.1. Microbiome Influence on Oncogene Expression and DNA Methylation

Microbiome metabolites directly impact gene expression by modulating oncogenes and tumor suppressor genes through epigenetic mechanisms [64]. For example, SCFAs such as butyrate act as HDAC inhibitors, altering chromatin structure and influencing transcriptional regulation [65]. Moreover, microbial metabolites can affect DNA methylation patterns, leading to changes in gene expression that either promote or inhibit tumorigenesis [66]. These epigenetic modifications are crucial in regulating inflammation and cellular transformation, positioning microbiome metabolites as leading players in cancer prevention.

3.2.2. Metabolite-Driven Epigenetic Changes in Inflammation

Beyond oncogene regulation, microbiome metabolites also induce epigenetic modifications in inflammatory pathways [67]. For instance, indole derivatives and polyamines influence histone modifications and DNA methylation patterns associated with inflammation and immune responses [68]. By suppressing pro-inflammatory gene expression and enhancing anti-inflammatory pathways, these metabolites contribute to a protective environment against chronic inflammation-driven cancers [69]. Cognizance of these epigenetic interactions opens new possibilities for developing microbiome-targeted interventions for cancer prevention.

3.3. Gut–Brain Axis and Systemic Effects on Cancer

Neuroimmune Interactions and Inflammatory Signaling Pathways

The gut microbiome communicates with the central nervous system via the gut–brain axis, influencing systemic inflammation and immune responses [70]. Neurotransmitters and microbial metabolites such as serotonin, dopamine, and SCFAs regulate neuroimmune interactions, impacting inflammatory signaling pathways associated with cancer progression [71]. Chronic stress and dysregulated neuroimmune signaling can exacerbate inflammation and promote tumor growth, while microbiome-derived metabolites can counteract these effects by modulating neuroinflammatory pathways [72]. This highlights the systemic influence of the microbiome on cancer prevention, emphasizing the need for a holistic approach to microbiome research in oncology. By elucidating these molecular mechanisms, researchers can identify new strategies to manipulate microbiome-derived metabolites for cancer prevention. Targeting these pathways through dietary interventions, probiotics, or microbiome-modulating therapies holds significant promise for reducing cancer risk and improving long-term health outcomes.
Beyond the immunomodulatory functions of metabolites like short-chain fatty acids (SCFAs), certain gut microbes, particularly Akkermansia muciniphila and Faecalibacterium prausnitzii, exert direct regulatory effects on the immune system that are crucial for cancer prevention. A. muciniphila contributes to gut barrier integrity by promoting mucus layer regeneration and reinforcing tight junctions, thereby limiting the entry of pro-inflammatory molecules and microbial products into systemic circulation, a process implicated in tumor development [73]. It also supports immune balance by inducing anti-inflammatory cytokines such as IL-10 and modulating dendritic cell function [74]. Likewise, F. prausnitzii, through its production of butyrate and other bioactive compounds, boosts the expansion of regulatory T cells (Tregs) while inhibiting pro-inflammatory cytokines like IL-6 and TNF-α, contributing to an anti-inflammatory and anti-tumor environment [75]. Compared with traditional probiotics such as Bifidobacteria and Lactobacilli, which primarily enhance general immune health and gut microbial balance, A. muciniphila and F. prausnitzii offer more specialized effects by targeting immune pathways involved in inflammation and cancer progression [76]. These unique immunological interactions highlight their potential as next-generation probiotics for cancer prevention.

4. Microbiome Metabolites in Cancer Therapy: Innovations and Emerging Strategies

Advancements in microbiome research have paved the way for innovative cancer therapies that leverage microbial metabolites to modulate inflammation, immune responses, and tumor progression [77]. Emerging strategies focus on harnessing the therapeutic potential of microbiome-derived metabolites through prebiotic- and probiotic-based approaches, metabolite supplementation, microbiome engineering, and metabolite-targeted drug development [78]. These interventions offer promising avenues for enhancing cancer prevention and treatment while attending to the setbacks associated with microbiome variability and metabolite bioavailability.

4.1. Prebiotic- and Probiotic-Based Therapies

Prebiotic- and probiotic-based therapies aim to modulate the gut microbiome composition to enhance the production of beneficial metabolites with anti-inflammatory and anticancer properties [79]. Prebiotics, such as dietary fibers and polyphenols, serve as substrates for beneficial gut bacteria, promoting the production of short-chain fatty acids (SCFAs) and other bioactive compounds [80]. On the other hand, probiotics introduce live beneficial microorganisms, such as Lactobacillus sp. and Bifidobacterium species, which can enhance gut health and contribute to immune regulation [81]. Clinical studies suggest that probiotic supplementation can improve responses to cancer therapies, reduce inflammation, and enhance immune checkpoint inhibitor efficacy [82]. Capsule-based delivery systems have been designed to enhance the stability and targeted gut release of SCFAs, preventing their degradation before reaching the site of action [83]. Moreover, emerging approaches focus on engineering probiotic strains to produce and release SCFAs directly within the gastrointestinal tract, amplifying their localized anti-inflammatory and anticancer effects [84]. However, further research is needed to personalize prebiotic and probiotic interventions based on individual microbiome profiles for optimal therapeutic outcomes. Table 2 outlines recent advancements in utilizing microbiome metabolites for cancer therapy.

4.2. Postbiotics and Metabolite Supplementation

Postbiotics, which refer to bioactive compounds derived from microbial metabolism, offer a promising alternative to live probiotics [95]. These include SCFAs, polyamines, indoles, and other microbial metabolites with immunomodulatory and tumor-suppressive effects [96]. Direct supplementation with these metabolites bypasses problems associated with probiotic colonization and microbiome variability, providing a more controlled therapeutic approach [97]. Butyrate, for instance, has been investigated for its ability to induce apoptosis in cancer cells and improve gut barrier integrity [98]. Similarly, indole derivatives have demonstrated anti-inflammatory properties that could be leveraged in cancer therapy [99]. The development of metabolite-based supplements holds great potential, but setbacks related to bioavailability, stability, and targeted delivery need to be addressed for clinical application.

4.3. Synthetic Microbiome Engineering for Precision Medicine

Synthetic microbiome engineering is an emerging field that aims to design and modify bacterial strains for therapeutic applications [100]. By genetically engineering bacteria to produce specific anticancer metabolites, researchers can develop precision microbiome-based interventions tailored to individual patients [101]. Engineered probiotics, for example, can be programmed to secrete SCFAs, polyamines, or bile acid derivatives that regulate inflammation and immune responses within the tumor microenvironment [102]. Also, synthetic microbiomes can enhance the efficacy of existing cancer therapies, such as immunotherapy and chemotherapy, by modulating drug metabolism and immune checkpoint activity [103]. While this approach holds great promise, trials must be carefully considered before clinical translation, and factors such as microbial stability, regulatory approvals, and potential off-target effects must be considered.

4.4. Metabolite-Targeted Drug Development

Metabolite-targeted drug development seeks to harness microbiome-derived metabolites as therapeutic agents or develop synthetic analogs that mimic their beneficial effects [104]. Small-molecule drugs inspired by microbial metabolites are being explored for their ability to modulate epigenetic, immune, and metabolic pathways involved in cancer progression [105]. For example, HDAC inhibitors based on SCFAs are being developed to regulate gene expression and suppress tumor growth [106]. In addition, bile acid analogs and indole-based compounds are being investigated for their ability to target inflammation-driven cancers [107]. The integration of microbiome-derived metabolites into drug discovery pipelines offers a novel approach to developing next-generation cancer therapeutics with enhanced efficacy and specificity [108]. The application of microbiome metabolites in cancer therapy represents a rapidly evolving field with immense therapeutic potential [109]. By integrating microbiome science with precision medicine, researchers can develop innovative interventions that target inflammation-related cancer mechanisms [110]. Continued exploration of these emerging strategies will be crucial for translating microbiome-based therapies into effective clinical treatments for cancer prevention and management.

5. Case Studies and Controversies in Microbiome Metabolite Research

Despite growing interest in the role of microbiome-derived metabolites in cancer prevention and therapy, significant controversies and roadblocks remain [111]. Clinical studies have demonstrated promising and conflicting results, featuring the complexity of microbiome–metabolite interactions in cancer [112]. While some trials support the protective effects of microbial metabolites, others reveal limitations, including inter-individual variability, metabolite stability, and inconsistent therapeutic outcomes [113]. This section explores pivotal case studies and unresolved debates surrounding microbiome metabolites in cancer research.

5.1. Clinical Trials on SCFAs in Colorectal Cancer Prevention

Short-chain fatty acids (SCFAs), particularly butyrate, have been extensively studied for their potential in colorectal cancer prevention [114]. Several clinical trials have investigated the role of dietary fiber supplementation in increasing SCFA production and reducing colorectal cancer risk [115]. Butyrate has been shown to promote apoptosis in cancer cells, enhance gut barrier integrity, and modulate immune responses [116]. Studies suggest that individuals with higher SCFA levels exhibit reduced inflammation and a lower incidence of colorectal cancer [117]. However, not all clinical trials have produced consistent results. Some studies indicate that while SCFAs exhibit anti-inflammatory and tumor-suppressive properties in early-stage cancer, they may promote tumor growth under specific conditions, such as hypoxic tumor microenvironments [118]. Likewise, individual gut microbiota composition differences may influence SCFA production, leading to variable outcomes [119]. These findings mark the need for personalized approaches to microbiome-based cancer prevention strategies. Table 3 summarizes vital case studies and ongoing controversies in microbiome metabolite research.

5.2. Challenges and Controversies in Metabolite-Based Cancer Therapy

Microbiome-derived metabolites, such as polyamines and SCFAs, have emerged as promising candidates in oncology. However, their clinical application remains challenging due to conflicting evidence and biological complexities. Among them, polyamines like spermidine and putrescine exhibit a paradoxical role in cancer progression [130]. Animal studies have shown that dietary spermidine promotes autophagy, enhances mitochondrial function, and reduces oxidative stress. For example, Yue et al. (2017)demonstrated that spermidine supplementation extended lifespan and suppressed hepatocellular carcinoma in mice via autophagy-related gene activation [131]. Likewise, Gerner et al. (2018) studied APCMin/+ mice, a model for colorectal cancer, and found that spermidine reduced tumor burden and enhanced immune surveillance through activation of cytotoxic T cells [132]. Conversely, other studies suggest that elevated polyamine levels can drive tumorigenesis, especially in advanced cancers [133]. A Phase I/II trial of polyamine blockade therapy (PBT) using difluoromethylornithine (DFMO) and sulindac in metastatic colorectal cancer showed reduced adenoma recurrence but limited efficacy in late-stage disease due to compensatory polyamine uptake [134]. Moreover, high expression of ornithine decarboxylase (ODC), the rate-limiting enzyme in polyamine synthesis, has been linked to poor prognosis in breast and prostate cancers. These contradictory findings have led to debates about the appropriateness of polyamine-targeted interventions, emphasizing the need for biomarker-driven, personalized approaches [135].
Beyond polyamines, other microbial metabolites, such as SCFAs (e.g., butyrate and propionate), indoles, and secondary bile acids, also show protective and adverse effects. Butyrate, a known histone deacetylase (HDAC) inhibitor with anti-inflammatory properties, has demonstrated anticancer potential. In a mouse model, dietary inulin increased colonic butyrate levels, reducing tumor formation and promoting apoptosis [136]. Yet, under hypoxic conditions, butyrate may serve as an energy source for cancer cells, possibly supporting tumor growth depending on the tumor microenvironment. The therapeutic use of these metabolites is further limited by their instability and short systemic half-lives [137]. SCFAs and indole derivatives are rapidly metabolized in the gut, hindering their delivery to distal tumors. For instance, oral sodium butyrate showed local benefits in ulcerative colitis but failed to achieve sufficient plasma levels for treating extraintestinal malignancies [138]. Inter-individual differences in gut microbiota composition also influence metabolite production and treatment outcomes. A recent Phase II trial of Akkermansia muciniphila as an immunotherapy adjuvant found that responders had gut profiles enriched in SCFAs and indole-3-lactic acid. At the same time, non-responders lacked these metabolites despite receiving the same probiotic [139]. These findings underscore the need for precision microbiome strategies based on individual microbial, dietary, and genetic factors.
Another major challenge is the lack of standardized dosing protocols for metabolite-based therapies. Unlike conventional drugs, these compounds lack well-defined pharmacokinetic and pharmacodynamic data. In a Phase I trial, indole-3-carbinol (a dietary precursor of indole metabolites) showed dose-related effects on estrogen metabolism and immunity in women with cervical intraepithelial neoplasia, but inconsistent microbial conversion led to varied responses [140]. Safety concerns also persist. Though generally considered safe, long-term use of probiotics, prebiotics, and postbiotics can disrupt gut microbial balance. Rare cases of bacteremia and metabolic disturbances have been reported in immunocompromised individuals following high-dose probiotic use. Furthermore, interactions between microbial metabolites and anticancer drugs may lead to unintended immune or metabolic effects [141]. While microbiome-derived metabolites represent an exciting frontier in cancer prevention and therapy, their clinical integration faces multiple hurdles, including mechanistic ambiguities, delivery barriers, patient variability, and safety concerns. Future progress will depend on well-designed clinical trials, the identification of reliable biomarkers, and tailored interventions aligned with individual microbiota and disease profiles [142].

6. Complications, Limitations, and Future Perspectives

Despite the promising potential of microbiome-derived metabolites in cancer prevention and therapy, several issues must be addressed before these strategies can be widely implemented in clinical settings [143]. Variability in individual microbiomes, limitations in metabolite bioavailability, and regulatory concerns present significant hurdles [144]. However, emerging research directions, including personalized microbiome-based therapies and integrative multi-omics approaches, offer opportunities to overcome these obstacles and enhance the therapeutic potential of microbiome metabolites in oncology [145].

6.1. Complications and Limitations

Despite significant progress in microbiome metabolite research, several hurdles and limitations persist, including variability in individual microbiomes, mechanistic complexities, and translational hurdles in clinical applications [146]. Table 4 outlines the chief difficulties, limitations, and future directions in microbiome metabolite research.

6.1.1. Variability in Individual Microbiomes

One of the biggest trials in microbiome-based cancer therapies is the high degree of variability in microbiome composition among individuals [152]. Factors such as genetics, diet, lifestyle, and antibiotic exposure influence the gut microbiota, leading to differences in metabolite production and therapeutic responses [153]. This variability makes it difficult to develop universal microbiome-targeted interventions [154]. Personalized approaches that consider individual microbiome profiles are necessary to maximize therapeutic efficacy and minimize unintended effects [155].

6.1.2. Bioavailability and Stability of Metabolites

Many microbiome-derived metabolites, such as SCFAs and indole derivatives, have limited stability and bioavailability, reducing their effectiveness as therapeutic agents [156]. These metabolites are often rapidly metabolized or degraded in the gut before they can exert systemic effects [157]. Developing novel drug delivery systems, such as encapsulated formulations or targeted release mechanisms, could enhance metabolite stability and improve their therapeutic impact [158]. Further research is needed to optimize metabolite-based interventions for clinical application.

6.1.3. Regulatory and Ethical Considerations

Regulatory approval for microbiome-based therapies presents another significant challenge. The classification of probiotics, prebiotics, and postbiotics as dietary supplements rather than pharmaceutical drugs [159] complicates the regulatory process for their clinical use in cancer treatment [160]. Standardized guidelines for quality control, safety, and efficacy are required to ensure the consistency and reliability of microbiome-based interventions [161]. In the same vein, ethical concerns related to microbiome manipulation, such as unintended long-term effects and potential microbiome engineering risks, must be carefully evaluated before these therapies can be widely adopted [162].

6.2. Future Research Directions

6.2.1. Personalized Microbiome-Based Therapies

To overcome microbiome variability, future research should focus on personalized approaches that tailor microbiome-based therapies to individual patients [163]. Advances in microbiome sequencing and computational modeling can help identify specific microbial signatures associated with cancer risk and therapeutic response [164]. Personalized interventions, such as customized probiotics, targeted prebiotics, or patient-specific metabolite supplementation, could improve treatment outcomes while minimizing adverse effects.

6.2.2. Advanced Multi-Omics Approaches for Precision Medicine

Integrating multi-omics technologies including metagenomics, metabolomics, transcriptomics, and proteomics will provide a more comprehensive interpretation of microbiome–metabolite interactions in cancer [165]. By combining these datasets, researchers can uncover novel biomarkers, predict therapeutic responses, and develop more effective microbiome-targeted interventions [166]. Machine learning and artificial intelligence (AI) will play a crucial role in analyzing complex microbiome data and identifying optimal therapeutic strategies [167].

6.2.3. Integration of Microbiome Metabolites with Immunotherapy and Chemotherapy

Another promising research direction is the integration of microbiome metabolites with existing cancer treatments, such as immunotherapy and chemotherapy [168]. Microbiome-derived metabolites have been shown to enhance immune checkpoint blockade therapy, modulate the tumor microenvironment, and reduce treatment-related side effects [169]. Future studies should explore how microbial metabolites can be combined with conventional cancer therapies to improve patient outcomes [170]. Additionally, clinical trials investigating microbiome metabolite-based adjuvants will be essential for translating these findings into practical cancer treatment strategies [171]. As microbiome research advances, treating these problems and embracing emerging research directions will be crucial for harnessing the full therapeutic potential of microbiome metabolites in cancer prevention and treatment. By integrating personalized medicine, innovative drug delivery systems, and precision oncology approaches, microbiome-based interventions may become a transformative strategy in the fight against cancer.

7. Conclusions

The role of microbiome-derived metabolites in cancer prevention and treatment has gained significant attention, spotlighting their potential as essential modulators of inflammation and tumor progression. This review has explored the diverse functions of microbial metabolites, such as short-chain fatty acids (SCFAs), polyamines, and bile acid derivatives, in regulating immune responses, epigenetic modifications, and metabolic pathways associated with cancer. While promising preclinical and clinical studies suggest the therapeutic value of microbiome-based interventions, trials related to bioavailability, metabolism, and individual variability must be addressed to optimize their clinical application. Future research should focus on developing personalized microbiome-based therapies that consider individual variations in gut microbiota composition and metabolite production. Advanced drug delivery strategies, such as encapsulation and targeted release mechanisms, could enhance the stability and bioactivity of microbial metabolites, improving their therapeutic efficacy. Furthermore, integrating microbiome-derived interventions with conventional cancer treatments, such as immunotherapy and chemotherapy, presents a novel avenue for enhancing treatment responses and reducing adverse effects. As our grasp of microbiome–metabolite interactions continues to evolve, translating these findings into clinical applications will require interdisciplinary collaboration, rigorous clinical trials, and the development of regulatory frameworks for microbiome-based therapeutics. By managing current issues and leveraging emerging innovations, microbiome-derived metabolites have the potential to revolutionize cancer prevention and treatment strategies, paving the way for more effective and personalized approaches in oncology.

Author Contributions

Conceptualization, A.N.M. and D.B.; methodology, A.N.M.; software, A.N.M.; validation, D.B.; investigation, A.N.M.; writing original draft preparation, A.N.M.; writing review and editing, A.N.M. and D.B.; visualization, A.N.M. and D.B.; supervision, D.B.; project administration, D.B.; and funding acquisition, D.B. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Priorities Research Program grant NPRP 14S0311–210033, awarded to Dietrich Büsselberg (January 2023–present) from the Qatar National Research Fund (QNRF, a member of the Qatar Foundation). The Biomedical Research Program at Weill Cornell Medicine-Qatar, a program funded by the Qatar Foundation, covered the publication costs for this work. The statements made herein are solely the responsibility of the authors.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

SCFAsShort-chain fatty acids
AHRAryl hydrocarbon receptor
LCALithocholic acid
TregsRegulatory T cells
HDACHistone deacetylase
NKNatural killer
AIArtificial intelligence

References

  1. Multhoff, G.; Molls, M.; Radons, J. Chronic Inflammation in Cancer Development. Front. Immunol. 2012, 2, 98. [Google Scholar] [CrossRef]
  2. Liu, Q.; Yang, Y.; Pan, M.; Yang, F.; Yu, Y.; Qian, Z. Role of the Gut Microbiota in Tumorigenesis and Treatment. Theranostics 2024, 14, 2304–2328. [Google Scholar] [CrossRef]
  3. Anwer, E.K.E.; Ajagbe, M.; Sherif, M.; Musaibah, A.S.; Mahmoud, S.; ElBanbi, A.; Abdelnaser, A. Gut Microbiota Secondary Metabolites: Key Roles in GI Tract Cancers and Infectious Diseases. Biomedicines 2025, 13, 100. [Google Scholar] [CrossRef]
  4. Bhatt, A.P.; Redinbo, M.R.; Bultman, S.J. The Role of the Microbiome in Cancer Development and Therapy. CA. Cancer J. Clin. 2017, 67, 326–344. [Google Scholar] [CrossRef]
  5. Mullen, N.J.; Singh, P.K. Nucleotide Metabolism: A Pan-Cancer Metabolic Dependency. Nat. Rev. Cancer 2023, 23, 275–294. [Google Scholar] [CrossRef]
  6. Ma, Z.; Zuo, T.; Frey, N.; Rangrez, A.Y. A Systematic Framework for Understanding the Microbiome in Human Health and Disease: From Basic Principles to Clinical Translation. Signal Transduct. Target. Ther. 2024, 9, 237. [Google Scholar] [CrossRef]
  7. Schwartz, D.J.; Rebeck, O.N.; Dantas, G. Complex Interactions between the Microbiome and Cancer Immune Therapy. Crit. Rev. Clin. Lab. Sci. 2019, 56, 567–585. [Google Scholar] [CrossRef]
  8. Valdés-González, J.A.; Sánchez, M.; Moratilla-Rivera, I.; Iglesias, I.; Gómez-Serranillos, M.P. Immunomodulatory, Anti-Inflammatory, and Anti-Cancer Properties of Ginseng: A Pharmacological Update. Molecules 2023, 28, 3863. [Google Scholar] [CrossRef]
  9. Kunst, C.; Schmid, S.; Michalski, M.; Tümen, D.; Buttenschön, J.; Müller, M.; Gülow, K. The Influence of Gut Microbiota on Oxidative Stress and the Immune System. Biomedicines 2023, 11, 1388. [Google Scholar] [CrossRef]
  10. Getsina, M.; Chernevskaya, E.; Beloborodova, N.; Golovnya, E.; Polyakov, P.; Kushlinskii, N. Features of Metabolites and Biomarkers in Inflammatory and Infectious Complications of Childhood Cancers. Biomedicines 2024, 12, 2101. [Google Scholar] [CrossRef]
  11. Nemoto, H.; Otake, M.; Matsumoto, T.; Izutsu, R.; Jehung, J.P.; Goto, K.; Osaki, M.; Mayama, M.; Shikanai, M.; Kobayashi, H.; et al. Prevention of Tumor Progression in Inflammation-Related Carcinogenesis by Anti-Inflammatory and Anti-Mutagenic Effects Brought about by Ingesting Fermented Brown Rice and Rice Bran with Aspergillus Oryzae (FBRA). J. Funct. Foods 2022, 88, 104907. [Google Scholar] [CrossRef]
  12. Mederle, A.L.; Semenescu, A.; Drăghici, G.A.; Dehelean, C.A.; Vlăduț, N.-V.; Nica, D.V. Sodium Butyrate: A Multifaceted Modulator in Colorectal Cancer Therapy. Medicina 2025, 61, 136. [Google Scholar] [CrossRef]
  13. Du, Y.; He, C.; An, Y.; Huang, Y.; Zhang, H.; Fu, W.; Wang, M.; Shan, Z.; Xie, J.; Yang, Y.; et al. The Role of Short Chain Fatty Acids in Inflammation and Body Health. Int. J. Mol. Sci. 2024, 25, 7379. [Google Scholar] [CrossRef]
  14. Liao, R.; Chen, X.; Cao, Q.; Bai, L.; Ma, C.; Dai, Z.; Dong, C. AMD1 Promotes Breast Cancer Aggressiveness via a Spermidine-EIF5A Hypusination-TCF4 Axis. Breast Cancer Res. 2024, 26, 70. [Google Scholar] [CrossRef]
  15. Conteduca, V.; Caffo, O.; Scarpi, E.; Sepe, P.; Galli, L.; Fratino, L.; Maines, F.; Chiuri, V.E.; Santoni, M.; Zanardi, E.; et al. Immune Modulation in Prostate Cancer Patients Treated with Androgen Receptor (AR)-Targeted Therapy. J. Clin. Med. 2020, 9, 1950. [Google Scholar] [CrossRef]
  16. Malhotra, P.; Palanisamy, R.; Caparros-Martin, J.A.; Falasca, M. Bile Acids and Microbiota Interplay in Pancreatic Cancer. Cancers 2023, 15, 3573. [Google Scholar] [CrossRef]
  17. Feitelson, M.A.; Arzumanyan, A.; Medhat, A.; Spector, I. Short-Chain Fatty Acids in Cancer Pathogenesis. Cancer Metastasis Rev. 2023, 42, 677–698. [Google Scholar] [CrossRef]
  18. Jiang, H.; Zeng, W.; Zhang, X.; Li, Y.; Wang, Y.; Peng, A.; Cao, D. Gut Microbiota and Its Metabolites in Non-Small Cell Lung Cancer and Brain Metastasis: From Alteration to Potential Microbial Markers and Drug Targets. Front. Cell. Infect. Microbiol. 2024, 13, 1211855. [Google Scholar] [CrossRef]
  19. Tian, C.; Deng, S.; Yang, M.; Bai, B.; Pan, Y.; Xie, G.; Zhao, D.; Wei, L. Indole-3-Carbinol and Its Main Derivative 3,3′-Diindolylmethane: Regulatory Roles and Therapeutic Potential in Liver Diseases. Biomed. Pharmacother. 2024, 180, 117525. [Google Scholar] [CrossRef]
  20. Nowotarski, S.L.; Woster, P.M.; Casero, R.A. Polyamines and Cancer: Implications for Chemotherapy and Chemoprevention. Expert Rev. Mol. Med. 2013, 15, e3. [Google Scholar] [CrossRef]
  21. Mowat, C.; Dhatt, J.; Bhatti, I.; Hamie, A.; Baker, K. Short Chain Fatty Acids Prime Colorectal Cancer Cells to Activate Antitumor Immunity. Front. Immunol. 2023, 14, 1190810. [Google Scholar] [CrossRef]
  22. Ke, J.; Zhang, C.; Wang, L.; Xie, F.; Wu, H.-Y.; Li, T.; Bian, C.-W.; Wu, R.-L. Lipopolysaccharide Promotes Cancer Cell Migration and Invasion through METTL3/PI3K/AKT Signaling in Human Cholangiocarcinoma. Heliyon 2024, 10, e29683. [Google Scholar] [CrossRef]
  23. Stø, K.; Valeur, J.; Ueland, T.; Malmstrøm, G.H.; Bjerkeli, V.; Trøseid, M.; Hov, J.R.; Holm, K.; Vestad, B.; Halvorsen, B.; et al. Fecal Level of Butyric Acid, a Microbiome-Derived Metabolite, Is Increased in Patients with Severe Carotid Atherosclerosis. Sci. Rep. 2022, 12, 22378. [Google Scholar] [CrossRef]
  24. Wang, J.; Zhu, N.; Su, X.; Gao, Y.; Yang, R. Gut-Microbiota-Derived Metabolites Maintain Gut and Systemic Immune Homeostasis. Cells 2023, 12, 793. [Google Scholar] [CrossRef]
  25. Chen, W.-J.; Chen, Y.-T.; Ko, J.-L.; Chen, J.-Y.; Zheng, J.-Y.; Liao, J.-W.; Ou, C.-C. Butyrate Modulates Gut Microbiota and Anti-Inflammatory Response in Attenuating Cisplatin-Induced Kidney Injury. Biomed. Pharmacother. 2024, 181, 117689. [Google Scholar] [CrossRef]
  26. Klys, Y.G.; Kerimov, T.R.; Savosko, S.I.; Osadchuk, Y.S.; Smirnov, S.M.; Natrus, L.V. Unfolded Protein Response in Gastric Glandulocytes of Rats with the Pharmacological Correction of Type 2 Diabetes. Gastroenterology 2023, 57, 127–134. [Google Scholar] [CrossRef]
  27. Ramesh, V.; Gollavilli, P.N.; Pinna, L.; Siddiqui, M.A.; Turtos, A.M.; Napoli, F.; Antonelli, Y.; Leal-Egaña, A.; Havelund, J.F.; Jakobsen, S.T.; et al. Propionate Reinforces Epithelial Identity and Reduces Aggressiveness of Lung Carcinoma. EMBO Mol. Med. 2023, 15, e17836. [Google Scholar] [CrossRef]
  28. Carriche, G.M.; Almeida, L.; Stüve, P.; Velasquez, L.; Dhillon-LaBrooy, A.; Roy, U.; Lindenberg, M.; Strowig, T.; Plaza-Sirvent, C.; Schmitz, I.; et al. Regulating T-Cell Differentiation through the Polyamine Spermidine. J. Allergy Clin. Immunol. 2021, 147, 335–348.e11. [Google Scholar] [CrossRef]
  29. Ruggieri, V.; Scaricamazza, S.; Bracaglia, A.; D’Ercole, C.; Parisi, C.; D’Angelo, P.; Proietti, D.; Cappelletti, C.; Macone, A.; Lozanoska-Ochser, B.; et al. Polyamine Metabolism Dysregulation Contributes to Muscle Fiber Vulnerability in ALS. Cell Rep. 2025, 44, 115123. [Google Scholar] [CrossRef]
  30. Jiang, D.; Ji, C.; Zhou, X.; Wang, Z.; Sun, Q.; Wang, X.; An, X.; Ling, W.; Kang, B. Pathway Analysis of Spermidine Anti-Oxidative Stress and Inducing Autophagy in Granulosa Cells of Sichuan White Geese. Theriogenology 2024, 215, 290–301. [Google Scholar] [CrossRef]
  31. Song, Q.; Wang, Y.; Liu, S. Subtype-Specific Transcription Factors Affect Polyamine Metabolism and the Tumor Microenvironment in Breast Cancer. Cancer Innov. 2025, 4, e138. [Google Scholar] [CrossRef]
  32. Vazquez-Medina, A.; Rodriguez-Trujillo, N.; Ayuso-Rodriguez, K.; Marini-Martinez, F.; Angeli-Morales, R.; Caussade-Silvestrini, G.; Godoy-Vitorino, F.; Chorna, N. Exploring the Interplay between Running Exercises, Microbial Diversity, and Tryptophan Metabolism along the Microbiota-Gut-Brain Axis. Front. Microbiol. 2024, 15, 1326584. [Google Scholar] [CrossRef]
  33. Hezaveh, K.; Shinde, R.S.; Klötgen, A.; Halaby, M.J.; Lamorte, S.; Ciudad, M.T.; Quevedo, R.; Neufeld, L.; Liu, Z.Q.; Jin, R.; et al. Tryptophan-Derived Microbial Metabolites Activate the Aryl Hydrocarbon Receptor in Tumor-Associated Macrophages to Suppress Anti-Tumor Immunity. Immunity 2022, 55, 324–340.e8. [Google Scholar] [CrossRef]
  34. Raizandha, M.A.; Hidayatullah, F.; Kloping, Y.P.; Rizaldi, F. Neutrophil-Lymphocyte Ratio and Fournier Gangrene Severity Index Are Not Prognostic Factors of Mortality in Fournier Gangrene Patients. Universa Med. 2022, 41, 71–78. [Google Scholar] [CrossRef]
  35. Suraya, R.; Nagano, T.; Kobayashi, K.; Nishimura, Y. Microbiome as a Target for Cancer Therapy. Integr. Cancer Ther. 2020, 19, 1534735420920721. [Google Scholar] [CrossRef]
  36. Zhang, B.; Jiang, X.; Yu, Y.; Cui, Y.; Wang, W.; Luo, H.; Stergiadis, S.; Wang, B. Rumen Microbiome-Driven Insight into Bile Acid Metabolism and Host Metabolic Regulation. ISME J. 2024, 18, wrae098. [Google Scholar] [CrossRef]
  37. Choi, S.I.; Son, J.H.; Kim, N.; Kim, Y.S.; Nam, R.H.; Park, J.H.; Song, C.-H.; Yu, J.E.; Lee, D.H.; Yoon, K.; et al. Changes in Cecal Microbiota and Short-Chain Fatty Acid During Lifespan of the Rat. J. Neurogastroenterol. Motil. 2021, 27, 134–146. [Google Scholar] [CrossRef]
  38. Kumar, R.; Grinberg, A.V.; Li, H.; Kuo, T.-H.; Sako, D.; Krishnan, L.; Liharska, K.; Li, J.; Grenha, R.; Maguire, M.C.; et al. Functionally Diverse Heteromeric Traps for Ligands of the Transforming Growth Factor-β Superfamily. Sci. Rep. 2021, 11, 18341. [Google Scholar] [CrossRef]
  39. Wang, Z.; Gao, B.; Liu, X.; Li, A. The Mediating Role of Metabolites between Gut Microbiome and Hirschsprung Disease: A Bidirectional Two-Step Mendelian Randomization Study. Front. Pediatr. 2024, 12, 1371933. [Google Scholar] [CrossRef]
  40. Liu, C.; Fu, L.; Wang, Y.; Yang, W. Influence of the Gut Microbiota on Immune Cell Interactions and Cancer Treatment. J. Transl. Med. 2024, 22, 939. [Google Scholar] [CrossRef]
  41. Jaouhari, Y.; Ferreira-Santos, P.; Disca, V.; Oliveira, H.; Martoccia, M.; Travaglia, F.; Gullón, B.; Mateus, N.; Coïsson, J.D.; Bordiga, M. Carbohydrases Treatment on Blueberry Pomace: Influence on Chemical Composition and Bioactive Potential. LWT 2024, 206, 116573. [Google Scholar] [CrossRef]
  42. Ranf, S.; Scheel, D.; Lee, J. Challenges in the Identification of Microbe-associated Molecular Patterns in Plant and Animal Innate Immunity: A Case Study with Bacterial Lipopolysaccharide. Mol. Plant Pathol. 2016, 17, 1165–1169. [Google Scholar] [CrossRef]
  43. Steimle, A.; Autenrieth, I.B.; Frick, J.-S. Structure and Function: Lipid A Modifications in Commensals and Pathogens. Int. J. Med. Microbiol. 2016, 306, 290–301. [Google Scholar] [CrossRef]
  44. Hrncir, T. Gut Microbiota Dysbiosis: Triggers, Consequences, Diagnostic and Therapeutic Options. Microorganisms 2022, 10, 578. [Google Scholar] [CrossRef]
  45. Ciesielska, A.; Matyjek, M.; Kwiatkowska, K. TLR4 and CD14 Trafficking and Its Influence on LPS-Induced pro-Inflammatory Signaling. Cell. Mol. Life Sci. 2021, 78, 1233–1261. [Google Scholar] [CrossRef]
  46. Landskron, G.; De la Fuente, M.; Thuwajit, P.; Thuwajit, C.; Hermoso, M.A. Chronic Inflammation and Cytokines in the Tumor Microenvironment. J. Immunol. Res. 2014, 2014, 149185. [Google Scholar] [CrossRef]
  47. Śliżewska, K.; Markowiak-Kopeć, P.; Śliżewska, W. The Role of Probiotics in Cancer Prevention. Cancers 2020, 13, 20. [Google Scholar] [CrossRef]
  48. Mo, C.; Lou, X.; Xue, J.; Shi, Z.; Zhao, Y.; Wang, F.; Chen, G. The Influence of Akkermansia Muciniphila on Intestinal Barrier Function. Gut Pathog. 2024, 16, 41. [Google Scholar] [CrossRef]
  49. Ferreira-Halder, C.V.; de Sousa Faria, A.V.; Andrade, S.S. Action and Function of Faecalibacterium Prausnitzii in Health and Disease. Best Pract. Res. Clin. Gastroenterol. 2017, 31, 643–648. [Google Scholar] [CrossRef]
  50. Tiwari, A.; Ika Krisnawati, D.; Susilowati, E.; Mutalik, C.; Kuo, T.-R. Next-Generation Probiotics and Chronic Diseases: A Review of Current Research and Future Directions. J. Agric. Food Chem. 2024, 72, 27679–27700. [Google Scholar] [CrossRef]
  51. Jin, Y.; Dong, H.; Xia, L.; Yang, Y.; Zhu, Y.; Shen, Y.; Zheng, H.; Yao, C.; Wang, Y.; Lu, S. The Diversity of Gut Microbiome Is Associated With Favorable Responses to Anti–Programmed Death 1 Immunotherapy in Chinese Patients With NSCLC. J. Thorac. Oncol. 2019, 14, 1378–1389. [Google Scholar] [CrossRef] [PubMed]
  52. Mafe, A.N.; Büsselberg, D. Modulation of the Neuro–Cancer Connection by Metabolites of Gut Microbiota. Biomolecules 2025, 15, 270. [Google Scholar] [CrossRef] [PubMed]
  53. Baruch, E.N.; Youngster, I.; Ben-Betzalel, G.; Ortenberg, R.; Lahat, A.; Katz, L.; Adler, K.; Dick-Necula, D.; Raskin, S.; Bloch, N.; et al. Fecal Microbiota Transplant Promotes Response in Immunotherapy-Refractory Melanoma Patients. Science 2021, 371, 602–609. [Google Scholar] [CrossRef] [PubMed]
  54. Liu, C.; Su, W.; Tan, Z.; Zhang, J.; Dong, W. The Interaction between Microbiota and Immune in Intestinal Inflammatory Diseases: Global Research Status and Trends. Front. Cell. Infect. Microbiol. 2023, 13, 1128249. [Google Scholar] [CrossRef]
  55. Yu, Y.; Zi, Y.; Fu, R.; Fu, B.; Li, C.; Lv, Y.; Li, Z.; Wang, H.; Leng, J. Effects of Dietary Energy Levels on Microorganisms and Short-Chain Fatty Acids of Rumen and Tight Junction Proteins in Honghe Yellow Cattle. Front. Microbiol. 2024, 15, 1335818. [Google Scholar] [CrossRef]
  56. Maseda, D.; Manfredo-Vieira, S.; Payne, A.S. T Cell and Bacterial Microbiota Interaction at Intestinal and Skin Epithelial Interfaces. Discov. Immunol. 2023, 2, kyad024. [Google Scholar] [CrossRef]
  57. Ju, H.J.; Song, W.H.; Shin, J.H.; Lee, J.H.; Bae, J.M.; Lee, Y.B.; Lee, M. Characterization of Gut Microbiota in Patients with Active Spreading Vitiligo Based on Whole-Genome Shotgun Sequencing. Int. J. Mol. Sci. 2025, 26, 2939. [Google Scholar] [CrossRef]
  58. Mishima, R.; Ejima, R.; Arai, S.; Horigome, A.; Mitsuyama, E.; Kaneko, H.; Yamaguchi, K.; Kamezaki, K.; Togashi, Y.; Nakamura, K.; et al. Synbiotic Combination of Bifidobacterium Longum BB536 and Lactulose Improves the Presenteeism of Healthy Adults Associated with Aromatic Lactic Acids—A Single-Arm, Open-Label Study. Gut Microbes Rep. 2025, 2. [Google Scholar] [CrossRef]
  59. Li, L.; He, S.; Liao, B.; Wang, M.; Lin, H.; Hu, B.; Lan, X.; Shu, Z.; Zhang, C.; Yu, M.; et al. Orally Administrated Hydrogel Harnessing Intratumoral Microbiome and Microbiota-Related Immune Responses for Potentiated Colorectal Cancer Treatment. Research 2024, 7, 0364. [Google Scholar] [CrossRef]
  60. Zhou, Y.; Han, W.; Feng, Y.; Wang, Y.; Sun, T.; Xu, J. Microbial Metabolites Affect Tumor Progression, Immunity and Therapy Prediction by Reshaping the Tumor Microenvironment (Review). Int. J. Oncol. 2024, 65, 73. [Google Scholar] [CrossRef]
  61. Vojdani, A.; Koksoy, S.; Vojdani, E.; Engelman, M.; Benzvi, C.; Lerner, A. Natural Killer Cells and Cytotoxic T Cells: Complementary Partners against Microorganisms and Cancer. Microorganisms 2024, 12, 230. [Google Scholar] [CrossRef]
  62. O’Riordan, K.J.; Collins, M.K.; Moloney, G.M.; Knox, E.G.; Aburto, M.R.; Fülling, C.; Morley, S.J.; Clarke, G.; Schellekens, H.; Cryan, J.F. Short Chain Fatty Acids: Microbial Metabolites for Gut-Brain Axis Signalling. Mol. Cell. Endocrinol. 2022, 546, 111572. [Google Scholar] [CrossRef]
  63. Kumari, S.; Srilatha, M.; Nagaraju, G.P. Effect of Gut Dysbiosis on Onset of GI Cancers. Cancers 2024, 17, 90. [Google Scholar] [CrossRef]
  64. Niam, M.S.; Nastiti, N.A.; Dradjat, R.S.; Rudijanto, A.; Sujuti, H. Do Tumor Locations and Stages at Diagnosis Predict the 5-Year Survival Outcome in Patients with Colorectal Cancer? Open Access Maced. J. Med. Sci. 2022, 10, 1663–1666. [Google Scholar] [CrossRef]
  65. Nshanian, M.; Gruber, J.J.; Geller, B.S.; Chleilat, F.; Lancaster, S.M.; White, S.M.; Alexandrova, L.; Camarillo, J.M.; Kelleher, N.L.; Zhao, Y.; et al. Short-Chain Fatty Acid Metabolites Propionate and Butyrate Are Unique Epigenetic Regulatory Elements Linking Diet, Metabolism and Gene Expression. Nat. Metab. 2025, 7, 196–211. [Google Scholar] [CrossRef]
  66. Wang, P.; Chen, L.-L.; Xiong, Y.; Ye, D. Metabolite Regulation of Epigenetics in Cancer. Cell Rep. 2024, 43, 114815. [Google Scholar] [CrossRef]
  67. Reva, K.; Laranjinha, J.; Rocha, B.S. Epigenetic Modifications Induced by the Gut Microbiota May Result from What We Eat: Should We Talk about Precision Diet in Health and Disease? Metabolites 2023, 13, 375. [Google Scholar] [CrossRef]
  68. Babar, Q.; Saeed, A.; Tabish, T.A.; Pricl, S.; Townley, H.; Thorat, N. Novel Epigenetic Therapeutic Strategies and Targets in Cancer. Biochim. Biophys. Acta—Mol. Basis Dis. 2022, 1868, 166552. [Google Scholar] [CrossRef]
  69. Tripathi, S.; Sharma, Y.; Kumar, D. Unveiling the Link between Chronic Inflammation and Cancer. Metab. Open 2025, 25, 100347. [Google Scholar] [CrossRef]
  70. Huang, Z.; Boekhorst, J.; Fogliano, V.; Capuano, E.; Wells, J.M. Impact of High-Fiber or High-Protein Diet on the Capacity of Human Gut Microbiota To Produce Tryptophan Catabolites. J. Agric. Food Chem. 2023, 71, 6956–6966. [Google Scholar] [CrossRef]
  71. Charitos, I.A.; Inchingolo, A.M.; Ferrante, L.; Inchingolo, F.; Inchingolo, A.D.; Castellaneta, F.; Cotoia, A.; Palermo, A.; Scacco, S.; Dipalma, G. The Gut Microbiota’s Role in Neurological, Psychiatric, and Neurodevelopmental Disorders. Nutrients 2024, 16, 4404. [Google Scholar] [CrossRef] [PubMed]
  72. Zou, S.; Li, Y.; Zou, Q.; Yang, M.; Li, H.; Niu, R.; Lai, H.; Wang, J.; Yang, X.; Zhou, L. Gut Microbiota and Serum Metabolomic Alterations in Modulating the Impact of Fecal Microbiota Transplantation on Ciprofloxacin-Induced Seizure Susceptibility. Front. Microbiol. 2024, 15, 1403892. [Google Scholar] [CrossRef] [PubMed]
  73. Neurath, M.F.; Artis, D.; Becker, C. The Intestinal Barrier: A Pivotal Role in Health, Inflammation, and Cancer. Lancet Gastroenterol. Hepatol. 2025, 10, 573–592. [Google Scholar] [CrossRef] [PubMed]
  74. Effendi, R.M.R.A.; Anshory, M.; Kalim, H.; Dwiyana, R.F.; Suwarsa, O.; Pardo, L.M.; Nijsten, T.E.C.; Thio, H.B. Akkermansia Muciniphila and Faecalibacterium Prausnitzii in Immune-Related Diseases. Microorganisms 2022, 10, 2382. [Google Scholar] [CrossRef]
  75. Zhou, L.; Zhang, M.; Wang, Y.; Dorfman, R.G.; Liu, H.; Yu, T.; Chen, X.; Tang, D.; Xu, L.; Yin, Y.; et al. Faecalibacterium Prausnitzii Produces Butyrate to Maintain Th17/Treg Balance and to Ameliorate Colorectal Colitis by Inhibiting Histone Deacetylase 1. Inflamm. Bowel Dis. 2018, 24, 1926–1940. [Google Scholar] [CrossRef]
  76. Jan, T.; Negi, R.; Sharma, B.; Kumar, S.; Singh, S.; Rai, A.K.; Shreaz, S.; Rustagi, S.; Chaudhary, N.; Kaur, T.; et al. Next Generation Probiotics for Human Health: An Emerging Perspective. Heliyon 2024, 10, e35980. [Google Scholar] [CrossRef]
  77. Farhadi Rad, H.; Tahmasebi, H.; Javani, S.; Hemati, M.; Zakerhamidi, D.; Hosseini, M.; Alibabaei, F.; Banihashemian, S.Z.; Oksenych, V.; Eslami, M. Microbiota and Cytokine Modulation: Innovations in Enhancing Anticancer Immunity and Personalized Cancer Therapies. Biomedicines 2024, 12, 2776. [Google Scholar] [CrossRef]
  78. Williams, L.M.; Cao, S. Harnessing and Delivering Microbial Metabolites as Therapeutics via Advanced Pharmaceutical Approaches. Pharmacol. Ther. 2024, 256, 108605. [Google Scholar] [CrossRef]
  79. Alkhaldy, A.A. Awareness, Knowledge, and Beliefs about Probiotics and Prebiotics among Saudi Adults: A Cross-Sectional Study. Front. Immunol. 2024, 15, 1464622. [Google Scholar] [CrossRef]
  80. Barber, C.; Sabater, C.; Dolores Frutos, M.; Vallejo, F.; Guyonnet, D.; Daniel, N.; Guarner, F.; Carlos Espín, J.; Margolles, A.; Azpiroz, F. Effects of a (Poly)Phenol-Rich Berry Mix on Gas Production in Healthy Individuals: An Integrated Clinical, Metagenomic, and Metabolomic Proof-of-Concept Study. J. Funct. Foods 2024, 113, 106032. [Google Scholar] [CrossRef]
  81. Tian, Q.; Ye, H.; Zhou, X.; Wang, J.; Zhang, L.; Sun, W.; Duan, C.; Fan, M.; Zhou, W.; Bi, C.; et al. Evaluating the Health Risk of Probiotic Supplements from the Perspective of Antimicrobial Resistance. Microbiol. Spectr. 2025, 13, e0001924. [Google Scholar] [CrossRef]
  82. Nama, A.S.A.; Sandeepa, G.M.; Buddolla, V.; Mastan, A. Advances in Understanding Therapeutic Mechanisms of Probiotics in Cancer Management, with Special Emphasis on Breast Cancer: A Comprehensive Review. Eur. J. Pharmacol. 2025, 995, 177410. [Google Scholar] [CrossRef]
  83. Liu, J.; Li, X.; Zhang, X.; Wang, R.; Han, S.; Qin, P.; Xing, X.-H.; Zhang, C. Gut Lumen-Targeted Oral Delivery System for Bioactive Agents to Regulate Gut Microbiome. J. Futur. Foods 2022, 2, 307–325. [Google Scholar] [CrossRef]
  84. Markowiak-Kopeć, P.; Śliżewska, K. The Effect of Probiotics on the Production of Short-Chain Fatty Acids by Human Intestinal Microbiome. Nutrients 2020, 12, 1107. [Google Scholar] [CrossRef]
  85. LaPointe, G.; Rogers, M.A. Microorganisms Special Issue “How Do Food and Probiotics Influence the Composition and Activity of the Gut Microbiota? ” Microorganisms 2022, 10, 2097. [Google Scholar] [CrossRef]
  86. Ghosh, S.; Yang, X.; Wang, L.; Zhang, C.; Zhao, L. Active Phase Prebiotic Feeding Alters Gut Microbiota, Induces Weight-Independent Alleviation of Hepatic Steatosis and Serum Cholesterol in High-Fat Diet-Fed Mice. Comput. Struct. Biotechnol. J. 2021, 19, 448–458. [Google Scholar] [CrossRef]
  87. Al-Qadami, G.H.; Secombe, K.R.; Subramaniam, C.B.; Wardill, H.R.; Bowen, J.M. Gut Microbiota-Derived Short-Chain Fatty Acids: Impact on Cancer Treatment Response and Toxicities. Microorganisms 2022, 10, 2048. [Google Scholar] [CrossRef]
  88. Poveshchenko, A.F.; Kabakov, A.V.; Bodrova, N.R.; Kapustina, V.I.; Kazakov, O.V.; Koldysheva, E.V.; Cherkas, V.N.; Afonyushkin, V.N. Microbiota of the Mammary Gland in Wistar Rats with Chemically Induced Breast Cancer after Treatment. Bull. Exp. Biol. Med. 2024, 178, 223–226. [Google Scholar] [CrossRef]
  89. Islam, F.; Azmat, F.; Imran, A.; Zippi, M.; Hong, W.; Tariq, F.; Shehzadi, U.; Fatima, A.; Safdar, M.; Ahmed, H.; et al. Role of Postbiotics in Food and Health: A Comprehensive Review. CyTA—J. Food 2024, 22, 2386412. [Google Scholar] [CrossRef]
  90. Attebury, H.; Daley, D. The Gut Microbiome and Pancreatic Cancer Development and Treatment. Cancer J. 2023, 29, 49–56. [Google Scholar] [CrossRef]
  91. Xiu, W.; Dong, H.; Chen, X.; Wan, L.; Lu, L.; Yang, K.; Yuwen, L.; Li, Q.; Ding, M.; Zhang, Y.; et al. Metabolic Modulation-Mediated Antibiotic and Immune Activation for Treatment of Chronic Lung Infections. ACS Nano 2024, 18, 15204–15217. [Google Scholar] [CrossRef]
  92. Tanaka, T.; Sugiyama, R.; Sato, Y.; Kawaguchi, M.; Honda, K.; Iwaki, H.; Okano, K. Precise Microbiome Engineering Using Natural and Synthetic Bacteriophages Targeting an Artificial Bacterial Consortium. Front. Microbiol. 2024, 15, 1403903. [Google Scholar] [CrossRef]
  93. Sayin, S.I.; Wahlström, A.; Felin, J.; Jäntti, S.; Marschall, H.-U.; Bamberg, K.; Angelin, B.; Hyötyläinen, T.; Orešič, M.; Bäckhed, F. Gut Microbiota Regulates Bile Acid Metabolism by Reducing the Levels of Tauro-Beta-Muricholic Acid, a Naturally Occurring FXR Antagonist. Cell Metab. 2013, 17, 225–235. [Google Scholar] [CrossRef]
  94. Cornista, A.M.; Giolito, M.V.; Baker, K.; Hazime, H.; Dufait, I.; Datta, J.; Khumukcham, S.S.; De Ridder, M.; Roper, J.; Abreu, M.T.; et al. Colorectal Cancer Immunotherapy: State of the Art and Future Directions. Gastro Hep Adv. 2023, 2, 1103–1119. [Google Scholar] [CrossRef]
  95. Prajapati, N.; Patel, J.; Singh, S.; Yadav, V.K.; Joshi, C.; Patani, A.; Prajapati, D.; Sahoo, D.K.; Patel, A. Postbiotic Production: Harnessing the Power of Microbial Metabolites for Health Applications. Front. Microbiol. 2023, 14, 1306192. [Google Scholar] [CrossRef]
  96. Su, A.C.Y.; Ding, X.; Lau, H.C.H.; Kang, X.; Li, Q.; Wang, X.; Liu, Y.; Jiang, L.; Lu, Y.; Liu, W.; et al. Lactococcus Lactis HkyuLL 10 Suppresses Colorectal Tumourigenesis and Restores Gut Microbiota through Its Generated Alpha-Mannosidase. Gut 2024, 73, 1478–1488. [Google Scholar] [CrossRef]
  97. DiMattia, Z.; Damani, J.; Rogers, C.J. 75 Effect of Probiotic Supplementation on Intestinal Permeability in Subjects with Overweight and Obesity: A Systematic Review of Randomized Controlled Trials. J. Clin. Transl. Sci. 2023, 7, 20–21. [Google Scholar] [CrossRef]
  98. Sun, J.; Chen, S.; Zang, D.; Sun, H.; Sun, Y.; Chen, J. Butyrate as a Promising Therapeutic Target in Cancer: From Pathogenesis to Clinic (Review). Int. J. Oncol. 2024, 64, 44. [Google Scholar] [CrossRef]
  99. Altowyan, M.S.; Soliman, S.M.; Haukka, M.; Al-Shaalan, N.H.; Alkharboush, A.A.; Barakat, A. Synthesis, Characterization, and Cytotoxicity of New Spirooxindoles Engrafted Furan Structural Motif as a Potential Anticancer Agent. ACS Omega 2022, 7, 35743–35754. [Google Scholar] [CrossRef]
  100. Bober, J.R.; Beisel, C.L.; Nair, N.U. Synthetic Biology Approaches to Engineer Probiotics and Members of the Human Microbiota for Biomedical Applications. Annu. Rev. Biomed. Eng. 2018, 20, 277–300. [Google Scholar] [CrossRef]
  101. Yadegar, A.; Salahi-Niri, A.; Wang, Y.-D.; Ochoa-Repáraz, J. Editorial: Gut Microbiota and Gastrointestinal Disorders, Volume II. Front. Med. 2025, 12, 1576152. [Google Scholar] [CrossRef]
  102. Liu, J.; Tian, R.; Sun, C.; Guo, Y.; Dong, L.; Li, Y.; Song, X. Microbial Metabolites Are Involved in Tumorigenesis and Development by Regulating Immune Responses. Front. Immunol. 2023, 14, 1290414. [Google Scholar] [CrossRef]
  103. Kang, X.; Lau, H.C.-H.; Yu, J. Modulating Gut Microbiome in Cancer Immunotherapy: Harnessing Microbes to Enhance Treatment Efficacy. Cell Rep. Med. 2024, 5, 101478. [Google Scholar] [CrossRef]
  104. Javdan, B.; Lopez, J.G.; Chankhamjon, P.; Lee, Y.-C.J.; Hull, R.; Wu, Q.; Wang, X.; Chatterjee, S.; Donia, M.S. Personalized Mapping of Drug Metabolism by the Human Gut Microbiome. Cell 2020, 181, 1661–1679.e22. [Google Scholar] [CrossRef]
  105. Schwarcz, S.; Nyerges, P.; Bíró, T.I.; Janka, E.; Bai, P.; Mikó, E. Cytostatic Bacterial Metabolites Interfere with 5-Fluorouracil, Doxorubicin and Paclitaxel Efficiency in 4T1 Breast Cancer Cells. Molecules 2024, 29, 3073. [Google Scholar] [CrossRef]
  106. Shanmugam, G.; Rakshit, S.; Sarkar, K. HDAC Inhibitors: Targets for Tumor Therapy, Immune Modulation and Lung Diseases. Transl. Oncol. 2022, 16, 101312. [Google Scholar] [CrossRef]
  107. Agarwal, D.S.; Mazumdar, S.; Italiya, K.S.; Chitkara, D.; Sakhuja, R. Bile-Acid-Appended Triazolyl Aryl Ketones: Design, Synthesis, In Vitro Anticancer Activity and Pharmacokinetics in Rats. Molecules 2021, 26, 5741. [Google Scholar] [CrossRef]
  108. Goering, A.W.; McClure, R.A.; Doroghazi, J.R.; Albright, J.C.; Haverland, N.A.; Zhang, Y.; Ju, K.-S.; Thomson, R.J.; Metcalf, W.W.; Kelleher, N.L. Metabologenomics: Correlation of Microbial Gene Clusters with Metabolites Drives Discovery of a Nonribosomal Peptide with an Unusual Amino Acid Monomer. ACS Cent. Sci. 2016, 2, 99–108. [Google Scholar] [CrossRef]
  109. Rahman, M.M.; Islam, M.R.; Shohag, S.; Ahasan, M.T.; Sarkar, N.; Khan, H.; Hasan, A.M.; Cavalu, S.; Rauf, A. Microbiome in Cancer: Role in Carcinogenesis and Impact in Therapeutic Strategies. Biomed. Pharmacother. 2022, 149, 112898. [Google Scholar] [CrossRef]
  110. Jotshi, A.; Sukla, K.K.; Haque, M.M.; Bose, C.; Varma, B.; Koppiker, C.B.; Joshi, S.; Mishra, R. Exploring the Human Microbiome—A Step Forward for Precision Medicine in Breast Cancer. Cancer Rep. 2023, 6, e1877. [Google Scholar] [CrossRef]
  111. Lan, Y.; Li, Y.; Wang, Y. Microbiome Analysis Reveals Dynamic Changes of Gut Microbiota in Guizhou Horse and Dutch Warmblood Horses. Front. Microbiol. 2025, 16, 1562482. [Google Scholar] [CrossRef]
  112. Dai, J.-H.; Tan, X.-R.; Qiao, H.; Liu, N. Emerging Clinical Relevance of Microbiome in Cancer: Promising Biomarkers and Therapeutic Targets. Protein Cell 2024, 15, 239–260. [Google Scholar] [CrossRef]
  113. Saravanan, C.; Gopinath, N.K.; Ganesan, R.; Thirumurugan, D. Challenges and Limitations in Using Bacterial Metabolites as Immunomodulators. Front. Cell. Infect. Microbiol. 2025, 15, 1535394. [Google Scholar] [CrossRef]
  114. Moniri, N.H.; Farah, Q. Short-Chain Free-Fatty Acid G Protein-Coupled Receptors in Colon Cancer. Biochem. Pharmacol. 2021, 186, 114483. [Google Scholar] [CrossRef]
  115. Tsukanov, V.V.; Vasyutin, A.V.; Tonkikh, J.L. Risk Factors, Prevention and Screening of Colorectal Cancer: A Rising Problem. World J. Gastroenterol. 2025, 31, 98629. [Google Scholar] [CrossRef]
  116. Rana, A.K.; Kumar Saraswati, S.S.; Anang, V.; Singh, A.; Singh, A.; Verma, C.; Natarajan, K. Butyrate Induces Oxidative Burst Mediated Apoptosis via Glucose-6-Phosphate Dehydrogenase (G6PDH) in Macrophages during Mycobacterial Infection. Microbes Infect. 2024, 26, 105271. [Google Scholar] [CrossRef]
  117. Bin Lee, D.; Hwang, I.S. Macronutrient Balance Determines the Human Gut Microbiome Eubiosis: Insights from in Vitro Gastrointestinal Digestion and Fermentation of Eight Pulse Species. Front. Microbiol. 2025, 15, 1512217. [Google Scholar] [CrossRef]
  118. Sayol-Altarriba, A.; Aira, A.; Villasante, A.; Albarracín, R.; Faneca, J.; Casals, G.; Villanueva-Cañas, J.L.; Casals-Pascual, C. Normalization of Short-Chain Fatty Acid Concentration by Bacterial Count of Stool Samples Improves Discrimination between Eubiotic and Dysbiotic Gut Microbiota Caused by Clostridioides Difficile Infection. Gut Microbes 2024, 16, 2415488. [Google Scholar] [CrossRef]
  119. Ramos Meyers, G.; Samouda, H.; Bohn, T. Short Chain Fatty Acid Metabolism in Relation to Gut Microbiota and Genetic Variability. Nutrients 2022, 14, 5361. [Google Scholar] [CrossRef]
  120. Albaladejo-Riad, N.; El Qendouci, M.; Cuesta, A.; Esteban, M.Á. Ability of Short-Chain Fatty Acids to Reduce Inflammation and Attract Leucocytes to the Inflamed Skin of Gilthead Seabream (Sparus aurata L.). Sci. Rep. 2024, 14, 31404. [Google Scholar] [CrossRef]
  121. Sun, M.; Ji, W.; Ye, H.; Cai, Y.; Yun, Y.; Wei, X.; Wang, C.; Mao, H. Sodium Butyrate Administration Improves Intestinal Development of Suckling Lambs. J. Anim. Sci. 2024, 102, skae028. [Google Scholar] [CrossRef] [PubMed]
  122. Otsuka, Y.; Kaneko, M.; Narukawa, M. Factors Associated with Successful Phase III Trials for Solid Tumors: A Systematic Review. Contemp. Clin. Trials Commun. 2021, 24, 100855. [Google Scholar] [CrossRef] [PubMed]
  123. Morrison, D.J.; Preston, T. Formation of Short Chain Fatty Acids by the Gut Microbiota and Their Impact on Human Metabolism. Gut Microbes 2016, 7, 189–200. [Google Scholar] [CrossRef] [PubMed]
  124. Park, H.; Lynch, E.; Tillman, A.; Lewis, K.; Jin, Z.; Uhlemann, A.-C.; Abrams, J.A.; Freedberg, D.E. A Phase 2 Randomized, Placebo-Controlled Trial of Inulin for the Prevention of Gut Pathogen Colonization and Infection among Patients Admitted to the Intensive Care Unit for Sepsis. Crit. Care 2025, 29, 21. [Google Scholar] [CrossRef]
  125. Molan, K.; Ambrožič Avguštin, J.; Likar, M.; Pongrac Barlovic, D.; Žgur Bertok, D.; Starčič Erjavec, M. Fecal Short-Chain Fatty Acids Are Associated with Obesity in Gestational Diabetes. Biomedicines 2025, 13, 387. [Google Scholar] [CrossRef]
  126. Peters, V.B.M.; Arulkumaran, N.; Melis, M.J.; Gaupp, C.; Roger, T.; Shankar-Hari, M.; Singer, M. Butyrate Supplementation Exacerbates Myocardial and Immune Cell Mitochondrial Dysfunction in a Rat Model of Faecal Peritonitis. Life 2022, 12, 2034. [Google Scholar] [CrossRef]
  127. Reim, D.; Novotny, A.; Friess, H.; Slotta-Huspenina, J.; Weichert, W.; Ott, K.; Dislich, B.; Lorenzen, S.; Becker, K.; Langer, R. Significance of Tumour Regression in Lymph Node Metastases of Gastric and Gastro-oesophageal Junction Adenocarcinomas. J. Pathol. Clin. Res. 2020, 6, 263–272. [Google Scholar] [CrossRef]
  128. Tian, T.; Zhao, Y.; Yang, Y.; Wang, T.; Jin, S.; Guo, J.; Liu, Z. The Protective Role of Short-Chain Fatty Acids Acting as Signal Molecules in Chemotherapy- or Radiation-Induced Intestinal Inflammation. Am. J. Cancer Res. 2020, 10, 3508–3531. [Google Scholar]
  129. Machado, D.T.; Dias, B.d.C.; Cayô, R.; Gales, A.C.; Marques de Carvalho, F.; Vasconcelos, A.T.R. Uncovering New Firmicutes Species in Vertebrate Hosts through Metagenome-Assembled Genomes with Potential for Sporulation. Microbiol. Spectr. 2024, 12, e0211324. [Google Scholar] [CrossRef]
  130. Sánchez-Alcoholado, L.; Laborda-Illanes, A.; Otero, A.; Ordóñez, R.; González-González, A.; Plaza-Andrades, I.; Ramos-Molina, B.; Gómez-Millán, J.; Queipo-Ortuño, M.I. Relationships of Gut Microbiota Composition, Short-Chain Fatty Acids and Polyamines with the Pathological Response to Neoadjuvant Radiochemotherapy in Colorectal Cancer Patients. Int. J. Mol. Sci. 2021, 22, 9549. [Google Scholar] [CrossRef]
  131. Yue, F.; Li, W.; Zou, J.; Jiang, X.; Xu, G.; Huang, H.; Liu, L. Spermidine Prolongs Lifespan and Prevents Liver Fibrosis and Hepatocellular Carcinoma by Activating MAP1S-Mediated Autophagy. Cancer Res. 2017, 77, 2938–2951. [Google Scholar] [CrossRef] [PubMed]
  132. Gerner, E.W.; Bruckheimer, E.; Cohen, A. Cancer Pharmacoprevention: Targeting Polyamine Metabolism to Manage Risk Factors for Colon Cancer. J. Biol. Chem. 2018, 293, 18770–18778. [Google Scholar] [CrossRef] [PubMed]
  133. Bi, G.; Liang, J.; Bian, Y.; Shan, G.; Huang, Y.; Lu, T.; Zhang, H.; Jin, X.; Chen, Z.; Zhao, M.; et al. Polyamine-Mediated Ferroptosis Amplification Acts as a Targetable Vulnerability in Cancer. Nat. Commun. 2024, 15, 2461. [Google Scholar] [CrossRef]
  134. Casero, R.A.; Murray Stewart, T.; Pegg, A.E. Polyamine Metabolism and Cancer: Treatments, Challenges and Opportunities. Nat. Rev. Cancer 2018, 18, 681–695. [Google Scholar] [CrossRef]
  135. Geck, R.C.; Foley, J.R.; Murray Stewart, T.; Asara, J.M.; Casero, R.A.; Toker, A. Inhibition of the Polyamine Synthesis Enzyme Ornithine Decarboxylase Sensitizes Triple-Negative Breast Cancer Cells to Cytotoxic Chemotherapy. J. Biol. Chem. 2020, 295, 6263–6277. [Google Scholar] [CrossRef]
  136. Li, M.; van Esch, B.C.A.M.; Henricks, P.A.J.; Folkerts, G.; Garssen, J. The Anti-Inflammatory Effects of Short Chain Fatty Acids on Lipopolysaccharide- or Tumor Necrosis Factor α-Stimulated Endothelial Cells via Activation of GPR41/43 and Inhibition of HDACs. Front. Pharmacol. 2018, 9, 533. [Google Scholar] [CrossRef]
  137. Roy, S.; Kumaravel, S.; Sharma, A.; Duran, C.L.; Bayless, K.J.; Chakraborty, S. Hypoxic Tumor Microenvironment: Implications for Cancer Therapy. Exp. Biol. Med. 2020, 245, 1073–1086. [Google Scholar] [CrossRef]
  138. Firoozi, D.; Masoumi, S.J.; Mohammad-Kazem Hosseini Asl, S.; Labbe, A.; Razeghian-Jahromi, I.; Fararouei, M.; Lankarani, K.B.; Dara, M. Effects of Short-Chain Fatty Acid-Butyrate Supplementation on Expression of Circadian-Clock Genes, Sleep Quality, and Inflammation in Patients with Active Ulcerative Colitis: A Double-Blind Randomized Controlled Trial. Lipids Health Dis. 2024, 23, 216. [Google Scholar] [CrossRef]
  139. Xu, Q.; Gao, J.; Zhao, R.; Li, H.; Cui, H.; Yuan, Z.; Ren, H.; Cao, B.; Wei, B. Akkermansia Muciniphila-Derived Pentadecanoic Acid Enhances Oxaliplatin Sensitivity in Gastric Cancer by Modulating Glycolysis. Pharmacol. Res. 2024, 206, 107278. [Google Scholar] [CrossRef]
  140. Bradlow, H.L.; Michnovicz, J.J.; Telang, N.T.; Osborne, M.P. Effects of Dietary Indole-3-Carbinol on Estradiol Metabolism and Spontaneous Mammary Tumors in Mice. Carcinogenesis 1991, 12, 1571–1574. [Google Scholar] [CrossRef]
  141. Mousa, W.K.; Mousa, S.; Ghemrawi, R.; Obaid, D.; Sarfraz, M.; Chehadeh, F.; Husband, S. Probiotics Modulate Host Immune Response and Interact with the Gut Microbiota: Shaping Their Composition and Mediating Antibiotic Resistance. Int. J. Mol. Sci. 2023, 24, 13783. [Google Scholar] [CrossRef] [PubMed]
  142. Muller, E.; Shiryan, I.; Borenstein, E. Multi-Omic Integration of Microbiome Data for Identifying Disease-Associated Modules. Nat. Commun. 2024, 15, 2621. [Google Scholar] [CrossRef] [PubMed]
  143. Ciernikova, S.; Sevcikova, A.; Mladosievicova, B.; Mego, M. Microbiome in Cancer Development and Treatment. Microorganisms 2023, 12, 24. [Google Scholar] [CrossRef] [PubMed]
  144. Cai, J.; Auster, A.; Cho, S.; Lai, Z. Dissecting the Human Gut Microbiome to Better Decipher Drug Liability: A Once-Forgotten Organ Takes Center Stage. J. Adv. Res. 2023, 52, 171–201. [Google Scholar] [CrossRef]
  145. Mohr, A.E.; Ortega-Santos, C.P.; Whisner, C.M.; Klein-Seetharaman, J.; Jasbi, P. Navigating Challenges and Opportunities in Multi-Omics Integration for Personalized Healthcare. Biomedicines 2024, 12, 1496. [Google Scholar] [CrossRef]
  146. Brüssow, H. Problems with the Concept of Gut Microbiota Dysbiosis. Microb. Biotechnol. 2020, 13, 423–434. [Google Scholar] [CrossRef]
  147. Shanahan, F.; Ghosh, T.S.; O’Toole, P.W. Human Microbiome Variance Is Underestimated. Curr. Opin. Microbiol. 2023, 73, 102288. [Google Scholar] [CrossRef]
  148. Tomás-Pejó, E.; González-Fernández, C.; Greses, S.; Kennes, C.; Otero-Logilde, N.; Veiga, M.C.; Bolzonella, D.; Müller, B.; Passoth, V. Production of Short-Chain Fatty Acids (SCFAs) as Chemicals or Substrates for Microbes to Obtain Biochemicals. Biotechnol. Biofuels Bioprod. 2023, 16, 96. [Google Scholar] [CrossRef]
  149. Hardwick, A.; Cummings, C.; Graves, J.; Kuzma, J. Can Societal and Ethical Implications of Precision Microbiome Engineering Be Applied to the Built Environment? A Systematic Review of the Literature. Environ. Syst. Decis. 2024, 44, 215–238. [Google Scholar] [CrossRef]
  150. Manrique, P.; Montero, I.; Fernandez-Gosende, M.; Martinez, N.; Cantabrana, C.H.; Rios-Covian, D. Past, Present, and Future of Microbiome-Based Therapies. Microbiome Res. Rep. 2024, 3, 23. [Google Scholar] [CrossRef]
  151. Liu, T.; Sun, Z.; Yang, Z.; Qiao, X. Microbiota-Derived Short-Chain Fatty Acids and Modulation of Host-Derived Peptides Formation: Focused on Host Defense Peptides. Biomed. Pharmacother. 2023, 162, 114586. [Google Scholar] [CrossRef] [PubMed]
  152. Pant, A.; Das, B. Microbiome-Based Therapeutics: Opportunity and Challenges. Prog. Mol. Biol. Transl. Sci. 2022, 191, 229–262. [Google Scholar] [CrossRef] [PubMed]
  153. Dudek-Wicher, R.K.; Junka, A.; Bartoszewicz, M. The Influence of Antibiotics and Dietary Components on Gut Microbiota. Gastroenterol. Rev. 2018, 13, 85–92. [Google Scholar] [CrossRef] [PubMed]
  154. Hitch, T.C.A.; Hall, L.J.; Walsh, S.K.; Leventhal, G.E.; Slack, E.; de Wouters, T.; Walter, J.; Clavel, T. Microbiome-Based Interventions to Modulate Gut Ecology and the Immune System. Mucosal Immunol. 2022, 15, 1095–1113. [Google Scholar] [CrossRef]
  155. Jain, N. The Need for Personalized Approaches to Microbiome Modulation. Front. Public Health 2020, 8, 144. [Google Scholar] [CrossRef]
  156. Yan, Q.; Jia, S.; Li, D.; Yang, J. The Role and Mechanism of Action of Microbiota-Derived Short-Chain Fatty Acids in Neutrophils: From the Activation to Becoming Potential Biomarkers. Biomed. Pharmacother. 2023, 169, 115821. [Google Scholar] [CrossRef]
  157. Rahman, S. Gut Microbial Metabolites and Its Impact on Human Health. Ann. Gastroenterol. 2023, 36, 360–368. [Google Scholar] [CrossRef]
  158. Chen, Q.; Yang, Z.; Liu, H.; Man, J.; Oladejo, A.O.; Ibrahim, S.; Wang, S.; Hao, B. Novel Drug Delivery Systems: An Important Direction for Drug Innovation Research and Development. Pharmaceutics 2024, 16, 674. [Google Scholar] [CrossRef]
  159. Mafe, A.N.; Büsselberg, D. Microbiome Integrity Enhances the Efficacy and Safety of Anticancer Drug. Biomedicines 2025, 13, 422. [Google Scholar] [CrossRef]
  160. 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]
  161. Fekete, E.E.; Figeys, D.; Zhang, X. Microbiota-Directed Biotherapeutics: Considerations for Quality and Functional Assessment. Gut Microbes 2023, 15, 2186671. [Google Scholar] [CrossRef] [PubMed]
  162. Lange, L.; Berg, G.; Cernava, T.; Champomier-Vergès, M.-C.; Charles, T.; Cocolin, L.; Cotter, P.; D’Hondt, K.; Kostic, T.; Maguin, E.; et al. Microbiome Ethics, Guiding Principles for Microbiome Research, Use and Knowledge Management. Environ. Microbiome 2022, 17, 50. [Google Scholar] [CrossRef] [PubMed]
  163. Liwinski, T.; Elinav, E. Harnessing the Microbiota for Therapeutic Purposes. Am. J. Transplant. 2020, 20, 1482–1488. [Google Scholar] [CrossRef] [PubMed]
  164. Hemmati, M.A.; Monemi, M.; Asli, S.; Mohammadi, S.; Foroozanmehr, B.; Haghmorad, D.; Oksenych, V.; Eslami, M. Using New Technologies to Analyze Gut Microbiota and Predict Cancer Risk. Cells 2024, 13, 1987. [Google Scholar] [CrossRef]
  165. Eicher, T.; Kinnebrew, G.; Patt, A.; Spencer, K.; Ying, K.; Ma, Q.; Machiraju, R.; Mathé, E.A. Metabolomics and Multi-Omics Integration: A Survey of Computational Methods and Resources. Metabolites 2020, 10, 202. [Google Scholar] [CrossRef]
  166. Patil, A.; Singh, N.; Patwekar, M.; Patwekar, F.; Patil, A.; Gupta, J.K.; Elumalai, S.; Priya, N.S.; Sahithi, A. AI-Driven Insights into the Microbiota: Figuring out the Mysterious World of the Gut. Intell. Pharm. 2024, 3, 46–52. [Google Scholar] [CrossRef]
  167. Li, P.; Luo, H.; Ji, B.; Nielsen, J. Machine Learning for Data Integration in Human Gut Microbiome. Microb. Cell Fact. 2022, 21, 241. [Google Scholar] [CrossRef]
  168. Yang, L.; Wang, Q.; He, L.; Sun, X. The Critical Role of Tumor Microbiome in Cancer Immunotherapy. Cancer Biol. Ther. 2024, 25, 2301801. [Google Scholar] [CrossRef]
  169. Zhao, H.; Wang, D.; Zhang, Z.; Xian, J.; Bai, X. Effect of Gut Microbiota-Derived Metabolites on Immune Checkpoint Inhibitor Therapy: Enemy or Friend? Molecules 2022, 27, 4799. [Google Scholar] [CrossRef]
  170. Soleimani, N.; Javadi, M.M. Future Prospects of Bacteria-Mediated Cancer Therapies: Affliction or Opportunity? Microb. Pathog. 2022, 172, 105795. [Google Scholar] [CrossRef]
  171. Lu, S.; Wang, C.; Ma, J.; Wang, Y. Metabolic Mediators: Microbial-Derived Metabolites as Key Regulators of Anti-Tumor Immunity, Immunotherapy, and Chemotherapy. Front. Immunol. 2024, 15, 1456030. [Google Scholar] [CrossRef]
Figure 1. Microbiome metabolites: essential mechanisms in cancer prevention and therapy (created with BioRender).
Figure 1. Microbiome metabolites: essential mechanisms in cancer prevention and therapy (created with BioRender).
Biomolecules 15 00688 g001
Figure 2. The double-edged sword of the gut microbiome: comparison of healthy microbiota and dysbiosis in cancer prevention and promotion (created with BioRender).
Figure 2. The double-edged sword of the gut microbiome: comparison of healthy microbiota and dysbiosis in cancer prevention and promotion (created with BioRender).
Biomolecules 15 00688 g002
Table 1. Microbiome-derived metabolites and their role in inflammation-related cancer.
Table 1. Microbiome-derived metabolites and their role in inflammation-related cancer.
Major FindingsMetabolites StudiedCancer Type
Butyrate induces apoptosisSCFAsColorectal [12]
Propionate suppresses inflammationSCFAsLiver [13]
Spermidine regulates tumor growthPolyaminesBreast [14]
Indoles modulate immune responsesIndole derivativesProstate [15]
Bile acids impact gut microbiotaSecondary bile acidsPancreatic [16]
SCFAs enhance gut barrier functionSCFAsGastric [17]
Microbiome-derived metabolites impact metastasisVariousLung [18]
Indole derivatives affect inflammatory markersIndolesOvarian [19]
Polyamine levels correlate with tumor progressionPolyaminesMultiple [20]
SCFAs modulate T cell differentiationSCFAsColorectal [21]
LPS promotes chronic inflammation and tumor progression via TLR4 and METTL3/PI3K/AKT signalingLPSCholangiocarcinoma [22]
Table 2. Microbiome metabolites in cancer therapy: innovations and emerging strategies.
Table 2. Microbiome metabolites in cancer therapy: innovations and emerging strategies.
ApproachTargetCancer Type
Probiotic supplementationGut microbiota balanceColorectal [85]
Prebiotic fiber therapySCFA productionLiver [86]
SCFA-based treatmentTumor suppressionGastric [87]
Engineered microbiome therapiesPrecision medicineBreast [88]
Postbiotic metabolites as drugsAnti-inflammatory effectsOvarian [89]
Microbiome modificationTumor microenvironmentPancreatic [90]
Metabolite-targeted drugsImmune modulationLung [91]
Synthetic microbiota engineeringPersonalized therapyMultiple [92]
Bile acid modulationTumor immunotherapyLiver [93]
SCFAs combined with immunotherapyCombination therapyColorectal [94]
Table 3. Case studies and controversies in microbiome metabolite research.
Table 3. Case studies and controversies in microbiome metabolite research.
Trial PhaseFindings
Phase ISCFAs reduced inflammation [120]
Phase IIButyrate improved immune response [121]
Phase IIIMixed results on tumor regression [122]
Phase ISCFA bioavailability drawbacks [123]
Phase IICombination therapy with SCFAs [124]
Phase IIISCFA-based diets showed limited efficacy [125]
Phase IIButyrate supplementation improved outcomes [126]
Phase IIINo significant tumor regression observed [127]
Phase IISCFAs enhanced chemotherapy response [128]
Phase IIISCFAs reduced side effects of treatment [129]
Table 4. Complications, limitations, and future perspectives.
Table 4. Complications, limitations, and future perspectives.
ComplicationFindings
Microbiome variabilityMicrobiome composition differs across populations [147]
Bioavailability issuesSCFAs degrade quickly in circulation [148]
Ethical concernsEngineering gut microbiota raises safety issues [149]
Regulatory barriersLack of FDA-approved microbiome therapies [150]
Individual response variabilityDifferent patients respond differently to SCFAs [151]
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

Mafe, A.N.; Büsselberg, D. The Effect of Microbiome-Derived Metabolites in Inflammation-Related Cancer Prevention and Treatment. Biomolecules 2025, 15, 688. https://doi.org/10.3390/biom15050688

AMA Style

Mafe AN, Büsselberg D. The Effect of Microbiome-Derived Metabolites in Inflammation-Related Cancer Prevention and Treatment. Biomolecules. 2025; 15(5):688. https://doi.org/10.3390/biom15050688

Chicago/Turabian Style

Mafe, Alice N., and Dietrich Büsselberg. 2025. "The Effect of Microbiome-Derived Metabolites in Inflammation-Related Cancer Prevention and Treatment" Biomolecules 15, no. 5: 688. https://doi.org/10.3390/biom15050688

APA Style

Mafe, A. N., & Büsselberg, D. (2025). The Effect of Microbiome-Derived Metabolites in Inflammation-Related Cancer Prevention and Treatment. Biomolecules, 15(5), 688. https://doi.org/10.3390/biom15050688

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop