Nurah EkhlaqueFebruary 18, 2025
Tag: Personalized Medicine , Microbiome , Drug Metabolism
The microbiome supports digestion, nutrient absorption, and regulates drug metabolism. It also influences treatment responses and affects pharmacokinetics and pharmacodynamics. The enzymatic activity of gut bacteria can enhance drug efficacy or cause toxicity. Understanding these interactions is essential for advancing personalized medicine.
Microbial enzymes significantly influence drug processing in the human body. The study of microbiome drug metabolism highlights how gut bacteria introduce an additional layer of complexity, complementing liver enzymes traditionally seen as the primary agents of drug metabolism. Microbial biotransformation can involve processes such as reduction, hydrolysis, and demethylation, fundamentally altering a drug’s chemical structure and activity.
A well-known example of how gut bacteria affect drug metabolism is the interaction between Eggerthella lenta and the cardiac drug digoxin, commonly used to treat heart failure and irregular heartbeats. E. lenta can reduce the drug’s effectiveness by converting it into an inactive form through an enzyme called cardiac glycoside reductase (Cgr). Research by Haiser et al. (2013) showed that this inactivation process is controlled by the cgr operon in E. lenta. The study also found that increasing dietary arginine suppresses the activity of the cgr operon, helping to maintain digoxin’s therapeutic effect. This suggests that dietary strategies, such as arginine supplementation, could improve drug response and treatment outcomes for patients taking digoxin.¹
Drug absorption and bioavailability are essential factors that determine how much of a medication reaches the bloodstream and exerts its therapeutic effect. The gut microbiota plays a crucial role in modulating these processes. In some cases, microbial activity enhances drug efficacy, while in others, it may inhibit absorption and reduce effectiveness.
Sulfasalazine is a prodrug commonly used in the treatment of inflammatory bowel disease (IBD). Its therapeutic efficacy depends on activation by gut microbiota, which cleave sulfasalazine into its active components: 5-aminosalicylic acid (5-ASA) and sulfapyridine. This microbial activation is essential for the drug's anti-inflammatory effects. In individuals with a dysbiotic gut microbiome, this activation process may be impaired, leading to reduced drug efficacy. Personalized microbiome-based therapies are being explored to enhance drug activation and improve outcomes for patients with such gut profiles. ²
While gut bacteria can enhance drug efficacy, they may also produce toxic metabolites. Irinotecan, a chemotherapy drug for colorectal cancer, is converted to SN-38 in the liver and later inactivated to SN-38 glucuronide (SN-38G) for excretion. However, gut bacterial β-glucuronidase can reactivate SN-38 from SN-38G, causing severe gastrointestinal toxicity, including diarrhea and mucosal damage.³
Preclinical studies show that inhibiting bacterial β-glucuronidase reduces gut toxicity without affecting irinotecan’s anticancer efficacy.⁴ Targeted enzyme inhibitors effectively prevent SN-38 reactivation, improving chemotherapy safety. Advances in understanding β-glucuronidase structure have enabled the design of selective inhibitors to reduce irinotecan-induced toxicity and enhance treatment success.
Drug metabolism by the microbiome is well-known, but its influence on individual responses has only recently been recognized. Variations in gut microbiota explain why two patients on identical medications can have vastly different reactions, highlighting the potential for personalized medicine.
Individual Variability and Precision Medicine: The unique composition of an individual's gut microbiome significantly affects drug metabolism. For example, gut microbiota variability can lead to contrasting outcomes in patients receiving the same medication.⁵ Advances in metagenomic sequencing, which analyze the microbiome’s collective genetic material, are helping researchers predict drug responses and improve treatment decisions. By identifying specific microbial genes and pathways involved in drug metabolism, personalized strategies can be developed to enhance drug efficacy and reduce adverse effects.⁶ This approach marks a crucial step toward precision medicine, where treatments are customized to an individual’s microbial profile, reducing drug-related complications and improving therapeutic success.
Microbiome-Guided Drug Development: The pharmaceutical industry is increasingly looking to the microbiome as a target for novel therapies. Microbiome-based interventions, such as faecal microbiota transplantation (FMT), probiotics, and engineered microbial consortia, are being tested in clinical trials for a wide range of conditions, from cancer immunotherapy enhancement to antibiotic-associated colitis treatment.⁷
Researchers are developing gut microbiome therapies to adjust gut bacteria and improve the effectiveness of treatments. Recent market analysis estimates the global microbiome therapeutics market will grow from $919.4 million in 2023 to $21.5 billion by 2030 at a CAGR of 56.9%. This rapid growth is driven by rising gastrointestinal disorders, increasing focus on gut health, and advances in microbiome-targeted therapies. As clinical evidence and regulatory clarity improve, these innovations are reshaping treatment strategies for chronic diseases such as diabetes and inflammatory bowel disease (IBD).
● Microbiota Modulation in Chemotherapy
Cancer treatment is an area where the influence of the microbiome is especially clear. Studies show that certain gut bacteria can either enhance or hinder responses to immune checkpoint inhibitors, a breakthrough in cancer immunotherapy. For example, Faecalibacterium prausnitzii has been linked to better responses to anti-PD-1 therapy, while an overabundance of Bacteroides fragilis may reduce its effectiveness. To address these variations, treatments that target the microbiome, such as prebiotics, probiotics, and selective antibiotics, are being explored to improve therapy response.
● Probiotic Therapy in Inflammatory Diseases
For conditions like inflammatory bowel disease (IBD), probiotics have moved beyond simply promoting gut health to becoming supportive treatments alongside standard drug therapy. Escherichia coli Nissle 1917, for instance, has been shown to improve remission rates when combined with traditional anti-inflammatory medications, highlighting its potential as a therapeutic addition.
● Reducing Drug Toxicity through Enzyme Inhibition
In cases like irinotecan treatment, developing inhibitors that specifically target gut bacterial enzymes such as β-glucuronidase offers an effective way to reduce side effects without compromising the drug’s cancer-fighting ability. This approach could be applied to other drugs with microbial-associated toxicities, strengthening the connection between microbiology and pharmacology for safer therapies.
The exploration of the human microbiome as a diagnostic tool is transforming clinical practice. By analyzing microbial DNA, clinicians can detect patterns associated with various health conditions, including drug resistance, inflammatory diseases, and mental health disorders. For instance, specific microbial signatures have been identified as early indicators of colorectal cancer and type 2 diabetes.
In colorectal cancer (CRC), studies reveal that patients often exhibit significant changes in their gut microbiota compared to healthy individuals. These changes may involve variations in bacterial diversity, abundance, and composition. Although using gut microbiota as a diagnostic biomarker for CRC shows potential, more research is required to confirm its accuracy and establish standardized testing protocols.⁸
In type 2 diabetes, research shows it is linked to a microbial signature marked by a decrease in butyrate-producing bacteria. This imbalance may play a role in the development of diabetes and presents potential for microbiome-based diagnostics.⁹ Moreover, the gut microbiome also affects mental health. Evidence suggests that gut microbiota significantly contributes to the immunological mechanisms underlying mental disorders, particularly psychotic disorders and mood disorders. Dysbiosis, an imbalance in gut microbiota, is linked to increased inflammation and disrupted stress responses through the hypothalamic-pituitary-adrenal (HPA) axis, contributing to mood disorders and psychosis.¹⁰ However, the underlying mechanisms remain unclear and require further research. Although microbiome-based diagnostics are still emerging, they hold significant potential. With advances in artificial intelligence and machine learning, these tools may revolutionize disease prediction and monitoring, allowing for earlier intervention and more precise therapies.
The gut microbiome is a growing focus in precision medicine. Advances in metagenomics and next-generation sequencing (NGS) help clinicians predict drug responses and develop treatments based on individual microbial profiles, improving therapy effectiveness and minimizing adverse reactions. Emerging microbiome-targeted strategies, including probiotics, prebiotics, and enzyme inhibitors, show significant potential for enhancing drug efficacy and reducing toxicity. Machine learning and multi-omics approaches are refining these treatments, enabling early disease prediction and customized solutions. Despite its potential, integrating microbiome science into clinical practice remains challenging. Factors such as microbial diversity, diet, genetics, and the absence of standardized protocols complicate implementation.
However, technological advances are expanding clinical applications.The future of medicine will likely depend on microbiome research to improve drug response and disease management, transforming healthcare with personalized, microbiome-focused therapies.
The gut microbiome plays a key role in drug metabolism and personalized medicine. Advances in sequencing and microbiome-guided therapies are improving care and reducing adverse reactions. Its integration into mainstream medicine will transform healthcare and enhance personalized therapies.
1. Haiser, Henry J., et al. ‘Predicting and Manipulating Cardiac Drug Inactivation by the Human Gut Bacterium Eggerthella Lenta’. Science (New York, N.Y.), vol. 341, no. 6143, July 2013, pp. 295–98. PubMed Central, https://doi.org/10.1126/science.1235872.
2. Lima, Svetlana F., et al. ‘The Gut Microbiome Regulates the Clinical Efficacy of Sulfasalazine Therapy for IBD-Associated Spondyloarthritis’. Cell Reports Medicine, vol. 5, no. 3, Feb. 2024, p. 101431. PubMed Central, https://doi.org/10.1016/j.xcrm.2024.101431.
3. Wallace, Bret D., et al. ‘Structure and Inhibition of Microbiome β-Glucuronidases Essential to the Alleviation of Cancer Drug Toxicity’. Chemistry & Biology, vol. 22, no. 9, Sept. 2015, pp. 1238–49. PubMed Central, https://doi.org/10.1016/j.chembiol.2015.08.005
4. Bhatt, Aadra P., et al. ‘Targeted Inhibition of Gut Bacterial β-Glucuronidase Activity Enhances Anticancer Drug Efficacy’. Proceedings of the National Academy of Sciences, vol. 117, no. 13, Mar. 2020, pp. 7374–81. DOI.org (Crossref), https://doi.org/10.1073/pnas.1918095117.
5. Zhao, Qing, et al. ‘Drug-Microbiota Interactions: An Emerging Priority for Precision Medicine’. Signal Transduction and Targeted Therapy, vol. 8, no. 1, Oct. 2023, pp. 1–27. www.nature.com, https://doi.org/10.1038/s41392-023-01619-w.
6. Javdan, Bahar, et al. ‘Personalized Mapping of Drug Metabolism by the Human Gut Microbiome’. Cell, vol. 181, no. 7, June 2020, pp. 1661-1679.e22. PubMed Central, https://doi.org/10.1016/j.cell.2020.05.001.
7. Soto Chervin, C., and T. F. Gajewski. ‘Microbiome-Based Interventions: Therapeutic Strategies in Cancer Immunotherapy’. Immuno-Oncology Technology, vol. 8, Nov. 2020, pp. 12–20. PubMed Central, https://doi.org/10.1016/j.iotech.2020.11.001.
8. Herlo, Lucian-Flavius, et al. ‘Gut Microbiota Signatures in Colorectal Cancer as a Potential Diagnostic Biomarker in the Future: A Systematic Review’. International Journal of Molecular Sciences, vol. 25, no. 14, July 2024, p. 7937. PubMed Central, https://doi.org/10.3390/ijms25147937.
9. Noureldein, Mohamed, et al. ‘Intestinal Microbiota Regulates Diabetes and Cancer Progression by IL-1β and NOX4 Dependent Signaling Cascades’. Cellular and Molecular Life Sciences, vol. 79, no. 9, Aug. 2022, p. 502. Springer Link, https://doi.org/10.1007/s00018-022-04485-x.
10. Rogers, G. B., et al. ‘From Gut Dysbiosis to Altered Brain Function and Mental Illness: Mechanisms and Pathways’. Molecular Psychiatry, vol. 21, no. 6, June 2016, pp. 738–48. www.nature.com, https://doi.org/10.1038/mp.2016.50.
Nurah Ekhlaque is a freelance medical writer with a Master’s in Biotechnology from Guru Ghasidas University, India. With over three years of experience, she specialises in crafting research-based, engaging content for the healthcare and life sciences sectors.
Her research experience includes working as a Research Assistant at Saarland University, Germany, and as a trainee at AIIMS, India, where she developed expertise in molecular biology techniques like immunohistochemistry and confocal imaging. In addition to writing, Nurah mentors aspiring medical writers, guiding them to create effective healthcare content.
She is currently a freelance pharma writer for Sinoexpo PharmaSources.com, where she focuses on innovations and trends in healthcare and biotechnology, offering insights into advancements shaping the industry.
Contact Us
Tel: (+86) 400 610 1188
WhatsApp/Telegram/Wechat: +86 13621645194
Follow Us: