banner

News

Jul 18, 2023

Drug

Signal Transduction and Targeted Therapy volume 8, Article number: 386 (2023) Cite this article

1226 Accesses

22 Altmetric

Metrics details

Individual variability in drug response (IVDR) can be a major cause of adverse drug reactions (ADRs) and prolonged therapy, resulting in a substantial health and economic burden. Despite extensive research in pharmacogenomics regarding the impact of individual genetic background on pharmacokinetics (PK) and pharmacodynamics (PD), genetic diversity explains only a limited proportion of IVDR. The role of gut microbiota, also known as the second genome, and its metabolites in modulating therapeutic outcomes in human diseases have been highlighted by recent studies. Consequently, the burgeoning field of pharmacomicrobiomics aims to explore the correlation between microbiota variation and IVDR or ADRs. This review presents an up-to-date overview of the intricate interactions between gut microbiota and classical therapeutic agents for human systemic diseases, including cancer, cardiovascular diseases (CVDs), endocrine diseases, and others. We summarise how microbiota, directly and indirectly, modify the absorption, distribution, metabolism, and excretion (ADME) of drugs. Conversely, drugs can also modulate the composition and function of gut microbiota, leading to changes in microbial metabolism and immune response. We also discuss the practical challenges, strategies, and opportunities in this field, emphasizing the critical need to develop an innovative approach to multi-omics, integrate various data types, including human and microbiota genomic data, as well as translate lab data into clinical practice. To sum up, pharmacomicrobiomics represents a promising avenue to address IVDR and improve patient outcomes, and further research in this field is imperative to unlock its full potential for precision medicine.

Why does a drug with a well-response for one patient, but not for the next, or in some cases, lead to serious adverse drug reactions (ADRs), namely individual variability in drug response (IVDR)? The answers are complicated and multi-faceted. First of all, therapeutic response rates of different drugs caused by IVDR differ. According to a survey, cyclooxygenase-2 (COX-2) inhibitors exhibit the most remarkable response rate (80%) among drugs used to treat various diseases, while tumor chemotherapy displays the lowest response rate (25%).1 Furthermore, the response rates for various other medications varied from 50–75%, indicating that a significant proportion of patients did not benefit from these treatments.1 Secondly, the rate of ADRs caused by IVDR is not the same thing. ADRs are the joint occurrence and can have a substantial impact on morbidity, mortality, and healthcare costs.2,3,4,5 For example, ADRs accounted for approximately 4.93% of emergency hospitalizations in China, with a majority of them being preventable (around 73.7%).6 However, IVDR makes the prevention of ADRs tricky. These facts underscore the urgent need to better understand IVDR to meet the highly effective drug or prevent ADRs.

Precision medicine aims to develop tailored medical interventions on the basis of an individual’s genetic, environmental, and lifestyle elements.7,8,9 However, IVDR is a major barrier to the implementation of precision medicine, as it poses a challenge to predict drug efficacy and identify patients who are at risk for ADRs.10,11,12,13 Consequently, it is crucial to develop improved biomarkers that can predict drug efficacy and toxicity in order to successfully implement precision medicine.

Pharmacogenetics focuses on identifying genetic variants that influence drug metabolism, transport, pharmacodynamics (PD), and others.14,15,16,17,18,19 Pharmacogenomics is a broader field that considers whole genome and its impact on drug response.11,13,20,21,22,23,24,25,26 Despite the progress made in pharmacogenetics and pharmacogenomics, studies to determine IVDR have shown that human genome contributes anywhere between 20% and 95% of the variability (this percentage may vary depending on the disease model).27 Hence, genetic factors alone are insufficient to explain IVDR, and additional factors (such as diversity in gut microbiota spectrum) may also be important.28,29,30,31,32,33,34

Gut microbiota, which is a bifunctional and heterogeneous ecosystem, often described as a ‘metabolic organ’, contains over 100 trillion microbes and 5 million genes, making it much larger than human gene count (~150 times).35,36,37,38,39,40,41,42,43,44,45 Due to various host-related factors, gut microbiota composition and structure vary greatly between individuals and over time as well as external factors (e.g., drugs, diet, and environment).46,47,48,49,50,51,52 Studies conducted on both mice and humans have demonstrated that drug intake has significant effects on the structure of gut microbiota.53,54,55 In recent years, gut microbiota has also been mandated for IVDR, gut microbiota can alter drug PD and pharmacokinetics (PK) by direct transforming the drug or modulating the metabolism or immune system of the host.42,54,56,57,58

The term pharmacomicrobiomics has been coined to describe the interactions between gut microbiota and drug response that can alter PK (i.e., changes in drug absorption, distribution, metabolism, excretion, and its plasma drug concentration dynamics) or PD (i.e., changes in drug targets or biological pathways resulting in differential susceptibility of the organism to pharmacological effects).59,60,61,62,63,64,65 Understanding this interaction can help develop microbiota-targeted approaches to enhance drug efficacy and reduce ADRs.66,67,68 Clearly, pharmacomicrobiomics is becoming an integral part of the advances in precision medicine, and modifying gut microbiota could be a highly appealing option for managing the efficiency and safety of drugs at an individualistic level.58,61 This review outlines pharmacomicrobiomics advances in human diseases to better understand the impact of IVDR in future precision medicine.

IVDR is a complex and multifactorial phenomenon that can result from a combination of factors, as shown in Fig. 1, including (1) Genetics: Variations in individual genes encoding drug-metabolizing enzymes, transporters, and targets can affect their metabolism and response to drugs, leading to IVDR.69,70,71,72,73 (2) Age: Younger and older patients may experience different drug responses due to differences in their physiological function, such as changes in liver and kidney function, body composition, and hormone levels.74,75,76,77,78 For example, elderly patients may have reduced renal function and hepatic metabolism, leading to altered PK.79,80,81 (3) Gender: Biological dissimilarities between men and women can influence drug response.82,83,84,85,86,87 Women may have higher drug concentrations due to disparities in body composition and hormone levels, which will affect drug metabolism and clearance.88,89,90,91 (4) Lifestyle: Factors such as diet, exercise, smoking, and alcohol intake can affect drug response by changing the way drugs are metabolized and eliminated from the body.92,93,94,95 (5) Disease state: Patients with certain diseases or conditions may experience IVDR due to changes in organ function, altered drug metabolism, or altered drug receptor expression.96,97,98,99 For example, drug metabolism may be reduced in patients with liver diseases and drug clearance may be reduced in patients with kidney diseases.100,101,102,103 (6) Drug-drug interactions: Concomitant use of multiple medications may result in drug-drug interactions that alter drug absorption, distribution, metabolism, and excretion (ADME).104,105,106,107 (7) Environmental factors: Exposure to environmental toxins and pollutants can affect drug metabolism and clearance, potentially leading to IVDR.108,109,110,111,112 (8) Gut microbiota: Gut microbiota may also affect drug PK and PD, and variations in the composition of gut microbiota between individuals contribute to IVDR.42,56,57,66,113,114,115 Overall, understanding these factors is critical to optimizing drug therapy and minimising the risk of ADRs.

Causes of the individual variability in drug response (IVDR)

In the early 1900s, Archibald Garrod, a renowned physiologist from England, proposed that genetic factors may contribute to inborn errors in drug metabolism and IVDR.116,117 Since then, numerous studies have demonstrated that genetic factors can explain as much as 95% of the inter-individual variability in drug PK and PD for specific drugs or drug classes118,119,120,121 (Table 1). In the late 1950s, it was discovered that inherited glucose-6-phosphate dehydrogenase deficiency could lead to serious hemolysis in patients administered primaquine, an anti-malarial drug, primarily in African-Americans and minimally in Caucasians of North, West, and East European heritage.122,123,124 Subsequently, in 1959, Vogel introduced the term pharmacogenetics to define IVDR.125 The evolution (from genetics to genomics) and application of genome-wide strategies has led to the emergence of the term pharmacogenomics, with the potential to target specific drug therapies to genetically defined patient subpopulations and to establish novel diseases and therapeutic classifications at the molecular and phenotypic level.15,20,21,22,126,127,128

IVDR poses a serious challenge in clinical management, and genetic polymorphisms play a vital role in this variability.16,17,25 Genetic variations in genes associated with drug transport, metabolism, and targets can change the individual’s sensitivity to treatment, resulting in variable drug response.121,129,130,131,132,133,134,135,136,137 The function of transporters in controlling the transfer of drugs and their metabolites into and out of cells is critical to drug efficacy and toxicity.138,139 In the case of pro-drugs, metabolism is essential to prevent ADRs caused by elevated plasma drug levels in some patients. However, environmental factors can also influence drug metabolism, efficacy, and toxicity, leading to IVDR.18,140 Despite the important influences of genetic variation on drug response, the complexity of environment and microbiota may also constrain the prediction of drug response on the basis of genomic diversity alone.141 Hence, it is crucial to have a comprehensive comprehension of the intricate interaction between genetic and external factors to develop precision medicine strategies that optimize drug therapy for individual patients.

IVDR can also be attributed to the presence of variant alleles of drug-metabolizing enzymes, transporters, and targets in different populations.142,143,144 Allelic variation of target genes in the study population is essential for the advancement of personalized tumor treatment.145,146,147,148,149,150 However, the frequency of unique variant alleles can vary significantly between races, making it difficult to incorporate pedigree into clinical decision-making because it confounds drug response.25 In addition, PK differences between drug-metabolizing enzyme and transporter populations can affect IVDR and raise the risk of ADRs (e.g., thiopurines, allopurinol, and carbamazepine).143,151 A lot of ADRs are linked to specific phenotypes of drug metabolites, such as hypersensitivity reactions triggered by sulfonamides.25,152,153,154 A more in-depth mechanistic and phenotypic study of drug metabolism and individual differences between patients may help to guide treatment based on unique characteristics and the application of predictive models to avoid ADRs.25

The Human Genome Project (HGP) was concluded in 2003, but human genome contains fewer coding genes, and variations in genetic, epigenetic, and regulators are insufficient to describe IVDR in the phenotype, with limited utility for precision medicine155 (Fig. 2). The focus has shifted to gut microbiota to study its composition, variation, and function in understanding the spectrum of human phenotypic variation, including its impact on well-being, immune system, and drug response. In comparison to human genome, gut microbiota is remarkably adaptive and mobile, similar to a cloud whose components and genetic pool are unclear at every moment in time and space156 (Table 1). It can be partially or completely swapped out and is exposed to various factors that influence its evolution, including immune responses, phage attacks, diet, toxins, anti-biotics, and others.157,158 Because of the abilities of gut microbiota to influence drug response and disease progression, targeted manipulation of gut microbiota may improve drug efficacy and reduce drug-drug interactions.141 Mobility of gut microbiota is not limited to movement within inter-individuals, but can also occur within intra-individuals due to factors such as spatial, transient, seasonal, hormonal, and nutritional changes or the presence of multiple drugs.159 The diversity of gut microbiota gives it a remarkable metabolic capacity that exceeds even that of the host.160,161,162 Specifically, gut microbiota can produce a range of metabolic responses to drugs and xenobiotics, resulting in both direct effects on drug metabolism and toxicity, and indirect effects on host metabolic enzymes/transporters/immune system, these responses can ultimately affect IVDR.45,163 Gut microbiota can directly affect drug metabolism in several manners, including generating enzymes that degrade or catalyze a drug molecule, competing with drug molecules for a metabolic enzyme, altering the metabolic levels of drugs in the host, or generating enzymes that stimulate metabolites originally derived from the diet.59,164 The presence of microbiota-encoded enzymes represents a potentially valuable intermediate target for PK modification that may ultimately improve clinical response.44 In turn, drug administration may also affect microbiota metabolism or growth, leading to variations in the structure and function of microbiota.63 Changes in a pathological state, metabolic enzyme expression, and drug transporter expression may result in gut microbiota producing compounds that affect drug PK and PD.165

Timeline of the historical milestone for the development of pharmacomicrobiomics in the past eight decades

In addition, human genome and gut microbiota work in tandem to influence personal metabolism and contribute to IVDR.59 Through anatomical and physiological linkages (e.g., the intestinal barrier), they exchange metabolically active molecules and beneficial host-immune interactions.164,166 This metabolic axis of gut microbiota links human genome to microbiota ecosystem as a genetic or epigenetic basis for up-keeping human immune and nutrition functions.166,167,168,169 Interactions between human genome and the diversity of gut microbiota genomes lead to the synthesis of compounds benefiting human healthcare, e.g., amino acids (AAs), short-chain fatty acids (SCFAs), and bile acids (BAs).165,170 Genes encoded by gut microbiota greatly enhance the metabolic potential of human, contributing as much as 36% of small molecule metabolism in human blood.59 Therefore, the measurement of large numbers of metabolites is necessary to better report and manage the multitude of diseases and treatment efficacy issues.171

In summary, understanding the influence of human genome heterogeneity and gut microbiota spectrum diversity on IVDR is a key component in developing and improving precision medicine while minimizing ADRs.59 In addition, modeling changes in gut microbiota will help to understand xenobiotic metabolism, thereby accurately predicting drug responses by taking into account genetic diversity and metabolic interactions between host and pertinent microorganisms.59

The microbiome encompasses the genetic material of various microorganisms, such as bacteria, fungi, protozoa, and viruses, that inhabit both the external and internal regions of the host.172,173,174 The term microbiome can denote the genetic make-up of the microbiota or the entire genome of all its members.59,175,176 Microbiome can also refer to the total environment that microorganisms inhabit.177 Meanwhile, microbiomics is a scientific discipline that utilizes high-throughput molecular approaches to study microbial communities.178,179,180

The Human Microbiome Project (HMP) is a collaborative program initiated by the National Institutes of Health (NIH) in 2007 to explore in depth the diversity and functional of the human microbiota181 (Fig. 2). The project aims to apply high-throughput sequence techniques to characterize microbiota from multiple body sites in human, to investigate the importance of microbiota in IVDR, and to initiate novel research tools and resources for the scientific community. The HMP involved a consortium of more than 200 researchers across multiple institutions, whose findings greatly enhance our knowledge of microbiota and its impact on human health and IVDR. The project was completed in 2012, and its data and resources continue to be used in ongoing microbiota research. The second phase of the HMP started in 2014 and is still in progress.182 With the support of the HMP, numerous revolutionary findings have significantly enhanced our understanding of microbiota-associated disorders. The discovery of these results has led researchers to reevaluate the origin and development of human diseases and utilize the unique capabilities of the microbiota to create innovative diagnostic methods and more accurate treatments. Furthermore, numerous projects such as the Earth Microbiome Project (EMP), Integrative Human Microbiome Plan (iHMP), and Metagenomics of the Human Intestinal Gut (MetaHIT) have been initiated to further analyze the composition and functions of microbiota, resulting in the accumulation of extensive microbiota data represented as phylogenetic or functional composition profiles.41,183,184,185 Meanwhile, it has also facilitated the emergence and development of new microbiota-related fields of discipline.

Recently, the research of drug-microbiota interactions has become a systematically developed field, due to the increasing fascination with the variation of gut microbiota.62,186,187,188,189 This field has given rise to various sub-disciplines, including pharmacomicrobiomics, pharmacometabonomics, and pharmacometagenomics.159,190,191,192,193 These sub-disciplines aim to study the effect of microbiota on PK and PD, and to explore how microbiota variation affects IVDR in human diseases194,195,196,197 (Fig. 3). In addition, these areas explore systems-level studies of microbial metabolites and their diagnostic and implications for pharmacological properties of drugs.57,198,199

An overview of pharmacomicrobiomics is a rapidly growing field that seeks to understand the complex interactions between drugs, microbiota, and host physiology

In fact, during the 1930s, several research teams reported that prontosil had a beneficial effect on mice infections of various strains. But the outcomes were considered atypical regarding the lack of anti-bacterial activity of prontosil in vitro200 (Fig. 2). Nevertheless, sulfanilamide, a metabolite of prontosil, exhibited potent anti-bacterial effects in vivo, suggesting that the anti-bacterial properties of prontosil stemmed from this derivative.200,201 Subsequently, numerous sulfonamide compounds have been synthesized and used to treat bacterial infections, and many studies of drug-microbiota interactions have sprouted among them. Until 2010 when it was first proposed, the term ‘pharmacomicrobiomics’ was used to describe the effects of alterations in microbiota composition and function on IVDR and host genetic190 (Fig. 3). While the interactions between drugs and individual microbiota have been studied for some time, advanced and accurate meta-omics technologies, including 16 S ribosomal RNA (rRNA) amplicon, 18 S rRNA amplicon, ITS, MiSeq, HiSeq, and whole-metagenome shotgun sequencing have facilitated a deeper exploration into the phylogeny, interactions, and functions of microbiota species.202,203,204,205,206 By using these technologies, the development of pharmacomicrobiomics has made significant progress, providing a comprehensive analysis of the connection between drugs and the microbiota.

To study the effects of gut microbiota on drugs, terms such as pharmacometabolomics and pharmacometabonomics can be used in addition to pharmacomicrobiomics.141,191,207,208 These fields focus on the systems-level study of metabolites from microbiota, integrating genomics and metabolomics.198,209,210,211,212 While primarily concerned with microbiota variation, these fields also study the ultimate metabolic products of host-microbiota interactions, namely the metabolites that are generated as a result of drug metabolizing in humans and with its co-occurring microbiota.208,213

Pharmacometagenomics is another term used to study IVDR by integrating the analysis of microbiota genomes and human genetics.193 While metagenomics studies the species and gene pool of ecosystems, pharmacometagenomics specifically examines shotgun sequencing or chance microbiota genome analysis, and it needs to be distinguished from amplicon analysis, which merely determines the phylogenetic tree of the living beings. What characterizes pharmacometagenomics is that it integrates microbiota composition (by 16 S or 18 S amplicon sequencing) and potential function (by metagenomics, metaproteomics, metatranscriptomics, and metabolomics), all elements of microbiomics. The integration of multi-omic approaches is essential, as it can be difficult to solely predict function from microbiota.214

The emerging field connecting the composition and activity of microbiota to prevalent diseases and drugs has spawned numerous promising discoveries in recent years. Association studies have revealed bidirectional effects between various drugs and gut microbiota in patients with gastrointestinal disorders (e.g., inflammatory bowel diseases and colorectal cancer) and other system diseases (e.g., cardiovascular and metabolism disorders, autoimmunity diseases, and psychiatric disorders). These findings have important implications for understanding IVDR and developing precision medicine approaches tailored to individual microbiota composition and function. We will discuss these findings in greater detail below (Fig. 4).

Examples of drug-microbiota interactions with well-defined mechanisms. Created with BioRender.com

In the context of cancer, gut microbiota is known to be critical for the pathogenesis, progression and IVDR of several types of cancers.215,216 Numerous studies have investigated the potential of pharmacomicrobiomics to improve tumor treatment outcomes (Table 2). Here, we describe the mechanisms by which several representative drugs and gut microbiota interact in cancer patients.

CTX is a pro-drug that needs activation, with its primary active ingredient being phosphoramide mustard, which creates crosslinks in inter and intra-strand deoxyribonucleic acid (DNA).217,218,219,220 T cells, particularly cytotoxic T cells, play a crucial role in targeting tumor cells in various cancers, and increased T cell infiltration into tumors has been linked to better patient outcomes.221,222,223,224 Previous microbiota studies have identified specific microbiota associated with improved anti-tumor T cell responses following tumor therapy.225 CTX activates anti-tumor immune response, thereby boosting the differentiation of anti-tumor T helper (Th)-1 and Th17 cells, exhausting pro-tumor regulatory T (Treg) cell populations, stimulating the secretion of various cytokines, and increasing the death of immunogenic tumor cells.226,227,228,229,230,231,232,233 Saxman et al. compared the efficacy of combined CTX with CTX alone and showed IVDR of 18.8% in the combined group compared with 6% in the other group.234 It is worth noting that CTX treatment results in increased intestinal permeability due to decreased function of tight junctions and adhesive junctions.235,236 This increased permeability is associated with higher levels of pathogenic strains, e.g., Escherichia coli (E. coli), Enterobacteraceae, Pseudomonas, and Enterococci in the gut.236 Furthermore, CTX treatment reduces fecal microbiota diversity and increases the Firmicutes-to-Bacteroidetes ratio.237 Several species and families were found in CTX-treated samples but not in untreated samples, e.g., Bacteroides acidifaciencs and Streptococcaceae, even though the implications of this are not well known.237

A study conducted in 2013 discovered that germ-free (GF) and antibiotic-treated mice had a noticeably decrease anti-tumor response to CTX treatment.235 In particular, CTX treatment resulted in increased Th17-mediated pro-inflammatory interleukin (IL) -17 secretion in specific pathogen-free (SPF) mice, but not in GF or antibiotic-treated mice. This research further revealed that the CTX-mediated translocation of gram-positive bacteria, e.g., Lactobacillus johnsonii and Enterococcus hirae (E. hirae), from the gut to the secondary lymphatic organs is vital to differentiation of CD4+ T cells.235 Co-administration of CTX and vancomycin had worse total anti-tumor activity than CTX monotherapy, emphasizing how essential elements of the microbiota influence the action of CTX.235 Further research has identified E. hirae, when translocated from the gut to the secondary lymphatic organs, increased the CD8+ T/Treg cells ratio within tumors.238 On the other hand, Barnesiella intestinihominis does not translocate but accumulates in the colon, stimulating CD8+ T cells and Th1 cells, thus enhancing the infiltration of interferon (IFN)-γ-producing γδT cells into tumors.238 The interaction between IVDR of CTX and the microbiota is complex, as CTX affects the intestinal barrier integrity, resulting in the translocation of pathogenic species, while being affected by commensal bacteria for pharmacological immune-mediated efficacy. Such discoveries emphasize the significance of considering gut microbiota in the use of CTX and other chemotherapeutic agents.

Chemotherapeutic agents such as CPT-11 are commonly for treating several cancers, however, their use can be limited by the occurrence of severe diarrhea as ADRs.239,240,241,242 This side effect can be attributed to the influence of gut microbiota, which can convert the less toxic SN-38-glucuronide (SN-38-G) metabolite back into the extremely toxic 7-ethyl-10-hydroxycamptothecin (SN-38), leading to intestinal damage.243,244 Specifically, certain strains (e.g., E. coli, Staphylococcus, and Clostridium) that express high levels of β-glucuronidase (GUSs), a microbial enzyme that eliminates SN-38-G, have been shown to exacerbate this problem.245 However, antibiotic administration to decrease the abundance of such strains has been investigated as a potential strategy to alleviate CPT-11-induced diarrhea.246,247

In clinical trials, the use of anti-biotics (e.g., levofloxacin, cholestyramine, and neomycin) has been evaluated in conjunction with CPT-11 treatment.246,247 As a result, the overall incidence of IVDR was significantly decreased from the 40% reported in earlier studies.246 Administering neomycin alongside CPT-11 reduced or eliminated diarrhea in six out of seven volunteers, according to another study.247 Although co-administration of these drugs leads to a reduction or elimination of diarrhea, concerns have been raised about the possible consequences for gut microbiota and the emergence of antibiotic resistance.248 Additionally, selective inhibition of microbial GUSs has been investigated as an alternative strategy.249,250 Repurposing existing drugs and designing new GUSs inhibitors have been explored, with promising results demonstrating effective reduction of diarrhea without compromising the anti-tumor effects of CPT-11.249,251,252,253

The administration of probiotics another potential strategy for mitigating CPT-11-induced diarrhea has also been investigated. Enterococcus faecium (E. faecium) and multi-strain probiotics (e.g., Lactobacillus and Bifidobacterium) have shown effectiveness in reducing GUSs activity and diarrhea in clinical studies.254,255 However, the potential risks and costs of probiotics must be carefully considered, particularly when administering them to immunocompromised individuals, as they could potentially increase the risk of superinfection.

In conclusion, the intervention of gut microbiota has shown potential in mitigating the ADRs associated with CPT-11 treatment. While the use of anti-biotics and probiotics has proven effective in reducing diarrhea, their potential modulation of gut microbiota and the occurrence of anti-biotic resistance must be carefully considered. Selective inhibition of microbial GUSs offers a promising alternative strategy to mitigate CPT-11-induced diarrhea. More in-depth studies are warranted to assess the effects and risks of such measures in larger patient populations.

Platinum-containing anti-neoplastic drugs, including oxaliplatin and cisplatin, have been used extensively to treat several cancers.256,257,258 These drugs work by interfering with the DNA replication process in tumor cells, leading to their death.259,260,261 However, the effectiveness of these drugs relies on the generation of reactive oxygen species (ROS), which cause oxidative injury to tumor cells. Interestingly, recent reports reveal that gut microbiota is crucial for the production of ROS and IVDR associated with these drugs.262,263 Specifically, gut microbiota, particularly gram-negative bacteria, regulate the myeloid differentiation primary response 88 (MYD88) related pathway to prepare myeloid cells for ROS release in the tumor undergoing treatment with oxaliplatin.263 This means that the integrity of the microbiota is necessary for the early cytotoxic effects of these drugs. Moreover, gut microbiota also plays a significant role in the late immunomodulatory effects of oxaliplatin.

For an effective anti-cancer immune response, immunogenic gut commensals are required, along with the antigenic properties of the apoptosis mediated by oxaliplatin. Immunogenic bacteria, such as non-enterotoxigenic Bacteroides fragilis and Erysipelotrichaceae, induce migrating dendritic cells (DCs) to transduce IL-1β and IL-12 responses to follicular Th cells.264 This interaction leads to increased IgG2b reaction and increased anti-cancer effector CD8+ T cells activity. It is important to mention that without immunogenic gut commensals, these immune responses are greatly diminished.

Moreover, metabolites such as SCFAs can also help to the improvement of the immune response to oxaliplatin.265 SCFAs, especially butyrate, are able to block histone deacetylases (HDCA) to upregulate the DNA transcription modulator inhibitor of DNA binding 2 (ID2).266 This, in turn, enhances the cytotoxic activity of CD8+ T cells through IL-12 signaling, promoting anti-tumor immunity generated by oxaliplatin. In conclusion, gut microbiota and their metabolites are major regulators of the anti-tumor immunity responses to platinum-based drugs.

ICIs have revolutionized tumor therapy by specifically modulating inhibitory receptors (e.g., CTLA-4, PD-1, LAG-3, and TIM-3) and ligands (PD-L1) presented on immune and tumor cells, ultimately stimulating anti-tumor IVDR.267,268,269 Emerging preclinical evidence also emphasizes the role of gut microbiota in modulating the IVDR of ICIs treatment by influencing the immune response.235,238,263

Studies using preclinical mouse models have shown that an effective anti-tumor response after anti-CTLA-4 antibody therapy requires intact commensal microbiota.270 Anti-biotic treatment or the absence of living microorganisms compromised the anti-tumor efficacy of anti-CTLA-4 antibodies.270 Recolonization experiments with specific strains isolates showed that the introduction of certain commensal bacteria (e.g., Bacteroides fragilis, Bacteroides thetaiotaomicron, and Burkholderia cepacia) was able to recover anti-tumor responses.270 Another study reported that Bifidobacterium was linked to improved anti-tumor efficacy of anti-PD-L1 antibodies, and that orally administered Bifidobacterium improved the anti-tumor efficacy of ICIs by promoting the role of DCs and activating cytotoxic CD8+ T cells.271 These findings indicate that specific commensal bacteria may enhance the anti-tumor efficacy of ICIs.

Further research aimed to identify specific strains in human microbiota that can enhance the effectiveness of anti-tumor treatment. Research on patients with metastatic melanoma, has shown that certain bacterial strains (e.g., Faecalibacterium prausnitzii and Gemmiger formicilis) are linked to a favorable reaction to anti-tumor antibodies, while other strains (e.g., Bacteroides) are linked to adverse linked to ADRs.272 In addition, a greater α-diversity and the existence of particular bacterial (e.g., Ruminococcaceae) are also connected to a positive reaction to anti-PD-1 antibodies.273 In non-small cell lung cancer or urothelial tumor patients with clinically effective to ICIs, Akkermansia muciniphila (A. muciniphila) and E. hirae were found to be more abundant than normal.274 Despite the fact that Bifidobacterium longum, Collinsella aerofaciens, and E. faecium were enriched in melanoma patients who were responsive to anti-PD-1 antibodies, Ruminococcus obeum and Roseburia intestinalis were enriched in non-responders.275 The transfer of microbiota from tumor patients responsive to ICIs to GF- or antibiotic-pretreated mice resulted in enhanced anti-tumor efficacy.273,274,275 In addition, co-administration of a mixture of 11 strains acquired by healthy volunteers was found to enhance the anti-tumor effects of ICIs in GF mice by vigorously inducing the production of IFN-γ by CD8+ T cells.276 Although B cells have been considered to play a potential role in ICIs therapy, the relationship between B cells responses and gut microbiota has not been extensively studied.277

Collectively, these studies indicate that the mouse and human gut microbiota may play a crucial role in regulating tumor IVDR on ICIs by modulating the host immune system. However, there is limited coherence in the key microbiota subgroups reported in these studies, which may be due to factors such as geographical and population differences, specific microbiota associations with certain tumor types, and other aspects of the microbiota that are currently understudied.278,279 To further explore these associations, larger clinical studies and deeper sequencing analyses may be necessary.280

Although targeting gut microbiota to enhance the effectiveness of ICIs efficacy is a relatively new strategy, recent progress has been made. Two early clinical studies using fecal microbiota transplant (FMT) showed the potential for improved responses to ICIs therapy.281,282 In one trial, 10 individuals with anti-PD-1-resistant metastatic melanoma were administered FMT from a donor who had previously achieved complete remission with anti-PD-1 therapy.281 This trial revealed that FMT, along with anti-PD-1 reinduction therapy, is both secure and viable for certain patients, leading to heightened immune activity within the tumor. Different research involved the use of FMT derived from melanoma patients who had shown a long-term response to treat patients with anti-PD-1-resistant melanoma.282 Among the 15 patients, 3 achieved objective responses and 3 had long-lasting stable disease. These findings indicate that manipulating gut microbiota could potentially alter the tumor microenvironment and counteract resistance to ICIs treatment. However, larger patient cohorts are needed to identify the ideal composition of FMT and the microbiota profiles of patients that will improve ICIs efficacy.

Cardiovascular diseases (CVDs) are responsible for 31% of global mortality, causing over 17.7 million deaths worldwide.283,284,285,286 Studies show that alterations in gut microbiota are linked to an increased risk of CVDs, e.g., hypertension, atherosclerosis (AS), and heart failure by affecting IVDR and inflammation.287,288,289,290,291 Pharmacomicrobiomics holds promise for understanding how microbiota affects CVDs and how drugs can be developed to better target microbiota.292 However, more work is necessary to truly unlock the intricate interactions between microbiota, drugs, and CVDs. Next, we describe the mechanisms of drug-microbiota interactions for patients with CVDs (Table 3).

Warfarin, a vitamin K antagonist, has become the mainline of oral anti-coagulant treatment for preventing and treating thromboembolic complications in 1954.293,294,295 However, its narrow therapeutic window and apparent IVDR prompted many studies to identify genetic factors that influence treatment outcomes.296,297,298 Through these studies, mutations predictive of therapeutic response have been identified in VKORC1 and CYP2C9, which encode metabolic enzymes associated with vitamin K and coumarin vitamin K antagonists, respectively.299,300 Nonetheless, approximately 35% of individuals with a delay in responding to warfarin have not been explained by these genetic factors.301 Recently, researchers have investigated the possible impact of gut microbiota response to warfarin, given the known association between gut microbiota and vitamin K metabolism. A trial of 200 patients with varying degrees of reaction to warfarin showed that Bacteroides, Escherichia Shigella, and Klebsiella were prominent in the lower-responders. Escherichia Shigella has the necessary enzymes to produce vitamin K, which may account for the weak response. In contrast, Enterococcus was connected to an increased reaction to warfarin.302 This research is the initial one to establish a connection between gut microbiota and IVDR to warfarin, a medicinal compound that has a limited therapeutic window and can result in bleeding or inefficiency if not dosed appropriately. Although these findings show promise, additional research is required to confirm and refine the results. Determining the impact of gut microbiota in warfarin response has the potential to improve individualized dosing strategies and optimize treatment outcomes.

Statins are commonly utilized for the first and second-line management of CVDs by reduction of low-density lipoprotein cholesterol (LDL-C), the so-called ‘bad’ cholesterol.303,304,305 By inhibiting 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA), statins mainly reduce the de novo synthesis of cholesterol in the liver.306 However, studies in recent decades have shown that statins have a number of pleiotropic effects, including anti-inflammation, anti-thrombosis, and anti-oxidation, which may be involved in their cardiovascular protective properties.307,308 Statins have been proven to inhibit the function of Rho kinase and Rac1 protein, which are associated with the progression of AS, a major risk factor for CVDs. Statins also activate peroxisome proliferator-activated receptor-gamma (PPAR-γ), a nuclear receptor that contributes to the regulation of inflammation, lipometabolism, and glycometabolism.308 However, the mechanisms underlying such effects of statins are poorly understood.

Furthermore, recent research indicates that statins can modify the structure of gut microbiota, leading to changes in the proportion of certain species. One study found that statin treatment dramatically boosted the amount of certain anti-inflammation-related species (e.g., Bacteroides, Butyricimonas, and Mucispirillum), which may help to ameliorate hyperglycemia.309 Meanwhile, another research suggests that statins could cause gut dysregulation by altering the complexity of the BAs pool via a pregnancy X receptor (PXR) based mechanism,310 potentially leading to adverse health effects.311 BAs are synthesized from cholesterol in the liver and are important in the digestion and absorption of dietary fats. Direct inhibition of de novo cholesterol formation by statins could inhibit BAs synthesis in the liver, thereby altering the BAs pool in the human gut. Alterations in the number and structure of BAs in the gut may directly or indirectly induce considerable and complex physiological reactions by altering gut microbiota.

Despite growing evidence that statins might be beneficial for gut microbiota, more studies are warranted to investigate the long-term effects of statin treatment on gut microbiota and whether these changes are consistently beneficial for CVDs. In addition, it is critical to evaluate the possible risks and benefits of statin treatment and IVDR when making treatment decisions for the prevention and management of CVDs.

Digoxin, also known as digitalis, is a drug that can enhance the pumping efficiency of damaged or weak hearts, leading to better clinical stability and exercise capacity.312,313 However, in lower-flow congestive heart failure, digoxin may be ineffective in 10% of patients, due to deactivation by specific strains, highlighting the significance of IVDR from humans and microbiota. The deactivation of digoxin is mediated by the transformation of digoxin to an inactivated state, dihydro-digoxin, by Eggerthella lenta (E. lenta).314,315,316,317 In another groundbreaking research, an operon coding for the E. lenta cytochrome, cardiac glycoside reductase (cgr), was identified to be transcriptionally activated by digoxin, suggesting a predictable microbiota biomarker for digoxin deactivation. The researchers also explored the potential that endogenous digoxin-related substances might have been chosen for inactivation and determined if sensible dietary interference could regulate the inactivation of digoxin in vivo. As previously understood, E. lenta proliferation relies on arginine, which not only improves proliferation but also prevents digoxin deactivation. Hence, increased arginine concentrations, either dietary or microbiota-derived, may be able to suppress this unwanted microbiota function.318,319 To test this hypothesis, the researchers performed an in vivo experiment using mice given two distinct diets: one devoid of any protein and the other containing 20% of total calorie intake from protein. The results showed that an increase in protein intake notably raised levels of digoxin in the blood and urine, but only in mice colonized with digoxin-reducing strains.318 Overall, this research offers a new understanding of how E. lenta deactivates digoxin and emphasizes the possible utilization of dietary interference to regulate this microbiota process. These findings could have significant clinical applications for individuals with low-flow congestive heart failure who may benefit from digoxin therapy. Furthermore, the identification of predictive microbiota biomarkers for drug inactivation could lead to the identification of personalized treatment guidelines on the basis of individual microbiota.

Captopril is a first-generation ACE-Is drug widely used to treat hypertension through the inhibition of the renin-angiotensin system (RAS) in centrally and peripherally sites.320,321 In addition to its anti-hypertensive effects, captopril has also been found to influence the composition of gut microbiota.322,323,324,325 Research has indicated that captopril maintains its anti-hypertensive activity after withdrawal. An animal experiment treating rats with captopril showed an increased abundance of Parabacteroides, Mucispirillum, and Allobaculum.326 In addition, captopril reduces neuronal inflammation in the autoregional area and diminishes sympathetic nerve drive, which balances the gut microbiota.327,328 Recent findings also suggest that mothers treated with captopril have a persistent anti-hypertensive response by reconstituting gut microbiota, improving gut pathologies and permeabilities, and restoring the dysregulated gut-brain axis in male offspring.329 For example, the presence of Clostridia and Clostridiales were higher in rats of maternal captopril. Pregnant rats treated with captopril exhibited a greater mean abundance of the order Clostridiales (e.g., Dehalobacterium, Oscillospira, Roseburia, and Coprococcus) in gut microbiota, in comparison to pregnant rats. As a result, captopril could affect gut microbiota growth and composition in humans, thereby altering the effectiveness of drugs.330 The effect of captopril on gut microbiota may be significant in managing hypertension and associated diseases.

Benazepril and Enalapril are second-generation of ACE-Is and are primarily for treating CVDs (e.g., arterial hypertension and congestive heart failure).331 These drugs work by inhibiting the activity of the angiotensin-converting enzyme (ACE), resulting in a lower generation of angiotensin II, a hormone that narrows the vessels and raises pressure.332,333 Recently, researchers have shown that both benazepril and enalapril have the capacity to positively influence gut microbiota.334,335,336,337 In rats, benazepril treatment was found to promote the restoration of gut microbiota structure by changing the balance of gut microbiota.338,339 Specifically, benazepril is primarily metabolized in the liver and transformed into diacid benazeprilat, which can affect gut microbiota. In contrast, enalapril has been shown to reduce blood levels of trimethylamine N-oxide (TMAO), a compound produced by the gut microbiota metabolizing certain nutrition.340,341 High concentrations of TMAO are linked to a higher incidence of CVDs. Enalapril influences plasma TMAO levels by modifying the composition of gut microbiota and controlling the excretion of methylamines in urine.337 These findings suggest that ACE-Is might be effective in improving gut microbiota composition and cardiovascular health. Therefore, by promoting the restoration of gut microbiota structure and reducing TMAO levels, benazepril and enalapril may help reduce the risk of CVDs.

The endocrine system is a complicated system of glands that produce hormones, which are responsible for regulating various bodily functions such as metabolism, growth, and development.342,343,344 Current evidence suggests that microbiota plays an important role in the regulation of the endocrine system and can influence the IVDR of endocrine disorders.345 The study of pharmacomicrobiomics in the endocrine system has the potential to enhance our knowledge of the underlying mechanisms of IVDR and to advance precision medicine based on the characteristics of microbiota. Further research in this field is necessary to fully realize the potential benefits of this approach. Below, we will list several cases of interaction between anti-diabetics and gut microbiota (Table 3).

Metformin (1,1-Metformin Hydrochloride), a widespread anti-diabetic drug, is also known to have other benefits, such as reducing obesity, and reducing the incidence of CVDs and cancers.346,347 However, the precise mechanisms by which metformin works to regulate glucose levels are not yet fully understood.347,348,349,350,351 While some theories suggest that metformin activates AMP-activated protein kinase (AMPK) and inhibits the mitochondrial respiratory chain, these completely account for all of its positive actions.352 Thus, researchers have investigated mechanisms underlying gut microbiota regulation of glucose metabolism and energy balance, particularly the potential involvement in IVDR.353,354,355 Recently, some work has highlighted how gut microbiota might influence the anti-hyperglycemic effect of metformin. For example, Wu et al. transplanted feces from untreated Type 2 Diabetes Mellitus (T2D) patients to GF mice and observed significant improvement in glucose tolerance compared to control mice. They identified the expression of A. muciniphila and its metal-binding proteins as key factors in metformin-microbiota interactions. This contributed to explaining earlier unresolved questions about the relationship between metformin effectiveness and metal balance.353 Another study by Sun et al. showed that the anti-hyperglycemic effect of metformin involves a new mechanism named the Bacteroides fragilis-glycoursodeoxycholic acid (GUDCA)-intestinal farnesoid X receptor (FXR) axis. This pathway relies on gut microbiota and involves the activation of intestinal FXR signaling to ameliorate metabolic dysfunction. Metformin was found to reduce Bacteroides fragilis, preventing GUDCA degradation.354

In addition to its anti-hyperglycemic effect, metformin demonstrated protective efficacy against CVDs through anti-inflammatory mechanisms including activating the AMPK pathway and inhibiting nuclear factor-κB (NF-κB) pathway.356,357,358 Growing evidence indicates that microbiota-mediated pathways might be implicated in the therapeutic effects of metformin. For example, changes in gut microbiota structure due to metformin treatment may lead to alterations in glucose, hormones, BAs pool, SCFAs, and immunity.351,352,359 Collectively, emerging evidence suggests that the gut plays a vital role in metformin therapy and that microbiota-mediated mechanisms involving changes in gut microbiota structure may be responsible for some of its pleiotropic effects. This has important implications for developing new therapies that target gut microbiota and their interaction with IVDRs.

AGIs are a class of drugs that alter the absorption and metabolism of carbohydrates in the small intestine.360,361 These drugs have been found to significantly affect the structure and variability of gut microbiota, which may have important implications for managing metabolism diseases (e.g., T2D). Acarbose and miglitol are two commonly prescribed AGIs that have been widely evaluated for their impact on gut microbiota. In vitro, experiments have shown that acarbose selectively inhibits the development of E. coli by inhibiting the maltose importer, while clinical trials have indicated that acarbose enhances the abundance of microbiota that produce SCFAs, e.g., Prevotella, Lactobacillus, and Faecalibacterium.362,363 SCFAs are known to be beneficial for metabolic health, and treatment with lactobacillus has been shown to reduce glycemia.364,365,366 Furthermore, a study demonstrated that Dialister, a taxon found in gut microbiota, was enhanced after acarbose therapy and had a negative correlation with Hemoglobin A1c (HbA1c), suggesting a possible role in controlling glucose metabolism.362 Acarbose has also been shown to increase Bifidobacterium and Lactobacillus, and reduce harmful Bacteroidaceae, Enterobacteriaceae, and lecithinase positive Clostridium in stool collected from people with hyperlipidemia or T2D.367,368

Miglitol, on the other hand, has anti-inflammatory properties in mice, inhibiting histological and molecular indicators of inflammatory reaction and decreasing intestinal transit time generated by high-fat and high-glucose diet (HFHSD).369 Consuming an energy-dense diet has been shown to increase Erysipelotrichaceae and Coriobacteriaceae, but miglitol has been found to reverse this effect. It has been hypothesized that the reduction in intestinal inflammation is connected to these alterations in gut microbiota. However, the effect of miglitol on the heterogeneity and structure of gut microbiota remains unknown and requires further study. Overall, the results of these findings indicate that AGIs, particularly acarbose, may be useful in reestablishing gut microbiota balance in individuals with T2D. By increasing SCFAs-producing microbiota and decreasing potentially harmful microbiota, these drugs can potentially impact metabolic health with positive effects. More studies are necessary to fully elucidate the IVDR of AGIs on gut microbiota and their impact on metabolic health.

Glucagon-like peptide-1 (GLP-1) is an incretin hormone produced by enteroendocrine cells as a result of dietary.370,371,372 Its role in enhancing glucose-induced insulin secretion, inhibiting glucagon secretion, and suppressing appetite and gastric emptying makes it a powerful target in treating diabetes and overweight.370,373 Recent studies have emphasized the function of gut microbiota in regulating satiety and glucose homeostasis by inducing GLP-1 secretion.374,375,376,377 Furthermore, GLP1-RAs (e.g., liraglutide), an emerging sub-class of anti-diabetic drugs, have been shown to modulate the intestinal microenvironment and alter gut microbiota composition.378,379,380,381,382 In general, the Firmicutes-to-Bacteroidetes ratio is considered an important indicator of gut microbiota structure. Studies have shown that liraglutide can modify gut microbiota to a leaner proportion in normally-weighed diabetes mice, resulting in a higher Firmicutes-to-Bacteroides ratio.378 Interestingly, another research demonstrated that liraglutide increased the Bacteroides-to-Firmicutes ratio to decrease body weight in both simply overweight and diabetes-overweight individuals independent of glycaemic condition.380 Such differences are probably attributed to varying levels of hyperglycemia and different model systems used. GLP1-RAs have been proven to profoundly change the structure of gut microbiota in diabetic male rats.379,383 These changes include selective enhancement of several SCFAs-producing microbiota, e.g., Bacteroides, Lachnospiraceae, and Bifidobacterium.379 Furthermore, GLP1-RAs can re-establish, at least in part, the homeostasis of gut microbiota.383

DPP4Is (e.g., sitagliptin and vildagliptin) are first-line hypoglycaemic drugs approved by the American Association of Clinical Endocrinologists for T2D.370,384 DPP4Is decrease blood glucose primarily via inhibition of GLP-1 degradation.385 Previous studies have suggested the DPP-4-related function of gut microbiota may be a goal for DPP4Is, potentially opening up novel therapeutic applications for DPP4Is to modulate gut microbiota discordance. Studies have shown that DPP4Is can improve glycaemic control through increased Bacteroidetes, thereby greatly reverting high-fat diet (HFD)-induced alterations in gut microbiota.386 The study of the effect of sitagliptin on gut microbiota showed decreased Bacteroidetes, and increased Firmicutes and Tenericutes. However, sitagliptin moderately restored microbiota imbalance and modified the abundance of SCFAs-producing microbiota in T2D rats.387 Similarly, vildagliptin administration was related to increased Bacteroidetes and decreased Firmicutes coupled with decreased Firmicutes-to-Bacteroidetes ratio in diabetic rats.388 Moreover, vildagliptin has been reported to produce benefits by modulating gut microbiota, leading to increased Lactobacilli and decreased Oscillibacter.385 To clarify the mechanisms of variation, researchers experimentally confirmed that vildagliptin inhibited Toll-like receptor (TLR) ligands in caecal content and restoration of antimicrobial peptide levels and ileum crypt depth.385 Studies have also shown that DPP4Is may indirectly reduce the secretion of pro-inflammatory cytokines in the liver through their effect on gut microbiota. Overall, these studies highlight the importance of DPP4Is on gut microbiota and reveal promising strategies to improve glucose homeostasis and IVDR.

The digestive system is one of the most extensive areas of pharmacomicrobiomics because it is home to complicated and varied microbiota that is critical in drug metabolism and efficacy.189,389 Gut microbiota has also been demonstrated to impact outcomes of drugs used to treat a variety of digestive diseases, e.g., inflammatory bowel diseases (IBD) and irritable bowel syndrome (IBS).390 Patients with IBD have been detected to have altered gut microbiota composition and function, which may affect IVDR. This has contributed to the advancement of microbiota-based treatments for IBD, e.g., FMT, which involves transplanting a healthy gut microbiota into a patient to restore microbial balance and improve treatment outcomes. Overall, the field of pharmacomicrobiomics in the digestive system has enormous potential for improving drug efficacy and reducing ADRs. However, deeper studies are necessary to better comprehend the complicated between gut microbiota and drugs, as well as to develop microbiota-based therapies for various digestive and metabolic disorders (Table 3).

SAS is a medication that was initially developed with the aim of treating inflammatory conditions caused by bacterial infections. However, its effectiveness was later discovered in treating ulcerative colitis (UC).391 The drug is composed of aminosalicylate and sulfapyridine, which are linked together by an azo bond. One of the unique properties of SAS is that it is not rapidly absorbed in the up-stream gastrointestinal tract. Instead, it is broken down into its components by gut microbiota in the colon. Sulfapyridine is then absorbed into the bloodstream, while mesalazine (5-aminosalicylic acid, 5-ASA) can be inactivated in the colon.392 A PK study in health subjects revealed that gut microbiota are critical in activating SAS. This discovery might explain why this drug is better at treating UC than Crohn’s disease (CD).393 The ability of gut microbiota SAS could be enhanced by administering probiotic strains (e.g., Lactobacillus acidophilus L10, Bifidobacterium lactis B94, and Streptococcus salivarius K12). In an in vitro trial, researchers incubated the contents of a rat colon with SAS alone or SAS combined with probiotics under anaerobic conditions.394 They found that the probiotics possessed azoreductase activity, allowing them to metabolize SAS. The samples incubated with SAS plus probiotics had a greater level of 5-ASA and sulfapyridine restored. Despite SAS being an efficient and inexpensive therapy for UC, some patients have reported ADRs when using it, such as nausea, skin rash, and anorexia.395 This may be caused by the metabolism of SAS through gut microbiota. As a result, SAS is less popular than other treatments for UC.

PPIs are widespread drugs for the treatment of acid-induced diseases including peptic ulcers, gastro-oesophageal reflux, dyspepsia, gastro-duodenopathy, and bleeding.396,397,398 Due to their effectiveness and safety properties, PPIs therapy has expanded rapidly in the last decades. Controversies have arisen, particularly relating to the safety profile and possible ADRs of chronic therapy with PPIs. Up to 70% of PPIs prescriptions have been estimated to be unneeded.399 Once started, the original indication for PPIs is rarely re-evaluated, with later withdrawal attempts resulting in needless long-term use.400,401

A large-scale patient-based clinical trial in the Netherlands shown that PPIs are among the drugs in greatest correlation with reduced gut microbiota diversity and taxonomic changes.402 Studies analyzing 16 S data from various cohorts found that PPI use led to decreased abundance of gut microbiota and increased oral microbiota.403,404,405 For example, Enterobacteriaceae, Enterococcaceae, and Lactobacillaceae increased, Ruminococcaceae and Bifidobacteriaceae decreased, while alterations towards typically oral microbiota are represented by Rothia dentocariosa, Rothia mucilaginosa, Actinomyces, and Micrococcaceae increased.404 In addition, the observed changes appear to be a class-efficacy of PPIs, with increased doses linked to greater microbiota changes.115 Recent research has shown that PPIs use is highly relevant to 24 taxa and 133 pathways, with predictable function alterations including increased biosynthesis of fatty acid, lipid biosynthesis, and L-arginine, and degradation of purine de-oxyribonucleoside.115 Another in vitro experiment testing direct affected by popular drugs on gut microbiota demonstrated significant alterations in growing speeds, meaning that bonding of PPIs to bacterial H+/K+ ATPases may mediate the directly influence.406 The reduction in gastric acidity caused by PPIs allows oral microbiota to colonize gut microbiota, resulting in altered taxonomic homeostasis and potentially increased susceptibility to intestinal infections, e.g., Clostridium difficile (C. difficile), Campylobacter, and Salmonella.407,408,409,410 Moreover, PPIs initiation and withdrawal may cause alterations in gut microbiota, potentially exacerbating liver cirrhosis.411 Furthermore, long-term PPIs use, particularly in childhood, can cause permanent alterations in the growing gut microbiota, potentially leading to obesity later in life.412 Although PPIs are widely recognized as harmless and efficient, medical practitioners should reassess the extensive, long-term impact and OTC accessibility. The rapid and widespread shift to PPIs use has led to variations in changes in gut microbiota, affecting up to one-fifth of the population. Therefore, it is important to meticulously evaluate the long-term impact of PPIs-induced alterations on gut microbiota, especially during early development, and their potential effects on health and disease in later life.

NSAIDs are generally used to alleviate pain and inflammation, but their use can cause ADRs, such as stomach ulcers and damage to the mucous membrane of the small intestine.413,414 This is due to the fact that the bacterial glucuronidase metabolizes NSAIDs in the intestine, similar to the case of CPT-11, which converts the drug into toxic metabolites that damage the intestinal mucosa.415 The glucuronides of NSAIDs produced in the liver reach the intestine via the bile, where bacterial GUSs hydrolyse them into aglycones.416 These aglycones are then reabsorbed and converted by cytochrome P450 into potentially cytotoxic intermediates that cause intestinal toxicity.417 Like CPT-11, a recent study demonstrated that Inh1 (Inhibitor 1), a novel microbe-specific GUSs inhibitor, can reduce the intestinal ADRs of diclofenac.418 This suggests that inhibiting GUSs is a promising approach to reducing the adverse effects of certain drugs. While gut microbiota produces GUSs with beneficial functions to the organism, opportunist or intestine-pathogenic species, e.g., Clostridium perfrengens and E. coli, are responsible for the de-glucuronidation of drugs producing harmful metabolites. This is due to intracellular variations of GUSs, including variations in conformation, hydrophobicity, and mobility.419 The enhancing potential of bacterial GUSs-induced ADRs, especially gut injury, is not unique to NSAIDs and CPT-11. Other drugs including Regorafenib, an anti-tumor tyrosine kinase inhibitor, and venotonic flavonoids, have also been shown to be substrates for GUSs.420,421,422 Such evidence emphasizes the value of understanding the importance of bacterial enzymes in IVDR to develop strategies to reduce ADRs.

Although pharmacomicrobiomics research is still in its infancy, preliminary studies indicate that microbiota may play a key role in regulating IVDR in the nervous system. One example of this is the use of probiotics to enhance the effectiveness of anti-depressant medication.423 Several researches have indicated that certain strains of gut microbiota can produce neurotransmitters, e.g., serotonin and gamma-aminobutyric acid (GABA), which are known to regulate mood and anxiety. By supplementing these strains with probiotics, the researchers hope to increase the availability of these neurotransmitters and thereby improve the response to treatment with anti-depressants. Another area of interest is the effect of the microbiota on neurodegenerative diseases, e.g., Parkinson’s disease, for which the commonly used therapeutic drug is levodopa (Table 3).

Levodopa is a drug for the treatment of Parkinson’s disease, a neurological disorder described by tremors, stiffness and difficulty in moving.424,425 Levodopa is taken orally and must be reabsorbed through the small intestine to reach the brain, where it is transformed into dopamine by the tyrosine decarboxylases (tyrDCs). The effectiveness of the drug depends on its bioavailability to the brain, and the drug is usually used alongside inhibitors of catechol metabolic (e.g., carbidopa and entacapone) to inhibit extra-brain metabolism. Emerging evidence suggests that gut microbiota can metabolize levodopa. Certain species, e.g., Enterococcus faecalis (E. faecalis) and Lactobacillus, have been found to contain tyrDCs.426,427 However, this microbiota metabolism reduces the amount of levodopa that is available to the brain, resulting in decreased drug efficacy. Additionally, microbiota metabolism of levodopa can also lead to ADRs, as E. lenta and other ten species can further convert dopamine to m-tyramine, which can cause hypertensive crisis.427

It is interesting to note that gut microbiota can also directly metabolize carbidopa and entacapone. For example, E. faecalis has been shown to metabolize both levodopa and entacapone with 98.9% efficiency.42 In turn, entacapone would reduce some species, e.g., Ruminococcus torques, which can still metabolize entacapone with 84% efficient.42,406 Such findings illustrate the complex interactions between gut microbiota and IVDR, which can influence drug efficacy and safety. While the field of pharmacomicrobiomics in the nervous system is still in its early stages, it has the opportunity to broaden our insight into drug-microbiota interactions and lead to the emergence of novel therapeutic strategies for neurological and psychiatric disorders.

The locomotor system is an indispensable part of the human body, consisting of bones, muscles, and joints, which work together to allow movement.428 The health of the locomotor system can be affected by various diseases, e.g., osteoarthritis (OA), rheumatoid arthritis (RA), and osteoporosis, which can result in pain, stiffness, and reduced mobility. In recent years, a growing focus has been placed on the impact of gut microbiota in the pathology and treatment of these diseases.429 One example of such a drug is methotrexate (MTX), which is commonly used to treat RA (Table 3). Evidence indicates that gut microbiota can influence the effect of MTX, the details of which will be described later.

Another example is NSAIDs, which are frequently applied to treat pain and inflammation related to various locomotor system disorders. However, these drugs can also cause gastrointestinal side effects, such as ulcers and bleeding. Research indicates that gut microbiota plays a crucial role in the metabolism of NSAIDs and can affect their efficacy and safety. Similar to the description of NSAIDs in digestive system diseases above, this will not be repeated.

MTX is a potent cytotoxic drug used to treat autoimmune diseases (e.g., RA).430 Its underlying action is based on inhibiting dihydrofolate reductase and thymidylate synthetase, thereby preventing de novo synthesis of pyrimidines and purines. While MTX is effective, it also has a high incidence of ADRs, including gastrointestinal, hematological, nephrotoxicity, and hepatotoxicity.431 One potential factor that may impact the response to MTX is gut microbiota. Research has indicated that MTX treatment could modify the composition of gut microbiota, decreasing the abundance of enterobacterial, especially E. faecium, and increasing the abundance of Lachnospiraceae.432 However, the decrease in beneficial strains (e.g., Ruminococcaceae, Bacteroidetes, and Bacteroides fragilis) may accentuate the gastrointestinal ADRs, particularly intestinal mucositis.433,434

Interestingly, the association between MTX and gut microbiota is bidirectional.435,436 The variability of gut microbiota may affect MTX therapy responses. The study found that patients with greater gut microbiota diversity, who had statistically abundant Prevotella maculosa, responded better to MTX.436 Despite the promising results, the response rate to MTX has highly IVDR, between 10% and 80%, with just 40% of patients obtaining a therapy blood concentration, and the ADRs are significant.60,437 Genetic factors have been explored to predict the IVDR to MTX, but they have not achieved clinical relevance.438,439,440 More investigations are necessary to assess the feasibility of using gut microbiota as a biomarker for MTX response and to determine its clinical significance. Nevertheless, promising evidence indicates that gut microbiota may play an important role in the PD of MTX, and further exploration may contribute to the emergence of personalized therapy for patients.

The genitourinary system is an essential system for reproduction and waste excretion.441 Recently, evidence indicates that microbiota of the genitourinary system affects the efficacy and toxicity of several drugs, leading to the emergence of a field of pharmacomicrobiomics in this system. In particular, genitourinary microbiota has been shown to affect the effectiveness of androgen deprivation therapy (ADT), a frequent therapy for prostate cancer. ADT typically involves the use of androgen synthetic inhibitor (e.g., abiraterone acetate) or androgen receptor antagonist (e.g., enzalutamide, degarelix acetate) (Table 3).

Recently research has revealed the influence of ADT on the immune system and gut microbiota, and how this affects the progression of prostate cancer (PCa).442 ABI is a widely used drug in ADT, and research by Terrisse et al. underlined the crucial role of thymus-dependent T cells in regulating PCa progression, as CD4+ and CD8+ T cells consumption leads to a partial decrease in tumor development controlled by IVDR.443 Furthermore, the beneficial gut microbiota, e.g., A. muciniphila and Lachnospiraceae, were depleted in patients treated with ABI. However, this depletion could be reversed by the effects of the drug, which remodels the gut microbiota and promotes the proliferation of anti-inflammatory A. muciniphila, thereby elevating microbiota production of vitamin K2.444 Furthermore, the research suggested that specific gut microbiota, such as those with higher α-diversity and A. muciniphila, were related to better efficacy.444,445 Further studies by Sfanos et al. found that the genitourinary microbiota composition of men taking oral androgen-targeted therapy (ATT) differed from those taking Gonadotropin-releasing hormone (GnRH) agonists or antagonists monotherapy or not taking ADT. Species able to synthesize steroids, e.g., A. muciniphila and Ruminoccocaceae, were enriched in microbiota of men treated with orally ATT, which could affect disease progression and immunotherapies.445

Another study showed that gut microbiota could regulate the levels of circulated sexual hormones via effects on human cells, but also by direct biotransformation or synthesis, thereby promoting ADT resistances.446 PCa patients demonstrated higher levels of strains e.g., A. muciniphila and Ruminoccocaceae, which are capable of synthesizing steroid hormones via CYP17A1-like bacterial enzymes. Treating cultivated bacteria with CYP17A1 inhibitor ABI inhibited androgen biosynthesis. However, the response of the microbiota to reduced androgen levels leading to the expansion of strains capable of synthesizing androgens, has yet to be elucidated. In conclusion, the ability of the microbiota to regulate circulating sex hormones and the expansion of bacteria capable of synthesizing androgens can drive resistance to ADT. Future studies are warranted to clarify the intricate interactions between gut microbiota, PCa, and therapy.

In the respiratory system, microbiota plays an important role in maintaining health by modulating immunity, protecting against pathogens, and maintaining the mucosal barrier.447,448,449 Studies have shown that drugs may influence the composition and function of respiratory microbiota, and conversely, microbiota may impact drug metabolism and response. For example, anti-biotics can reduce microbiota diversity and enhance the growth of opportunistic pathogens that can cause chronic respiratory diseases. Inhaled corticosteroids have been shown to increase microbiota diversity and decrease the abundance of pathogenic bacteria, which may contribute to their therapeutic efficacy. Furthermore, the respiratory microbiota can impact drug delivery to the lungs. One of the best-known examples is anti-tuberculosis (anti-TB) therapy with the anti-biotic isoniazid (INH), rifampin, and pyrazinamide (PZA) (Table 3).

The impact of anti-TB anti-biotics on the structure of gut microbiota has been studied in mice infected by Mycobacterium tuberculosis.450,451 The study found that treatment with INH, RIF, and PZA for three months led to significant and persistent eco-dysbiosis in a mouse model. The composition of gut microbiota was altered, with decreased Clostridiales and Lactobacillus, while increasing Porphyromonadaceae, Bacteroidetes, and Proteobacteria.452 The study further showed that a single dose of RIF decreased microbiota diversity, while either INH or PZA alone resulted in alterations to microbiota composition compared to gut microbiota of mice not treated with anti-biotics. Specifically, INH was found to enrich Gordonibacter, while PZA was found to enrich Marvinbryantia.406,452,453 Although TB infection did not dramatically alter gut microbiota composition, it was found to modulate the mucosal immune response. In humans, first-line anti-TB pharmacotherapy seems to have a minimal impact on gut microbiota.454 However, some strains have been demonstrated to regulate immunological responses in the host, such as Bacteroides, which produce polysaccharides that increase Treg cell responses and mediate mucosal tolerance.455 Prevotella has been associated with enhanced Th17 cells-mediated inflammation, while Lactobacillus promotes innate and adaptive immunological responses.456,457

Studies have also found that anti-TB drugs can reduce the microbiota necessary for intestinal homeostasis, increasing the risk of recurrent TB.458 T cells in previously treated patients also recognize type-2 epitopes poorly compared to patients without TB. The type 2 epitopes in the human gut are associated with mycobacteria. Multi-year trials of anti-TB treatment can contribute to a deeper understanding of the importance of microbiota in the progression of TB. A combination of anti-TB anti-biotics and host modifiers that target the host pathway may reduce treatment time and severity of TB, as well as the risk of reinfection. Emerging studies on host-pathogen correlations, host immunology, and host-targeted therapies indicate that this approach may be beneficial in the treatment of TB.459

Pharmacomicrobiomics is an emerging area that aims to comprehend the intricate relationships among drugs, microbiota, and host physiology.460 Understanding the multiple mechanisms of action in pharmacomicrobiomics is critical to the advancement of effective and safe treatment strategies that take into account the multiple mechanisms of interaction between IVDR and microbiota.

Gut microbiota is known to affect the effectiveness and acceptability of drugs by enzymatic modification structure and alteration of bioactivity or toxicity. A list of specific families of enzymes encoded by the microbiota and their effects on drugs is presented in Table 4. Conclusively, gut microbiota can impact IVDR in several ways, including (1) Alteration of drug metabolism: Microbiota can influence the activities of enzymes that metabolize drugs within the body, which can influence the efficacy and side effects of drugs.461,462 For instance, the microbiota can produce beta-lactamases that break down beta-lactam anti-biotics, rendering them ineffective.463,464,465,466 (2) Induction of drug resistance: Microbiota can develop resistance to drugs over time, making them less effective in treating infections.282,467 This resistance may be due to mutations in the DNA of microbiota or the acquisition of resistance genes from others. (3) Modification of drug targets: Microbiota can modify the drug targets in the body, making them less susceptible to the drug’s effects. For example, the microbiota can modify the target site of fluoroquinolone anti-biotics, thereby reducing their ability to bind and inhibit the enzyme.468,469 (4) Interaction with PK: Microbiota can also impact PK, which can affect drug efficacy and safety. For example, gut microbiota can modify the ADME of drugs, altering their therapeutic effects and toxicity.470 (5) Destruction of the host microbiota: The use of anti-biotics can interfere with the natural ecosystem of the host’s microbiota, leading to the overgrowth of pathogenic species (e.g., C. difficile).471,472,473 This can cause infections and other ADRs, which can impact drug safety. (6) Modulation of host immune response: Gut microbiota can modulate immune function in the host, which may influence the activity and toxicity of drugs that target the immune system.474 For example, gut microbiota can modulate the effectiveness of ICIs in tumor treatment by modulating the host immune response.61,475,476,477,478 (7) Regulation of gut barrier function: Gut microbiota may influence the integrity of the gut barrier, thereby affecting drug absorption and gut-host interactions.479,480 For example, gut microbiota can regulate the level of tight junction protein expression in the intestine, thereby affecting drug absorption.

In summary, microbiota can impact IVDR through a variety of complex and multifaceted mechanisms. A deeper insight into such relationships may help in the discovery of more beneficial and safer drugs, as well as in the management of IVDR.

There has been an increasing emphasis on the influence of drugs on gut microbiota and its influence on human healthcare. In a significant and comprehensive study, the authors evaluated the anti-microbial properties of more than 1000 drugs, among them 835 targeted agents that affect human cells.406 Of these, 24% showed anti-bacterial effects, influencing the proliferation of a minimum of 50% of bacterial strains tested. To sum up, several mechanisms are involved by which drugs affect gut microbiota, (1) Direct anti-biotic effects: Anti-biotics can selectively kill off certain microbiota, including both harmful and beneficial species.392,481,482 This can lead to imbalances in gut microbiota, which can have negative effects on human health. (2) Altered gut motility: The speed at which food passes through the intestine affects the proliferation and function of gut microbiota. Certain drugs, such as opioid painkillers, can slow down gut motility, which can lead to overgrowth of harmful bacteria.483,484 This may result in gastrointestinal symptoms like constipation and bloating, and in some cases can cause infections (e.g., C. difficile).485 (3) Modulation of immune function: Gut microbiota has a key function in modulating gut immunity. Several drugs, such as ICIs, can affect gut immunity which in turn can affect gut microbiota.486 For example, ICIs can decrease gut inflammation, which can lead to alterations in gut microbiota.487 (4) Changes in pH: The pH value in the intestine affects the growth and survival of different types of bacteria. Some drugs, such as PPIs used to treat acid reflux, can change the pH value of the gut, which affects the proliferation of different microbiota, thereby affecting the overall composition of gut microbiota.404 (5) Interference with microbial metabolism: Gut microbiota has a vital function in metabolizing compounds in the gut. Several drugs, such as NSAIDs, can interfere with microbial metabolism, which may have an effect on gut microbiota. In particular, NSAIDs can interfere with gut microbiota metabolism of BAs, leading to changes in gut microbial community.488 (6) Dietary changes: Certain drugs, such as laxatives or anti-diarrheal agents, can change the dietary environment in the gut.489,490 This may influence gut microbiota by changing the availability of nutrients and other compounds that gut microbiota use to grow and survive.

It’s important to note that a drug’s impact on gut microbiota can be either positive or negative. For example, certain probiotics or prebiotics are able to actively regulate gut microbiota, which may have a positive effect on the individual’s well-being.491 However, taking anti-biotics or other drugs that negatively affect gut microbiota can result in dysbiosis, which negatively affects human health. Therefore, it is essential to carefully evaluate the potential impact of drugs on gut microbiota and to develop personalized drug treatments that take into account an individual’s gut microbiota composition and function.

This review section identifies challenges and offers strategies for professionals, clinicians, and scientists involved in microbiota research and its influence on human well-being.

Pharmacomicrobiomics is a fast-developing discipline that aims to comprehend the intricate interaction between gut microbiota and drug response. While there have been significant advancements in this area, several challenges remain: (1) Lack of standardized methodologies: Currently, there is a lack of standardization in sample collection, sequencing, and data analysis in pharmacomicrobiomics studies. This creates a challenge in comparing findings among different studies and hinders the development of guidelines for clinical practice. (2) Complexity of gut microbiota: Gut microbiota is an intricate and ever-changing ecosystem that is impacted by various elements, including diet, lifestyle, and host genetics. It is difficult to disentangle the effects of these factors from drug-induced changes in the microbiota. (3) IVDR: There is significant inter-individual variability in gut microbiota, which can impact drug-microbiota interactions. (4) Limited understanding of mechanisms: Despite increasing evidence of drug-microbiota interactions, there is still a lack of comprehensive understanding regarding the mechanisms behind these interactions. It is also unclear how these interactions vary across different drugs and different patients. (5) Clinical translation: Although the potential clinical applications of pharmacomicrobiomics are promising, there are still many obstacles to be overcome before these findings can be translated into clinical practice. For example, the development of microbiota-based biomarkers for drug response will require large-scale validation studies and regulatory approval. (6) Ethical considerations: As with any field of personalized medicine, there are ethical considerations when using microbiota data to make clinical decisions. These include issues around privacy, data sharing, and potential discrimination based on microbiota characteristics. (7) Sample size: Numerous pharmacomicrobiomics studies often have limited sample sizes, potentially restricting their statistical power and the ability to generalize findings. Additional research of greater scale is required to confirm and expand upon preliminary results. (8) Diversity of populations: Most studies in pharmacomicrobiomics have been conducted on Western populations, and there is a lack of diversity in terms of ethnicity, geography, and lifestyle factors. This limits our understanding of how drug-microbiota interactions may vary across different populations. (9) Confounding factors: Various elements, such as diet, age, sex, and environmental exposures, can have an impact on gut microbiota and confuse the interpretation of the results, making it challenging to distinguish the effects of the drug from other factors.

Overall, addressing these challenges will require collaboration between researchers, clinicians, and industry partners to develop standardized methodologies, improve our understanding of the mechanisms underlying drug-microbiota interactions, and ensure that findings are translated into safe and effective clinical practice.

In light of these facts, to overcome these challenges, several strategies have been proposed.

To advance research in the field, it is important to provide practitioners and researchers with a ready-made database of selected knowledge. Current web resources are highlighted in Table 5. However, such databases are scarce, which can lead to valuable time being wasted and hinder research efforts. Therefore, it is necessary to establish new databases or contribute to existing ones as a member of a volunteer team, a crowdsourcing participant, or a crowdfunding supporter. In particular, it is important to link the microbiota reference genome databases, the microbiota-diseases relationships databases, and the microbiota-drug association prediction databases to the pharmacomicrobiomic data resource.

The methods for investigating pharmacomicrobiomics are identical to the overall techniques employed to decipher the microbiota at various levels, including its taxonomic and genomic makeup, gene inventory, functional capacity, the tangible manifestation of its genes (at the RNA or protein level), and ultimately, its true functionality is indicated by the amalgamation of metabolites derived from the microbiota. Therefore, the analysis of drug-microbiota interactions can be effectively conducted using various techniques including amplicon sequencing (16 S or 18 S rRNA subunit), shotgun microbiota sequencing or metagenomics, metatranscriptomics, metaproteomics, as well as metabolomics and metabonomics, as listed in Fig. 5.

Multi-omics strategies facilitating pharmacomicrobiomics towards human precision medicine

These high-throughput data need to be modeled by artificial intelligence (AI) algorithms using advanced computational techniques.492 Machine learning is a sub-field of AI in which machines can acquire knowledge from data without the need for explicit programming.199 Machine learning includes both supervised and unsupervised algorithms. Classification or prediction tasks often use supervised algorithms, while clustering tasks often use unsupervised algorithms to group data based on similarity. The application of these methods could effectively address the needs of clustering and predicting patients, as well as discovering new biomarkers.493 The integrated analysis of multi-omics data has been enhanced by advances in machine learning. These approaches consist mainly of concatenation, modeling, and transformation-based techniques.

It is essential to develop predictive models and software that simulate drug-microbiota interactions to reduce research time and costs. A major limitation in developing such models is the need for aggregated evidence to drive reliable assumptions that can be applied for testing and validation. By combining chemoinformatics with genomic and proteomic similarities, new interactions between drugs and various microbial enzymes can be predicted. This would significantly facilitate the establishment of predictive models for drug-microbiota interactions.

Prebiotics, which are indigestible components of food, stimulate the development of beneficial microbiota in the gut, while probiotics are live microorganisms that can be beneficial to health when ingested in moderation.494 Both prebiotics and probiotics have the potential to modify the composition and activity of gut microbiota and potentially enhance drug efficacy and reduce ADRs.495 Several recognized effects include: anti-bacterial effects by modulating pathogens’ genetic code composition, inhibiting competition for pathogens’ binding domains, inhibiting pathogens’ virus code or proteome composition, and stimulating the immune response by increasing anti-inflammatory cytokines and rescuing and modulating pro-inflammatory compounds.495

FMT involves the transfer of fecal material from healthy donors to patients to restore gut microbiota.496 Initially, FMT was developed to treat C. difficile infection, more recently it has been highlighted as a promising approach to treating some complex digestive diseases.497,498 In addition to providing a diverse, stable gut microbiota, FMT also provides AAs, SCFAs, and BAs that assist in restoring normal gut physiological activity.499 Furthermore, the donor microbiota and their related anti-bacterial products, like adhesins, may be able to competitively match the site, inhibiting pathogenic microbiota colonization of the gut and preventing damage, which is a key action of FMT treatment.500

Bacteriophage therapy, a practice dating back to the 1940s, utilizes naturally-occurring viruses to combat bacterial infections but has recently been enhanced with bio-engineered phages and associated lysing proteins to fight multi-drug resistant microbiota.501 Phages infect and lyse bacteria, and have several advantages, such as minimal harm to humans, breakdown of biofilms, specificity to certain hosts, and self-replication.502,503,504 Additionally, phages play a crucial role in ecosystem regulation by controlling microbiota.505,506,507 Phage therapy involves the use of ‘phage cocktail’, a mixture of lytic phages proven effective against the targeted pathogen.502 However, there are challenges associated with the therapy, including the need to selectively induce lysogenic phages, persistence in the gut, and immune responses.508,509 Phage therapy must also avoid interfering with standard microbiota function and creating selective pressure on non-target microbiota, which can lead to resistant offspring and disrupt beneficial bacteria and host adaptation mechanisms.510 The field is still expanding and requires further exploration.

Human microbiota are valuable resources for identifying targets associated with various illnesses and IVDR. These biomarkers could potentially be used as effective replacements for procedures such as biomicroscopy and endoscopy, making them a promising tool for diagnosis and prediction.43,511 The microbiota releases substances and metabolites that can influence the occurrence, development, and outcome of disease, which highlights the value of microbiota biomarkers for individualized treatment.512,513 However, the inter-individual variability of the human microbiota, the emergence of new multi-drug resistance microbiota strains, and different microbiota drugs modify mechanisms hamper precision medicine. Nevertheless, precision medicine still holds the ideal opportunity for future theranostics, with a full understanding of the impact of the microbiota on IVDR allowing stratifying patients on the basis of identified biomarkers, microbiota types, and metabotypes.514,515

The integration of multiple high-throughput multi-omics datasets can provide a comprehensive profile of IVDR, both at a populational and single-cell level. The database includes five modules: amplicon sequencing (16 S or 18 S rRNA subunit), metagenomics, metatranscriptomics, metaproteomics, and Metabolomics/Metabonomics (Fig. 5). This approach allows the investigation into the genetic variation and the regulation of molecular pathways in diseases and facilitates novel therapeutic strategies. To support extensive IVDR research, an open and integrated database, called the IVDR Atlas, has been established. These modules provide researchers with the opportunity to explore extensive gene variation and modulation datasets generated by multi-omics sequencing.

Hopefully, the nascent discipline of pharmacomicrobiomics, which involves the study of drug-microbiota and drug-microenzyme interactions, is gaining traction due to the accessibility of on-line information, biomarker identification, and advances in methods, models, and software. The continuation of this pattern is anticipated through increased systematic screening, which will aid in the creation of new clinical practice recommendations and the implementation of existing guidelines. These developments, which are encouraged by the current advancements in technology, contribute to a growing tendency towards ‘precision medicine’ and the ‘One Health approach’. It is anticipated that the inclusion of microbiota typing and pharmacomicrobiomic testing in treatment protocols and drug labels will become customary, although the timing of this paradigm change is difficult to forecast. However, it is clear that the field is moving towards precision medicine, which is increasingly favored from an experimental, ethical, and economic point of view.

If you have reached this point, we cordially invite you to participate in these endeavors. To achieve the dream of developing precision medicine based on a thorough understanding of the human microbiota and a diverse set of tools for manipulating it, a collaborative effort across multiple traditional disciplines is essential. However, the challenges in this field are enormous, and our current knowledge is limited. Therefore, without a significant increase in research efforts, we will not be able to realize the whole value of pharmacomicrobiomic.

Spear, B. B., Heath-Chiozzi, M. & Huff, J. Clinical application of pharmacogenetics. Trends Mol. Med. 7, 201–204 (2001).

Article CAS PubMed Google Scholar

Lazarou, J., Pomeranz, B. H. & Corey, P. N. Incidence of adverse drug reactions in hospitalized patients: a meta-analysis of prospective studies. JAMA 279, 1200–1205 (1998).

Article CAS PubMed Google Scholar

Pirmohamed, M. et al. Adverse drug reactions as cause of admission to hospital: prospective analysis of 18 820 patients. BMJ 329, 15–19 (2004).

Article PubMed PubMed Central Google Scholar

Feitosa Ramos, S. et al. Adverse drug reactions to anti-infectives in hospitalized children: a multicenter study in Brazil. J. Pediatr. Infect. Dis. Soc. 12, 76–82 (2023).

Article Google Scholar

Jiang, H. et al. Adverse drug reactions and correlations with drug-drug interactions: a retrospective study of reports from 2011 to 2020. Front. Pharm. 13, 923939 (2022).

Article CAS Google Scholar

Zhang, Y. et al. Emergency hospitalizations for adverse drug events in China: clinical pharmacists’ approach to assessment and categorization. Pharmacoepidemiol. Drug Saf. 30, 636–643 (2021).

Article CAS PubMed Google Scholar

Konig, I. R., Fuchs, O., Hansen, G., von Mutius, E. & Kopp, M. V. What is precision medicine? Eur. Respir. J. 50, 1700391 (2017).

Article PubMed Google Scholar

Ramaswami, R., Bayer, R. & Galea, S. Precision medicine from a public health perspective. Annu. Rev. Public Health 39, 153–168 (2018).

Article PubMed Google Scholar

Prasad, R. B. & Groop, L. Precision medicine in type 2 diabetes. J. Intern. Med. 285, 40–48 (2019).

Article CAS PubMed Google Scholar

Buitelaar, J. et al. Toward precision medicine in ADHD. Front. Behav. Neurosci. 16, 900981 (2022).

Article CAS PubMed PubMed Central Google Scholar

Ahmed, S., Zhou, Z., Zhou, J. & Chen, S. Q. Pharmacogenomics of drug metabolizing enzymes and transporters: relevance to precision medicine. Genom. Proteom. Bioinforma. 14, 298–313 (2016).

Article CAS Google Scholar

Clarke, J. D. & Cherrington, N. J. Nonalcoholic steatohepatitis in precision medicine: unraveling the factors that contribute to individual variability. Pharm. Ther. 151, 99–106 (2015).

Article CAS Google Scholar

Zhou, Y., Tremmel, R., Schaeffeler, E., Schwab, M. & Lauschke, V. M. Challenges and opportunities associated with rare-variant pharmacogenomics. Trends Pharm. Sci. 43, 852–865 (2022).

Article CAS PubMed Google Scholar

Chang, W. C., Tanoshima, R., Ross, C. J. D. & Carleton, B. C. Challenges and opportunities in implementing pharmacogenetic testing in clinical settings. Annu. Rev. Pharm. Toxicol. 61, 65–84 (2021).

Article CAS Google Scholar

Johnson, J. A. & Cavallari, L. H. Pharmacogenetics and cardiovascular disease-implications for personalized medicine. Pharm. Rev. 65, 987–1009 (2013).

Article CAS PubMed PubMed Central Google Scholar

Limdi, N. A. et al. Warfarin pharmacogenetics: a single VKORC1 polymorphism is predictive of dose across 3 racial groups. Blood 115, 3827–3834 (2010).

Article CAS PubMed PubMed Central Google Scholar

Davies, S. M. et al. Pharmacogenetics of minimal residual disease response in children with B-precursor acute lymphoblastic leukemia: a report from the Children’s Oncology Group. Blood 111, 2984–2990 (2008).

Article CAS PubMed PubMed Central Google Scholar

Roses, A. D. Pharmacogenetics in drug discovery and development: a translational perspective. Nat. Rev. Drug Discov. 7, 807–817 (2008).

Article CAS PubMed Google Scholar

Swen, J. J. et al. A 12-gene pharmacogenetic panel to prevent adverse drug reactions: an open-label, multicentre, controlled, cluster-randomised crossover implementation study. Lancet 401, 347–356 (2023).

Article CAS PubMed Google Scholar

Haidar, C. E., Crews, K. R., Hoffman, J. M., Relling, M. V. & Caudle, K. E. Advancing pharmacogenomics from single-gene to preemptive testing. Annu Rev. Genom. Hum. Genet 23, 449–473 (2022).

Article Google Scholar

Lavertu, A. et al. Pharmacogenomics and big genomic data: from lab to clinic and back again. Hum. Mol. Genet 27, R72–R78 (2018).

Article CAS PubMed PubMed Central Google Scholar

Weinshilboum, R. M. & Wang, L. Pharmacogenomics: precision medicine and drug response. Mayo Clin. Proc. 92, 1711–1722 (2017).

Article CAS PubMed Google Scholar

Roden, D. M. et al. Pharmacogenomics: challenges and opportunities. Ann. Intern. Med. 145, 749–757 (2006).

Article PubMed PubMed Central Google Scholar

Eichelbaum, M., Ingelman-Sundberg, M. & Evans, W. E. Pharmacogenomics and individualized drug therapy. Annu. Rev. Med. 57, 119–137 (2006).

Article CAS PubMed Google Scholar

Evans, W. E. & Johnson, J. A. Pharmacogenomics: the inherited basis for interindividual differences in drug response. Annu Rev. Genom. Hum. Genet 2, 9–39 (2001).

Article CAS Google Scholar

Pirmohamed, M. Pharmacogenomics: current status and future perspectives. Nat. Rev. Genet 24, 350–362 (2023).

Article CAS PubMed Google Scholar

Kalow, W., Tang, B. K. & Endrenyi, L. Hypothesis: comparisons of inter- and intra-individual variations can substitute for twin studies in drug research. Pharmacogenetics 8, 283–289 (1998).

Article CAS PubMed Google Scholar

Lindell, A. E., Zimmermann-Kogadeeva, M. & Patil, K. R. Multimodal interactions of drugs, natural compounds and pollutants with the gut microbiota. Nat. Rev. Microbiol. 20, 431–443 (2022).

Article CAS PubMed Google Scholar

Nie, P. et al. Gut microbiome interventions in human health and diseases. Med. Res. Rev. 39, 2286–2313 (2019).

Article PubMed Google Scholar

Hou, K. et al. Microbiota in health and diseases. Signal Transduct. Target Ther. 7, 135 (2022).

Article PubMed PubMed Central Google Scholar

Steiner, H. E. et al. Role of the gut microbiome in cardiovascular drug response: the potential for clinical application. Pharmacotherapy 42, 165–176 (2022).

Article PubMed Google Scholar

Zhou, C. B., Zhou, Y. L. & Fang, J. Y. Gut microbiota in cancer immune response and immunotherapy. Trends Cancer 7, 647–660 (2021).

Article CAS PubMed Google Scholar

Roviello, G., Iannone, L. F., Bersanelli, M., Mini, E. & Catalano, M. The gut microbiome and efficacy of cancer immunotherapy. Pharm. Ther. 231, 107973 (2022).

Article CAS Google Scholar

Lee, K. A. et al. Cross-cohort gut microbiome associations with immune checkpoint inhibitor response in advanced melanoma. Nat. Med. 28, 535–544 (2022).

Article CAS PubMed PubMed Central Google Scholar

Scepanovic, P. et al. A comprehensive assessment of demographic, environmental, and host genetic associations with gut microbiome diversity in healthy individuals. Microbiome 7, 130 (2019).

Article PubMed PubMed Central Google Scholar

Zheng, P. et al. Gut microbial signatures can discriminate unipolar from bipolar depression. Adv. Sci. (Weinh.) 7, 1902862 (2020).

CAS PubMed Google Scholar

Vinje, S., Stroes, E., Nieuwdorp, M. & Hazen, S. L. The gut microbiome as novel cardio-metabolic target: the time has come! Eur. Heart J. 35, 883–887 (2014).

Article PubMed Google Scholar

Wei, M. Y. et al. The microbiota and microbiome in pancreatic cancer: more influential than expected. Mol. Cancer 18, 97 (2019).

Article PubMed PubMed Central Google Scholar

Robertson, R. C. et al. Maternal omega-3 fatty acids regulate offspring obesity through persistent modulation of gut microbiota. Microbiome 6, 95 (2018).

Article PubMed PubMed Central Google Scholar

Gill, S. R. et al. Metagenomic analysis of the human distal gut microbiome. Science 312, 1355–1359 (2006).

Article CAS PubMed PubMed Central Google Scholar

Qin, J. et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature 464, 59–65 (2010).

Article CAS PubMed PubMed Central Google Scholar

Zimmermann, M., Zimmermann-Kogadeeva, M., Wegmann, R. & Goodman, A. L. Mapping human microbiome drug metabolism by gut bacteria and their genes. Nature 570, 462–467 (2019).

Article CAS PubMed PubMed Central Google Scholar

Penalver Bernabe, B., Cralle, L. & Gilbert, J. A. Systems biology of the human microbiome. Curr. Opin. Biotechnol. 51, 146–153 (2018).

Article CAS PubMed Google Scholar

Enright, E. F., Gahan, C. G., Joyce, S. A. & Griffin, B. T. The impact of the gut microbiota on drug metabolism and clinical outcome. Yale J. Biol. Med. 89, 375–382 (2016).

CAS PubMed PubMed Central Google Scholar

Kinross, J. M., Darzi, A. W. & Nicholson, J. K. Gut microbiome-host interactions in health and disease. Genome Med. 3, 14 (2011).

Article PubMed PubMed Central Google Scholar

Rodriguez, J. M. et al. The composition of the gut microbiota throughout life, with an emphasis on early life. Micro. Ecol. Health Dis. 26, 26050 (2015).

Google Scholar

Piazzon, M. C. et al. Under control: how a dietary additive can restore the gut microbiome and proteomic profile, and improve disease resilience in a marine teleostean fish fed vegetable diets. Microbiome 5, 164 (2017).

Article PubMed PubMed Central Google Scholar

Shkoporov, A. N. et al. Reproducible protocols for metagenomic analysis of human faecal phageomes. Microbiome 6, 68 (2018).

Article PubMed PubMed Central Google Scholar

Montassier, E. et al. CLOUD: a non-parametric detection test for microbiome outliers. Microbiome 6, 137 (2018).

Article PubMed PubMed Central Google Scholar

Huang, L. et al. Analysis of microbiota in elderly patients with Acute Cerebral Infarction. PeerJ 7, e6928 (2019).

Article PubMed PubMed Central Google Scholar

Jiang, H. et al. Impact of host intraspecies genetic variation, diet, and age on bacterial and fungal intestinal microbiota in tigers. Microbiologyopen 9, e1050 (2020).

Article PubMed PubMed Central Google Scholar

Visconti, A. et al. Interplay between the human gut microbiome and host metabolism. Nat. Commun. 10, 4505 (2019).

Article PubMed PubMed Central Google Scholar

Chua, L. L. et al. Reduced microbial diversity in adult survivors of childhood acute lymphoblastic leukemia and microbial associations with increased immune activation. Microbiome 5, 35 (2017).

Article PubMed PubMed Central Google Scholar

Jackson, M. A. et al. Gut microbiota associations with common diseases and prescription medications in a population-based cohort. Nat. Commun. 9, 2655 (2018).

Article PubMed PubMed Central Google Scholar

Heirali, A. A. et al. The effects of inhaled aztreonam on the cystic fibrosis lung microbiome. Microbiome 5, 51 (2017).

Article PubMed PubMed Central Google Scholar

Javdan, B. et al. Personalized mapping of drug metabolism by the human gut microbiome. Cell 181, 1661–1679.e22 (2020).

Article CAS PubMed PubMed Central Google Scholar

Weersma, R. K., Zhernakova, A. & Fu, J. Interaction between drugs and the gut microbiome. Gut 69, 1510–1519 (2020).

Article CAS PubMed Google Scholar

Alexander, J. L. et al. Gut microbiota modulation of chemotherapy efficacy and toxicity. Nat. Rev. Gastroenterol. Hepatol. 14, 356–365 (2017).

Article CAS PubMed Google Scholar

Saad, R., Rizkallah, M. R. & Aziz, R. K. Gut Pharmacomicrobiomics: the tip of an iceberg of complex interactions between drugs and gut-associated microbes. Gut Pathog. 4, 16 (2012).

Article CAS PubMed PubMed Central Google Scholar

Scher, J. U., Nayak, R. R., Ubeda, C., Turnbaugh, P. J. & Abramson, S. B. Pharmacomicrobiomics in inflammatory arthritis: gut microbiome as modulator of therapeutic response. Nat. Rev. Rheumatol. 16, 282–292 (2020).

Article PubMed Google Scholar

Ting, N. L., Lau, H. C. & Yu, J. Cancer pharmacomicrobiomics: targeting microbiota to optimise cancer therapy outcomes. Gut 71, 1412–1425 (2022).

Article PubMed Google Scholar

Panebianco, C., Andriulli, A. & Pazienza, V. Pharmacomicrobiomics: exploiting the drug-microbiota interactions in anticancer therapies. Microbiome 6, 92 (2018).

Article PubMed PubMed Central Google Scholar

Doestzada, M. et al. Pharmacomicrobiomics: a novel route towards personalized medicine? Protein Cell 9, 432–445 (2018).

Article CAS PubMed PubMed Central Google Scholar

Hassan, R. et al. Drug response in association with pharmacogenomics and pharmacomicrobiomics: towards a better personalized medicine. Brief. Bioinform 22, bbaa292 (2021).

Article PubMed Google Scholar

Sharma, A., Buschmann, M. M. & Gilbert, J. A. Pharmacomicrobiomics: the holy grail to variability in drug response? Clin. Pharm. Ther. 106, 317–328 (2019).

Article Google Scholar

Tsunoda, S. M., Gonzales, C., Jarmusch, A. K., Momper, J. D. & Ma, J. D. Contribution of the gut microbiome to drug disposition, pharmacokinetic and pharmacodynamic variability. Clin. Pharmacokinet. 60, 971–984 (2021).

Article CAS PubMed PubMed Central Google Scholar

Lee, H. L. et al. Targeted approaches for in situ gut microbiome manipulation. Genes (Basel) 9, 351 (2018).

Article PubMed Google Scholar

Wong, W. F. & Santiago, M. Microbial approaches for targeting antibiotic-resistant bacteria. Micro. Biotechnol. 10, 1047–1053 (2017).

Article Google Scholar

El Rouby, N., Lima, J. J. & Johnson, J. A. Proton pump inhibitors: from CYP2C19 pharmacogenetics to precision medicine. Expert Opin. Drug Metab. Toxicol. 14, 447–460 (2018).

Article CAS PubMed PubMed Central Google Scholar

Hauser, A. S. et al. Pharmacogenomics of GPCR drug targets. Cell 172, 41–54 e19 (2018).

Article CAS PubMed PubMed Central Google Scholar

Chaudhry, S. R. et al. Pharmacogenetic prediction of individual variability in drug response based on CYP2D6, CYP2C9 and CYP2C19 genetic polymorphisms. Curr. Drug Metab. 15, 711–718 (2014).

Article CAS PubMed Google Scholar

Muszkat, M. Interethnic differences in drug response: the contribution of genetic variability in beta adrenergic receptor and cytochrome P4502C9. Clin. Pharm. Ther. 82, 215–218 (2007).

Article CAS Google Scholar

Kamath, A. et al. Role of drug transporters in elucidating inter-individual variability in pediatric chemotherapy-related toxicities and response. Pharmaceuticals 15, 990 (2022).

Article CAS PubMed PubMed Central Google Scholar

Joharatnam-Hogan, N., Shiu, K. K. & Khan, K. Challenges in the treatment of gastric cancer in the older patient. Cancer Treat. Rev. 85, 101980 (2020).

Article PubMed Google Scholar

Murgo, A. J. & Espinoza-Delgado, I. Development of novel anticancer agents in older patients: pharmacokinetic, pharmacodynamic, and other considerations. Cancer J. 11, 481–487 (2005).

Article CAS PubMed Google Scholar

Schwartz, J. B. The current state of knowledge on age, sex, and their interactions on clinical pharmacology. Clin. Pharm. Ther. 82, 87–96 (2007).

Article CAS Google Scholar

Deneer, V. H. & van Hemel, N. M. Is antiarrhythmic treatment in the elderly different? a review of the specific changes. Drugs Aging 28, 617–633 (2011).

Article CAS PubMed Google Scholar

Besir, F. H. et al. Enoxaparin-associated giant retroperitoneal hematoma in pulmonary embolism treatment. N. Am. J. Med. Sci. 3, 524–526 (2011).

Article PubMed PubMed Central Google Scholar

Spirk, D. et al. Predictors of in-hospital mortality in elderly patients with acute venous thrombo-embolism: the SWIss Venous ThromboEmbolism Registry (SWIVTER). Eur. Heart J. 33, 921–926 (2012).

Article PubMed Google Scholar

Saltalamacchia, G., Frascaroli, M., Bernardo, A. & Quaquarini, E. Renal and cardiovascular toxicities by new systemic treatments for prostate cancer. Cancers (Basel) 12, 1750 (2020).

Article CAS PubMed Google Scholar

Masnoon, N., Shakib, S., Kalisch-Ellett, L. & Caughey, G. E. What is polypharmacy? A systematic review of definitions. BMC Geriatr. 17, 230 (2017).

Article PubMed PubMed Central Google Scholar

Scherholz, M. L., Rao, R. T. & Androulakis, I. P. Modeling inter-sex and inter-individual variability in response to chronopharmacological administration of synthetic glucocorticoids. Chronobiol. Int. 37, 281–296 (2020).

Article CAS PubMed Google Scholar

Conforti, F. et al. Sex-based differences in response to anti-PD-1 or PD-L1 treatment in patients with non-small-cell lung cancer expressing high PD-L1 levels. A systematic review and meta-analysis of randomized clinical trials. ESMO Open 6, 100251 (2021).

Article CAS PubMed PubMed Central Google Scholar

Federman, D. D. The biology of human sex differences. N. Engl. J. Med. 354, 1507–1514 (2006).

Article CAS PubMed Google Scholar

Soldin, O. P. & Mattison, D. R. Sex differences in pharmacokinetics and pharmacodynamics. Clin. Pharmacokinet. 48, 143–157 (2009).

Article CAS PubMed PubMed Central Google Scholar

Scandlyn, M. J., Stuart, E. C. & Rosengren, R. J. Sex-specific differences in CYP450 isoforms in humans. Expert Opin. Drug Metab. Toxicol. 4, 413–424 (2008).

Article CAS PubMed Google Scholar

Rodenburg, E. M., Stricker, B. H. & Visser, L. E. Sex-related differences in hospital admissions attributed to adverse drug reactions in the Netherlands. Br. J. Clin. Pharm. 71, 95–104 (2011).

Article Google Scholar

Calcagno, A. et al. Tenofovir plasma concentrations according to companion drugs: a cross-sectional study of HIV-positive patients with normal renal function. Antimicrob. Agents Chemother. 57, 1840–1843 (2013).

Article CAS PubMed PubMed Central Google Scholar

Cerrato, E. et al. Cardiovascular disease in HIV patients: from bench to bedside and backwards. Open Heart 2, e000174 (2015).

Article PubMed PubMed Central Google Scholar

Anderson, G. D. Sex and racial differences in pharmacological response: where is the evidence? Pharmacogenetics, pharmacokinetics, and pharmacodynamics. J. Women’s Health (Larchmt.) 14, 19–29 (2005).

Article Google Scholar

Anderson, G. D. Gender differences in pharmacological response. Int. Rev. Neurobiol. 83, 1–10 (2008).

Article PubMed Google Scholar

Siguret, V., Pautas, E. & Gouin-Thibault, I. Warfarin therapy: influence of pharmacogenetic and environmental factors on the anticoagulant response to warfarin. Vitam. Horm. 78, 247–264 (2008).

Article CAS PubMed Google Scholar

Hirsh, J. et al. Oral anticoagulants: mechanism of action, clinical effectiveness, and optimal therapeutic range. Chest 119, 8S–21S (2001).

Article CAS PubMed Google Scholar

Cavallari, L. H. & Limdi, N. A. Warfarin pharmacogenomics. Curr. Opin. Mol. Ther. 11, 243–251 (2009).

CAS PubMed Google Scholar

Park, J. N., Lee, J. S., Noh, M. Y. & Sung, M. K. Association between usual vitamin K intake and anticoagulation in patients under warfarin therapy. Clin. Nutr. Res. 4, 235–241 (2015).

Article PubMed PubMed Central Google Scholar

Guaraldi, G. et al. Efficacy and safety of atazanavir in patients with end-stage liver disease. Infection 37, 250–255 (2009).

Article CAS PubMed Google Scholar

Greenblatt, D. J., Shader, R. I. & Lofgren, S. Rational psycho-pharmacology for patients with medical diseases. Annu Rev. Med. 27, 407–420 (1976).

Article CAS PubMed Google Scholar

Lv, C., Lv, J., Liu, Y., Liu, Q. & Zou, D. Pediatric pharmaceutical care with anti-infective medication in a patient with acute hematogenous osteomyelitis caused by methicillin-resistant Staphylococcus aureus. Int J. Immunopathol. Pharm. 34, 2058738420925713 (2020).

Article CAS Google Scholar

Waldinger, R., Weinberg, G. & Gitman, M. Local anesthetic toxicity in the geriatric population. Drugs Aging 37, 1–9 (2020).

Article PubMed Google Scholar

Rosmarin, D. M., Lebwohl, M., Elewski, B. E. & Gottlieb, A. B., National Psoriasis, F. Cyclosporine and psoriasis: 2008 National Psoriasis Foundation Consensus Conference. J. Am. Acad. Dermatol. 62, 838–853 (2010).

Article PubMed Google Scholar

Lewis, J. H. & Stine, J. G. Review article: prescribing medications in patients with cirrhosis - a practical guide. Aliment Pharm. Ther. 37, 1132–1156 (2013).

Article CAS Google Scholar

Verbeeck, R. K. Pharmacokinetics and dosage adjustment in patients with hepatic dysfunction. Eur. J. Clin. Pharm. 64, 1147–1161 (2008).

Article CAS Google Scholar

Macpherson, A. J., Heikenwalder, M. & Ganal-Vonarburg, S. C. The liver at the nexus of host-microbial interactions. Cell Host Microbe 20, 561–571 (2016).

Article CAS PubMed Google Scholar

Mani, R. & Pollard, J. R. Antiepileptic drugs and other medications: what interactions may arise? Curr. Treat. Options Neurol. 11, 253–261 (2009).

Article PubMed Google Scholar

Huang, L., Lizak, P., Dvorak, C. C., Aweeka, F. & Long-Boyle, J. Simultaneous determination of fludarabine and clofarabine in human plasma by LC-MS/MS. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 960, 194–199 (2014).

Article CAS Google Scholar

Relling, M. V. et al. Adverse effect of anticonvulsants on efficacy of chemotherapy for acute lymphoblastic leukaemia. Lancet 356, 285–290 (2000).

Article CAS PubMed Google Scholar

Rendic, S. Summary of information on human CYP enzymes: human P450 metabolism data. Drug Metab. Rev. 34, 83–448 (2002).

Article CAS PubMed Google Scholar

O’Malley, M., Healy, P., Daignault, S. & Ramnath, N. Cigarette smoking and gemcitabine-induced neutropenia in advanced solid tumors. Oncology 85, 216–222 (2013).

Article PubMed Google Scholar

Hamilton, M. et al. Effects of smoking on the pharmacokinetics of erlotinib. Clin. Cancer Res. 12, 2166–2171 (2006).

Article CAS PubMed Google Scholar

Hughes, A. N. et al. Overcoming CYP1A1/1A2 mediated induction of metabolism by escalating erlotinib dose in current smokers. J. Clin. Oncol. 27, 1220–1226 (2009).

Article CAS PubMed PubMed Central Google Scholar

van der Bol, J. M. et al. Cigarette smoking and irinotecan treatment: pharmacokinetic interaction and effects on neutropenia. J. Clin. Oncol. 25, 2719–2726 (2007).

Article PubMed Google Scholar

Kanai, M. et al. A history of smoking is inversely correlated with the incidence of gemcitabine-induced neutropenia. Ann. Oncol. 20, 1397–1401 (2009).

Article CAS PubMed Google Scholar

Dikeocha, I. J., Al-Kabsi, A. M., Miftahussurur, M. & Alshawsh, M. A. Pharmacomicrobiomics: Influence of gut microbiota on drug and xenobiotic metabolism. FASEB J. 36, e22350 (2022).

Article CAS PubMed Google Scholar

Enright, E. F., Griffin, B. T., Gahan, C. G. M. & Joyce, S. A. Microbiome-mediated bile acid modification: Role in intestinal drug absorption and metabolism. Pharm. Res. 133, 170–186 (2018).

Article CAS Google Scholar

Vich Vila, A. et al. Impact of commonly used drugs on the composition and metabolic function of the gut microbiota. Nat. Commun. 11, 362 (2020).

Article CAS PubMed PubMed Central Google Scholar

Garrod, A. E. The incidence of alkaptonuria: a study in chemical individuality. 1902 [classical article]. Yale J. Biol. Med. 75, 221–231 (2002).

PubMed PubMed Central Google Scholar

Perlman, R. L. & Govindaraju, D. R. Archibald E. Garrod: the father of precision medicine. Genet Med. 18, 1088–1089 (2016).

Article CAS PubMed Google Scholar

Evans, W. E. & Relling, M. V. Moving towards individualized medicine with pharmacogenomics. Nature 429, 464–468 (2004).

Article CAS PubMed Google Scholar

Evans, W. E. & McLeod, H. L. Pharmacogenomics-drug disposition, drug targets, and side effects. N. Engl. J. Med. 348, 538–549 (2003).

Article CAS PubMed Google Scholar

Weinshilboum, R. Inheritance and drug response. N. Engl. J. Med. 348, 529–537 (2003).

Article PubMed Google Scholar

Dawed, A. Y. et al. Pharmacogenomics of GLP-1 receptor agonists: a genome-wide analysis of observational data and large randomised controlled trials. Lancet Diabetes Endocrinol. 11, 33–41 (2023).

Article CAS PubMed Google Scholar

Beutler, E. Drug-induced hemolytic anemia. Pharm. Rev. 21, 73–103 (1969).

CAS PubMed Google Scholar

Beutler, E. The hemolytic effect of primaquine and related compounds: a review. Blood 14, 103–139 (1959).

Article CAS PubMed Google Scholar

Beutler, E., Dern, R. J. & Alving, A. S. The hemolytic effect of primaquine. III. A study of primaquine-sensitive erythrocytes. J. Lab Clin. Med. 44, 177–184 (1954).

CAS PubMed Google Scholar

Vogel, F. Moderne probleme der humangenetik. Ergebn. Inn. Med. Kinderheilk 12, 52–125 (1959).

Google Scholar

Xu, H. et al. Multiplexed SNP genotyping using the Qbead system: a quantum dot-encoded microsphere-based assay. Nucleic Acids Res. 31, e43 (2003).

Article PubMed PubMed Central Google Scholar

Gibbs, R. A. The Human Genome Project changed everything. Nat. Rev. Genet 21, 575–576 (2020).

Article CAS PubMed PubMed Central Google Scholar

Nebert, D. W., Zhang, G. & Vesell, E. S. From human genetics and genomics to pharmacogenetics and pharmacogenomics: past lessons, future directions. Drug Metab. Rev. 40, 187–224 (2008).

Article CAS PubMed PubMed Central Google Scholar

Ruiz, C., Zitnik, M. & Leskovec, J. Identification of disease treatment mechanisms through the multiscale interactome. Nat. Commun. 12, 1796 (2021).

Article CAS PubMed PubMed Central Google Scholar

Ming, X. & Laing, B. Bioconjugates for targeted delivery of therapeutic oligonucleotides. Adv. Drug Deliv. Rev. 87, 81–89 (2015).

Article CAS PubMed PubMed Central Google Scholar

Higgins, G. A., Williams, A. M., Ade, A. S., Alam, H. B. & Athey, B. D. Druggable transcriptional networks in the human neurogenic epigenome. Pharm. Rev. 71, 520–538 (2019).

Article CAS PubMed PubMed Central Google Scholar

Varga, J., Dobson, L., Remenyi, I. & Tusnady, G. E. TSTMP: target selection for structural genomics of human transmembrane proteins. Nucleic Acids Res. 45, D325–D330 (2017).

Article CAS PubMed Google Scholar

Somogyi, A. A. & Phillips, E. Genomic testing as a tool to optimise drug therapy. Aust. Prescr. 40, 101–104 (2017).

Article PubMed PubMed Central Google Scholar

Tam, H. K. et al. Allosteric drug transport mechanism of multidrug transporter AcrB. Nat. Commun. 12, 3889 (2021).

Article CAS PubMed PubMed Central Google Scholar

van de Ven, R. et al. A role for multidrug resistance protein 4 (MRP4; ABCC4) in human dendritic cell migration. Blood 112, 2353–2359 (2008).

Article PubMed PubMed Central Google Scholar

Oslin, D. W. et al. Effect of pharmacogenomic testing for drug-gene interactions on medication selection and remission of symptoms in major depressive disorder: the PRIME care randomized clinical trial. JAMA 328, 151–161 (2022).

Article CAS PubMed PubMed Central Google Scholar

Tardif, J. C. et al. Pharmacogenetics-guided dalcetrapib therapy after an acute coronary syndrome: the dal-GenE trial. Eur. Heart J. 43, 3947–3956 (2022).

Article CAS PubMed PubMed Central Google Scholar

Tsutsumi, K. et al. Structures of the wild-type MexAB-OprM tripartite pump reveal its complex formation and drug efflux mechanism. Nat. Commun. 10, 1520 (2019).

Article PubMed PubMed Central Google Scholar

Nigam, S. K. What do drug transporters really do? Nat. Rev. Drug Discov. 14, 29–44 (2015).

Article CAS PubMed Google Scholar

Surendiran, A., Pradhan, S. C. & Adithan, C. Role of pharmacogenomics in drug discovery and development. Indian J. Pharm. 40, 137–143 (2008).

Article CAS Google Scholar

Clayton, T. A., Baker, D., Lindon, J. C., Everett, J. R. & Nicholson, J. K. Pharmacometabonomic identification of a significant host-microbiome metabolic interaction affecting human drug metabolism. Proc. Natl Acad. Sci. USA 106, 14728–14733 (2009).

Article CAS PubMed PubMed Central Google Scholar

Zhou, H. H., Koshakji, R. P., Silberstein, D. J., Wilkinson, G. R. & Wood, A. J. Racial differences in drug response. Altered sensitivity to and clearance of propranolol in men of Chinese descent as compared with American whites. N. Engl. J. Med. 320, 565–570 (1989).

Article CAS PubMed Google Scholar

Yasuda, S. U., Zhang, L. & Huang, S. M. The role of ethnicity in variability in response to drugs: focus on clinical pharmacology studies. Clin. Pharm. Ther. 84, 417–423 (2008).

Article CAS Google Scholar

Man, M. et al. Genetic variation in metabolizing enzyme and transporter genes: comprehensive assessment in 3 major East Asian subpopulations with comparison to Caucasians and Africans. J. Clin. Pharm. 50, 929–940 (2010).

Article CAS Google Scholar

Scharfe, C. P. I., Tremmel, R., Schwab, M., Kohlbacher, O. & Marks, D. S. Genetic variation in human drug-related genes. Genome Med. 9, 117 (2017).

Article PubMed PubMed Central Google Scholar

Bernard, E. et al. Implications of TP53 allelic state for genome stability, clinical presentation and outcomes in myelodysplastic syndromes. Nat. Med. 26, 1549–1556 (2020).

Article CAS PubMed PubMed Central Google Scholar

Taylor, C. A. et al. IL7 genetic variation and toxicity to immune checkpoint blockade in patients with melanoma. Nat. Med. 28, 2592–2600 (2022).

Article CAS PubMed PubMed Central Google Scholar

Funnell, T. et al. Single-cell genomic variation induced by mutational processes in cancer. Nature 612, 106–115 (2022).

Article CAS PubMed PubMed Central Google Scholar

Martinez-Ruiz, C. et al. Genomic-transcriptomic evolution in lung cancer and metastasis. Nature 616, 543–552 (2023).

Article CAS PubMed PubMed Central Google Scholar

Naranbhai, V. et al. HLA-A*03 and response to immune checkpoint blockade in cancer: an epidemiological biomarker study. Lancet Oncol. 23, 172–184 (2022).

Article CAS PubMed Google Scholar

Wang, C. W., Preclaro, I. A. C., Lin, W. H. & Chung, W. H. An updated review of genetic associations with severe adverse drug reactions: translation and implementation of pharmacogenomic testing in clinical practice. Front. Pharm. 13, 886377 (2022).

Article CAS Google Scholar

Khan, D. A., Knowles, S. R. & Shear, N. H. Sulfonamide hypersensitivity: fact and fiction. J. Allergy Clin. Immunol. Pract. 7, 2116–2123 (2019).

Article CAS PubMed Google Scholar

Chow, T. G. & Khan, D. A. Sulfonamide hypersensitivity. Clin. Rev. Allergy Immunol. 62, 400–412 (2022).

Article CAS PubMed Google Scholar

Neuman, M. G. et al. Immunopathogenesis of hypersensitivity syndrome reactions to sulfonamides. Transl. Res. 149, 243–253 (2007).

Article CAS PubMed Google Scholar

Relman, D. A. & Falkow, S. The meaning and impact of the human genome sequence for microbiology. Trends Microbiol. 9, 206–208 (2001).

Article CAS PubMed Google Scholar

Maringanti, V. S., Bucci, V. & Gerber, G. K. MDITRE: scalable and interpretable machine learning for predicting host status from temporal microbiome dynamics. mSystems 7, e0013222 (2022).

Article PubMed Google Scholar

Pflughoeft, K. J. & Versalovic, J. Human microbiome in health and disease. Annu. Rev. Pathol. 7, 99–122 (2012).

Article CAS PubMed Google Scholar

Geurtsen, J. et al. Expression of the lipopolysaccharide-modifying enzymes PagP and PagL modulates the endotoxic activity of Bordetella pertussis. Infect. Immun. 74, 5574–5585 (2006).

Article CAS PubMed PubMed Central Google Scholar

ElRakaiby, M. et al. Pharmacomicrobiomics: the impact of human microbiome variations on systems pharmacology and personalized therapeutics. OMICS 18, 402–414 (2014).

Article CAS PubMed PubMed Central Google Scholar

Gutierrez Lopez, D. E., Lashinger, L. M., Weinstock, G. M. & Bray, M. S. Circadian rhythms and the gut microbiome synchronize the host’s metabolic response to diet. Cell Metab. 33, 873–887 (2021).

Article CAS PubMed Google Scholar

Zheng, P. et al. Gut microbiome remodeling induces depressive-like behaviors through a pathway mediated by the host’s metabolism. Mol. Psychiatry 21, 786–796 (2016).

Article CAS PubMed Google Scholar

Xue, M. Y., Sun, H. Z., Wu, X. H., Liu, J. X. & Guan, L. L. Multi-omics reveals that the rumen microbiome and its metabolome together with the host metabolome contribute to individualized dairy cow performance. Microbiome 8, 64 (2020).

Article CAS PubMed PubMed Central Google Scholar

Wilson, I. D. & Nicholson, J. K. Gut microbiome interactions with drug metabolism, efficacy, and toxicity. Transl. Res. 179, 204–222 (2017).

Article CAS PubMed Google Scholar

Koppel, N., Maini Rekdal, V. & Balskus, E. P. Chemical transformation of xenobiotics by the human gut microbiota. Science 356, eaag2770 (2017).

Article PubMed Google Scholar

Khalsa, J., Duffy, L. C., Riscuta, G., Starke-Reed, P. & Hubbard, V. S. Omics for understanding the gut-liver-microbiome axis and precision medicine. Clin. Pharm. Drug Dev. 6, 176–185 (2017).

Article CAS Google Scholar

Zhao, L. et al. Targeting the human genome-microbiome axis for drug discovery: inspirations from global systems biology and traditional Chinese medicine. J. Proteome Res. 11, 3509–3519 (2012).

Article CAS PubMed Google Scholar

Yachida, S. et al. Metagenomic and metabolomic analyses reveal distinct stage-specific phenotypes of the gut microbiota in colorectal cancer. Nat. Med. 25, 968–976 (2019).

Article CAS PubMed Google Scholar

Almeida, A. et al. A new genomic blueprint of the human gut microbiota. Nature 568, 499–504 (2019).

Article CAS PubMed PubMed Central Google Scholar

Qin, Y. et al. Combined effects of host genetics and diet on human gut microbiota and incident disease in a single population cohort. Nat. Genet 54, 134–142 (2022).

Article CAS PubMed PubMed Central Google Scholar

Li, Q. et al. Main active components of Jiawei Gegen Qinlian decoction protects against ulcerative colitis under different dietary environments in a gut microbiota-dependent manner. Pharm. Res. 170, 105694 (2021).

Article CAS Google Scholar

Kaddurah-Daouk, R. & Weinshilboum, R. Pharmacometabolomics Research Network Metabolomic signatures for drug response phenotypes: pharmacometabolomics enables precision medicine. Clin. Pharm. Ther. 98, 71–75 (2015).

Article CAS Google Scholar

Chakladar, J. et al. The liver microbiome is implicated in cancer prognosis and modulated by alcohol and hepatitis B. Cancers (Basel) 12, 1642 (2020).

Article CAS PubMed Google Scholar

Grierson, J., Flies, E. J., Bissett, A., Ammitzboll, H. & Jones, P. Which soil microbiome? Bacteria, fungi, and protozoa communities show different relationships with urban green space type and use-intensity. Sci. Total Environ. 863, 160468 (2023).

Article CAS PubMed Google Scholar

Wu, X., Xia, Y., He, F., Zhu, C. & Ren, W. Intestinal mycobiota in health and diseases: from a disrupted equilibrium to clinical opportunities. Microbiome 9, 60 (2021).

Article PubMed PubMed Central Google Scholar

Berg, G. et al. Microbiome definition re-visited: old concepts and new challenges. Microbiome 8, 103 (2020).

Article PubMed PubMed Central Google Scholar

El-Sayed, A., Aleya, L. & Kamel, M. Microbiota’s role in health and diseases. Environ. Sci. Pollut. Res. Int. 28, 36967–36983 (2021).

Article PubMed PubMed Central Google Scholar

Marchesi, J. R. & Ravel, J. The vocabulary of microbiome research: a proposal. Microbiome 3, 31 (2015).

Article PubMed PubMed Central Google Scholar

Rajendhran, J. & Gunasekaran, P. Microbial phylogeny and diversity: small subunit ribosomal RNA sequence analysis and beyond. Microbiol. Res. 166, 99–110 (2011).

Article CAS PubMed Google Scholar

Kumar, P. S. Microbiomics: were we all wrong before? Periodontol 2000 85, 8–11 (2021).

Article PubMed Google Scholar

Dominguez-Bello, M. G., Godoy-Vitorino, F., Knight, R. & Blaser, M. J. Role of the microbiome in human development. Gut 68, 1108–1114 (2019).

Article CAS PubMed Google Scholar

Turnbaugh, P. J. et al. The human microbiome project. Nature 449, 804–810 (2007).

Article CAS PubMed PubMed Central Google Scholar

Proctor, L. M. The National Institutes of Health Human Microbiome Project. Semin. Fetal Neonatal Med. 21, 368–372 (2016).

Article PubMed Google Scholar

Integrative, H.M.P.R.N.C. The Integrative Human Microbiome Project: dynamic analysis of microbiome-host omics profiles during periods of human health and disease. Cell Host Microbe 16, 276–289 (2014).

Article Google Scholar

Gilbert, J. A. et al. The Earth Microbiome Project: Meeting report of the “1 EMP meeting on sample selection and acquisition” at Argonne National Laboratory October 6 2010. Stand Genom. Sci. 3, 249–253 (2010).

Article Google Scholar

Gilbert, J. A., Jansson, J. K. & Knight, R. The Earth Microbiome project: successes and aspirations. BMC Biol. 12, 69 (2014).

Article PubMed PubMed Central Google Scholar

McCoubrey, L. E., Gaisford, S., Orlu, M. & Basit, A. W. Predicting drug-microbiome interactions with machine learning. Biotechnol. Adv. 54, 107797 (2022).

Article CAS PubMed Google Scholar

Pryor, R., Martinez-Martinez, D., Quintaneiro, L. & Cabreiro, F. The role of the microbiome in drug response. Annu. Rev. Pharm. Toxicol. 60, 417–435 (2020).

Article CAS Google Scholar

Kamath, S., Stringer, A. M., Prestidge, C. A. & Joyce, P. Targeting the gut microbiome to control drug pharmacomicrobiomics: the next frontier in oral drug delivery. Expert Opin. Drug Deliv. 7, 1–17 (2023).

Article Google Scholar

Becker, H. E. F., Demers, K., Derijks, L. J. J., Jonkers, D. & Penders, J. Current evidence and clinical relevance of drug-microbiota interactions in inflammatory bowel disease. Front. Microbiol. 14, 1107976 (2023).

Article PubMed PubMed Central Google Scholar

Rizkallah, M. R., Saad, R. & Aziz, R. K. The Human Microbiome Project, personalized medicine and the birth of pharmacomicrobiomics. Curr. Pharm. Pers. Med. 8, 182–193 (2010).

CAS Google Scholar

Clayton, T. A. et al. Pharmaco-metabonomic phenotyping and personalized drug treatment. Nature 440, 1073–1077 (2006).

Article CAS PubMed Google Scholar

Nicholson, J. K., Wilson, I. D. & Lindon, J. C. Pharmacometabonomics as an effector for personalized medicine. Pharmacogenomics 12, 103–111 (2011).

Article CAS PubMed Google Scholar

Nayak, R. R. & Turnbaugh, P. J. Mirror, mirror on the wall: which microbiomes will help heal them all? BMC Med. 14, 72 (2016).

Article PubMed PubMed Central Google Scholar

Balasopoulou, A., Patrinos, G. P. & Katsila, T. Pharmacometabolomics informs viromics toward precision medicine. Front. Pharm. 7, 411 (2016).

Article Google Scholar

Katsila, T., Balasopoulou, A., Tsagaraki, I. & Patrinos, G. P. Pharmacomicrobiomics informs clinical pharmacogenomics. Pharmacogenomics 20, 731–739 (2019).

Article CAS PubMed Google Scholar

Wilson, I. D. Drugs, bugs, and personalized medicine: pharmacometabonomics enters the ring. Proc. Natl Acad. Sci. USA 106, 14187–14188 (2009).

Article CAS PubMed PubMed Central Google Scholar

Fu, J. et al. Pharmacometabonomics: data processing and statistical analysis. Brief. Bioinform. 22, bbab138 (2021).

Article PubMed Google Scholar

Everett, J. R. From metabonomics to pharmacometabonomics: the role of metabolic profiling in personalized medicine. Front. Pharm. 7, 297 (2016).

Article Google Scholar

Sorbara, M. T. & Pamer, E. G. Microbiome-based therapeutics. Nat. Rev. Microbiol. 20, 365–380 (2022).

Article CAS PubMed Google Scholar

Wainwright, M. & Kristiansen, J. E. On the 75th anniversary of Prontosil. Dyes Pigments 88, 231–234 (2010).

Article Google Scholar

Bentley, R. Different roads to discovery; Prontosil (hence sulfa drugs) and penicillin (hence beta-lactams). J. Ind. Microbiol. Biotechnol. 36, 775–786 (2009).

Article CAS PubMed Google Scholar

Paranjape, K. et al. Unravelling the importance of the eukaryotic and bacterial communities and their relationship with Legionella spp. ecology in cooling towers: a complex network. Microbiome 8, 157 (2020).

Article CAS PubMed PubMed Central Google Scholar

Lepage, P. et al. A metagenomic insight into our gut’s microbiome. Gut 62, 146–158 (2013).

Article PubMed Google Scholar

Valles-Colomer, M. et al. Meta-omics in inflammatory bowel disease research: applications, challenges, and guidelines. J. Crohns Colitis 10, 735–746 (2016).

Article PubMed Google Scholar

Sha, G. et al. Integrated meta-omics study on rapid tylosin removal mechanism and dynamics of antibiotic resistance genes during aerobic thermophilic fermentation of tylosin mycelial dregs. Bioresour. Technol. 351, 127010 (2022).

Article CAS PubMed Google Scholar

McDaniel, E. A. et al. Prospects for multi-omics in the microbial ecology of water engineering. Water Res. 205, 117608 (2021).

Article CAS PubMed Google Scholar

Kaddurah-Daouk, R. & Weinshilboum, R. M. Pharmacometabolomics Research Network Pharmacometabolomics: implications for clinical pharmacology and systems pharmacology. Clin. Pharm. Ther. 95, 154–167 (2014).

Article CAS Google Scholar

Kaddurah-Daouk, R., Kristal, B. S. & Weinshilboum, R. M. Metabolomics: a global biochemical approach to drug response and disease. Annu. Rev. Pharm. Toxicol. 48, 653–683 (2008).

Article CAS Google Scholar

Nicholson, J. K. & Wilson, I. D. Opinion: understanding ‘global’ systems biology: metabonomics and the continuum of metabolism. Nat. Rev. Drug Discov. 2, 668–676 (2003).

Article CAS PubMed Google Scholar

Beger, R. D., Schmidt, M. A. & Kaddurah-Daouk, R. Current concepts in pharmacometabolomics, biomarker discovery, and precision medicine. Metabolites 10, 129 (2020).

Article CAS PubMed PubMed Central Google Scholar

Everett, J. R., Loo, R. L. & Pullen, F. S. Pharmacometabonomics and personalized medicine. Ann. Clin. Biochem. 50, 523–545 (2013).

Article PubMed Google Scholar

Everett, J. R. Pharmacometabonomics in humans: a new tool for personalized medicine. Pharmacogenomics 16, 737–754 (2015).

Article CAS PubMed Google Scholar

Rosato, A. et al. From correlation to causation: analysis of metabolomics data using systems biology approaches. Metabolomics 14, 37 (2018).

Article PubMed PubMed Central Google Scholar

McWhinney, S. R. & McLeod, H. L. Using germline genotype in cancer pharmacogenetic studies. Pharmacogenomics 10, 489–493 (2009).

Article CAS PubMed Google Scholar

Derosa, L. et al. Microbiota-centered interventions: the next breakthrough in immuno-oncology? Cancer Discov. 11, 2396–2412 (2021).

Article CAS PubMed Google Scholar

He, Y., Liu, Q. W., Liao, H. X. & Xu, W. W. Microbiota in cancer chemoradiotherapy resistance. Clin. Transl. Med. 11, e250 (2021).

Article CAS PubMed Google Scholar

Emadi, A., Jones, R. J. & Brodsky, R. A. Cyclophosphamide and cancer: golden anniversary. Nat. Rev. Clin. Oncol. 6, 638–647 (2009).

Article CAS PubMed Google Scholar

Gershwin, M. E., Goetzl, E. J. & Steinberg, A. D. Cyclophosphamide: use in practice. Ann. Intern. Med. 80, 531–540 (1974).

Article CAS PubMed Google Scholar

Patel, J. M. Metabolism and pulmonary toxicity of cyclophosphamide. Pharm. Ther. 47, 137–146 (1990).

Article CAS Google Scholar

Barnes, H., Holland, A. E., Westall, G. P., Goh, N. S. & Glaspole, I. N. Cyclophosphamide for connective tissue disease-associated interstitial lung disease. Cochrane Database Syst. Rev. 1, CD010908 (2018).

PubMed Google Scholar

Clemente, C. G. et al. Prognostic value of tumor infiltrating lymphocytes in the vertical growth phase of primary cutaneous melanoma. Cancer 77, 1303–1310 (1996).

3.0.CO;2-5" data-track-action="article reference" href="https://doi.org/10.1002%2F%28SICI%291097-0142%2819960401%2977%3A7%3C1303%3A%3AAID-CNCR12%3E3.0.CO%3B2-5" aria-label="Article reference 221" data-doi="10.1002/(SICI)1097-0142(19960401)77:73.0.CO;2-5">Article CAS PubMed Google Scholar

Zhang, L. et al. Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N. Engl. J. Med. 348, 203–213 (2003).

Article CAS PubMed Google Scholar

Katz, S. C. et al. T cell infiltrate predicts long-term survival following resection of colorectal cancer liver metastases. Ann. Surg. Oncol. 16, 2524–2530 (2009).

Article PubMed Google Scholar

Jamiyan, T. et al. Prognostic impact of a tumor-infiltrating lymphocyte subtype in triple negative cancer of the breast. Breast Cancer 27, 880–892 (2020).

Article PubMed Google Scholar

Roy, S. & Trinchieri, G. Microbiota: a key orchestrator of cancer therapy. Nat. Rev. Cancer 17, 271–285 (2017).

Article CAS PubMed Google Scholar

Viaud, S. et al. Cyclophosphamide induces differentiation of Th17 cells in cancer patients. Cancer Res. 71, 661–665 (2011).

Article CAS PubMed Google Scholar

Nakahara, T. et al. Cyclophosphamide enhances immunity by modulating the balance of dendritic cell subsets in lymphoid organs. Blood 115, 4384–4392 (2010).

Article CAS PubMed PubMed Central Google Scholar

Ghiringhelli, F. et al. CD4+CD25+ regulatory T cells suppress tumor immunity but are sensitive to cyclophosphamide which allows immunotherapy of established tumors to be curative. Eur. J. Immunol. 34, 336–344 (2004).

Article CAS PubMed Google Scholar

Bracci, L. et al. Cyclophosphamide enhances the antitumor efficacy of adoptively transferred immune cells through the induction of cytokine expression, B-cell and T-cell homeostatic proliferation, and specific tumor infiltration. Clin. Cancer Res. 13, 644–653 (2007).

Article CAS PubMed Google Scholar

Sistigu, A. et al. Immunomodulatory effects of cyclophosphamide and implementations for vaccine design. Semin. Immunopathol. 33, 369–383 (2011).

Article CAS PubMed Google Scholar

Hughes, E. et al. T-cell modulation by cyclophosphamide for tumour therapy. Immunology 154, 62–68 (2018).

Article CAS PubMed PubMed Central Google Scholar

Madondo, M. T., Quinn, M. & Plebanski, M. Low dose cyclophosphamide: mechanisms of T cell modulation. Cancer Treat. Rev. 42, 3–9 (2016).

Article PubMed Google Scholar

Zhao, C. et al. Post-transplant cyclophosphamide alters immune signatures and leads to impaired T cell reconstitution in allogeneic hematopoietic stem cell transplant. J. Hematol. Oncol. 15, 64 (2022).

Article CAS PubMed PubMed Central Google Scholar

Toulmonde, M., Demolis, P. & Houede, N. Salvage chemotherapy for hormone-refractory prostate cancer: association of Adriamycin and ifosfamide. Exp. Ther. Med. 1, 1005–1011 (2010).

Article CAS PubMed PubMed Central Google Scholar

Viaud, S. et al. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science 342, 971–976 (2013).

Article CAS PubMed PubMed Central Google Scholar

Yang, J., Liu, K. X., Qu, J. M. & Wang, X. D. The changes induced by cyclophosphamide in intestinal barrier and microflora in mice. Eur. J. Pharm. 714, 120–124 (2013).

Article CAS Google Scholar

Xu, X. & Zhang, X. Effects of cyclophosphamide on immune system and gut microbiota in mice. Microbiol. Res. 171, 97–106 (2015).

Article CAS PubMed Google Scholar

Daillere, R. et al. Enterococcus hirae and Barnesiella intestinihominis facilitate cyclophosphamide-induced therapeutic immunomodulatory effects. Immunity 45, 931–943 (2016).

Article CAS PubMed Google Scholar

Mego, M. et al. Prevention of irinotecan induced diarrhea by probiotics: a randomized double blind, placebo controlled pilot study. Complement Ther. Med. 23, 356–362 (2015).

Article PubMed Google Scholar

Bleiberg, H. CPT-11 in gastrointestinal cancer. Eur. J. Cancer 35, 371–379 (1999).

Article CAS PubMed Google Scholar

Dobi, A., Gasque, P., Guiraud, P. & Selambarom, J. Irinotecan (CPT-11) canonical anti-cancer drug can also modulate antiviral and pro-inflammatory responses of primary human synovial fibroblasts. Cells 10, 1431 (2021).

Article CAS PubMed PubMed Central Google Scholar

Bailly, C. Irinotecan: 25 years of cancer treatment. Pharm. Res. 148, 104398 (2019).

Article CAS Google Scholar

Zhao, J. et al. In situ activation of STING pathway with polymeric SN38 for cancer chemoimmunotherapy. Biomaterials 268, 120542 (2021).

Article CAS PubMed Google Scholar

Karas, S. et al. Optimal sampling strategies for irinotecan (CPT-11) and its active metabolite (SN-38) in cancer patients. AAPS J. 22, 59 (2020).

Article CAS PubMed Google Scholar

Stringer, A. M. et al. Faecal microflora and beta-glucuronidase expression are altered in an irinotecan-induced diarrhea model in rats. Cancer Biol. Ther. 7, 1919–1925 (2008).

Article CAS PubMed Google Scholar

Flieger, D. et al. Phase II clinical trial for prevention of delayed diarrhea with cholestyramine/levofloxacin in the second-line treatment with irinotecan biweekly in patients with metastatic colorectal carcinoma. Oncology 72, 10–16 (2007).

Article CAS PubMed Google Scholar

Kehrer, D. F. et al. Modulation of irinotecan-induced diarrhea by cotreatment with neomycin in cancer patients. Clin. Cancer Res. 7, 1136–1141 (2001).

CAS PubMed Google Scholar

Leibovici, L. et al. Antibiotic prophylaxis in neutropenic patients: new evidence, practical decisions. Cancer 107, 1743–1751 (2006).

Article CAS PubMed Google Scholar

Wallace, B. D. et al. Alleviating cancer drug toxicity by inhibiting a bacterial enzyme. Science 330, 831–835 (2010).

Article CAS PubMed PubMed Central Google Scholar

Kong, R. et al. Old drug new use-amoxapine and its metabolites as potent bacterial beta-glucuronidase inhibitors for alleviating cancer drug toxicity. Clin. Cancer Res. 20, 3521–3530 (2014).

Article CAS PubMed PubMed Central Google Scholar

Huang, P. T. et al. Enhancement of CPT-11 antitumor activity by adenovirus-mediated expression of beta-glucuronidase in tumors. Cancer Gene Ther. 18, 381–389 (2011).

Article CAS PubMed Google Scholar

Cheng, K. W. et al. Pharmacological inhibition of bacterial beta-glucuronidase prevents irinotecan-induced diarrhea without impairing its antitumor efficacy in vivo. Pharm. Res. 139, 41–49 (2019).

Article CAS Google Scholar

Chamseddine, A. N. et al. Intestinal bacterial beta-glucuronidase as a possible predictive biomarker of irinotecan-induced diarrhea severity. Pharm. Ther. 199, 1–15 (2019).

Article CAS Google Scholar

Gallotti, B. et al. Effects of dietary fibre intake in chemotherapy-induced mucositis in murine model. Br. J. Nutr. 126, 853–864 (2021).

Article CAS PubMed Google Scholar

Takasuna, K. et al. Involvement of beta-glucuronidase in intestinal microflora in the intestinal toxicity of the antitumor camptothecin derivative irinotecan hydrochloride (CPT-11) in rats. Cancer Res. 56, 3752–3757 (1996).

CAS PubMed Google Scholar

Songjang, W. et al. Tumor-promoting activity and proteomic profiling of cisplatin/oxaliplatin-derived DAMPs in cholangiocarcinoma cells. Int J. Mol. Sci. 23, 10540 (2022).

Article CAS PubMed PubMed Central Google Scholar

Skripova, V., Vlasenkova, R., Zhou, Y., Astsaturov, I. & Kiyamova, R. Identification of new regulators of pancreatic cancer cell sensitivity to oxaliplatin and cisplatin. Molecules 27, 1289 (2022).

Article CAS PubMed PubMed Central Google Scholar

Zhang, C., Xu, C., Gao, X. & Yao, Q. Platinum-based drugs for cancer therapy and anti-tumor strategies. Theranostics 12, 2115–2132 (2022).

Article PubMed PubMed Central Google Scholar

Yamada, Y. et al. Phase III study comparing oxaliplatin plus S-1 with cisplatin plus S-1 in chemotherapy-naive patients with advanced gastric cancer. Ann. Oncol. 26, 141–148 (2015).

Article CAS PubMed Google Scholar

Vaisman, A., Masutani, C., Hanaoka, F. & Chaney, S. G. Efficient translesion replication past oxaliplatin and cisplatin GpG adducts by human DNA polymerase eta. Biochemistry 39, 4575–4580 (2000).

Article CAS PubMed Google Scholar

Cruet-Hennequart, S., Glynn, M. T., Murillo, L. S., Coyne, S. & Carty, M. P. Enhanced DNA-PK-mediated RPA2 hyperphosphorylation in DNA polymerase eta-deficient human cells treated with cisplatin and oxaliplatin. DNA Repair (Amst.) 7, 582–596 (2008).

Article CAS PubMed Google Scholar

Lokody, I. Tumour microenvironment: bacterial balance affects cancer treatment. Nat. Rev. Cancer 14, 10 (2014).

Article CAS PubMed Google Scholar

Iida, N. et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science 342, 967–970 (2013).

Article CAS PubMed PubMed Central Google Scholar

Roberti, M. P. et al. Chemotherapy-induced ileal crypt apoptosis and the ileal microbiome shape immunosurveillance and prognosis of proximal colon cancer. Nat. Med. 26, 919–931 (2020).

Article CAS PubMed Google Scholar

Bachem, A. et al. Microbiota-derived short-chain fatty acids promote the memory potential of antigen-activated CD8(+) T cells. Immunity 51, 285–297 e5 (2019).

Article CAS PubMed Google Scholar

He, Y. et al. Gut microbial metabolites facilitate anticancer therapy efficacy by modulating cytotoxic CD8(+) T cell immunity. Cell Metab. 33, 988–1000 e7 (2021).

Article CAS PubMed Google Scholar

Qin, S. et al. Novel immune checkpoint targets: moving beyond PD-1 and CTLA-4. Mol. Cancer 18, 155 (2019).

Article PubMed PubMed Central Google Scholar

Andrews, L. P., Yano, H. & Vignali, D. A. A. Inhibitory receptors and ligands beyond PD-1, PD-L1 and CTLA-4: breakthroughs or backups. Nat. Immunol. 20, 1425–1434 (2019).

Article CAS PubMed Google Scholar

Burugu, S., Dancsok, A. R. & Nielsen, T. O. Emerging targets in cancer immunotherapy. Semin Cancer Biol. 52, 39–52 (2018).

Article CAS PubMed Google Scholar

Vetizou, M. et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 350, 1079–1084 (2015).

Article CAS PubMed PubMed Central Google Scholar

Sivan, A. et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 350, 1084–1089 (2015).

Article CAS PubMed PubMed Central Google Scholar

Chaput, N. et al. Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab. Ann. Oncol. 28, 1368–1379 (2017).

Article CAS PubMed Google Scholar

Gopalakrishnan, V. et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 359, 97–103 (2018).

Article CAS PubMed Google Scholar

Routy, B. et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 359, 91–97 (2018).

Article CAS PubMed Google Scholar

Matson, V. et al. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science 359, 104–108 (2018).

Article CAS PubMed PubMed Central Google Scholar

Tanoue, T. et al. A defined commensal consortium elicits CD8 T cells and anti-cancer immunity. Nature 565, 600–605 (2019).

Article CAS PubMed Google Scholar

Helmink, B. A. et al. B cells and tertiary lymphoid structures promote immunotherapy response. Nature 577, 549–555 (2020).

Article CAS PubMed PubMed Central Google Scholar

Poore, G. D. et al. Microbiome analyses of blood and tissues suggest cancer diagnostic approach. Nature 579, 567–574 (2020).

Article CAS PubMed PubMed Central Google Scholar

Costea, P. I. et al. Towards standards for human fecal sample processing in metagenomic studies. Nat. Biotechnol. 35, 1069–1076 (2017).

Article CAS PubMed Google Scholar

Tam, V. et al. Benefits and limitations of genome-wide association studies. Nat. Rev. Genet 20, 467–484 (2019).

Article CAS PubMed Google Scholar

Baruch, E. N. et al. Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients. Science 371, 602–609 (2021).

Article CAS PubMed Google Scholar

Davar, D. et al. Fecal microbiota transplant overcomes resistance to anti-PD-1 therapy in melanoma patients. Science 371, 595–602 (2021).

Article CAS PubMed PubMed Central Google Scholar

Jamison, D. T. et al. Disease Control Priorities in Developing Countries. 2nd edition. Washington (DC): The International Bank for Reconstruction and Development/ The World Bank (2006).

Ruan, Y. et al. Cardiovascular disease (CVD) and associated risk factors among older adults in six low-and middle-income countries: results from SAGE Wave 1. BMC Public Health 18, 778 (2018).

Article PubMed PubMed Central Google Scholar

Nitsa, A. et al. Vitamin D in cardiovascular disease. Vivo 32, 977–981 (2018).

Article CAS Google Scholar

Roth, G. A. et al. Global, regional, and national burden of cardiovascular diseases for 10 causes, 1990 to 2015. J. Am. Coll. Cardiol. 70, 1–25 (2017).

Article PubMed PubMed Central Google Scholar

Zuo, H. L. et al. Interactions of antithrombotic herbal medicines with Western cardiovascular drugs. Pharm. Res. 159, 104963 (2020).

Article CAS Google Scholar

Curini, L. & Amedei, A. Cardiovascular diseases and pharmacomicrobiomics: a perspective on possible treatment relevance. Biomedicines 9, 1338 (2021).

Article CAS PubMed PubMed Central Google Scholar

Kyoung, J., Atluri, R. R. & Yang, T. Resistance to antihypertensive drugs: is gut microbiota the missing link? Hypertension 79, 2138–2147 (2022).

Article CAS PubMed Google Scholar

Chakaroun, R. M., Olsson, L. M. & Backhed, F. The potential of tailoring the gut microbiome to prevent and treat cardiometabolic disease. Nat. Rev. Cardiol. 20, 217–235 (2023).

Article PubMed Google Scholar

Yang, Z. et al. Gut microbiota and hypertension: association, mechanisms and treatment. Clin. Exp. Hypertens. 45, 2195135 (2023).

Article PubMed Google Scholar

Chen, H. Q. et al. Pharmacomicrobiomics: exploiting the drug-microbiota interactions in antihypertensive treatment. Front. Med. (Lausanne) 8, 742394 (2021).

Article PubMed Google Scholar

Pirmohamed, M. Warfarin: almost 60 years old and still causing problems. Br. J. Clin. Pharm. 62, 509–511 (2006).

Article Google Scholar

Kimmel, S. E. Warfarin pharmacogenomics: current best evidence. J. Thromb. Haemost. 13, S266–S271 (2015).

Article CAS PubMed Google Scholar

Wu, S. et al. Warfarin and vitamin K epoxide reductase: a molecular accounting for observed inhibition. Blood 132, 647–657 (2018).

Article CAS PubMed PubMed Central Google Scholar

Czogalla, K. J. et al. Warfarin and vitamin K compete for binding to Phe55 in human VKOR. Nat. Struct. Mol. Biol. 24, 77–85 (2017).

Article CAS PubMed Google Scholar

Chen, X. et al. A novel vitamin K derived anticoagulant tolerant to genetic variations of vitamin K epoxide reductase. J. Thromb. Haemost. 19, 689–700 (2021).

Article CAS PubMed PubMed Central Google Scholar

Rishavy, M. A. et al. Warfarin alters vitamin K metabolism: a surprising mechanism of VKORC1 uncoupling necessitates an additional reductase. Blood 131, 2826–2835 (2018).

Article CAS PubMed PubMed Central Google Scholar

Wise, E. S. et al. The influence of VKORC1 and CYP2C9 mutations on warfarin response after total hip and knee arthroplasty. J. Orthop. 12, S145–S151 (2015).

Article PubMed PubMed Central Google Scholar

Jorgensen, A. L., FitzGerald, R. J., Oyee, J., Pirmohamed, M. & Williamson, P. R. Influence of CYP2C9 and VKORC1 on patient response to warfarin: a systematic review and meta-analysis. PLoS One 7, e44064 (2012).

Article CAS PubMed PubMed Central Google Scholar

Xue, L. et al. Theory-based pharmacokinetics and pharmacodynamics of S- and R-warfarin and effects on international normalized ratio: influence of body size, composition and genotype in cardiac surgery patients. Br. J. Clin. Pharm. 83, 823–835 (2017).

Article CAS Google Scholar

Wang, L. et al. The gut microbes, Enterococcus and Escherichia-Shigella, affect the responses of heart valve replacement patients to the anticoagulant warfarin. Pharm. Res. 159, 104979 (2020).

Article CAS Google Scholar

Ward, N. C., Watts, G. F. & Eckel, R. H. Statin toxicity. Circ. Res. 124, 328–350 (2019).

Article CAS PubMed Google Scholar

Lim, G. B. Statins outperform dietary supplements for LDL-C lowering. Nat. Rev. Cardiol. 20, 6 (2023).

PubMed Google Scholar

Dehnavi, S. et al. Statins and autoimmunity: state-of-the-art. Pharm. Ther. 214, 107614 (2020).

Article CAS Google Scholar

Arnett, D. K. et al. ACC/AHA guideline on the primary prevention of cardiovascular disease: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation 140, e596–e646 (2019).

PubMed PubMed Central Google Scholar

Zhou, Q. & Liao, J. K. Statins and cardiovascular diseases: from cholesterol lowering to pleiotropy. Curr. Pharm. Des. 15, 467–478 (2009).

Article CAS PubMed PubMed Central Google Scholar

Oesterle, A., Laufs, U. & Liao, J. K. Pleiotropic effects of statins on the cardiovascular system. Circ. Res. 120, 229–243 (2017).

Article CAS PubMed PubMed Central Google Scholar

Kim, J. et al. Alterations in gut microbiota by statin therapy and possible intermediate effects on hyperglycemia and hyperlipidemia. Front. Microbiol. 10, 1947 (2019).

Article PubMed PubMed Central Google Scholar

Caparros-Martin, J. A. et al. Statin therapy causes gut dysbiosis in mice through a PXR-dependent mechanism. Microbiome 5, 95 (2017).

Article PubMed PubMed Central Google Scholar

Tuteja, S. & Ferguson, J. F. Gut microbiome and response to cardiovascular drugs. Circ. Genom. Precis Med. 12, 421–429 (2019).

Article CAS PubMed PubMed Central Google Scholar

Campbell, T. J. & MacDonald, P. S. Digoxin in heart failure and cardiac arrhythmias. Med. J. Aust. 179, 98–102 (2003).

Article PubMed Google Scholar

Patocka, J., Nepovimova, E., Wu, W. & Kuca, K. Digoxin: pharmacology and toxicology-A review. Environ. Toxicol. Pharm. 79, 103400 (2020).

Article CAS Google Scholar

Haiser, H. J. et al. Predicting and manipulating cardiac drug inactivation by the human gut bacterium Eggerthella lenta. Science 341, 295–298 (2013).

Article CAS PubMed PubMed Central Google Scholar

Saha, J. R., Butler, V. P. Jr, Neu, H. C. & Lindenbaum, J. Digoxin-inactivating bacteria: identification in human gut flora. Science 220, 325–327 (1983).

Article CAS PubMed Google Scholar

Bo, J. et al. Eggerthella lenta bloodstream infections: two cases and review of the literature. Future Microbiol. 15, 981–985 (2020).

Article CAS PubMed Google Scholar

Kageyama, A., Benno, Y. & Nakase, T. Phylogenetic evidence for the transfer of Eubacterium lentum to the genus Eggerthella as Eggerthella lenta gen. nov., comb. nov. Int J. Syst. Bacteriol. 49, 1725–1732 (1999).

Article CAS PubMed Google Scholar

Haiser, H. J., Seim, K. L., Balskus, E. P. & Turnbaugh, P. J. Mechanistic insight into digoxin inactivation by Eggerthella lenta augments our understanding of its pharmacokinetics. Gut Microbes 5, 233–238 (2014).

Article PubMed PubMed Central Google Scholar

David, L. A. et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 505, 559–563 (2014).

Article CAS PubMed Google Scholar

Evered, M. D., Robinson, M. M. & Richardson, M. A. Captopril given intracerebroventricularly, subcutaneously or by gavage inhibits angiotensin-converting enzyme activity in the rat brain. Eur. J. Pharm. 68, 443–449 (1980).

Article CAS Google Scholar

Antunes, A. M., Guerrante, R. D., Avila, J. P., Lins Mendes, F. M. & Fierro, I. M. Case study of patents related to captopril, Squibb’s first blockbuster. Expert Opin. Ther. Pat. 26, 1449–1457 (2016).

Article CAS PubMed Google Scholar

Berecek, K. H., Reaves, P. & Raizada, M. Effects of early perturbation of the renin-angiotensin system on cardiovascular remodeling in spontaneously hypertensive rats. Vasc. Pharm. 42, 93–98 (2005).

Article CAS Google Scholar

Wu, J. N. & Berecek, K. H. Prevention of genetic hypertension by early treatment of spontaneously hypertensive rats with the angiotensin converting enzyme inhibitor captopril. Hypertension 22, 139–146 (1993).

Article CAS PubMed Google Scholar

Zhang, L., Edwards, D. G. & Berecek, K. H. Effects of early captopril treatment and its removal on plasma angiotensin converting enzyme (ACE) activity and arginine vasopressin in hypertensive rats (SHR) and normotensive rats (WKY). Clin. Exp. Hypertens. 18, 201–226 (1996).

Article PubMed Google Scholar

Wu, H., Lam, T. Y. C., Shum, T. F., Tsai, T. Y. & Chiou, J. Hypotensive effect of captopril on deoxycorticosterone acetate-salt-induced hypertensive rat is associated with gut microbiota alteration. Hypertens. Res. 45, 270–282 (2022).

Article CAS PubMed Google Scholar

Yang, T. et al. Sustained captopril-induced reduction in blood pressure is associated with alterations in gut-brain axis in the spontaneously hypertensive rat. J. Am. Heart Assoc. 8, e010721 (2019).

Article PubMed PubMed Central Google Scholar

Yang, T., Richards, E. M., Pepine, C. J. & Raizada, M. K. The gut microbiota and the brain-gut-kidney axis in hypertension and chronic kidney disease. Nat. Rev. Nephrol. 14, 442–456 (2018).

Article CAS PubMed PubMed Central Google Scholar

Santisteban, M. M., Kim, S., Pepine, C. J. & Raizada, M. K. Brain-gut-bone marrow axis: implications for hypertension and related therapeutics. Circ. Res. 118, 1327–1336 (2016).

Article CAS PubMed PubMed Central Google Scholar

Li, H. B., Yang, T., Richards, E. M., Pepine, C. J. & Raizada, M. K. Maternal treatment with captopril persistently alters gut-brain communication and attenuates hypertension of male offspring. Hypertension 75, 1315–1324 (2020).

Article CAS PubMed Google Scholar

Qi, Y., Aranda, J. M., Rodriguez, V., Raizada, M. K. & Pepine, C. J. Impact of antibiotics on arterial blood pressure in a patient with resistant hypertension—a case report. Int J. Cardiol. 201, 157–158 (2015).

Article PubMed PubMed Central Google Scholar

Angiotensin-Converting Enzyme Inhibitors. in LiverTox: Clinical and Research Information on Drug-Induced Liver Injury (Bethesda, 2012).

Wang, B. L., Pan, D. Q., Zhou, K. L., Lou, Y. Y. & Shi, J. H. Multi-spectroscopic approaches and molecular simulation research of the intermolecular interaction between the angiotensin-converting enzyme inhibitor (ACE inhibitor) benazepril and bovine serum albumin (BSA). Spectrochim. Acta A Mol. Biomol. Spectrosc. 212, 15–24 (2019).

Article CAS PubMed Google Scholar

Hou, F. F. et al. Efficacy and safety of benazepril for advanced chronic renal insufficiency. N. Engl. J. Med. 354, 131–140 (2006).

Article CAS PubMed Google Scholar

Senthong, V. et al. Intestinal microbiota-generated metabolite trimethylamine-N-oxide and 5-year mortality risk in stable coronary artery disease: the contributory role of intestinal microbiota in a COURAGE-like patient cohort. J. Am. Heart Assoc. 5, e002816 (2016).

Article PubMed PubMed Central Google Scholar

Li, X. S. et al. Gut microbiota-dependent trimethylamine N-oxide in acute coronary syndromes: a prognostic marker for incident cardiovascular events beyond traditional risk factors. Eur. Heart J. 38, 814–824 (2017).

CAS PubMed PubMed Central Google Scholar

Tang, W. H. et al. Increased trimethylamine N-oxide portends high mortality risk independent of glycemic control in patients with type 2 diabetes mellitus. Clin. Chem. 63, 297–306 (2017).

Article CAS PubMed Google Scholar

Konop, M. et al. Enalapril decreases rat plasma concentration of TMAO, a gut bacteria-derived cardiovascular marker. Biomarkers 23, 380–385 (2018).

Article CAS PubMed Google Scholar

Romano, K. A., Vivas, E. I., Amador-Noguez, D. & Rey, F. E. Intestinal microbiota composition modulates choline bioavailability from diet and accumulation of the proatherogenic metabolite trimethylamine-N-oxide. mBio 6, e02481 (2015).

Article PubMed PubMed Central Google Scholar

Craciun, S. & Balskus, E. P. Microbial conversion of choline to trimethylamine requires a glycyl radical enzyme. Proc. Natl Acad. Sci. USA 109, 21307–21312 (2012).

Article CAS PubMed PubMed Central Google Scholar

Nowinski, A. & Ufnal, M. Trimethylamine N-oxide: a harmful, protective or diagnostic marker in lifestyle diseases? Nutrition 46, 7–12 (2018).

Article CAS PubMed Google Scholar

Jaworska, K. et al. Hypertension in rats is associated with an increased permeability of the colon to TMA, a gut bacteria metabolite. PLoS One 12, e0189310 (2017).

Article PubMed PubMed Central Google Scholar

Qi, X., Yun, C., Pang, Y. & Qiao, J. The impact of the gut microbiota on the reproductive and metabolic endocrine system. Gut Microbes 13, 1–21 (2021).

Article PubMed Google Scholar

Cornejo-Pareja, I., Munoz-Garach, A., Clemente-Postigo, M. & Tinahones, F. J. Importance of gut microbiota in obesity. Eur. J. Clin. Nutr. 72, 26–37 (2019).

Article CAS PubMed Google Scholar

Pascale, A. et al. Microbiota and metabolic diseases. Endocrine 61, 357–371 (2018).

Article CAS PubMed Google Scholar

Neuman, H., Debelius, J. W., Knight, R. & Koren, O. Microbial endocrinology: the interplay between the microbiota and the endocrine system. FEMS Microbiol. Rev. 39, 509–521 (2015).

Article PubMed Google Scholar

Han, Y. et al. Effect of metformin on all-cause and cardiovascular mortality in patients with coronary artery diseases: a systematic review and an updated meta-analysis. Cardiovasc Diabetol. 18, 96 (2019).

Article PubMed PubMed Central Google Scholar

LaMoia, T. E. & Shulman, G. I. Cellular and molecular mechanisms of metformin action. Endocr. Rev. 42, 77–96 (2021).

Article PubMed Google Scholar

Giusti, L. et al. The protective action of metformin against pro-inflammatory cytokine-induced human islet cell damage and the mechanisms involved. Cells 11, 2465 (2022).

Article CAS PubMed PubMed Central Google Scholar

Sundelin, E., Jensen, J. B., Jakobsen, S., Gormsen, L. C. & Jessen, N. Metformin biodistribution: a key to mechanisms of action? J. Clin. Endocrinol. Metab. 105, dgaa332 (2020).

Article PubMed Google Scholar

Minamii, T., Nogami, M. & Ogawa, W. Mechanisms of metformin action: in and out of the gut. J. Diabetes Investig. 9, 701–703 (2018).

Article PubMed PubMed Central Google Scholar

Rena, G., Hardie, D. G. & Pearson, E. R. The mechanisms of action of metformin. Diabetologia 60, 1577–1585 (2017).

Article CAS PubMed PubMed Central Google Scholar

Foretz, M., Guigas, B. & Viollet, B. Understanding the glucoregulatory mechanisms of metformin in type 2 diabetes mellitus. Nat. Rev. Endocrinol. 15, 569–589 (2019).

Article CAS PubMed Google Scholar

Wu, H. et al. Metformin alters the gut microbiome of individuals with treatment-naive type 2 diabetes, contributing to the therapeutic effects of the drug. Nat. Med. 23, 850–858 (2017).

Article CAS PubMed Google Scholar

Sun, L. et al. Gut microbiota and intestinal FXR mediate the clinical benefits of metformin. Nat. Med. 24, 1919–1929 (2018).

Article CAS PubMed PubMed Central Google Scholar

Bryrup, T. et al. Metformin-induced changes of the gut microbiota in healthy young men: results of a non-blinded, one-armed intervention study. Diabetologia 62, 1024–1035 (2019).

Article CAS PubMed PubMed Central Google Scholar

Pollak, M. The effects of metformin on gut microbiota and the immune system as research frontiers. Diabetologia 60, 1662–1667 (2017).

Article CAS PubMed Google Scholar

Duca, F. A. et al. Metformin activates a duodenal Ampk-dependent pathway to lower hepatic glucose production in rats. Nat. Med. 21, 506–511 (2015).

Article CAS PubMed PubMed Central Google Scholar

Luo, F. et al. Metformin in patients with and without diabetes: a paradigm shift in cardiovascular disease management. Cardiovasc. Diabetol. 18, 54 (2019).

Article PubMed PubMed Central Google Scholar

Dalile, B., Van Oudenhove, L., Vervliet, B. & Verbeke, K. The role of short-chain fatty acids in microbiota-gut-brain communication. Nat. Rev. Gastroenterol. Hepatol. 16, 461–478 (2019).

Article PubMed Google Scholar

Agrawal, N., Sharma, M., Singh, S. & Goyal, A. Recent advances of alpha-glucosidase inhibitors: a comprehensive review. Curr. Top. Med. Chem. 22, 2069–2086 (2022).

Article CAS PubMed Google Scholar

Dhameja, M. & Gupta, P. Synthetic heterocyclic candidates as promising alpha-glucosidase inhibitors: an overview. Eur. J. Med. Chem. 176, 343–377 (2019).

Article CAS PubMed Google Scholar

Zhang, X. et al. Effects of acarbose on the gut microbiota of prediabetic patients: a randomized, double-blind, controlled crossover trial. Diabetes Ther. 8, 293–307 (2017).

Article CAS PubMed PubMed Central Google Scholar

Brunkhorst, C., Andersen, C. & Schneider, E. Acarbose, a pseudooligosaccharide, is transported but not metabolized by the maltose-maltodextrin system of Escherichia coli. J. Bacteriol. 181, 2612–2619 (1999).

Article CAS PubMed PubMed Central Google Scholar

Yadav, H., Jain, S. & Sinha, P. R. Effect of Dahi containing Lactococcus lactis on the progression of diabetes induced by a high-fructose diet in rats. Biosci. Biotechnol. Biochem. 70, 1255–1258 (2006).

Article CAS PubMed Google Scholar

Yadav, H., Jain, S. & Sinha, P. R. Antidiabetic effect of probiotic dahi containing Lactobacillus acidophilus and Lactobacillus casei in high fructose fed rats. Nutrition 23, 62–68 (2007).

Article PubMed Google Scholar

Kaushal, D. & Kansal, V. K. Dahi containing Lactobacillus acidophilus and Bifidobacterium bifidum improves phagocytic potential of macrophages in aged mice. J. Food Sci. Technol. 51, 1147–1153 (2014).

Article CAS PubMed Google Scholar

Su, B. et al. Acarbose treatment affects the serum levels of inflammatory cytokines and the gut content of bifidobacteria in Chinese patients with type 2 diabetes mellitus. J. Diabetes 7, 729–739 (2015).

Article CAS PubMed Google Scholar

Maruhama, Y. et al. Effects of a glucoside-hydrolase inhibitor (Bay g 5421) on serum lipids, lipoproteins and bile acids, fecal fat and bacterial flora, and intestinal gas production in hyperlipidemic patients. Tohoku J. Exp. Med. 132, 453–462 (1980).

Article CAS PubMed Google Scholar

Kishida, Y., Okubo, H., Ohno, H., Oki, K. & Yoneda, M. Effect of miglitol on the suppression of nonalcoholic steatohepatitis development and improvement of the gut environment in a rodent model. J. Gastroenterol. 52, 1180–1191 (2017).

Article CAS PubMed PubMed Central Google Scholar

Drucker, D. J. & Nauck, M. A. The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet 368, 1696–1705 (2006).

Article CAS PubMed Google Scholar

Meier, J. J. GLP-1 receptor agonists for individualized treatment of type 2 diabetes mellitus. Nat. Rev. Endocrinol. 8, 728–742 (2012).

Article CAS PubMed Google Scholar

Drucker, D. J. Mechanisms of action and therapeutic application of glucagon-like peptide-1. Cell Metab. 27, 740–756 (2018).

Article CAS PubMed Google Scholar

Baggio, L. L. & Drucker, D. J. Glucagon-like peptide-1 receptors in the brain: controlling food intake and body weight. J. Clin. Invest. 124, 4223–4226 (2014).

Article PubMed PubMed Central Google Scholar

Tolhurst, G. et al. Short-chain fatty acids stimulate glucagon-like peptide-1 secretion via the G-protein-coupled receptor FFAR2. Diabetes 61, 364–371 (2012).

Article CAS PubMed PubMed Central Google Scholar

Kimura, I. et al. The gut microbiota suppresses insulin-mediated fat accumulation via the short-chain fatty acid receptor GPR43. Nat. Commun. 4, 1829 (2013).

Article PubMed Google Scholar

Vettorazzi, J. F. et al. The bile acid TUDCA increases glucose-induced insulin secretion via the cAMP/PKA pathway in pancreatic beta cells. Metabolism 65, 54–63 (2016).

Article CAS PubMed Google Scholar

Aoki, R. et al. A proliferative probiotic Bifidobacterium strain in the gut ameliorates progression of metabolic disorders via microbiota modulation and acetate elevation. Sci. Rep. 7, 43522 (2017).

Article PubMed PubMed Central Google Scholar

Wang, L., Li, P., Tang, Z., Yan, X. & Feng, B. Structural modulation of the gut microbiota and the relationship with body weight: compared evaluation of liraglutide and saxagliptin treatment. Sci. Rep. 6, 33251 (2016).

Article CAS PubMed PubMed Central Google Scholar

Zhang, Q. et al. Featured article: structure moderation of gut microbiota in liraglutide-treated diabetic male rats. Exp. Biol. Med. (Maywood) 243, 34–44 (2018).

Article CAS PubMed Google Scholar

Zhao, L. et al. A glucagon-like peptide-1 receptor agonist lowers weight by modulating the structure of gut microbiota. Front. Endocrinol. (Lausanne) 9, 233 (2018).

Article PubMed Google Scholar

Charpentier, J. et al. Liraglutide targets the gut microbiota and the intestinal immune system to regulate insulin secretion. Acta Diabetol. 58, 881–897 (2021).

Article CAS PubMed Google Scholar

Shang, J. et al. Liraglutide-induced structural modulation of the gut microbiota in patients with type 2 diabetes mellitus. PeerJ 9, e11128 (2021).

Article PubMed PubMed Central Google Scholar

Yuan, X. et al. Identification of therapeutic effect of glucagon-like peptide 1 in the treatment of STZ-induced diabetes mellitus in rats by restoring the balance of intestinal flora. J. Cell Biochem. 119, 10067–10074 (2018).

Article CAS PubMed Google Scholar

Handelsman, Y. et al. American association of clinical endocrinologists and american college of endocrinology—clinical practice guidelines for developing a diabetes mellitus comprehensive care plan - 2015. Endocr. Pract. 21, 1–87 (2015).

Article PubMed PubMed Central Google Scholar

Olivares, M. et al. The potential role of the dipeptidyl peptidase-4-like activity from the gut microbiota on the host health. Front. Microbiol. 9, 1900 (2018).

Article PubMed PubMed Central Google Scholar

Liao, X. et al. Alteration of gut microbiota induced by DPP-4i treatment improves glucose homeostasis. EBioMedicine 44, 665–674 (2019).

Article PubMed PubMed Central Google Scholar

Yan, X., Feng, B., Li, P., Tang, Z. & Wang, L. Microflora disturbance during progression of glucose intolerance and effect of sitagliptin: an animal study. J. Diabetes Res. 2016, 2093171 (2016).

Article PubMed PubMed Central Google Scholar

Zhang, Q. et al. Vildagliptin increases butyrate-producing bacteria in the gut of diabetic rats. PLoS One 12, e0184735 (2017).

Article PubMed PubMed Central Google Scholar

Zhao, X. et al. Host-microbiota interaction-mediated resistance to inflammatory bowel disease in pigs. Microbiome 10, 115 (2022).

Article CAS PubMed PubMed Central Google Scholar

Alshehri, D. et al. Dysbiosis of gut microbiota in inflammatory bowel disease: current therapies and potential for microbiota-modulating therapeutic approaches. Bosn. J. Basic Med. Sci. 21, 270–283 (2021).

CAS PubMed PubMed Central Google Scholar

Nilsson, A. et al. Olsalazine versus sulphasalazine for relapse prevention in ulcerative colitis: a multicenter study. Am. J. Gastroenterol. 90, 381–387 (1995).

CAS PubMed Google Scholar

Kim, D. H. Gut microbiota-mediated drug-antibiotic interactions. Drug Metab. Dispos. 43, 1581–1589 (2015).

Article CAS PubMed Google Scholar

Azad Khan, A. K., Guthrie, G., Johnston, H. H., Truelove, S. C. & Williamson, D. H. Tissue and bacterial splitting of sulphasalazine. Clin. Sci. (Lond.) 64, 349–354 (1983).

Article CAS PubMed Google Scholar

Lee, H. J., Zhang, H., Orlovich, D. A. & Fawcett, J. P. The influence of probiotic treatment on sulfasalazine metabolism in rat. Xenobiotica 42, 791–797 (2012).

Article CAS PubMed Google Scholar

Crouwel, F., Buiter, H. J. C. & de Boer, N. K. Gut microbiota-driven drug metabolism in inflammatory bowel disease. J. Crohns Colitis 15, 307–315 (2020).

Article PubMed PubMed Central Google Scholar

Savarino, V. et al. The appropriate use of proton-pump inhibitors. Minerva Med 109, 386–399 (2018).

Article PubMed Google Scholar

Graham, D. Y. & Tansel, A. Interchangeable use of proton pump inhibitors based on relative potency. Clin. Gastroenterol. Hepatol. 16, 800–808.e7 (2018).

Article CAS PubMed Google Scholar

Pinto-Sanchez, M. I., Yuan, Y., Bercik, P. & Moayyedi, P. Proton pump inhibitors for functional dyspepsia. Cochrane Database Syst. Rev. 3, CD011194 (2017).

PubMed Google Scholar

Forgacs, I. & Loganayagam, A. Overprescribing proton pump inhibitors. BMJ 336, 2–3 (2008).

Article PubMed PubMed Central Google Scholar

Akram, F., Huang, Y., Lim, V., Huggan, P. J. & Merchant, R. A. Proton pump inhibitors: are we still prescribing them without valid indications? Australas. Med. J. 7, 465–470 (2014).

Article PubMed PubMed Central Google Scholar

Eid, S. M. et al. Patterns and predictors of proton pump inhibitor overuse among academic and non-academic hospitalists. Intern Med. 49, 2561–2568 (2010).

Article PubMed Google Scholar

Bonder, M. J. et al. The influence of a short-term gluten-free diet on the human gut microbiome. Genome Med. 8, 45 (2016).

Article PubMed PubMed Central Google Scholar

Jackson, M. A. et al. Proton pump inhibitors alter the composition of the gut microbiota. Gut 65, 749–756 (2016).

Article PubMed Google Scholar

Imhann, F. et al. Proton pump inhibitors affect the gut microbiome. Gut 65, 740–748 (2016).

Article CAS PubMed Google Scholar

Freedberg, D. E. et al. Proton pump inhibitors alter specific taxa in the human gastrointestinal microbiome: a crossover trial. Gastroenterology 149, 883–885.e9 (2015).

Article CAS PubMed Google Scholar

Maier, L. et al. Extensive impact of non-antibiotic drugs on human gut bacteria. Nature 555, 623–628 (2018).

Article CAS PubMed PubMed Central Google Scholar

Buffie, C. G. et al. Precision microbiome reconstitution restores bile acid mediated resistance to Clostridium difficile. Nature 517, 205–208 (2015).

Article CAS PubMed Google Scholar

Buffie, C. G. & Pamer, E. G. Microbiota-mediated colonization resistance against intestinal pathogens. Nat. Rev. Immunol. 13, 790–801 (2013).

Article CAS PubMed PubMed Central Google Scholar

Leonard, J., Marshall, J. K. & Moayyedi, P. Systematic review of the risk of enteric infection in patients taking acid suppression. Am. J. Gastroenterol. 102, 2047–2056 (2007).

Article PubMed Google Scholar

Baur, D. et al. Effect of antibiotic stewardship on the incidence of infection and colonisation with antibiotic-resistant bacteria and Clostridium difficile infection: a systematic review and meta-analysis. Lancet Infect. Dis. 17, 990–1001 (2017).

Article PubMed Google Scholar

Bajaj, J. S. et al. Proton pump inhibitor initiation and withdrawal affects gut microbiota and readmission risk in cirrhosis. Am. J. Gastroenterol. 113, 1177–1186 (2018).

Article CAS PubMed Google Scholar

Stark, C. M., Susi, A., Emerick, J. & Nylund, C. M. Antibiotic and acid-suppression medications during early childhood are associated with obesity. Gut 68, 62–69 (2019).

Article CAS PubMed Google Scholar

Harirforoosh, S., Asghar, W. & Jamali, F. Adverse effects of nonsteroidal antiinflammatory drugs: an update of gastrointestinal, cardiovascular and renal complications. J. Pharm. Pharm. Sci. 16, 821–847 (2013).

Article PubMed Google Scholar

Sostres, C., Gargallo, C. J. & Lanas, A. Nonsteroidal anti-inflammatory drugs and upper and lower gastrointestinal mucosal damage. Arthritis Res. Ther. 15, S3 (2013).

Article PubMed PubMed Central Google Scholar

Higuchi, K. et al. Present status and strategy of NSAIDs-induced small bowel injury. J. Gastroenterol. 44, 879–888 (2009).

Article PubMed Google Scholar

Petruzzelli, M. et al. Intestinal mucosal damage caused by non-steroidal anti-inflammatory drugs: role of bile salts. Clin. Biochem. 40, 503–510 (2007).

Article CAS PubMed Google Scholar

Boelsterli, U. A., Redinbo, M. R. & Saitta, K. S. Multiple NSAID-induced hits injure the small intestine: underlying mechanisms and novel strategies. Toxicol. Sci. 131, 654–667 (2013).

Article CAS PubMed Google Scholar

Yauw, S. T. K. et al. Microbial glucuronidase inhibition reduces severity of diclofenac-induced anastomotic leak in rats. Surg. Infect. (Larchmt.) 19, 417–423 (2018).

Article PubMed Google Scholar

Dashnyam, P. et al. beta-Glucuronidases of opportunistic bacteria are the major contributors to xenobiotic-induced toxicity in the gut. Sci. Rep. 8, 16372 (2018).

Article PubMed PubMed Central Google Scholar

Ervin, S. M. et al. Targeting regorafenib-induced toxicity through inhibition of gut microbial beta-glucuronidases. ACS Chem. Biol. 14, 2737–2744 (2019).

Article CAS PubMed PubMed Central Google Scholar

Yang, B., Liu, H., Yang, J., Gupta, V. K. & Jiang, Y. New insights on bioactivities and biosynthesis of flavonoid glycosides. Trends Food Sci. Technol. 79, 116–124 (2018).

Article CAS Google Scholar

Falony, G. et al. Population-level analysis of gut microbiome variation. Science 352, 560–564 (2016).

Article CAS PubMed Google Scholar

Dinan, T. G. & Cryan, J. F. The microbiome-gut-brain axis in health and disease. Gastroenterol. Clin. North Am. 46, 77–89 (2017).

Article PubMed Google Scholar

LeWitt, P. A. Levodopa therapy for Parkinson’s disease: pharmacokinetics and pharmacodynamics. Mov. Disord. 30, 64–72 (2015).

Article CAS PubMed Google Scholar

Marsot, A., Guilhaumou, R., Azulay, J. P. & Blin, O. Levodopa in Parkinson’s disease: a review of population pharmacokinetics/pharmacodynamics analysis. J. Pharm. Pharm. Sci. 20, 226–238 (2017).

Article CAS PubMed Google Scholar

van Kessel, S. P. et al. Gut bacterial tyrosine decarboxylases restrict levels of levodopa in the treatment of Parkinson’s disease. Nat. Commun. 10, 310 (2019).

Article PubMed PubMed Central Google Scholar

Maini Rekdal, V., Bess, E. N., Bisanz, J. E., Turnbaugh, P. J. & Balskus, E. P. Discovery and inhibition of an interspecies gut bacterial pathway for Levodopa metabolism. Science 364, eaau6323 (2019).

Article PubMed Google Scholar

Musil, V., Blankova, A. & Baca, V. A plea for an extension of the anatomical nomenclature: the locomotor system. Bosn. J. Basic Med. Sci. 18, 117–125 (2018).

PubMed PubMed Central Google Scholar

Peretti, S. et al. The Yin-Yang pharmacomicrobiomics on treatment response in inflammatory arthritides: a narrative review. Genes (Basel) 14, 89 (2022).

Article PubMed Google Scholar

Zhao, S. et al. Drug repurposing by siderophore conjugation: synthesis and biological evaluation of siderophore-methotrexate conjugates as antibiotics. Angew. Chem. Int. Ed. Engl. 61, e202204139 (2022).

Article CAS PubMed Google Scholar

Romao, V. C., Lima, A., Bernardes, M., Canhao, H. & Fonseca, J. E. Three decades of low-dose methotrexate in rheumatoid arthritis: can we predict toxicity? Immunol. Res. 60, 289–310 (2014).

Article CAS PubMed Google Scholar

Ramos-Romero, S. et al. Mechanistically different effects of fat and sugar on insulin resistance, hypertension, and gut microbiota in rats. Am. J. Physiol. Endocrinol. Metab. 314, E552–E563 (2018).

Article CAS PubMed Google Scholar

Dunn, C. J. & Peters, D. H. Metformin. A review of its pharmacological properties and therapeutic use in non-insulin-dependent diabetes mellitus. Drugs 49, 721–749 (1995).

Article CAS PubMed Google Scholar

Zhou, B. et al. Induction and amelioration of methotrexate-induced gastrointestinal toxicity are related to immune response and gut microbiota. EBioMedicine 33, 122–133 (2018).

Article PubMed PubMed Central Google Scholar

Sayers, E., MacGregor, A. & Carding, S. R. Drug-microbiota interactions and treatment response: relevance to rheumatoid arthritis. AIMS Microbiol. 4, 642–654 (2018).

Article CAS PubMed PubMed Central Google Scholar

Zhang, X. et al. The oral and gut microbiomes are perturbed in rheumatoid arthritis and partly normalized after treatment. Nat. Med. 21, 895–905 (2015).

Article CAS PubMed Google Scholar

Goodman, S. M., Cronstein, B. N. & Bykerk, V. P. Outcomes related to methotrexate dose and route of administration in patients with rheumatoid arthritis: a systematic literature review. Clin. Exp. Rheumatol. 33, 272–278 (2015).

PubMed Google Scholar

Halilova, K. I. et al. Markers of treatment response to methotrexate in rheumatoid arthritis: where do we stand? Int. J. Rheumatol. 2012, 978396 (2012).

Article PubMed PubMed Central Google Scholar

Angelis-Stoforidis, P., Vajda, F. J. & Christophidis, N. Methotrexate polyglutamate levels in circulating erythrocytes and polymorphs correlate with clinical efficacy in rheumatoid arthritis. Clin. Exp. Rheumatol. 17, 313–320 (1999).

CAS PubMed Google Scholar

Dervieux, T. et al. Polyglutamation of methotrexate with common polymorphisms in reduced folate carrier, aminoimidazole carboxamide ribonucleotide transformylase, and thymidylate synthase are associated with methotrexate effects in rheumatoid arthritis. Arthritis Rheum. 50, 2766–2774 (2004).

Article CAS PubMed Google Scholar

Clemens, J. Q. Infection and inflammation of the genitourinary tract. J. Urol. 208, 455 (2022).

Article PubMed Google Scholar

Choi, E., Buie, J., Camacho, J., Sharma, P. & de Riese, W. T. W. Evolution of androgen deprivation therapy (ADT) and its new emerging modalities in prostate cancer: an update for practicing urologists, clinicians and medical providers. Res. Rep. Urol. 14, 87–108 (2022).

CAS PubMed PubMed Central Google Scholar

Terrisse, S. et al. Immune system and intestinal microbiota determine efficacy of androgen deprivation therapy against prostate cancer. J. Immunother. Cancer 10, e004191 (2022).

Article PubMed PubMed Central Google Scholar

Daisley, B. A. et al. Abiraterone acetate preferentially enriches for the gut commensal Akkermansia muciniphila in castrate-resistant prostate cancer patients. Nat. Commun. 11, 4822 (2020).

Article CAS PubMed PubMed Central Google Scholar

Sfanos, K. S., Yegnasubramanian, S., Nelson, W. G. & De Marzo, A. M. The inflammatory microenvironment and microbiome in prostate cancer development. Nat. Rev. Urol. 15, 11–24 (2018).

Article PubMed Google Scholar

McCulloch, J. A. & Trinchieri, G. Gut bacteria enable prostate cancer growth. Science 374, 154–155 (2021).

Article CAS PubMed Google Scholar

Dickson, R. P., Erb-Downward, J. R., Martinez, F. J. & Huffnagle, G. B. The microbiome and the respiratory tract. Annu Rev. Physiol. 78, 481–504 (2016).

Article CAS PubMed Google Scholar

Unger, S. A. & Bogaert, D. The respiratory microbiome and respiratory infections. J. Infect. 74, S84–S88 (2017).

Article PubMed Google Scholar

Man, W. H., de Steenhuijsen Piters, W. A. & Bogaert, D. The microbiota of the respiratory tract: gatekeeper to respiratory health. Nat. Rev. Microbiol. 15, 259–270 (2017).

Article CAS PubMed PubMed Central Google Scholar

Mi, J. et al. The research progress in immunotherapy of tuberculosis. Front. Cell Infect. Microbiol. 11, 763591 (2021).

Article CAS PubMed PubMed Central Google Scholar

Bermingham, W. H. et al. Practical management of suspected hypersensitivity reactions to anti-tuberculosis drugs. Clin. Exp. Allergy 52, 375–386 (2022).

Article PubMed Google Scholar

Namasivayam, S. et al. Longitudinal profiling reveals a persistent intestinal dysbiosis triggered by conventional anti-tuberculosis therapy. Microbiome 5, 71 (2017).

Article PubMed PubMed Central Google Scholar

Naidoo, C. C. et al. The microbiome and tuberculosis: state of the art, potential applications, and defining the clinical research agenda. Lancet Respir. Med. 7, 892–906 (2019).

Article PubMed Google Scholar

Wipperman, M. F. et al. Antibiotic treatment for Tuberculosis induces a profound dysbiosis of the microbiome that persists long after therapy is completed. Sci. Rep. 7, 10767 (2017).

Article PubMed PubMed Central Google Scholar

Shen, Y. et al. Outer membrane vesicles of a human commensal mediate immune regulation and disease protection. Cell Host Microbe 12, 509–520 (2012).

Article CAS PubMed PubMed Central Google Scholar

Larsen, J. M. The immune response to Prevotella bacteria in chronic inflammatory disease. Immunology 151, 363–374 (2017).

Article CAS PubMed PubMed Central Google Scholar

Wells, J. M. Immunomodulatory mechanisms of lactobacilli. Micro. Cell Fact. 10, S17 (2011).

Article Google Scholar

Scriba, T. J. et al. Differential recognition of mycobacterium tuberculosis-specific epitopes as a function of tuberculosis disease history. Am. J. Respir. Crit. Care Med. 196, 772–781 (2017).

Article CAS PubMed PubMed Central Google Scholar

Hawn, T. R., Matheson, A. I., Maley, S. N. & Vandal, O. Host-directed therapeutics for tuberculosis: can we harness the host? Microbiol. Mol. Biol. Rev. 77, 608–627 (2013).

Article PubMed PubMed Central Google Scholar

Caballero-Flores, G., Pickard, J. M. & Nunez, G. Microbiota-mediated colonization resistance: mechanisms and regulation. Nat. Rev. Microbiol. 21, 347–360 (2023).

Article CAS PubMed Google Scholar

Gong, X. et al. The interactions between gut microbiota and bioactive ingredients of traditional Chinese medicines: a review. Pharm. Res. 157, 104824 (2020).

Article CAS Google Scholar

Hayase, E. & Jenq, R. R. Role of the intestinal microbiome and microbial-derived metabolites in immune checkpoint blockade immunotherapy of cancer. Genome Med. 13, 107 (2021).

Article PubMed PubMed Central Google Scholar

Peng, Y. et al. Role of gut microbiota in travel-related acquisition of extended spectrum beta-lactamase-producing Enterobacteriaceae. J. Travel Med. 28, taab022 (2021).

Article PubMed Google Scholar

He, Z. et al. Gut microbiota-derived ursodeoxycholic acid from neonatal dairy calves improves intestinal homeostasis and colitis to attenuate extended-spectrum beta-lactamase-producing enteroaggregative Escherichia coli infection. Microbiome 10, 79 (2022).

Article CAS PubMed PubMed Central Google Scholar

Smet, A. et al. Broad-spectrum beta-lactamases among Enterobacteriaceae of animal origin: molecular aspects, mobility and impact on public health. FEMS Microbiol. Rev. 34, 295–316 (2010).

Article CAS PubMed Google Scholar

Piewngam, P. et al. Composition of the intestinal microbiota in extended-spectrum beta-lactamase-producing Enterobacteriaceae carriers and non-carriers in Thailand. Int J. Antimicrob. Agents 53, 435–441 (2019).

Article CAS PubMed Google Scholar

McCulloch, J. A. et al. Intestinal microbiota signatures of clinical response and immune-related adverse events in melanoma patients treated with anti-PD-1. Nat. Med. 28, 545–556 (2022).

Article CAS PubMed PubMed Central Google Scholar

Munier, A. L. et al. Comparative dynamics of the emergence of fluoroquinolone resistance in staphylococci from the nasal microbiota of patients treated with fluoroquinolones according to their environment. Int J. Antimicrob. Agents 46, 653–659 (2015).

Article CAS PubMed Google Scholar

de Lastours, V. & Fantin, B. Impact of fluoroquinolones on human microbiota. Focus on the emergence of antibiotic resistance. Future Microbiol. 10, 1241–1255 (2015).

Article PubMed Google Scholar

Klunemann, M. et al. Bioaccumulation of therapeutic drugs by human gut bacteria. Nature 597, 533–538 (2021).

Article PubMed PubMed Central Google Scholar

Abt, M. C., McKenney, P. T. & Pamer, E. G. Clostridium difficile colitis: pathogenesis and host defence. Nat. Rev. Microbiol. 14, 609–620 (2016).

Article CAS PubMed PubMed Central Google Scholar

O’Grady, K., Knight, D. R. & Riley, T. V. Antimicrobial resistance in Clostridioides difficile. Eur. J. Clin. Microbiol. Infect. Dis. 40, 2459–2478 (2021).

Article PubMed Google Scholar

Gil, F. & Paredes-Sabja, D. Acyldepsipeptide antibiotics as a potential therapeutic agent against Clostridium difficile recurrent infections. Future Microbiol. 11, 1179–1189 (2016).

Article CAS PubMed Google Scholar

Liu, X., Chen, Y., Zhang, S. & Dong, L. Gut microbiota-mediated immunomodulation in tumor. J. Exp. Clin. Cancer Res. 40, 221 (2021).

Article PubMed PubMed Central Google Scholar

Simpson, R. C. et al. Diet-driven microbial ecology underpins associations between cancer immunotherapy outcomes and the gut microbiome. Nat. Med. 28, 2344–2352 (2022).

Article CAS PubMed Google Scholar

Cheng, W. Y., Wu, C. Y. & Yu, J. The role of gut microbiota in cancer treatment: friend or foe? Gut 69, 1867–1876 (2020).

Article CAS PubMed Google Scholar

Li, X., Zhang, S., Guo, G., Han, J. & Yu, J. Gut microbiome in modulating immune checkpoint inhibitors. EBioMedicine 82, 104163 (2022).

Article CAS PubMed PubMed Central Google Scholar

Lu, Y. et al. Gut microbiota influence immunotherapy responses: mechanisms and therapeutic strategies. J. Hematol. Oncol. 15, 47 (2022).

Article PubMed PubMed Central Google Scholar

Paone, P. & Cani, P. D. Mucus barrier, mucins and gut microbiota: the expected slimy partners? Gut 69, 2232–2243 (2020).

Article CAS PubMed Google Scholar

Takiishi, T., Fenero, C. I. M. & Camara, N. O. S. Intestinal barrier and gut microbiota: shaping our immune responses throughout life. Tissue Barriers 5, e1373208 (2017).

Article PubMed PubMed Central Google Scholar

Ianiro, G., Tilg, H. & Gasbarrini, A. Antibiotics as deep modulators of gut microbiota: between good and evil. Gut 65, 1906–1915 (2016).

Article CAS PubMed Google Scholar

Becattini, S., Taur, Y. & Pamer, E. G. Antibiotic-induced changes in the intestinal microbiota and disease. Trends Mol. Med. 22, 458–478 (2016).

Article CAS PubMed PubMed Central Google Scholar

Akbarali, H. I. & Dewey, W. L. Gastrointestinal motility, dysbiosis and opioid-induced tolerance: is there a link? Nat. Rev. Gastroenterol. Hepatol. 16, 323–324 (2019).

Article PubMed PubMed Central Google Scholar

Kurz, A. & Sessler, D. I. Opioid-induced bowel dysfunction: pathophysiology and potential new therapies. Drugs 63, 649–671 (2003).

Article CAS PubMed Google Scholar

Mora, A. L. et al. Moderate to high use of opioid analgesics are associated with an increased risk of Clostridium difficile infection. Am. J. Med. Sci. 343, 277–280 (2012).

Article PubMed Google Scholar

Zhuang, Y. P. et al. Gut microbiota interactions with antitumor immunity in colorectal cancer: from understanding to application. Biomed. Pharmacother. 165, 115040 (2023).

Article CAS PubMed Google Scholar

Pezo, R. C., Wong, M. & Martin, A. Impact of the gut microbiota on immune checkpoint inhibitor-associated toxicities. Ther. Adv. Gastroenterol. 12, 1756284819870911 (2019).

Article CAS Google Scholar

Bai, Z. et al. Aspirin ameliorates atherosclerotic immuno-inflammation through regulating the Treg/Th17 axis and CD39-CD73 adenosine signaling via remodeling the gut microbiota in ApoE(-/-) mice. Int. Immunopharmacol. 120, 110296 (2023).

Article CAS PubMed Google Scholar

Kolodziejczyk, A. A., Zheng, D. & Elinav, E. Diet-microbiota interactions and personalized nutrition. Nat. Rev. Microbiol. 17, 742–753 (2019).

Article CAS PubMed Google Scholar

Zmora, N., Suez, J. & Elinav, E. You are what you eat: diet, health and the gut microbiota. Nat. Rev. Gastroenterol. Hepatol. 16, 35–56 (2019).

Article CAS PubMed Google Scholar

Sanders, M. E., Merenstein, D. J., Reid, G., Gibson, G. R. & Rastall, R. A. Probiotics and prebiotics in intestinal health and disease: from biology to the clinic. Nat. Rev. Gastroenterol. Hepatol. 16, 605–616 (2019).

Article PubMed Google Scholar

Lim, H. et al. Artificial intelligence approaches to human-microbiome protein-protein interactions. Curr. Opin. Struct. Biol. 73, 102328 (2022).

Article CAS PubMed Google Scholar

Cammarota, G. et al. Gut microbiome, big data and machine learning to promote precision medicine for cancer. Nat. Rev. Gastroenterol. Hepatol. 17, 635–648 (2020).

Article PubMed Google Scholar

Vitetta, L., Coulson, S., Linnane, A. W. & Butt, H. The gastrointestinal microbiome and musculoskeletal diseases: a beneficial role for probiotics and prebiotics. Pathogens 2, 606–626 (2013).

Article CAS PubMed PubMed Central Google Scholar

Bermudez-Brito, M., Plaza-Diaz, J., Munoz-Quezada, S., Gomez-Llorente, C. & Gil, A. Probiotic mechanisms of action. Ann. Nutr. Metab. 61, 160–174 (2012).

Article CAS PubMed Google Scholar

Gupta, S., Allen-Vercoe, E. & Petrof, E. O. Fecal microbiota transplantation: in perspective. Ther. Adv. Gastroenterol. 9, 229–239 (2016).

Article Google Scholar

Ser, H. L., Letchumanan, V., Goh, B. H., Wong, S. H. & Lee, L. H. The use of fecal microbiome transplant in treating human diseases: too early for poop? Front. Microbiol. 12, 519836 (2021).

Article PubMed PubMed Central Google Scholar

Khoruts, A. Fecal microbiota transplantation-early steps on a long journey ahead. Gut Microbes 8, 199–204 (2017).

Article PubMed PubMed Central Google Scholar

van Nood, E., Speelman, P., Nieuwdorp, M. & Keller, J. Fecal microbiota transplantation: facts and controversies. Curr. Opin. Gastroenterol. 30, 34–39 (2014).

Article PubMed Google Scholar

Borody, T. J. & Campbell, J. Fecal microbiota transplantation: techniques, applications, and issues. Gastroenterol. Clin. North Am. 41, 781–803 (2012).

Article PubMed Google Scholar

Li, M. et al. Pro- and anti-inflammatory effects of short chain fatty acids on immune and endothelial cells. Eur. J. Pharm. 831, 52–59 (2018).

Article CAS Google Scholar

Lin, D. M., Koskella, B. & Lin, H. C. Phage therapy: an alternative to antibiotics in the age of multi-drug resistance. World J. Gastrointest. Pharm. Ther. 8, 162–173 (2017).

Article Google Scholar

Bourdin, G. et al. Coverage of diarrhoea-associated Escherichia coli isolates from different origins with two types of phage cocktails. Micro. Biotechnol. 7, 165–176 (2014).

Article CAS Google Scholar

Ackermann, H. W. The first phage electron micrographs. Bacteriophage 1, 225–227 (2011).

Article PubMed PubMed Central Google Scholar

Rakhuba, D. V., Kolomiets, E. I., Dey, E. S. & Novik, G. I. Bacteriophage receptors, mechanisms of phage adsorption and penetration into host cell. Pol. J. Microbiol. 59, 145–155 (2010).

Article CAS PubMed Google Scholar

Motlagh, A. M., Bhattacharjee, A. S. & Goel, R. Biofilm control with natural and genetically-modified phages. World J. Microbiol Biotechnol. 32, 67 (2016).

Article PubMed Google Scholar

Ramachandran, G. & Bikard, D. Editing the microbiome the CRISPR way. Philos. Trans. R. Soc. Lond. B Biol. Sci. 374, 20180103 (2019).

Article CAS PubMed PubMed Central Google Scholar

Reyes, A., Wu, M., McNulty, N. P., Rohwer, F. L. & Gordon, J. I. Gnotobiotic mouse model of phage-bacterial host dynamics in the human gut. Proc. Natl Acad. Sci. USA 110, 20236–20241 (2013).

Article CAS PubMed PubMed Central Google Scholar

Lusiak-Szelachowska, M., Weber-Dabrowska, B., Jonczyk-Matysiak, E., Wojciechowska, R. & Gorski, A. Bacteriophages in the gastrointestinal tract and their implications. Gut Pathog. 9, 44 (2017).

Article PubMed PubMed Central Google Scholar

Divya Ganeshan, S. & Hosseinidoust, Z. Phage therapy with a focus on the human microbiota. Antibiotics (Basel) 8, 131 (2019).

Article PubMed Google Scholar

Ren, Z. et al. Gut microbiome analysis as a tool towards targeted non-invasive biomarkers for early hepatocellular carcinoma. Gut 68, 1014–1023 (2019).

Article CAS PubMed Google Scholar

Scott, A. J. et al. International Cancer Microbiome Consortium consensus statement on the role of the human microbiome in carcinogenesis. Gut 68, 1624–1632 (2019).

Article CAS PubMed Google Scholar

Rao, B. C. et al. Human microbiome is a diagnostic biomarker in hepatocellular carcinoma. Hepatobiliary Pancreat. Dis. Int. 19, 109–115 (2020).

Article CAS PubMed Google Scholar

Kuntz, T. M. & Gilbert, J. A. Introducing the microbiome into precision medicine. Trends Pharm. Sci. 38, 81–91 (2017).

Article CAS PubMed Google Scholar

Rizkallah, M. R., Saad, R. & Aziz, R. K. The Human Microbiome Project, personalized medicine and the birth of phar macomicrobiomics. Curr. Pharm. Pers. Med. 8, 182–193 (2010).

CAS Google Scholar

Download references

We thank the other members of the Department of Clinical Pharmacology (Xiangya Hospital, Central South University) for pivotal discussions and critical feedback on the manuscript.

This work was supported by the National Key Research and Development Program (No. 2021YFA1301200), the National Scientific Foundation of China (No. 82373961, 82073945, 82373960, and 81974513), the Natural Science Foundation of Hunan Province (grant 2022JJ80113, 2022JJ80097).

Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, PR China

Qing Zhao, Yao Chen, Weihua Huang, Honghao Zhou & Wei Zhang

Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, PR China

Qing Zhao, Yao Chen, Weihua Huang & Honghao Zhou

Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, PR China

Qing Zhao, Yao Chen, Weihua Huang & Honghao Zhou

National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, PR China

Qing Zhao, Yao Chen, Weihua Huang & Honghao Zhou

The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, PR China

Wei Zhang

The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, PR China

Wei Zhang

Central Laboratory of Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, Changsha, 410013, PR China

Wei Zhang

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

H.Z. and W.Z.: conceptualization and supervision. Q.Z.: writing the original draft and drawing the diagrams. Y.C.: review and revision. W.H.: modification. All authors have read and approved the article.

Correspondence to Wei Zhang.

The authors declare no competing interests.

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and Permissions

Zhao, Q., Chen, Y., Huang, W. et al. Drug-microbiota interactions: an emerging priority for precision medicine. Sig Transduct Target Ther 8, 386 (2023). https://doi.org/10.1038/s41392-023-01619-w

Download citation

Received: 31 October 2022

Revised: 20 July 2023

Accepted: 24 August 2023

Published: 09 October 2023

DOI: https://doi.org/10.1038/s41392-023-01619-w

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

SHARE