Translate this page into:
Role of microbiota in colorectal cancer: From pathogenesis to treatment
* Corresponding author E-mail address: eltanani@rakmhsu.ac.ae (M. El-Tanani), satyam@rakmhsu.ac.ae (S.M. Satyam)
-
Received: ,
Accepted: ,
Abstract
A balance between the gut bacteria is crucial for the development and management of cancer. An unhealthy gut microbial community leads to cancer growth by causing inflammation and DNA damage, and affecting the immune system response. Certain bacteria, such as Fusobacterium nucleatum, increase the risk of colorectal cancer (CRC), whereas microbial by-products influence inflammation and the spread of cancer cells. Recognizing these processes is vital in developing approaches to prevent and treat cancer. This review aims to comprehensively examine the role of gut microbiota in CRC pathogenesis, diagnosis, and treatment, with a focus on microbial-based interventions such as probiotics, prebiotics, and fecal microbiota transplantation (FMT).
Recent studies have shown that microbial patterns could serve as indicators for detecting and predicting the stages of CRC without the need for invasive procedures, such as traditional screening. Specifically, Fusobacterium nucleatum levels have been found to increase in cancer cases, suggesting a direct connection between microbial presence and disease advancement. Treatments that focus on adjusting the gut microbiota through methods such as probiotics and FMT have the potential to improve treatment outcomes and minimize adverse reactions. Studies have shown that FMT can enhance the results of chemotherapy in cancer treatment programs for patients with colon cancer by incorporating microbiota-based strategies into the treatment protocols.
Despite these possibilities, in this field of study and research on microbiota composition and environmental factors, there are still some hurdles owing to variations among individuals and diverse surroundings that require attention. Standardizing the investigative methods used to study microbiota is essential to achieve results that can be effectively replicated and applied in clinical settings. Moving forward, it would be beneficial for researchers to concentrate on conducting extensive validation studies and creating customized treatments based on microbiota analysis to seamlessly incorporate this aspect into regular CRC care routines.
In conclusion, the study of gut bacteria is promising in the field of cancer research and therapy. Ongoing collaboration and research are crucial for maximizing the effectiveness of microbiota-focused treatments to enhance cancer prevention, detection, and treatment outcomes. This study offers an examination of the existing knowledge and future pathways for utilizing gut bacteria to manage cancer.
Keywords
Cancer immunotherapy
Colorectal cancer (CRC)
Fecal microbiota transplantation (FMT)
Fusobacterium nucleatum
Gut microbiota
GRAPHICAL ABSTRACT
1. Introduction
The vast collection of organisms known as microbiota, mainly located in the gut, is crucial for promoting wellness and for preventing illnesses. This diverse array of microorganisms includes bacteria, viruses, fungi, and protozoa. Beneficially engaging the body by aiding digestion, regulating the immune system, and safeguarding it against harmful pathogens. The regulation of microbiota is essential for health. However, dysbiosis disrupts the community balance. It can lead to various diseases such as inflammatory bowel disease (IBD), obesity, diabetes, and cancer (Acevedo-Román et al., 2024).
Colorectal cancer (CRC) ranks highly among the causes of morbidity and mortality linked to cancer globally and is a public health issue (Morgan et al., 2023). Globally, CRC accounts for approximately 10% of all cancer cases, with an estimated 1.9 million new cases and 935,000 deaths in 20202. CRC is the third most diagnosed cancer worldwide, with significant regional variations in incidence rates. The occurrence and fatality rates of CRC differ among regions and demographics owing to factors such as dietary habits, lifestyle choices, genetic predisposition, and environmental influences. For instance, high-income countries report higher CRC rates compared to low-income regions, largely due to differences in dietary habits, lifestyle, and access to screening (Sawicki et al., 2021; Morgan et al., 2023). Risk factors such as obesity, smoking, and genetic predisposition further contribute to the global burden of CRC (Conti et al., 2020). With the progress in screening methods and therapies, CRC remains a health challenge that calls for a more profound comprehension of its causes and the creation of new treatment approaches (Conti et al., 2020; Sawicki et al., 2021).
New studies have focused on the role of gut microbiota in causing CRC (Wong and Yu 2019). Recent findings indicate that an imbalance in gut microbes may play a role in the development of cancer by triggering inflammation and creating harmful substances that can lead to cancer (Rebersek 2021). Specifically, identified strains such as Fusobacterium nucleatum have been linked to the risk of CRC (Kasai et al., 2016; Young et al., 2021). This emphasizes the role of microbiota in cancer progression. Studies indicate that Fusobacterium nucleatum is found more frequently in cancer tissues than in healthy tissues. This connection has been linked to the stages of the disease and worse outcomes for patients (Ou et al., 2022; Cao et al., 2024).
Growing evidence on the impact of microbiota on CRC treatment is becoming increasingly apparent. Research has shown that the gut microbiota can influence the effectiveness and side effects of chemotherapy and immunotherapies, which opens up possibilities for interventions based on microbiota (Zhao et al., 2023). Microbiota-based interventions, such as FMT, have shown promising results, with some studies reporting a 30-40% improvement in treatment outcomes when combined with chemotherapy11.
In addition, the intricate connection between gut bacteria and CRC has revealed the possibility of identifying and predicting disease progression. Scientists are currently investigating these patterns as indicators of early CRC spotting and are tracking their development. These indicators offer information about the disease stage and probable outcome, thus assisting in tailoring treatment strategies (Liu et al., 2023). For instance, distinct microbial configurations have been associated with the cancer stage, presenting a non-invasive approach for evaluating illness (Liu et al., 2023).
The intricate connection between microbiota and CRC underscores the significance of research and cooperation in this area. As we expand our understanding of the subject matter, it is vital to consider the differences and external factors that affect the microbiota composition. Future research will involve creating tailored therapies based on microbiota and incorporating examinations into CRC care. By utilizing the knowledge of the microbiota, we have the potential to unveil approaches to prevent the detection and treatment of cancer, which could lead to better patient outcomes and advancements in public health overall (Shi et al., 2024; Tito et al., 2024). This review aims to comprehensively examine the role of gut microbiota in CRC pathogenesis, diagnosis, and treatment, with a focus on microbial-based interventions such as probiotics, prebiotics, and FMT.
2. Methodologies used in microbiota studies
Studies included in this review were selected based on the following criteria: (1) focus on gut microbiota and CRC, (2) use of advanced sequencing technologies (e.g., 16S rRNA, metagenomics), and (3) availability of clinical or experimental data. Excluded studies were those that did not provide clear methodologies or lacked relevance to CRC pathogenesis or treatment. Knowledge of the methods utilized to study microbiota plays a role in comprehending the outcomes and their significance in cancer. Different methods have been used to examine gut microbiota, including the strengths and weaknesses of each approach.
The 16S rRNA sequencing method focuses on the 16S rRNA gene found in bacteria that have regions with variations that enable the identification of different bacterial species. The popularity of this technique stems from its cost efficiency and its ability to examine diversity in samples. Nevertheless, its drawback lies in its limited ability to distinguish closely related species and to provide insights into the functions of the microbiota (Johnson et al., 2019).
Metagenomics differs from 16S rRNA sequencing in that it entails sequencing all DNA in a sample to detect an array of microorganisms, such as bacteria and viruses, as well as fungi and protozoa (Yachida et al., 2019). This method provides a glimpse into the capabilities of microbiota by identifying genes associated with metabolic processes. Although metagenomics provides a thorough scope for analysis, it is expensive and requires bioinformatics software for data interpretation (Qureshi et al., 2023).
Metatranscriptomics involves sequencing RNA transcripts from samples to reveal details of metabolic processes and gene expression profiles within the microbial community. This method aids in understanding the behavior of the microbiota and their responses to environmental conditions. Nevertheless, metatranscriptomics is intricate, expensive, and requires top-notch RNA samples (Ojala et al., 2023).
Metabolomics focuses on examining the metabolites generated by microbiota to gain insights into processes in the gut environment. This approach can identify the metabolites associated with CRC development and offer a practical assessment of microbial functions. Nevertheless, it relies on tools such as mass spectrometry and is affected by diverse external elements, posing challenges for standardization (Terrón-Camero et al., 2022; Kong et al., 2023).
Each of these methods has its advantages and drawbacks to consider. 16S rRNA sequencing works well for exploring diversity at first glance; however, metagenomics and metatranscriptomics delve deeper into the genetic and functional capabilities of the microbial community. Metabolomics offers proof of activity but demands precise sample management and sophisticated analytical methods. When these techniques are combined strategically, a comprehensive understanding of the significance of the microbiota in CRC has emerged (Table 1).
Method | Strengths | Limitations |
---|---|---|
16S rRNA Sequencing | Cost-effective; good for assessing microbial diversity | Limited resolution for closely related species; no functional insights |
Metagenomics | Provides comprehensive genetic information; identifies functional potential | Expensive; requires advanced bioinformatics tools |
Metatranscriptomics | Reveals gene expression profiles; insights into microbial activity | Complex; requires high-quality RNA samples |
Metabolomics | Identifies metabolic by-products; links microbiota to CRC progression | Sensitive to external factors, requires precise sample handling |
3. The microbiota and CRC pathogenesis
Disrupted balance of microbes in the gut microbiota, known as dysbiosis, is now widely acknowledged as a player in the development of cancer. Dysbiosis is characterized by a significant shift in the Firmicutes to Bacteroidetes ratio (often <1.5:1) and a reduction in microbial diversity (Shannon index <3.0), accompanied by decreased short-chain fatty acid (SCFA) production (<5 mM) (Johnson et al., 2019; Kang et al., 2023). This disturbance can occur because of factors such as diet choice and antimicrobial use, as changes in lifestyle habits can promote the proliferation of harmful bacteria while reducing the diversity of beneficial microbes. Such an imbalance may lead to inflammation of the gut, a recognized risk factor for CRC (Zhao et al., 2023). Chronic inflammation promotes cancer growth by promoting cell growth, preventing cell death, and facilitating changes. Additionally, the inflamed environment leads to levels of oxygen and nitrogen compounds that can damage DNA and escalate cancer formation of cancer (Kim et al., 2023).
Certain types of bacteria are associated with a heightened risk of cancer. For example, Fusobacterium nucleatum and Bacteroides fragilis are frequently associated with CRC progression (Kasai et al., 2016; Young et al., 2021). One noteworthy example is Fusobacterium nucleatum, which has attracted interest for its association with CRC. This bacterium is believed to play a role in cancer development via various mechanisms. Fusobacterium nucleatum can attach to infiltrating cells and influence tumor formation by altering the immune response of the host. It triggers the release of substances and chemicals that can worsen inflammation and foster an environment conducive to tumor growth. Moreover, Fusobacterium nucleatum can suppress the function of killer cells and other immune cells. This allows cancer cells to evade detection by the system and keep growing unchecked (Ranjbar et al., 2021; Kim et al., 2023; Tong et al., 2023; Martin-Gallausiaux et al., 2024). While some studies suggest that Fusobacterium nucleatum is a key driver of CRC, others have reported conflicting findings, indicating that its role may vary depending on the patient’s genetic background and microbial community structure (Chen et al., 2020; Ranjbar et al., 2021).
Microbial by-products are essential for regulating inflammation, DNA integrity, and the growth of cancer cells. SCFAs such as butyrate are produced at levels of 10-20 mM in the colon, contributing to anti-inflammatory and anti-carcinogenic effects (Kang et al., 2023). When there is an imbalance in the gut flora, harmful substances such as bile acids and hydrogen sulfide may be produced, leading to increased inflammation and DNA damage (Shin et al., 2023). Metabolites have the potential to disturb balance within cells and contribute to changes that support the growth of cancer cells. The intricate relationship between metabolites and cells underscores the influence of gut bacteria on CRC development (Fig. 1), indicating a promising avenue for addressing these relationships through treatment approaches (Libiad et al., 2019; Kim 2021; Shin et al., 2023). In CRC patients, butyrate levels drop significantly to <5 mM, while secondary bile acids such as deoxycholic acid (DCA) increase from <1 μM in healthy individuals to >10 μM in CRC patients (Campbell et al., 2020). These changes occur progressively over weeks to months, correlating with disease progression (Sadler et al., 2020).

- Microbiota in CRC Pathogenesis. The gut microbiota plays a pivotal role in CRC initiation and progression through multiple mechanisms. Toxins and genotoxins produced by pathogenic bacteria induce DNA damage in intestinal epithelial cells (IECs), contributing to CRC development. Infections caused by harmful bacteria and their metabolic by-products activate tumor-associated myeloid cells (TAMCs), triggering inflammation that supports tumor growth. During inflammation, immune cells release ROS and reactive nitrogen species (RNS), leading to additional DNA damage and promoting CRC progression. Biofilms further contribute to CRC by activating interleukin-6 (IL-6) and its downstream effector, signal transducer and activator of transcription 3 (STAT3). Pathogenic bacteria and their virulence factors adhere to epithelial cells, promoting tumorigenesis. Key molecular players involved include STAT3 (a transcriptional activator), NF-κB (a transcription factor regulating DNA transcription), ROS and RNS (mediators of oxidative stress), cell adhesion molecules (CAMs), FadA (an adhesion factor from Fusobacterium), T cell immunoreceptor with Ig and ITIM domains (TIGIT, an immune regulatory molecule), and PCWBR22 (a protein repeat potentially involved in cell wall interactions).
3.1 Mechanism of microbiome on CRC
Gut microbial community affects CRC development. This influence is carried out through pathways, such as creating by-products and toxin secretion, influencing the immune system, and forming biofilms in the gut lining layers. Knowledge of these processes is essential for designing treatments to reduce CRC progression of CRC (Yang et al., 2020; Kang et al., 2023).
3.1.1 Bacterial metabolites
Metabolites produced by bacteria are crucial for the onset of CRC and affect its growth and progression. SCFAs, such as butyrate and acetate, generated from fibers by intestinal bacteria, play a vital role in gut health maintenance (Joshi and Durden 2019; Kang et al., 2023). Butyrate is specifically recognized for its ability to reduce inflammation and combat growth. This substance acts as an energy source for colon cells. Supports maintenance of the intestinal barrier and triggers cell death in CRC cells. It also hampers cell growth to shield against cancer and aids in managing the systemic response to lower persistent inflammation, which is a notable contributor to CRC risk. Nevertheless, a decline in bacteria that produce these substances due to an imbalance can weaken these defense mechanisms and enhance CRC progression (Joshi and Durden 2019; Liang et al., 2022).
In addition, microorganisms that transform bile acids produce secondary bile acids, which are associated with the promotion of CRC. These by-products, such as DCA and lithocholic acid (LCA), can cause DNA damage, oxidative stress, and inflammation, thereby forming a setting ripe for cancer development (Qin et al., 2024). An uptick in these bile acids can stem from a fat diet, which is associated with an imbalance in the gut flora and heightened generation of by-products. Maintaining a balance between SCFAs and harmful secondary bile acids is vital for colon health. The use of methods to boost SCFA production through adjustments in fibers and prebiotics can play a key role in preserving this equilibrium (Pandey and Umar 2021). Supplementation with SCFA-producing bacteria such as Lactobacillus and Bifidobacterium can also prove this. Recognizing the dual functions of bacterial by-products in CRC highlights the significance of managing the gut microbiota for cancer prevention and treatment, enabling promising strategies to reduce the risk of CRC and enhance patient outcomes (Pradeep Prasanna and Charalampopoulos 2019).
3.1.2 Bacterial toxins
The involvement of toxins is crucial in the development of CRC, as they cause direct damage to DNA and trigger inflammation, while also affecting the body’s immune response. Colibactin, a bacterial toxin linked to CRC, is produced by Escherichia coli. Colibactin acts as a genotoxin that induces DNA damage by breaking DNA strands and causing mutations and instability in the genome, which are important factors in the development of cancer. E. coli strains that produce colibactin are commonly found in the colon lining of CRC patients with CRC highlighting their significance in initiating and advancing tumors (Xue et al., 2019; Pleguezuelos-Manzano et al., 2020). The presence of Bacteroides fragilis can also contribute to this situation by generating Bacteroides fragilis toxin (BFT), which disrupts the barriers in the gut lining and triggers inflammation while encouraging cell growth. These actions of BFT involve breaking down the cadherin protein to activate the β-catenin signaling pathway, which is essential for the development of CRC. Repeated exposure to BFT over time leads to an environment that promotes inflammation, which in turn, facilitates the development of tumors (Wu et al., 2006; Purcell et al., 2022).
Another important bacterial toxin linked to cancer is lipopolysaccharide (LPS), which is found in the membranes of gram-negative bacteria. LPS can induce reactions by interacting with toll-like receptor-4 (TLR4) on immune cells and triggering the production of inflammatory cytokines. Prolonged inflammation caused by LPS exposure creates an environment that is conducive to cancer cell proliferation and survival. Ongoing inflammation triggers DNA damage and cell growth, which worsen the development of CRC (De Waal et al., 2020; Rajamanickam et al., 2020). The significance of poisons in cancer underscores the importance of tailored treatments and preventive measures that target microbial imbalances. Their cancer-causing effects on the health of the digestive system can be enhanced by decreasing the number of bacteria that produce toxins, employing antibiotic probiotics, or altering dietary habits (Shamekhi et al., 2020; Aleman et al., 2023).
The role of toxins in the development and progression of CRC is significant, involving a multifaceted mechanism that includes DNA damage initiation and progression, as well as chronic inflammation and immune modulation as key factors. Colibactin is a critical component in this process. Genotoxins synthesized by strains of Escherichia coli (E. coli) can disrupt DNA strands, causing mutations and genomic instability. Both of these pivotal aspects drive the growth of cancer cells (Dziubańska-Kusibab et al., 2020). In healthy individuals, the gut microbiota typically maintains a Firmicutes to Bacteroidetes ratio of approximately 2:1, with a Shannon diversity index ranging from 3.5 to 5.0 (Johnson et al., 2019). Recent research has revealed that patients’ colon linings often contain large quantities of E. coli strains that produce colibactin, and CRC highlights their crucial role in the development and progression of tumors in the colon area (Chen et al., 2022). In addition, BFT, produced by Bacteroides fragilis bacteria, directly causes CRC by damaging the gut lining and causing inflammation by activating the β-catenin signaling pathway. These harmful substances cause an imbalance in gut microbiota equilibrium, pushing it towards an environment that supports cancer growth (Haghi et al., 2019; Winston and Theriot 2020).
Furthermore, the membranes of gram-negative bacteria contain LPS, which drives chronic inflammation and aids in CRC progression. LPS interacts with TLR4 in cells and triggers the ongoing release of inflammatory cytokines (Gu et al., 2019). Long-term LPS-triggered inflammation exacerbates DNA damage. This facilitates the survival and proliferation of cancer cells, thereby creating a microenvironment that promotes cancer development. Exploring the functions of these toxins provides opportunities for developing treatments that could help lessen their negative impacts, perhaps by focusing on the bacteria themselves or by adjusting the inflammatory reactions they incite (Elkrief et al., 2019).
3.2 Immune modulation
The ability of the gut microbiota to influence CRC development is crucial as it affects both tumor advancement and the body’s immune response. Harmful bacteria can alter the system to create an environment conducive to tumor growth. For example, certain types of Fusobacterium nucleatum can hinder the function of killer cells (NK0 cells and T-cells), weakening the body’s capacity to identify and eradicate cancer cells. This mechanism of evading the immune system enables cancer cells to grow uncontrollably and plays a role in tumor formation (Chavakis et al., 2019; Parhi et al., 2020; Serna et al., 2020). Moreover, long-term inflammation caused by infection and imbalance in gut bacteria can result in the production of inflammatory proteins such as IL-6, TNF alpha, and IL-1beta. These substances, which cause inflammation, promote DNA damage, cell growth, and blood vessel development, thereby establishing an environment conducive to the advancement of cancer (Fields et al., 2019).
However, friendly bacteria in the gut can enhance the ability of this system to combat cancer. This shows how gut microbes play a role in regulating the system. Some probiotics and friendly bacteria can trigger the production of inflammatory signaling molecules such as IL-10 and TGF beta (Mirlekar 2022). This helps maintain balance and reduce inflammation. These beneficial bacteria can also increase the performance of killer T and natural killer cells. This enhances the effectiveness of the system in locating and destroying cells. Methods that target gut bacteria to promote microbes in the body can increase the effectiveness of immunotherapy in CRC patients. Techniques such as probiotics and prebiotics, along with FMT, rebalance the microbiota to enhance the body’s immunity against tumors and enhance treatment results (Fig. 2). Gut bacteria can provide opportunities for preventing and treating CRC effectively (Vendrik et al., 2020; Wang et al., 2021).

- Therapeutic Approaches for CRC Management (a) Probiotics aid in CRC prevention and treatment through three key mechanisms. First, they strengthen gut barrier integrity by enhancing mucin production, upregulating tight junction proteins (TJPs), and promoting epithelial repair. Second, they modulate immune responses by activating dendritic cells (DCs), reducing T helper 17 (Th17) cells, increasing regulatory T (Treg) cells, and polarizing macrophages toward the anti-inflammatory M2 phenotype, thereby mitigating colonic inflammation. Third, probiotics provide colonization resistance by inhibiting pathogenic bacteria through antimicrobial peptide (AMP) secretion, lowering luminal pH, and directly interacting with pathogens. Additionally, FMT restores healthy gut microbiota and improves gut barrier function. (b) Prebiotics support CRC management by selectively promoting the growth and activity of beneficial probiotic bacteria, facilitating fermentation processes, preventing pathogen colonization, and being absorbed into the gut to exert anti-inflammatory effects. Conversely, postbiotics may specifically induce cytotoxic effects on tumor cells while protecting intestinal epithelial cells by preventing apoptosis of healthy cells.
3.3 Biofilm formation
Biofilm creation involves the process by which bacteria attach to a surface, generate substances outside the cells that surround the group, and establish a setting. This process plays a role in cancer formation by encouraging long-term inflammation and shielding bacteria from immune responses while bolstering bacterial survival (Zhao et al., 2023). For example, bacteria adhere to the mucosal surface of the colon via adhesion proteins and hair-like structures. Following their growth phase and subsequent clustering into microcolonies, bacteria establish the foundation for biofilm formation. With the time and maturity of the biofilm structure, bacteria release substances- extracellular polymeric substance (EPS) comprising various compounds such as polysaccharides, proteins, lipids, and DNA outside the cell membrane (Chiba et al., 2022). This intricate matrix protects the community by shielding it from responses and antibiotic attacks. A stable environment within the biofilm facilitates the survival of bacteria as they manage to evade host defenses, leading to prolonged inflammation. Sustained inflammatory reactions trigger the production of inflammatory agents and reactive oxygen species (ROS), which can cause DNA damage and potentially support processes that lead to carcinogenesis (Hart et al., 2019). In CRC, biofilms formed by Fusobacterium nucleatum and Escherichia coli create a protective extracellular matrix that shields bacteria from immune responses and antibiotics. This biofilm-mediated protection leads to chronic inflammation, DNA damage, and tumorigenesis (Barzegari et al., 2020; Tuck et al., 2022). Disrupting biofilm formation through targeted therapies, such as probiotics and biofilm-degrading enzymes, could offer a novel approach to CRC treatment (Tytgat et al., 2019).
Bacteria that form biofilms tend to become more harmful and resistant to antibiotics, which can create an environment conducive to tumor growth in CRC. The protective EPS matrix shields bacteria from attacks, leading to prolonged inflammation and tumor formation (Barzegari et al., 2020). Additionally, biofilm presence can change the balance of gut bacteria by favoring the growth of species such as Fusobacterium nucleatum and Escherichia coli, upsetting microbiome harmony, and supporting CRC development. Furthermore, bacteria that form biofilms can create genotoxins such as colibactin, which directly damage the DNA of cells in the colon and result in mutations and instability. Managing biofilm production in cancer involves approaches to disrupt the biofilm structure and improve the elimination of bacteria, including biofilm treatment, probiotics, prebiotics, and targeted medical interventions. Learning about and targeting the processes involved in biofilm formation can lead to the development of more successful preventive and treatment measures against CRC (Tytgat et al., 2019; Tuck et al., 2022).
Bacteria in the gut play a role in how the immune system can identify and combat cancer cells. Some bacteria even help cancer evade the immune system detection process, which is a key factor in cancer development and spread (Blachier et al., 2021). For example, certain types of Fusobacterium nucleatum are known to effectively prevent natural defenses of the body, such as NK cells and T cells, from attacking tumor cells. By weakening this response, cancer cells can thrive and easily form tumors. Persistent inflammation over time is frequently caused by an imbalanced gut microbiota and results in the release of cytokines like IL-6, TNF-Α, and IL-1Β. These cytokines not only contribute to DNA damage but also stimulate the formation of new blood vessels and tumor cell growth. This indicates that focusing on bacterial regulatory impacts could lead to new approaches to enhance the immune ability of the body to combat cancer naturally (Mann and Maughan 2020).
Structured microbial communities known as biofilms play an important role in the development of CRC. They prolong inflammation and protect bacteria from the immune system and antibiotics in the gut, such as Fusobacterium nucleatum and E. coli. This allows these bacteria to grow and causes inflammation to worsen over time. As they mature, biofilms produce substances, such as EPS, which form a protective matrix that aids in creating a cancer-promoting environment (Singh et al., 2019). The presence of biofilms in the gut has been linked to DNA damage levels, as they generate genotoxins such as colibactin, which accelerates the mutation of colon cells. Innovative treatment approaches may concentrate on dismantling biofilm formation to make bacteria more susceptible to responses and antibiotic therapies, potentially halting the progression of CRC (Wilson et al., 2019).
4. Microbiota as modifiers of CRC risk factors
Dietary and lifestyle choices, including antimicrobial use, significantly influence the composition and function of gut bacteria, thereby modulating CRC risk (Dudek-Wicher et al., 2018). Our diet, lifestyle habits, and antimicrobial use significantly influence the composition and function of the bacteria in our guts. High-fat diets increase the abundance of bile acid-producing bacteria by 2-3-fold, while fiber-rich diets enhance SCFA-producing bacteria by 1.5- 2-fold (Kiriyama and Nochi 2021). These dietary changes can alter the Firmicutes to Bacteroidetes ratio, with high-fat diets reducing it to <1.5:1 and fiber-rich diets maintaining it at >2:1 (Sánchez-Alcoholado et al., 2020). Consuming diets rich in processed meats and fats but lacking in fiber has been associated with an imbalance in gut bacteria, which can potentially spur the development of CRC. Such eating patterns can lead to an increase in bacteria that produce substances such as bile acids and hydrogen sulfide, which can trigger inflammation and the formation of cancerous cells (Dudek-Wicher et al., 2018; Chen et al., 2020; Kiriyama and Nochi 2021).
Recent studies have quantified the risk posed by eating red or processed meat and revealed that participants with the highest intake of red meat had a 30% increased risk of CRC, and those with the highest intake of processed meat had a 40% increased risk (Stern et al., 2024). Calcium from dairy products is thought to lower CRC risk by binding bile acids and free fatty acids in the colon, reducing their ability to damage epithelial cells. This action also indirectly supports a more favorable microbial environment by limiting substrates that harmful bacteria might otherwise use to produce carcinogenic metabolites. The Million Women Study, which tracked over 540,000 women for an average of 16 years, demonstrated that an additional 300 mg of calcium per day, roughly the calcium content of one cup of milk, is associated with a 17% reduction in CRC risk (Papier et al., 2025). This protective association was specific to most dairy sources (except for cheese and ice cream), underscoring the potential importance of calcium’s biochemical role in modulating both bile acid metabolism and microbial composition. Moderate to heavy alcohol consumption is associated with 1.2- to 1.5-fold increased risks of cancers of the colon and rectum compared with no alcohol consumption (Fedirko et al., 2011). Dietary fiber is fermented by colonic bacteria to produce short-chain fatty acids (SCFAs) such as butyrate. Butyrate supports colonocyte energy metabolism, helps maintain gut barrier integrity, and exerts anti-inflammatory effects. Fiber also shifts the microbial community toward beneficial bacteria. A meta-analysis reported that for every 10 g/day increase in dietary fiber intake, the risk of CRC decreased by about 10% (RR ≈ 0.90, 95% CI 0.85–0.95) (Aune et al., 2011). Diets that include plenty of fruit, vegetables, and whole grains help maintain a healthy microbiota that produces beneficial SCFAs known for their anti-inflammatory and anticarcinogenic effects. Factors such as activity and stress levels play a role in shaping the gut microbiota and can affect the risk of CRC (Sánchez-Alcoholado et al., 2020).
People with a family background of CRC or changes like those in the APC or MUTYH genes have an increased likelihood of developing the condition. For instance, certain types of bacteria can engage with pathways linked to CRC and affect tumor growth. Studies have revealed that bacteria in the gut can influence the expression of genes and epigenetic changes that can either promote or hinder cancer development in the body. It is crucial to understand how elements and the gut microbiota interact to develop customized approaches to prevent and treat CRC (Salvucci et al., 2022).
The gut microbiome is crucial for processing carcinogens and protective substances that influence the risk of cancer. Bacteria in the gut can transform nutrients from food and toxins into beneficial or harm substances (Wang et al., 2023). For example, certain gut bacteria can convert hydrocarbons (PAHs) from charred meat into compounds that can cause mutations, leading to DNA damage and cancer. Beneficial bacteria can break down fibers into SCFAs, protecting CRC by supporting the proper functioning of epithelial cells and reducing inflammation. Moreover, microbiota can affect the way chemopreventive compounds, such as polyphenols from fruits and vegetables, are metabolized, enhancing their ability to protect against CRC. The dynamic relationship between the gut microbiota and these metabolic processes highlights the importance of maintaining the microbiota to prevent CRC (Xu et al., 2023).
5. Microbiota in CRC diagnosis and prognosis
Microbial patterns are increasingly being recognized as indicators of detection and tracking. These patterns consist of levels linked to different stages of disease progression. Advancements in sequencing technologies have allowed researchers to identify microbiota compositions associated with CRC that can aid in detection efforts (Araujo et al., 2022). For example, increased levels of species such as Fusobacterium nucleatum and Bacteroides fragilis have been observed in patients compared to those who are not affected by the disease. The detection of these microorganisms in fecal or tissue samples can be used as an invasive method to detect CRC (Salvucci et al., 2022). This could potentially replace the need for procedures, such as a colonoscopy. Microbiota-based diagnostics, such as stool tests for Fusobacterium nucleatum, offer a non-invasive alternative to colonoscopy, with a sensitivity of 85% and specificity of 90% in early CRC detection (Salvucci et al., 2022). While colonoscopy remains the gold standard with a sensitivity of 95%, microbiota-based tests are more cost-effective (Rye et al., 2022).
In CRC, there is an increase in harmful bacterial levels along with a decline in beneficial bacterial populations within the gut flora. These alterations in the microbiota may mirror the influence of tumors on the gut environment and advance their growth (Wang et al., 2023). Furthermore, the variety and complexity of the gut community have been correlated with these results, where decreased microbial variety is usually linked to worse forecasts. These findings imply that microbial profiles may not only assist in identifying CRC but also offer predictive information that may influence treatment choices (Xu et al., 2023).
Microbiota-based diagnostics show promise for future healthcare applications, and it is essential to address these obstacles and constraints before they can be widely used in settings. One significant hurdle is the nature of the microbiota composition across individuals, which can be affected by factors such as diet, lifestyle, location, and genetics. This diversity makes it challenging to identify indicators of CRC (Araujo et al., 2022). Additionally, discrepancies in how samples are collected, processed, and analyzed can introduce inconsistencies in microbiota data, making it difficult to reliably replicate findings. Developing methods and creating procedures is crucial for converting microbiota studies into reliable diagnostic tools (Table 2). Furthermore, extensive validation research involving demographics is necessary to verify the practicality of microbiota-driven diagnostics and guarantee their efficacy across patient categories (Mo et al., 2020).
Parameter | Traditional screening methods | Gut microbiota-based diagnostics |
---|---|---|
Screening methods | Colonoscopy (COL), Fecal Occult Blood Test (FOBT), Fecal Immunochemical Test (FIT), Computed Tomography Colonography (CTC), Sigmoidoscopy (SIG) | Microbial Biomarker Analysis (MBA), Metagenomic Sequencing (mNGS), Quantitative Polymerase Chain Reaction-based Fecal Microbiota Profiling (qPCR-FMP) |
Prognostic value | Primarily based on tumor staging, genetic markers, and histopathology | Microbial shifts correlate with colorectal cancer (CRC) progression, offering potential for earlier prognostic insights |
Key biomarkers | KRAS, BRAF mutations, Microsatellite Instability (MSI), Carcinoembryonic Antigen (CEA), Cancer Antigen 19-9 (CA19-9) | Elevated levels of Fusobacterium nucleatum (F. nucleatum), Bacteroides fragilis (B. fragilis), Escherichia coli (E. coli), and dysbiosis patterns in gut microbiota |
Sensitivity & specificity | High sensitivity and specificity in COL; FOBT/FIT has lower sensitivity (∼70–80%) | Varies; microbial markers have promising specificity (∼80–90%) and sensitivity (∼75–85%), but require further validation |
Non-invasiveness | COL and SIG are invasive; FOBT/FIT are non-invasive | Entirely non-invasive, stool sample-based testing |
Early detection capability | Effective for early detection but depends on compliance (COL every 10 years, FIT annually) | Potential for early detection via microbial shifts before structural tumor changes occur |
Patient compliance | Moderate; COL has low compliance due to discomfort and preparation requirements | High; stool-based microbiota tests are easy to perform at home |
Turnaround time | COL: Immediate; FOBT/FIT: Few days | Microbial sequencing: 2–7 days |
Cost-effectiveness |
- COL: High cost (US $2,000– $3,000 per procedure) but one-time for 10 years - FOBT/FIT: Low cost (US $20–$50 per test, annually) - CTC: ∼ US $500–$1,500 |
- Microbiome sequencing: ∼ US $200–$500 per test - PCR-based fecal tests: ∼ US $50–$150 - Potential for lower costs with widespread implementation |
Limitations |
- Invasiveness (COL) - Risk of complications (bleeding, perforation) - Requires skilled personnel |
- Microbiome variability (diet, medications influence results) - Standardization and validation still in progress - Need for larger cohort studies |
Future potential | COL remains the gold standard, but non-invasive tests (e.g., FIT, stool DNA) are improving | Could complement or even replace traditional methods with further validation and cost reduction |
Gut microbiota-based diagnostics show promise as a non-invasive, cost-effective alternative for CRC screening. While colonoscopy remains the gold standard for accuracy, microbial biomarkers could improve early detection and prognosis, reducing the need for invasive procedures. Recently, there has been growing interest in using gut microbiota as a tool for diagnosing and predicting CRC. Identification of strains as indicators for early detection and disease advancement in CRC is becoming more prominent. Analysis of tissue samples from CRC patients has consistently shown the levels of Fusobacterium nucleatum and Bacteroides fragilis (Mo et al., 2020). This non-invasive approach could offer a means to detect CRC compared to screening methods, such as colonoscopies, suggesting a more comfortable option that could increase screening participation rates. These distinct microorganism patterns possess the capability to revolutionize the detection and treatment of early CRC, thereby facilitating interventions at an earlier stage (Wu et al., 2021).
Recent research has shown that the types of microbes’ present vary throughout the CRC stages. Late-stage illnesses tend to involve bacteria, with fewer helpful bacterial communities in the gut microbiome than in the earlier stages of disease progression. This shift in balance at each stage not only aligns with the advancement of the disease but can also provide valuable clues regarding a patient’s prognosis (Quan et al., 2023). For instance, a decrease in diversity often indicates poor outcomes. The analysis of profiles not only aids in diagnosis but also directs treatment decisions and predicts survival rates. Nevertheless, implementing these findings in world settings poses difficulties because of the variations in gut bacterial composition among people, which are affected by dietary habits, genetics, way of life, and location. Maintaining uniformity in the collection and analysis of samples is crucial to tackle these obstacles and establish microbiota-driven diagnostics as a reliable tool for managing CRC (Cai et al., 2023).
6. Microbiota-based therapies
The exploration of microbiota-focused treatments in trials for CRC has become increasingly popular and promising for enhancing patient recovery (Table 3). These treatments, such as probiotics and prebiotics, along with fecal microbiota transplantation (FMT), aim to improve the effectiveness of CRC treatment and minimize its effects. Rigorous clinical studies have assessed the safety and success rate of these interventions (Ting et al., 2022). For example, research exploring the efficacy of different types has revealed encouraging findings in shaping the balance of gut bacteria, decreasing inflammation, and potentially hindering tumor development. Similarly, therapies involving prebiotics that support the proliferation of microbes are undergoing trials to assess their impact on improving gut well-being and outcomes related to cancer (Selvamani et al., 2022).
Type of clinical evidence | Population | Intervention | Key findings |
---|---|---|---|
Meta-analysis of RCTs (Randomized Controlled Trials) (Amitay et al., 2020) | 6,812 CRC patients | Probiotics/synbiotics (perioperative) |
Perioperative probiotics/synbiotics administration was associated with lower infection incidence (OR = 0.34, P < 0.001), lower diarrheal incidence (OR = 0.38, P < 0.001) Faster return to normal gut function (MD -0.66 days, P < 0.001), shorter postoperative antibiotics use (MD -0.64 days, P < 0.001), lower incidence of septicemia (OR = 0.31, P < 0.001), shorter length of hospital stay (MD -0.41 days, P = 0.110) |
Umbrella meta-analysis (Han et al., 2024) | 11518 CRC patients | Efficacy of probiotics on outcomes of CRC patients |
Probiotics administration resulted in a statistically significant reduction in the incidence of total infections (RR: 0.40, 95% CI: 0.31-0.51) Decreased risk of surgical site infections (RR: 0.56, 95% CI: 0.49-0.63), pneumonia (RR: 0.38, 95% CI: 0.30-0.48), urinary tract infections (RR: 0.44, 95%CI: 0.31-0.61), bacteremia (RR: 0.41, 95%CI: 0.30-0.56), and sepsis (RR: 0.35, 95% CI: 0.25-0.44) |
Prospective cohort study (Cui et al., 2023) | 45 CRC patients | Perioperative FMT + nutritional support |
Shorter hospital stays in FMT group (8.0±4.3 days vs. 11.2±5.4 days, t = 2.157, P = 0.037) Faster passage of flatus/bowel movement (2.2±3.2 days vs. 3.9±2.3 days, t = 2.072, P = 0.044) Lower incidence of abdominal distension (3/19 vs. 48% [11/23], P = 0.048 and diarrhea (3/19 vs. 52.2% [12/23], P = 0.023) |
Diagnostic validation clinical trial (Wong et al., 2017) | 309 CRC/advanced adenoma patients | Fusobacterium (Fn) + FIT (Fecal immunochemical test) |
Fn + FIT improved sensitivity (92.3% vs 73.1%, P < 0.001) Higher AUC (Area Under Curve) for CRC detection (0.95 vs. 0.86, P < 0.001) compared to FIT alone |
Phase II trial (Zhao et al., 2023) | 20 CRC patients | FMT + Tislelizumab + Fruquintinib |
Combined treatment showed 20% ORR (Objective Response Rate), 95% DCR (Disease Control Rate), and 60% CBR (Clinical Benefit Rate) Suggests FMT may enhance treatment efficacy |
Phase I trial (De Ana et al., 2024) | 14 CRC-MSS (Microsatellite-Stable CRC) patients | VE800 (11-strain Bacterial Consortium) + Nivolumab |
VE800 + Nivolumab was well-tolerated in CRC-MSS patients Safety: No dose-limiting toxicities ORR: 18% in the CRC-MSS cohort |
A ground-breaking method, known as FMT, is currently being studied for the treatment of CRC. The idea behind FMT involves transferring matters from donors to patients with CRC to help restore balanced and diverse gut microbiota. These initial trials have shown results in reducing tumor size and enhancing the efficacy of chemotherapy treatments (Chen et al., 2023). Further research is ongoing to validate these outcomes and to develop procedures for using FMT in the treatment of patients with CRC. Moreover, FMT has been studied as a treatment to improve the effectiveness of immunotherapy and reduce the side effects of treatment.
While certain studies show positive changes, results could exist; others point out issues such as differences in how individual microbiota react and difficulties in making treatments uniform. Real-life stories and personal accounts also demonstrate the advantages of therapies based on microbiota by highlighting cases in which patients saw responses, treatment, and fewer negative effects (Hao et al., 2021). Nonetheless, these treatments are still being researched, and further comprehensive clinical trials are required to confirm their efficacy and safety. Incorporating microbiota analysis into procedures shows the potential for tailoring CRC treatment to individuals; however, it requires addressing existing obstacles and guaranteeing consistent and reliable results among various patient groups (Chen et al., 2023).
Emerging treatments that focus on balancing gut microbiota show promise in improving CRC treatment outcomes and minimizing their effects. The use of probiotics and prebiotics is gaining interest because of their capacity to rebalance gut microbiota equilibrium, reduce inflammation, and support reactions (Sánchez-Alcoholado et al., 2020). Researchers have found that probiotics introduce bacteria into the gut, enhance the gut barrier function, promote immune response modulation, and hinder the proliferation of harmful bacteria. Prebiotics support the growth of bacteria in the gut and play a role in maintaining a healthy balance that promotes anti-inflammatory and anti-cancer properties within the body. Taking probiotics and prebiotics alongside therapies such as chemotherapy and immunotherapy could potentially improve their effectiveness by optimizing gut conditions (Dasari et al., 2020).
FMT, an intriguing advancement in microbiota-focused treatments, transfers individuals’ fecal matter to CRC patients to rebalance their microbial environment within the gut system (Rye et al., 2022). Early clinical studies indicated that FMT not only enhanced the diversity of gut microbiota but also improved the effectiveness of chemotherapy and decreased tumor size in patients. Furthermore, ongoing research is being conducted on using FMT to improve the efficiency of immunotherapies, positioning it as a method for treating CRC (Rye et al., 2022). There are still obstacles to overcome when establishing uniform FMT protocols and guaranteeing reliable outcomes in patients with different backgrounds and characteristics. We anticipate that as treatments continue to progress and develop in the field landscape, they will offer tailored approaches to care that improve CRC outcomes by considering the unique microbial composition of each patient (Rye et al., 2022).
7. Clinical applications of microbiota research in CRC
The integration of studies into medical applications has shown great potential for enhancing CRC treatment. In this section, we delve into methods for screening tools and treatment strategies inspired by research findings, in addition to personalized medicine advancements, including an overview of current clinical trials and their projected effects (Rye et al., 2022).
Microbial signs discovered in microbiota studies can be used as invasive indicators for CRC screening and early detection (Chen et al., 2021). One example is stool tests that check for bacteria such as Fusobacterium nucleatum and Bacteroides fragilis, which have shown promise as detection tools (Dasari et al., 2020). These microbiome indicators can support screening techniques such as colonoscopy and provide patients with an intrusive and convenient choice. By combining indicators with screening methods, the early detection rates of CRC can be enhanced. This improvement can result in an improved prognosis and increased survival rates (Abavisani et al., 2024).
Therapies using microbiota, such as probiotics and prebiotics, are being developed to enhance cancer treatment by influencing the gut microbiota. Probiotics and prebiotics work towards rejuvenating the gut environment by fostering the growth of bacteria that can help curb inflammation and hinder tumor development. FMT, a procedure in which fecal matter from individuals is transferred to patients with cancer, has the potential to restore microbial balance and improve treatment outcomes (Merrick et al., 2020; Rye et al., 2022). The effectiveness of these treatments is currently being studied in trials, along with therapies such as chemotherapy and immunotherapy.
Customizing care for CRC involves adapting treatment strategies based on the unique gut bacterial composition of an individual. Analysis of the microbiome can offer insights into how a patient may respond to treatments and potential adverse reactions (Chen et al., 2024; Kang et al., 2024). For instance. Individuals with varied and well-balanced gut microbiota may respond to immunotherapy. As a result, fewer side effects were observed. Developing personalized therapies based on gut bacteria involves combining data with information as well as details about diet and lifestyle choices. Enhancing treatment effectiveness while reducing negative impacts (Chen et al., 2022).
8. Critical insights
The gut microbiota plays an essential role in the development, diagnosis, and treatment of CRC. An imbalance in gut microbiota, known as dysbiosis, contributes to CRC progression through chronic inflammation, genotoxicity, immune modulation, and metabolic alterations. Specific bacterial strains, including Fusobacterium nucleatum, Bacteroides fragilis, and certain Escherichia coli strains, have been implicated in CRC due to their inflammatory properties, ability to cause DNA damage, and disruption of immune responses. These bacteria interact with host cells and the immune system, creating a tumor-friendly environment that facilitates disease progression.
Dysbiosis affects CRC development through various mechanisms. Some bacteria, such as Escherichia coli, produce colibactin, a genotoxin that causes DNA damage, leading to genomic instability and cancerous mutations. Fusobacterium nucleatum promotes CRC by suppressing immune surveillance and increasing inflammation, thereby helping tumors evade immune detection. Additionally, microbial metabolites play a dual role in CRC progression; while SCFAs like butyrate have protective effects, other metabolic by-products, such as secondary bile acids and hydrogen sulfide, contribute to DNA damage and tumor growth. Biofilm formation by certain bacteria further exacerbates the problem by creating a chronic inflammatory microenvironment that promotes bacterial persistence and resistance to treatment.
The composition of gut microbiota is increasingly being studied as a potential diagnostic and prognostic tool for CRC. Microbial biomarkers, including elevated levels of Fusobacterium nucleatum, Bacteroides fragilis, and Escherichia coli, have been detected in fecal and tissue samples of CRC patients, offering a non-invasive alternative to traditional screening methods. CRC progression is often associated with a decline in microbial diversity and an enrichment of pathogenic bacteria, which could serve as prognostic indicators. Furthermore, gut microbiota influences treatment outcomes, as patients with a diverse and balanced microbiome tend to respond better to immunotherapy, chemotherapy, and other targeted therapies, while those with dysbiosis may experience reduced efficacy and increased adverse effects. Given the influence of microbiota on CRC, therapeutic interventions targeting microbial balance are being explored to complement conventional treatments. Probiotics, such as Lactobacillus and Bifidobacterium, help restore gut equilibrium, reduce inflammation, and improve gut barrier integrity. Prebiotics, which serve as nutrients for beneficial bacteria, encourage the growth of protective microbial communities. FMT is emerging as a promising intervention, restoring microbial diversity and enhancing chemotherapy efficacy. Additional strategies, including dietary interventions that promote high-fiber intake, targeting pathogenic bacteria, and disrupting biofilms, are being investigated to improve CRC treatment outcomes. Addressing gut microbiota imbalances offers a promising avenue for CRC prevention, early detection, and personalized treatment strategies.
9. Challenges and future directions
The vast possibilities of using microbiota for various purposes are exciting; however, there are challenges to overcome, such as differences in microbiota composition between individuals and the necessity of established protocols. Current limitations in microbiota research include technical challenges in sample collection and analysis, lack of standardized protocols, high costs associated with advanced sequencing technologies, and implementation barriers in clinical settings. For instance, metagenomic sequencing can cost up to US $1,000 per sample, and the lack of standardized protocols leads to variability in results (Abavisani et al., 2024). Future research should prioritize the development of microbiota-based biomarkers, standardization of protocols, design of large-scale clinical trials, and integration of artificial intelligence for data analysis. Additionally, there is a need for cost-effective technologies to make microbiota-based diagnostics and therapies accessible globally (Ting et al., 2022; Abavisani et al., 2024). Collaboration across disciplines will play a role in translating microbiota research into practical clinical applications and revolutionizing the management of CRC.
10. Conclusion
In conclusion, the gut microbiota plays a pivotal role in CRC pathogenesis, offering new avenues for prevention, diagnosis, and treatment. Future research should focus on standardizing microbiota analysis methods, validating microbial biomarkers in diverse populations, and integrating microbiota-based therapies into clinical practice. Collaborative efforts across disciplines will be essential to translate these findings into effective, personalized CRC treatments, ultimately improving patient outcomes and reducing the global burden of CRC.
Utilizing our understanding of microbiota presents an opportunity to enhance cancer CRC outcomes. Researchers are investigating patterns as non-invasive indicators for early CRC detection and prognosis as alternatives to conventional screening methods. Moreover, microbial-based treatments, such as probiotics and FMT, have the potential to boost treatment effectiveness and minimize the effects. These approaches can be customized for each patient according to their microbiota profile, resulting in personalized and efficient treatment plans. To realize the benefits of microbiota-based treatments for cancer (CRC), ongoing research and partnerships are vital requirements. Creating methods for microbiota studies will guarantee repeatable results, thereby easing the transition from research outcomes to practical clinical applications. Extensive verification studies across demographics are necessary to validate the effectiveness of microbiota-centered diagnosis and treatment options. Furthermore, cross-disciplinary teamwork involving scientists, medical practitioners, and data analysts is key to incorporating analysis into the care of CRC patients.
Microbiomes present a territory for the research and treatment of CRC because delving deeper into the connection between the microbiome and CRC and creating interventions can enhance the prevention, diagnosis, and treatment of CRC. Moving forward, this area offers prospects for progress in health and patient outcomes. Collaboration and standardization within this field are crucial for unlocking the full potential of microbiota-driven treatments, which could fundamentally change how CRC leads to patient outcomes.
CRediT authorship contribution statement
Yahia El-Tanani: Literature search, data analysis, figure preparation, manuscript review, Mohamed El-Tanani: Concept, design, literature search, data analysis, manuscript preparation, Syed Arman Rabbani: Literature search, data analysis, figure preparation, manuscript review, Shakta Mani Satyam: Concept, design, literature search, data analysis, manuscript preparation, manuscript editing, manuscript review, Adil Farooq Wali: Literature search, data analysis, figure preparation, manuscript review, Alaa A. A. Aljabali: Literature search, data analysis, figure preparation, manuscript review, Manfredi Rizzo: Literature search, data analysis, figure preparation, manuscript review. All authors have read and agreed to the published version of the manuscript.
Declaration of competing interest
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
Data availability statement
The data generated and analyzed in this study are included in this article.
Declaration of Generative AI and AI-assisted technologies in the writing process
The authors confirm that there was no use of Artificial Intelligence (AI)-Assisted Technology for assisting in the writing or editing of the manuscript and no images were manipulated using AI.
References
- Deciphering the gut microbiome: The revolution of artificial intelligence in microbiota analysis and intervention. Current Research in Biotechnology. 2024;7:100211. https://doi.org/10.1016/j.crbiot.2024.100211
- [Google Scholar]
- Insights into gut dysbiosis: Inflammatory diseases, obesity, and restoration approaches. Int J Mol Sci. 2024;25:9715. https://doi.org/10.3390/ijms25179715
- [Google Scholar]
- Leaky gut and the ingredients that help treat it: A review. Molecules. 2023;28:619. https://doi.org/10.3390/molecules28020619
- [Google Scholar]
- Probiotic/Synbiotic Treatment and postoperative complications in colorectal cancer patients: Systematic review and meta-analysis of randomized controlled trials. Clin Transl Gastroenterol. 2020;11:e00268. https://doi.org/10.14309/ctg.0000000000000268
- [Google Scholar]
- Microbiota Modulation in patients with metabolic syndrome. Nutrients. 2022;14:4490. https://doi.org/10.3390/nu14214490
- [Google Scholar]
- Dietary fibre, whole grains, and risk of colorectal cancer: Systematic review and dose-response meta-analysis of prospective studies. BMJ. 2011;343:d6617. https://doi.org/10.1136/bmj.d6617
- [Google Scholar]
- The battle of probiotics and their derivatives against biofilms. Infect Drug Resist. 2020;13:659-672. https://doi.org/10.2147/IDR.S232982
- [Google Scholar]
- Production of hydrogen sulfide by the intestinal microbiota and epithelial cells and consequences for the colonic and rectal mucosa. Am J Physiol Gastrointest Liver Physiol. 2021;320:G125-G135. https://doi.org/10.1152/ajpgi.00261.2020
- [Google Scholar]
- Tumor bacterial markers diagnose the initiation and four stages of colorectal cancer. Front Cell Infect Microbiol. 2023;13:1123544. https://doi.org/10.3389/fcimb.2023.1123544
- [Google Scholar]
- Bacterial metabolism of bile acids promotes generation of peripheral regulatory T cells. Nature. 2020;581:475-479. https://doi.org/10.1038/s41586-020-2193-0
- [Google Scholar]
- Host-gut microbiota metabolic interactions and their role in precision diagnosis and treatment of gastrointestinal cancers. Pharmacol Res. 2024;207:107321. https://doi.org/10.1016/j.phrs.2024.107321
- [Google Scholar]
- Hematopoietic progenitor cells as integrative hubs for adaptation to and fine-tuning of inflammation. Nat Immunol. 2019;20:802-811. https://doi.org/10.1038/s41590-019-0402-5
- [Google Scholar]
- Clostridium butyricum, a butyrate-producing probiotic, inhibits intestinal tumor development through modulating Wnt signaling and gut microbiota. Cancer Lett. 2020;469:456-467. https://doi.org/10.1016/j.canlet.2019.11.019
- [Google Scholar]
- A commensal-encoded genotoxin drives restriction of Vibrio cholerae colonization and host gut microbiome remodeling. Proc Natl Acad Sci U S A. 2022;119:e2121180119. https://doi.org/10.1073/pnas.2121180119
- [Google Scholar]
- Fecal microbiota transplantation ameliorates type 2 diabetes via metabolic remodeling of the gut microbiota in db/db mice. BMJ Open Diabetes Res Care. 2023;11:e003282. https://doi.org/10.1136/bmjdrc-2022-003282
- [Google Scholar]
- Therapeutic approaches to colorectal cancer via strategies based on modulation of gut microbiota. Front Microbiol. 2022;13:945533. https://doi.org/10.3389/fmicb.2022.945533
- [Google Scholar]
- An altered fecal microbial profiling in rosacea patients compared to matched controls. J Formos Med Assoc. 2021;120:256-264. https://doi.org/10.1016/j.jfma.2020.04.034
- [Google Scholar]
- Resistant starch and the gut microbiome: Exploring beneficial interactions and dietary impacts. Food Chem X. 2024;21:101118. https://doi.org/10.1016/j.fochx.2024.101118
- [Google Scholar]
- Staphylococcus aureus utilizes environmental RNA as a building material in specific polysaccharide-dependent biofilms. NPJ Biofilms Microbiomes. 2022;8:17. https://doi.org/10.1038/s41522-022-00278-z
- [Google Scholar]
- Revisiting the impact of lifestyle on colorectal cancer risk in a gender perspective. Crit Rev Oncol Hematol. 2020;145:102834. https://doi.org/10.1016/j.critrevonc.2019.102834
- [Google Scholar]
- Analysis of short-term efficacy of perioperative fecal microbiota transplantation combined with nutritional support in patients with radiation-induced enteritis complicated by intestinal obstruction. Zhonghua wei Chang wai ke za zhi= Chinese Journal of Gastrointestinal Surgery.. 2023;26:955-962. https://doi.org/10.3760/cma.j.cn441530-20230816-00052
- [Google Scholar]
- ctDNA applications and integration in colorectal cancer: An NCI colon and rectal–anal task forces whitepaper. Nat Rev Clin Oncol. 2020;17:757-770. https://doi.org/10.1038/s41571-020-0392-0
- [Google Scholar]
- 607 CONSORTIUM-IO: a phase 1 study evaluating a combination of an 11-strain bacterial consortium (VE800) and nivolumab in treatment of select refractory or metastatic cancers. BMJ Specialist Journals. 2024
- [Google Scholar]
- Colorectal cancer is associated with increased circulating lipopolysaccharide, inflammation and hypercoagulability. Sci Rep. 2020;10:8777. https://doi.org/10.1038/s41598-020-65324-2
- [Google Scholar]
- The influence of antibiotics and dietary components on gut microbiota. Prz Gastroenterol. 2018;13:85-92. https://doi.org/10.5114/pg.2018.76005
- [Google Scholar]
- Colibactin DNA-damage signature indicates mutational impact in colorectal cancer. Nat Med. 2020;26:1063-1069. https://doi.org/10.1038/s41591-020-0908-2
- [Google Scholar]
- The intimate relationship between gut microbiota and cancer immunotherapy. Gut Microbes. 2019;10:424-428. https://doi.org/10.1080/19490976.2018.1527167
- [Google Scholar]
- Alcohol drinking and colorectal cancer risk: An overall and dose–response meta-analysis of published studies. Ann Oncol. 2011;22:1958-1972. https://doi.org/10.1093/annonc/mdq653
- [Google Scholar]
- Structural basis of IL-1 family cytokine signaling. Front Immunol. 2019;10:1412. https://doi.org/10.3389/fimmu.2019.01412
- [Google Scholar]
- Toll-Like receptor 4 signaling licenses the Cytosolic transport of lipopolysaccharide from bacterial outer membrane vesicles. Shock. 2019;51:256-265. https://doi.org/10.1097/SHK.0000000000001129
- [Google Scholar]
- The association between fecal enterotoxigenic b. fragilis with colorectal cancer. BMC Cancer. 2019;19:879. https://doi.org/10.1186/s12885-019-6115-1
- [Google Scholar]
- Preventive effect of probiotics on infections following colorectal cancer surgery: An umbrella meta-analysis. World J Gastrointest Surg. 2024;16:3546-3558. https://doi.org/10.4240/wjgs.v16.i11.3546
- [Google Scholar]
- Xiaoyaosan Improves antibiotic-induced depressive-like and anxiety-like behavior in mice through modulating the gut microbiota and regulating the NLRP3 inflammasome in the colon. Front Pharmacol. 2021;12:619103. https://doi.org/10.3389/fphar.2021.619103
- [Google Scholar]
- Microrheology and spatial heterogeneity of staphylococcus aureus biofilms modulated by hydrodynamic shear and biofilm-degrading enzymes. Langmuir. 2019;35:3553-3561. https://doi.org/10.1021/acs.langmuir.8b04252
- [Google Scholar]
- Evaluation of 16S rRNA gene sequencing for species and strain-level microbiome analysis. Nat Commun. 2019;10:5029. https://doi.org/10.1038/s41467-019-13036-1
- [Google Scholar]
- Combinatorial approach to improve cancer immunotherapy: Rational drug design strategy to simultaneously hit multiple targets to kill tumor cells and to activate the immune system. J Oncol. 2019;2019:5245034. https://doi.org/10.1155/2019/5245034
- [Google Scholar]
- Butyrate ameliorates colorectal cancer through regulating intestinal microecological disorders. Anticancer Drugs. 2023;34:227-237. https://doi.org/10.1097/CAD.0000000000001413
- [Google Scholar]
- Modulating gut microbiome in cancer immunotherapy: Harnessing microbes to enhance treatment efficacy. Cell Rep Med. 2024;5:101478. https://doi.org/10.1016/j.xcrm.2024.101478
- [Google Scholar]
- Comparison of human gut microbiota in control subjects and patients with colorectal carcinoma in adenoma: Terminal restriction fragment length polymorphism and next-generation sequencing analyses. Oncol Rep. 2016;35:325-333. https://doi.org/10.3892/or.2015.4398
- [Google Scholar]
- Control of lymphocyte functions by gut microbiota-derived short-chain fatty acids. Cell Mol Immunol. 2021;18:1161-1171. https://doi.org/10.1038/s41423-020-00625-0
- [Google Scholar]
- Fusobacterium nucleatum induces a tumor microenvironment with diminished adaptive immunity against colorectal cancers. Front Cell Infect Microbiol. 2023;13:1101291. https://doi.org/10.3389/fcimb.2023.1101291
- [Google Scholar]
- Physiological role of bile acids modified by the gut microbiome. Microorganisms. 2021;10:68. https://doi.org/10.3390/microorganisms10010068
- [Google Scholar]
- Integrated metagenomic and metabolomic analysis reveals distinct gut-microbiome-derived phenotypes in early-onset colorectal cancer. Gut. 2023;72:1129-1142. https://doi.org/10.1136/gutjnl-2022-327156
- [Google Scholar]
- Gut microbiota-derived butyrate regulates gut mucus barrier repair by activating the macrophage/WNT/ERK signaling pathway. Clin Sci (Lond). 2022;136:291-307. https://doi.org/10.1042/CS20210778
- [Google Scholar]
- Hydrogen sulfide perturbs mitochondrial bioenergetics and triggers metabolic reprogramming in colon cells. J Biol Chem. 2019;294:12077-12090. https://doi.org/10.1074/jbc.RA119.009442
- [Google Scholar]
- Identification of colorectal cancer progression-associated intestinal microbiome and predictive signature construction. J Transl Med. 2023;21:373. https://doi.org/10.1186/s12967-023-04119-1
- [Google Scholar]
- Fusobacterium nucleatum, rectal cancer and radiotherapy. Ann Oncol. 2020;31:1277-1278. https://doi.org/10.1016/j.annonc.2020.06.019
- [Google Scholar]
- Fusobacterium nucleatum promotes inflammatory and anti-apoptotic responses in colorectal cancer cells via ADP-heptose release and ALPK1/TIFA axis activation. Gut Microbes. 2024;16:2295384. https://doi.org/10.1080/19490976.2023.2295384
- [Google Scholar]
- Regulation, risk and safety of faecal microbiota transplant. Infect Prev Pract. 2020;2:100069. https://doi.org/10.1016/j.infpip.2020.100069
- [Google Scholar]
- Tumor promoting roles of IL-10, TGF-β, IL-4, and IL-35: Its implications in cancer immunotherapy. SAGE Open Med. 2022;10:20503121211069012. https://doi.org/10.1177/20503121211069012
- [Google Scholar]
- Meta-analysis of 16S rRNA microbial data identified distinctive and predictive microbiota dysbiosis in colorectal carcinoma Adjacent Tissue. mSystems. 2020;5:e00138-e00120. https://doi.org/10.1128/mSystems.00138-20
- [Google Scholar]
- Global burden of colorectal cancer in 2020 and 2040: Incidence and mortality estimates from GLOBOCAN. Gut. 2023;72:338-344. https://doi.org/10.1136/gutjnl-2022-327736
- [Google Scholar]
- Understanding human health through metatranscriptomics. Trends Mol Med. 2023;29:376-389. https://doi.org/10.1016/j.molmed.2023.02.002
- [Google Scholar]
- Fusobacterium nucleatum and colorectal cancer: From phenomenon to mechanism. Front Cell Infect Microbiol. 2022;12:1020583. https://doi.org/10.3389/fcimb.2022.1020583
- [Google Scholar]
- Microbiome in drug resistance to colon cancer. Curr Opin Physiol. 2021;23:100472. https://doi.org/10.1016/j.cophys.2021.100472
- [Google Scholar]
- Diet-wide analyses for risk of colorectal cancer: Prospective study of 12,251 incident cases among 542,778 women in the UK. Nat Commun. 2025;16:375. https://doi.org/10.1038/s41467-024-55219-5
- [Google Scholar]
- Breast cancer colonization by fusobacterium nucleatum accelerates tumor growth and metastatic progression. Nat Commun. 2020;11:3259. https://doi.org/10.1038/s41467-020-16967-2
- [Google Scholar]
- Mutational signature in colorectal cancer caused by genotoxic pks+ e. coli. Nature. 2020;580:269-273. https://doi.org/10.1038/s41586-020-2080-8
- [Google Scholar]
- Encapsulation in an alginate–goats’ milk–inulin matrix improves survival of probiotic Bifidobacterium in simulated gastrointestinal conditions and goats’ milk yoghurt. International Journal of Dairy Technology.. 2019;72:132-141. https://doi.org/10.1111/1471-0307.12568
- [Google Scholar]
- Enterotoxigenic Bacteroides fragilis activates IL-8 expression through Stat3 in colorectal cancer cells. Gut Pathog. 2022;14:16. https://doi.org/10.1186/s13099-022-00489-x
- [Google Scholar]
- Consistent signatures in the human gut microbiome of old- and young-onset colorectal cancer. Nat Commun. 2024;15:3396. https://doi.org/10.1038/s41467-024-47523-x
- [Google Scholar]
- Composition, function, and timing: Exploring the early-life gut microbiota in piglets for probiotic interventions. J Anim Sci Biotechnol. 2023;14:143. https://doi.org/10.1186/s40104-023-00943-z
- [Google Scholar]
- AI in drug discovery and its clinical relevance. Heliyon. 2023;9:e17575. https://doi.org/10.1016/j.heliyon.2023.e17575
- [Google Scholar]
- Selective targeting of the TLR4 co-receptor, MD2, prevents colon cancer growth and lung metastasis. Int J Biol Sci. 2020;16:1288-1302. https://doi.org/10.7150/ijbs.39098
- [Google Scholar]
- The dysbiosis signature of Fusobacterium nucleatum in colorectal cancer-cause or consequences? A systematic review. Cancer Cell Int. 2021;21:194. https://doi.org/10.1186/s12935-021-01886-z
- [Google Scholar]
- Gut microbiome and its role in colorectal cancer. BMC Cancer. 2021;21:1325. https://doi.org/10.1186/s12885-021-09054-2
- [Google Scholar]
- Fusobacterium nucleatum and Bacteroides fragilis detection in colorectal tumours: Optimal target site and correlation with total bacterial load. PLoS One. 2022;17:e0262416. https://doi.org/10.1371/journal.pone.0262416
- [Google Scholar]
- Short-chain fatty acids improve poststroke recovery via immunological mechanisms. J Neurosci. 2020;40:1162-1173. https://doi.org/10.1523/JNEUROSCI.1359-19.2019
- [Google Scholar]
- Patients with mesenchymal tumours and high fusobacteriales prevalence have worse prognosis in colorectal cancer (CRC) Gut. 2022;71:1600-1612. https://doi.org/10.1136/gutjnl-2021-325193
- [Google Scholar]
- The role of the gut microbiome in colorectal cancer development and therapy response. Cancers (Basel). 2020;12:1406. https://doi.org/10.3390/cancers12061406
- [Google Scholar]
- A review of colorectal cancer in terms of epidemiology, risk factors, development, symptoms and diagnosis. Cancers (Basel). 2021;13:2025. https://doi.org/10.3390/cancers13092025
- [Google Scholar]
- Efficacy of probiotics-based interventions as therapy for inflammatory bowel disease: A recent update. Saudi J Biol Sci. 2022;29:3546-3567. https://doi.org/10.1016/j.sjbs.2022.02.044
- [Google Scholar]
- Fusobacterium nucleatum persistence and risk of recurrence after preoperative treatment in locally advanced rectal cancer. Ann Oncol. 2020;31:1366-1375. https://doi.org/10.1016/j.annonc.2020.06.003
- [Google Scholar]
- An overview of yeast probiotics as cancer biotherapeutics: Possible clinical application in colorectal cancer. Clin Transl Oncol. 2020;22:1227-1239. https://doi.org/10.1007/s12094-019-02270-0
- [Google Scholar]
- Gut microbiota characteristics of colorectal cancer patients in Hubei, China, and differences with cohorts from other Chinese regions. Front Microbiol. 2024;15:1395514. https://doi.org/10.3389/fmicb.2024.1395514
- [Google Scholar]
- Roles of short-chain fatty acids in inflammatory bowel disease. Nutrients. 2023;15:4466. https://doi.org/10.3390/nu15204466
- [Google Scholar]
- Emerging applications of upconverting nanoparticles in intestinal infection and colorectal cancer. Int J Nanomedicine. 2019;14:1027-1038. https://doi.org/10.2147/IJN.S188887
- [Google Scholar]
- Genome-wide gene–environment interaction analyses to understand the relationship between red meat and processed meat intake and colorectal cancer risk. Cancer Epidemiology, Biomarkers & Prevention. 2024;33:400-410. https://doi.org/10.1158/1055-9965.epi-23-0717
- [Google Scholar]
- Comparison of metagenomics and metatranscriptomics tools: A guide to making the right choice. Genes (Basel). 2022;13:2280. https://doi.org/10.3390/genes13122280
- [Google Scholar]
- Cancer pharmacomicrobiomics: Targeting microbiota to optimise cancer therapy outcomes. Gut. 2022;71:1412-1425. https://doi.org/10.1136/gutjnl-2021-326264
- [Google Scholar]
- Microbiome confounders and quantitative profiling challenge predicted microbial targets in colorectal cancer development. Nat Med. 2024;30:1339-1348. https://doi.org/10.1038/s41591-024-02963-2
- [Google Scholar]
- Tubeimuside i improves the efficacy of a therapeutic fusobacterium nucleatum dendritic cell-based vaccine against colorectal cancer. Front Immunol. 2023;14:1154818. https://doi.org/10.3389/fimmu.2023.1154818
- [Google Scholar]
- Enhancing biocide efficacy: Targeting extracellular DNA for marine biofilm disruption. Microorganisms. 2022;10:1227. https://doi.org/10.3390/microorganisms10061227
- [Google Scholar]
- Bowel biofilms: tipping points between a healthy and compromised gut? Trends Microbiol. 2019;27:17-25. https://doi.org/10.1016/j.tim.2018.08.009
- [Google Scholar]
- Fecal microbiota transplantation in neurological disorders. Front Cell Infect Microbiol. 2020;10:98. https://doi.org/10.3389/fcimb.2020.00098
- [Google Scholar]
- Probiotics regulate gut microbiota: An effective method to improve immunity. Molecules. 2021;26:6076. https://doi.org/10.3390/molecules26196076
- [Google Scholar]
- Colorectal cancer and gut microbiota studies in China. Gut Microbes. 2023;15:2236364. https://doi.org/10.1080/19490976.2023.2236364
- [Google Scholar]
- The human gut bacterial genotoxin colibactin alkylates DNA. Science. 2019;363:eaar7785. https://doi.org/10.1126/science.aar7785
- [Google Scholar]
- Diversification of host bile acids by members of the gut microbiota. Gut Microbes. 2020;11:158-171. https://doi.org/10.1080/19490976.2019.1674124
- [Google Scholar]
- Quantitation of faecal fusobacterium improves faecal immunochemical test in detecting advanced colorectal neoplasia. Gut. 2017;66:1441-1448. https://doi.org/10.1136/gutjnl-2016-312766
- [Google Scholar]
- Gut microbiota in colorectal cancer: Mechanisms of action and clinical applications. Nat Rev Gastroenterol Hepatol. 2019;16:690-704. https://doi.org/10.1038/s41575-019-0209-8
- [Google Scholar]
- The bacteroides fragilis toxin binds to a specific intestinal epithelial cell receptor. Infect Immun. 2006;74:5382-5390. https://doi.org/10.1128/IAI.00060-06
- [Google Scholar]
- Identification of microbial markers across populations in early detection of colorectal cancer. Nat Commun. 2021;12:3063. https://doi.org/10.1038/s41467-021-23265-y
- [Google Scholar]
- The microbiome types of colorectal tissue are potentially associated with the prognosis of patients with colorectal cancer. Front Microbiol. 2023;14:1100873. https://doi.org/10.3389/fmicb.2023.1100873
- [Google Scholar]
- Structure elucidation of colibactin and its DNA cross-links. Science. 2019;365:eaax2685. https://doi.org/10.1126/science.aax2685
- [Google Scholar]
- Metagenomic and metabolomic analyses reveal distinct stage-specific phenotypes of the gut microbiota in colorectal cancer. Nat Med. 2019;25:968-976. https://doi.org/10.1038/s41591-019-0458-7
- [Google Scholar]
- Enteric short‐chain fatty acids promote proliferation of human neural progenitor cells. Journal of Neurochemistry.. 2020;154:635-646. https://doi.org/10.1111/jnc.14928
- [Google Scholar]
- Microbiome analysis of more than 2,000 NHS bowel cancer screening programme samples shows the potential to improve screening accuracy. Clin Cancer Res. 2021;27:2246-2254. https://doi.org/10.1158/1078-0432.CCR-20-3807
- [Google Scholar]
- Understanding bacterial biofilms: From definition to treatment strategies. Front Cell Infect Microbiol. 2023;13:1137947. https://doi.org/10.3389/fcimb.2023.1137947
- [Google Scholar]
- Role of the gut microbiota in anticancer therapy: From molecular mechanisms to clinical applications. Signal Transduct Target Ther. 2023;8:201. https://doi.org/10.1038/s41392-023-01406-7
- [Google Scholar]
- Fecal microbiota transplantation plus tislelizumab and fruquintinib in refractory microsatellite stable metastatic colorectal cancer: An open-label, single-arm, phase II trial (RENMIN-215) EClinicalMedicine. 2023;66:102315. https://doi.org/10.1016/j.eclinm.2023.102315
- [Google Scholar]