You're using an outdated browser. Please upgrade to a modern browser for the best experience.
Submitted Successfully!
Thank you for your contribution! You can also upload a video entry or images related to this topic. For video creation, please contact our Academic Video Service.
Version Summary Created by Modification Content Size Created at Operation
1 -- 4056 2022-05-05 14:57:04 |
2 update references and layout -147 word(s) 3909 2022-05-06 04:41:40 |

Video Upload Options

We provide professional Academic Video Service to translate complex research into visually appealing presentations. Would you like to try it?

Confirm

Are you sure to Delete?
Yes No
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Al-Ishaq, R.; Koklesova, L.; Kubatka, P.; Büsselberg, D. Immunomodulation by Gut Microbiome on Gastrointestinal Cancers. Encyclopedia. Available online: https://encyclopedia.pub/entry/22623 (accessed on 18 July 2025).
Al-Ishaq R, Koklesova L, Kubatka P, Büsselberg D. Immunomodulation by Gut Microbiome on Gastrointestinal Cancers. Encyclopedia. Available at: https://encyclopedia.pub/entry/22623. Accessed July 18, 2025.
Al-Ishaq, Raghad, Lenka Koklesova, Peter Kubatka, Dietrich Büsselberg. "Immunomodulation by Gut Microbiome on Gastrointestinal Cancers" Encyclopedia, https://encyclopedia.pub/entry/22623 (accessed July 18, 2025).
Al-Ishaq, R., Koklesova, L., Kubatka, P., & Büsselberg, D. (2022, May 05). Immunomodulation by Gut Microbiome on Gastrointestinal Cancers. In Encyclopedia. https://encyclopedia.pub/entry/22623
Al-Ishaq, Raghad, et al. "Immunomodulation by Gut Microbiome on Gastrointestinal Cancers." Encyclopedia. Web. 05 May, 2022.
Immunomodulation by Gut Microbiome on Gastrointestinal Cancers
Edit

Gastrointestinal cancer (GI) is a global health disease with a huge burden on a patient’s physical and psychological aspects of life and on health care providers. It is associated with multiple disease related challenges which can alter the patient’s quality of life and well-being. GI cancer development is influenced by multiple factors such as diet, infection, environment, and genetics. Although activating immune pathways and components during cancer is critical for the host’s survival, cancerous cells can target those pathways to escape and survive. As the gut microbiome influences the development and function of the immune system, research is conducted to investigate the gut microbiome–immune interactions, the underlying mechanisms, and how they reduce the risk of GI cancer. 

gut microbiome immune–gut interaction gastrointestinal cancer

1. Gastrointestinal Cancer

Globally, cancers are a significant cause of death and disability [1]. They are characterized by impaired homeostasis and cellular functions [2]. Cancers are classified based on the organ, tissue of origin, or the cancer cell’s molecular characteristics [3] and the development of cancers is influenced by environmental and genetic factors such as obesity, diet, smoking, and infections with pathogenic agents [4]. Gastrointestinal cancers (GI) are considered a major public health problem with challenging economic and medical burdens due to their high prevalence and mortality rate [5]. The symptoms and signs of GI cancers depend on the type of cancer (gastric cancer (GC), colorectal cancer (CRC), esophageal cancer (EC), pancreatic cancer (PC), and hepatocellular carcinoma (HCC)). They might include weight loss, abdominal pain, dysphagia, and anorexia [6] and the progression of GI cancers occurs in a multistage process. They result from uncontrolled cellular proliferation, the loss of apoptotic functions through the intrinsic and extrinsic apoptotic pathways, and the impairment of major pathways such as epithelial–mesenchymal transition (EMT), phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT), and nuclear factor-kappa (NF-κB) signaling pathways [7][8]. Efforts are required to understand GI-cancers’ underlying mechanisms through these specific impaired pathways.

2. The Immune System in Cancer Pathogenesis

The human immune system is defined as a group of cells that protect the body from foreign antigens such as toxins, microbes, viruses, and cancer cells [9]. The immune system has two lines of defense that complement each other; innate and adaptive immunity. Imbalance or defects in either line of defense could result in an inappropriate immune response in the body [10].
Cancer and the immune system have been widely discussed for a century [11]. The underlying mechanism between cancer cells and the immune system interaction involves three processes of how the immune system defends and protects the host; (i) the identification of non-self cells, (ii) the production of effector cells to specifically target the cancerous cells, and (iii) the development of immunological memory as a defense mechanism [12]. The role of immune cells in cancer includes both a pro-tumorigenic and an anti-tumorigenic function [11]. Inflammatory immune cells activation in cancer can present in different tumorigenesis stages and can lead to epigenetic modification, the induction of cancerous cellular proliferation, genomic instability, and the enhancement of a cancerous anti-apoptotic pathway, therefore, leading to cancer progression and dissemination [11]. During the pathogenesis of cancer, multiple components and pathways of innate and adaptive immunity are activated to identify cancerous cells and target their genetic and epigenetic alterations and modifications, thus leading to cancer elimination [13]. Such pathways include complement proteins activation aiding in cancer eradication, natural killer (NK) cells, cytotoxic immune cells which recognize and eliminate immunogenic cancerous cells, neutrophil protease activation, anti-tumor macrophages which display a pro-inflammatory like polarization playing a role in the elimination of immunogenic cancerous cells, CD4+ T-cells activation, the production of IL-22 promoting T-cells proliferation, and naïve B cells activation [14][15]. Despite these mechanisms, cancer can manage to overcome immune components as in the case of T-cells, in which cancerous cells can impair the functions of anti-tumor T-cells such as their ability to infiltrate the tumor survival, cytotoxicity, and proliferation abilities [15].
Advances in the development of immuno-oncology have changed the treatment of GI cancer. Multiple ongoing clinical trials evaluate the efficacy and safety of immunotherapy agents such as avelumab (anti-PD-L1) and relatlimab (anti-LAG3) in patients with advanced gastric cancer [16]. Additionally, as for CRC, two immune checkpoint inhibitors target programmed death-ligand 1 (PD-1) in metastatic cancer, namely, KEYNOTE 028 and CheckMate 142, with an objective response rate of 40% and 55%, respectively [17]. More are required to identify the common side effects of these treatments, to estimate the impact on patients with immunodeficiency, and to evaluate the role of gut microbiota in treatment utilization.

3. Gut Microbiota: Role in GI Cancer Immunity

In the human body, trillions of microorganisms, such as bacteria, viruses, fungi, and protozoan, are known as the microbiota [18]. The microbiota resides mainly on the respiratory and gastrointestinal tract’s mucosal surfaces with different concentrations and relative abundances [19]. Over time, changes in the microbiome composition occur due to internal or external factors such as lifestyle, genetics, geographical locations, and age, leading to significant variations between individuals [20]. The gut microbiome plays a role in protection from infections, vitamin production, and immune cells development and activity [21], but intestinal dysbiosis and the imbalance in the number of microbes and their diversity in the gut is linked to several pathogeneses such as cancer [22].
It has been reported that the impact of microbiota on the development, activities, and function of immune cells [23]. In mucosal sites, where the microbiota prominently resides, early B-lineage cells development occurs under the influence of extracellular signals from the microbiota [24]. Additionally, the gut microbiome promotes the differentiation of naïve T-cells into colonic Treg cells with unique T-cell receptors on their surfaces [25]. During the invasion of pathogenic bacteria, the gut microbiome promotes the activation of myeloid cells leading to cytokines production [23].
In cancer, the gut microbiome influences the anti-tumor immune response through (1) the induction of the T-cells response, (2) the engagement of a pattern recognition receptor that has pro-inflammatory effects, or (3) the mediation of specific metabolites, which can activate T-cell receptors [26]. Efforts are required to investigate and understand the underlying mechanisms between the gut microbiome and the immune system in the context of cancer and how those mechanisms can be utilized as targets for cancer therapy. Figure 1 summarizes the most common pathogens in the GI tract, their relative abundance, and reported immune regulations.

4. Microbiota–Immune Interactions: Role in GI Cancer Development

The gut microbiome plays a critical role in the pathogenesis of host diseases such as cancer [27]. As the gut microbiome is influenced by several factors such as diet, genetics, and lifestyle, its dysbiosis, either in the bacterial composition, bacterial bioactivity or diversity, can impair the balance of specific bacterial species and increase the abundance of inflammation-inducing species that can cause several diseases including inflammatory bowel disease and cancer [28]. The gut microbiome influences the host immune response to regulate cancer mechanisms such as progression, genetic instability, and the response to treatment [29]. Animal ones have reported that specific microbes such as Bacteroides fragilis and Escherichia coli can promote cancer development by releasing genotoxins, damaging the host DNA [30]. Additionally, the gut microbiome could impact the efficacy of cancer treatment, as seen in antibiotic-treated mice [31]. This suggests the critical role of intact microbiota in the gut for optimal treatment outcomes.
Additionally, the gut microbiome impacts the function of the mucosal B and T cells, which are essential for immune homeostasis as they inhibit the unregulated response to harmless antigens and preserve the mucosal barrier integrity in the intestine [32]. Disruption of the gut barrier facilitates the interaction between the immune cells and the microorganisms, resulting in cancer development through the induction of immunosuppressive or pro-inflammatory pathways [33]. Gut microbiome dysbiosis can influence cancer pathways by recruiting lymphocytes to the intestine, leading to cellular proliferation by activating the IL-6 pathway [34]. Moreover, TLRs upregulation can activate the nuclear factor (NF)-κB and JAK/STAT3, which are critical for immunosuppression and cellular proliferation [35].

4.1. Inflammation

Inflammation is associated with multiple diseases such as diabetes, cardiovascular disease, and multiple stages of cancer [36]. During cancer, acute inflammation is critical in the recruitment and accumulation of neutrophils, the stimulation of antigen presentation, and the maturation of dendritic cells leading to an anti-tumor response. Additionally, and during acute inflammation, the level of C-reactive protein and serum amyloid A protein (SAA), acute phase proteins can increase, with the latter being influenced by segmented filamentous bacteria. On the other hand, chronic inflammation is linked to different stages of cancer development, including transformation, promotion, proliferation, invasion, metastasis, survival, angiogenesis, and treatment resistance, with an accumulation of macrophages, lymphocytes, and plasma cells at the site [37]. In addition, chronic inflammation is considered a risk factor for gastrointestinal cancer development in patients with inflammatory bowel disease, as reports have illustrated a similar inflammatory microenvironment between cancer and inflammatory bowel diseases. Additionally, in both diseases, inflammatory cells produce similar mediators such as IL-6 and IL-12, which suggests the role played by the immune system in both diseases [38]. Damaged tissues in the body caused by either a physical or an ischemic injury, exposure to toxins, or an infection can result in an inflammatory response activation that is necessary to repair the damaged tissues [39]. An inflammatory response can become chronic when the causative agent of the inflammation persists, resulting in cellular proliferation and mutation, thus creating a suitable environment for cancer development [40]. Additionally, and due to chronic inflammation, host leukocytes such as macrophages, dendritic cells, and lymphocytes can be present in tumor areas. They can lead to immunosuppression and cancer growth by producing reactive oxygen species (ROS) that damage the intestinal epithelial cells’ DNA [39].
The gut microbiota in the intestine is usually segregated from the immune cells by a single layer of intestinal epithelial cells joined by tight junctions [41]. Dysbiosis in the gut can alter the permeability of the intestinal barrier, causing a disruption where commensal bacteria and their products can invade the mucosa, thus resulting in low-grade systemic inflammation. Due to that, inflammatory pathways such as Wnt and Notch are activated, affecting the mucosal epithelial cells, thus influencing immune homeostasis and increasing susceptibility to CRC [42]. After activating the myeloid differentiation factor 88 (MyD88), the invading commensal bacteria and their products interact with TLRs on tumor-infiltrating myeloid cells, leading to the production of inflammatory cytokines such as IL-23 activating the production of IL-6, IL-22, and IL-17A [43]. The production of those cytokines can eventually promote the activation of STAT3 and the nuclear factor-kB (NF-kB) signaling pathway [44]. The promoted activation of NF-kB signaling pathway by TLR-4 overexpression can induce COX-2 expression, a CRC biomarker, and an inflammation-associated gene in inflammatory bowel disease [45]. Figure 1 summarizes the interaction between the gut microbiome and the immune cells in GI cancer and its activation of inflammation. Meanwhile, another preclinical one documented that the activation of the inflammatory response significantly correlated with the disturbance of the gut microbiota and changes in the fecal metabolites [46]. It was found that these changes could be closely related to the occurrence of precancerous lesions of GC. The correlation analysis between inflammatory cytokines and gut microbiota/feces metabolites was evaluated in a N-methyl-N′-nitro-N-nitrosoguanidine multiple factors-induced rat model of GC. The results demonstrated a significant increase in pro-inflammatory serum cytokines such as IL-1β, IL-4, IL-6, IL-10, IFN-γ, TNF-α, and M-CSF.
Figure 1. Schematic representation of the immune—gut interactions during GI cancer and how it influences the inflammatory responses. Due to gut dysbiosis, the low level of short chain fatty acids can lead to the activation of inflammatory pathway, the production of cytokines and chemokines and the activation of STAT3 and NF-kB signaling pathways. “Created with BioRender.com”.
On the other hand, there was a significant decrease in the level of chemokine (C-X-C motif) ligand 1 (CXCL1) in the model group vs. controls. In this regard, the gut microbiota and fecal metabolic phenotype composition in the model group revealed that Lactobacillus and Bifidobacterium significantly increased. At the same time, Turicibacter, Romboutsia, Ruminococcaceae_UCG-014, Ruminococcaceae_UCG-005, and Ruminococcus_1 were significantly decreased compared to the control animals.

4.2. Cellular Proliferation

Cellular proliferation is a fundamental process essential for the development and hemostasis of the organism [47]. It is tightly regulated to ensure a precise and complete genome duplication [48]. Multiple factors, from DNA damage to growth factors, influence the process of DNA replication, especially the entering to the S phase of the cycle [49]. Cancer cells embody multiple characteristics that play a role in their survival and abnormal proliferation [50] and due to epigenetic changes and/or mutations, cancer cells are resistant to cellular proliferation regulators such as growth factors and hormones. Such changes promote the growth and survival of cancerous cells through the stimulation of proliferation pathways and the inhibition of apoptotic pathways [51]. Emerging evidence supports the gut microbiome’s role in influencing cellular proliferation in cancer through contact with immune cells, as seen in the case of Fusobacterium nucleatum, the most learned colon cancer-associated microorganism, which is enriched during cancer [52][53].
F. nucleatum is a commensal opportunistic anaerobic Gram-negative bacillus found mainly in the oral cavity. It is implicated in multiple diseases outside the oral cavity [54]. F. nucleatum plays a role in colon cancer progression and treatment with antibiotics such as metronidazole which reduces their load and cellular proliferation [55]. Additionally, F. nucleatum promotes cellular proliferation in CRC by binding FadA to E-cadherin, which mediates the bacteria’s attachment and invasion. This leads to the activation of β-catenin signaling and the increased expression of Wnt genes, transcription factors, and inflammatory genes, thus impacting T-cells infiltration levels [56][57]. On the other hand, some bacterial strains, such as Holdemanella biformis, are reduced during gut tumorigenesis, which is critical in blocking tumor proliferation [58]. H. biformis impacts cellular proliferation by mediating SCFA such as butyrate, which inhibits histone deacetylase (HDAC) activities by enhancing H3 histone acetylation and reducing the NFATC3 pathway [59].
Efforts are required to identify potential bacteria strains and their role in GI cancer development. Additionally, more research is necessary to assess the feasibility of maybe using specific strains as a treatment option for GI cancer. Figure 2 summarizes the role of the reported bacteria on GI cancer.
Figure 2. Schematic illustration of two pathways in which two bacteria Fusobacterium nucleatum and Holdemanella biformis facilitate cancer progression and cellular proliferation through FadA- E-cadherin interaction and short chain fatty acids (SCFA), respectively. (A) represent the proliferative example while (B) the anti-proliferative example. “Created with BioRender.com”.

4.3. Metastasis

Metastasis is defined as the expansion of the primary tumor, leading to secondary tumors distant from the original tumor [60]. Metastasis occurs in a multi-step process that includes the separation from the primary tumor, the invasion through the surrounding tissues, and the entry and survival in the circulation [61]. Understanding the mechanism of metastasis is of great importance to managing and treating cancer. Therefore, assessing the impact of the gut microbiome, a potential therapeutic option, and immune system interaction can provide some insights. F. nucleatum is linked to CRC development and progression [62]. The polymerase chain reaction quantification of F. nucleatum DNA in 181 colorectal cancer liver metastases specimens reported that the presence and the quantity of the bacteria is inversely associated with a lower CD8+ T-cells density. This could suggest the potential involvement of F. nucleatum in cancer metastasis [63]. Mechanistically and in CRC, tissues are overexpressing sugar residues Gal-GalNAc, which is recognized by the F. nucleatum adhesion molecule, Fab2, and which is critical in mediating hemagglutinin and co-aggregation functions. Mechanistically, F. nucleatum could promote metastasis by activating the TLR-4 pathways, upregulating a cytochrome p450 known as CYP2J2. The metabolite of this cytochrome, 12,13-EpOME, then activates EMT, thus promoting CRC metastasis in vitro [64].
Additionally, F. nucleatum can evade anti-cancer immune responses by mediating the recognition and binding of the same Fab2 adhesion molecule to a receptor known as TIGIT, overexpressed on natural killer cells and other lymphocytes. The mediated binding inhibits the functions of lymphocytes and natural killer cells, therefore, protecting F. nucleatum and promoting a pro-tumorigenic environment [65]. Figure 3 highlights the reported mechanisms in which F. nucleatum promotes GI cancer metastasis.
Figure 3. Summarizes the influence of Fusobacterium nucleatum on cancer metastasis either by targeting sugar residues Gal-GalNAc on cancerous cells or targeting receptors that are overexpressed on natural killer cells (NK). “Created with BioRender.com”.

4.4. Apoptosis

Apoptosis is a basic cellular mechanism that is essential in the development and homeostasis of the organism [66]. Distinct morphological changes characterize it, controlled by intracellular and extracellular signals regulated by the cell environment [67]. Intrinsic and extrinsic pathways are the two major apoptotic pathways where they process the stress signal and execute the death signal in the cell [68]. Both exogenous and endogenous agents such as physical trauma, infectious agents, radiation, and chemotherapeutic drugs can trigger apoptosis [69]. In cancer, downregulation of apoptosis by pro-survival proteins is necessary to maintain the phenotypic properties. Such alteration is observed in the anti-apoptotic Bcl-2 family, which is overexpressed frequently in solid tumors [70]. On the other hand, one analyzed the expression of human BCL-G, a member of the BCL-2 family in gastrointestinal conditions, and they reported that both variants were highly expressed in a healthy gut. At the same time, their m-RNA level was decreased in colorectal cancer and inflammatory bowel disease conditions [71]. Additionally, it was reported that the depletion of BCL-G affected the secretion of chemokines such as CCL5 thus illustrating a non-apoptotic function of the BCL-2 family. More are required to assess the role of the BCL-2 family in shaping the immune system, apoptosis, and maybe the regulation of chemokines.
The gut microbiome is a critical mediator of the host’s health by producing certain metabolites essential for immune system regulations [72]. Gut dysbiosis can reduce the beneficial bacteria responsible for producing SCFA, such as butyrate [73]. Butyrate plays a role in maintaining the intestinal barrier function and reducing inflammation in the colon, as they supply colonocytes with 70% of their required energy [74]. Additionally, the butyrate induces IL-18 expression in the colon, which is essential in suppressing colonic inflammation [75]. The administration of butyrate reduces cellular proliferation and pro-inflammatory cytokines production, such as IL-6, while promoting apoptosis [76]. Gut analysis of patients with colon cancer and ulcerative colitis showed a significant reduction in butyrate levels and the number of butyrate-producing bacteria in the colon [77]. During cancer, and when the gut is in dysbiosis, butyrate production is reduced, impacting the butyrate receptor’s activity, GPR109a, found in the colon. This reduces IL-18 and IL-22 production, reducing the mucosal tissue repair capabilities, thus impacting cellular apoptosis [78][79]. Another described the significant role of moxibustion, a traditional Chinese medicine, in inducing apoptosis of rat GC cells in vivo by regulating intestinal flora [80]. It was summarized that moxibustion delayed the GC metastasis possibly by lowering the abundance of Ruminococcaceae and Prevotellaceae bacteria (bacteria producing short-chain fatty acids in the gut) and enhancing the occurrence of probiotic Akkermansia in the rat intestine.
Additionally, butyrate induces apoptosis in CRC through the mitochondrial pathway and caspase 3 [81]. When the butyrate level is reduced, the expression of Bcl-2 anti-apoptotic family is enhanced, while the expression of Bax/Bak, cytochrome c is reduced [74]. Figure 4 summarizes the role of gut dysbiosis and butyrate production on cellular apoptosis during cancer.
Figure 4. Illustrations of the role of short-chain fatty acids specifical butyrate on cellular apoptosis. The figure highlights the Gpr109a receptor and the pathways that lead to the reduction of mucosal tissue repair and Caspase 3 activation. “Created with BioRender.com”.

5. Influence of Gut Microbiome on Immunotherapy

Current cancer treatments, including chemotherapy, surgery, endocrine therapy, and radiotherapy, are usually non-specific approaches. They frequently reach a refractory period, leading to treatment failure and disease recurrence [82][83]. Targeting the immune system and enhancing the patient’s immune system to attack the tumor can potentially be therapeutic [84]. Cancer immunotherapy is an alternative approach that utilizes specific components of a patient’s immune system to selectively target and eliminate tumor cells, thus mitigating the side effects of the currently used treatments [85]. Depending on the mechanism by which the therapy activates the immune response, immunotherapy can be passive, such as cell-based therapy and chimeric antigen receptor T cell therapy (CAR-T cell) or active, such as vaccination, immunostimulatory cytokines, and immune checkpoint inhibitors [86][87]. Immune checkpoint inhibitors are used as a treatment option to induce a T-cells mediated response against cancerous cells to selectively block the inhibitory checkpoint receptors manipulated by the tumor cell [88]. Types of inhibitory checkpoint receptors include programmed cell death protein 1 (PD-1), cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), T cell immunoglobulin and mucin protein 3 (TIM-3), and programmed cell death 1 ligand 1 (PD-L1) [89]. To treat CRC, immunomodulatory therapy such as CTLA4, PD-1, and PD-L1 is currently used to target selective checkpoint molecules and inhibit T-cell activation [90]. Despite this, 19 patients with unselected CRC did not demonstrate positive clinical responses when using Nivolumab, a monoclonal antibody that binds to PD-1 receptor [91]. The gut microbiome plays a role in stimulating and influencing immunotherapy against cancer [92]. The intestinal microbiota is an essential factor in providing an optimal CpG-oligonucleotide immunotherapy response which activates innate immune cells [93]. Moreover, the microbiome influences immunotherapy as a community, but specific microbes such as Bacteroides fragilis can enhance PD-1/PD-L1 and CTLA-4 immunotherapy as they activate Th1 cells [94]

6. Future of GI Cancer Treatment?

The gut microbiome can interfere directly or indirectly with current treatments such as chemotherapy and immunotherapy, which might impact a treatment’s outcome. Manipulating the gut microbiome composition using fecal microbiota transplantation or phytochemicals might improve therapeutic outcomes [95]. Fecal microbiota transplantation (FMT) is known as the transplantation of microbes from the gut of a healthy donor to a recipient either through the upper or lower gastrointestinal tract [96]. It was first documented in clinical use in 1958 to treat Clostridium difficile infection as it helped treat 80% of the affected patients [97]. The advantages of using FMT include its safety and its ability to restore intestinal microbial diversity [98]. Limited are available that investigates the role and the application of FMT in the context of GI cancer treatment. It was found that reported the effectiveness of FMT in mice receiving intestinal microbiota from wild mice, as the results showed better resistance to CRC [99]. Additionally, and on a different approach, the usage of phytochemicals for GI cancer treatment has recently gained attention. The bioactive plant-derived compounds generally have lower oral bioavailability due to poor aqueous solubility, and therefore, the gut microbiome is essential for the metabolism and absorption of bioactive compounds [100]. Several data support the role of 13 bioactive secondary compounds on GI cancer [101]. For example, lutein, an abundant fat-soluble bioactive compound found primarily in green leaved vegetables, was reported to significantly reduce aberrant crypt foci (ACF) in the colon of mice, thus reducing cellular proliferation [102]. Despite those reports that support potential treatments, research is much needed to investigate the potential synergetic effects between the currently used treatments and FMT or phytochemicals. Additionally, attention should be given to the required concentration and the appropriate delivery mode of FMT and phytochemicals to avoid toxicity and possible side effects. Moreover, looking at the role of gut enzymes in the metabolism and the utilization of those natural bioactive compounds, more is needed to investigate the underlying mechanisms played by those enzymes that might affect the treatment outcome, as shown in [103].

7. Conclusions

The gut microbiome plays an essential role in mediating the immune response, impacting its activities, development, and function. Generally, and during cancer, signature microbes in the gut influence the anti-tumor activities by producing specific metabolites or inducing T-cell responses. On the other hand, some reported bacterial species enhance cellular proliferation and metastasis during cancer and understanding those interactions in the context of cancer may provide potential therapeutic targets. Despite the advances in the field, more research is needed to understand the underlying mechanisms, investigate the impact on current treatments, and identify specific microbes and immune cells that might lead to this interaction. Additionally, clinical trials are essential to assess the influence ofimmune–gut interaction on immunotherapy treatment in clinical settings.

References

  1. Hassanzade, J.; Molavi, E.V.H.; Farahmand, M.; Rajaiifard, A.R. Incidence and Mortality Rate of Common Gastrointestinal Cancers in South of Iran, a Population Based Study. Iran. J. Cancer Prev. 2011, 4, 163–169.
  2. Machlowska, J.; Baj, J.; Sitarz, M.; Maciejewski, R.; Sitarz, R. Gastric Cancer: Epidemiology, Risk Factors, Classification, Genomic Characteristics and Treatment Strategies. Int. J. Mol. Sci. 2020, 21, 4012.
  3. Krieghoff-Henning, E.; Folkerts, J.; Penzkofer, A.; Weg-Remers, S. Cancer-an overview. Med. Mon. Pharm. 2017, 40, 48–54.
  4. Zali, H.; Rezaei-Tavirani, M.; Azodi, M. Gastric cancer: Prevention, risk factors and treatment. Gastroenterol. Hepatol. Bed Bench 2011, 4, 175–185.
  5. Rozen, P. Cancer of the gastrointestinal tract: Early detection or early prevention? Eur. J. Cancer Prev. 2020, 13, 71–75.
  6. Correa, P. Gastric cancer: Overview. Gastroenterol. Clin. N. Am. 2020, 42, 211–217.
  7. Igney, F.H.; Krammer, P.H. Death and anti-death: Tumour resistance to apoptosis. Nat. Rev. Cancer 2002, 2, 277–288.
  8. Qiao, L.; Wong, B.C. Targeting apoptosis as an approach for gastrointestinal cancer therapy. Drug Resist. Updates 2009, 12, 55–64.
  9. Turvey, S.E.; Broide, D.H. Innate immunity. J. Allergy Clin. Immunol. 2010, 125, S24–S32.
  10. Marshall, J.S.; Warrington, R.; Watson, W.; Kim, H.L. An introduction to immunology and immunopathology. Allergy Asthma Clin. Immunol. 2018, 14, 49.
  11. Adams, J.L.; Smothers, J.; Srinivasan, R.; Hoos, A. Big opportunities for small molecules in immuno-oncology. Nat. Rev. Drug Discov. 2015, 14, 603–622.
  12. Pandya, P.H.; Murray, M.E.; Pollok, K.E.; Renbarger, J.L. The Immune System in Cancer Pathogenesis: Potential Therapeutic Approaches. J. Immunol. Res. 2016, 2016, 4273943.
  13. Chen, D.S.; Mellman, I. Oncology meets immunology: The cancer-immunity cycle. Immunity 2013, 39, 1–10.
  14. Harris, T.J.; Drake, C.G. Primer on tumor immunology and cancer immunotherapy. J. Immunother. Cancer 2013, 1, 12.
  15. Gregory, A.D.; Houghton, A.M. Tumor-associated neutrophils: New targets for cancer therapy. Cancer Res. 2011, 71, 2411–2416.
  16. Kwak, Y.; Seo, A.N.; Lee, H.E.; Lee, H.S. Tumor immune response and immunotherapy in gastric cancer. J. Pathol. Transl. Med. 2020, 54, 20–33.
  17. Golshani, G.; Zhang, Y. Advances in immunotherapy for colorectal cancer: A review. Ther. Adv. Gastroenterol. 2020, 13, 1756284820917527.
  18. Garrett, W.S. Cancer and the microbiota. Science 2015, 348, 80–86.
  19. Nasr, R.; Shamseddine, A.; Mukherji, D.; Nassar, F.; Temraz, S. The Crosstalk between Microbiome and Immune Response in Gastric Cancer. Int. J. Mol. Sci. 2020, 21, 6586.
  20. Thaiss, C.A.; Zmora, N.; Levy, M.; Elinav, E. The microbiome and innate immunity. Nature 2016, 535, 65–74.
  21. Meng, C.; Bai, C.; Brown, T.D.; Hood, L.E.; Tian, Q. Human Gut Microbiota and Gastrointestinal Cancer. Genomics Proteomics Bioinformatics 2018, 16, 33–49.
  22. Pham, F.; Moinard-Butot, F.; Coutzac, C.; Chaput, N. Cancer and immunotherapy: A role for microbiota composition. Eur. J. Cancer 2021, 155, 145–154.
  23. Yu, Q.; Jia, A.; Li, Y.; Bi, Y.; Liu, G. Microbiota regulate the development and function of the immune cells. Int. Rev. Immunol. 2018, 37, 79–89.
  24. Wesemann, D.R.; Portuguese, A.J.; Meyers, R.M.; Gallagher, M.P.; Cluff-Jones, K.; Magee, J.M.; Alt, F.W. Microbial colonization influences early B-lineage development in the gut lamina propria. Nature 2013, 501, 112–115.
  25. Lathrop, S.K.; Bloom, S.M.; Rao, S.M.; Nutsch, K.; Lio, C.W.; Santacruz, N.; Hsieh, C.S. Peripheral education of the immune system by colonic commensal microbiota. Nature 2011, 478, 250–254.
  26. Zitvogel, L.; Ma, Y.; Raoult, D.; Kroemer, G.; Gajewski, T.F. The microbiome in cancer immunotherapy: Diagnostic tools and therapeutic strategies. Science 2018, 359, 1366–1370.
  27. Cenit, M.C.; Sanz, Y.; Codoner-Franch, P. Influence of gut microbiota on neuropsychiatric disorders. World J. Gastroenterol. 2017, 23, 5486–5498.
  28. Tajik, N.; Frech, M.; Schulz, O.; Schalter, F.; Lucas, S.; Azizov, V.; Durholz, K.; Steffen, F.; Omata, Y.; Rings, A. Targeting zonulin and intestinal epithelial barrier function to prevent onset of arthritis. Nat. Commun. 2020, 11, 1995.
  29. Panebianco, C.; Andriulli, A.; Pazienza, V. Pharmacomicrobiomics: Exploiting the drug-microbiota interactions in anticancer therapies. Microbiome 2018, 6, 92.
  30. Dzutsev, A.; Badger, J.H.; Perez-Chanona, E.; Roy, S.; Salcedo, R.; Smith, C.K.; Trinchieri, G. Microbes and Cancer. Annu. Rev. Immunol. 2017, 35, 199–228.
  31. Iida, N.; Dzutsev, A.; Stewart, C.A.; Smith, L.; Bouladoux, N.; Weingarten, R.A.; Molina, D.A.; Salcedo, R.; Back, T.; Cramer, S. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science 2013, 342, 967–970.
  32. Honda, K.; Littman, D.R. The microbiota in adaptive immune homeostasis and disease. Nature 2016, 535, 75–84.
  33. Tsuei, J.; Chau, T.; Mills, D.; Wan, Y.J. Bile acid dysregulation, gut dysbiosis, and gastrointestinal cancer. Exp. Biol. Med. 2014, 239, 1489–1504.
  34. Ge, Y.; Wang, X.; Guo, Y. Gut microbiota influence tumor development and Alter interactions with the human immune system. J. Exp. Clin. Cancer Res. 2021, 40, 42.
  35. Zhang, J.; Zhang, F.; Zhao, C.; Xu, Q.; Liang, C.; Yang, Y.; Wang, H.; Shang, Y.; Wang, Y.; Mu, X. Dysbiosis of the gut microbiome is associated with thyroid cancer and thyroid nodules and correlated with clinical index of thyroid function. Endocrine 2019, 64, 564–574.
  36. Aggarwal, B.B. Nuclear factor-kappaB: The enemy within. Cancer Cell 2004, 6, 203–208.
  37. Mantovani, A. Cancer: Inflammation by remote control. Nature 2005, 435, 752–753.
  38. Yao, H.; Wan, J.Y.; Zeng, J.; Huang, W.H.; Sava-Segal, C.; Li, L.; Yuan, C.S. Effects of compound K, an enteric microbiome metabolite of ginseng, in the treatment of inflammation associated colon cancer. Oncol. Lett. 2018, 15, 8339–8348.
  39. Singh, N.; Baby, D.; Rajguru, J.P.; Patil, P.B.; Thakkannavar, S.S.; Pujari, V.B. Inflammation and cancer. Ann. Afr. Med. 2019, 18, 121–126.
  40. Mantovani, A.; Allavena, P.; Sica, A.; Balkwill, F. Cancer-related inflammation. Nature 2008, 454, 36–44.
  41. Lee, T.C.; Huang, Y.C.; Lu, Y.Z.; Yeh, Y.C.; Yu, L.C. Hypoxia-induced intestinal barrier changes in balloon-assisted enteroscopy. J. Physiol. 2018, 596, 3411–3424.
  42. Tanaka, T.; Kohno, H.; Suzuki, R.; Hata, K.; Sugie, S.; Niho, N. Dextran sodium sulfate strongly promotes colorectal carcinogenesis in Apc (Min/+) mice: Inflammatory stimuli by dextran sodium sulfate results in development of multiple colonic neoplasms. Int. J. Cancer 2006, 118, 25–34.
  43. Song, X.; Gao, H.; Lin, Y.; Yao, Y.; Zhu, S.; Wang, J. Alterations in the Microbiota Drive Interleukin-17C Production from Intestinal Epithelial Cells to Promote Tumorigenesis. Immunity 2014, 40, 140–152.
  44. Cheng, Y.; Ling, Z.; Li, L. The Intestinal Microbiota and Colorectal Cancer. Front. Immunol. 2020, 11, 615056.
  45. Zou, S.; Fang, L.; Lee, M.H. Dysbiosis of gut microbiota in promoting the development of colorectal cancer. Gastroenterol. Rep. 2018, 6, 1–12.
  46. Chu, F.; Li, Y.; Meng, X.; Li, Y.; Li, T.; Zhai, M.; Ding, X. Gut Microbial Dysbiosis and Changes in Fecal Metabolic Phenotype in Precancerous Lesions of Gastric Cancer Induced With N-Methyl-N’-Nitro-N-Nitrosoguanidine, Sodium Salicylate, Ranitidine, and Irregular Diet. Front. Physiol. 2021, 12, 733979.
  47. Matson, J.P.; Cook, J.G. Cell cycle proliferation decisions: The impact of single cell analyses. FEBS J. 2017, 284, 362–375.
  48. Hallstrom, T.C.; Nevins, J.R. Balancing the decision of cell proliferation and cell fate. Cell Cycle 2009, 8, 532–535.
  49. Duronio, R.J.; Xiong, Y. Signaling pathways that control cell proliferation. Cold Spring Harb. Perspect. Biol. 2013, 5, a008904.
  50. Feitelson, M.A.; Arzumanyan, A.; Kulathinal, R.J.; Blain, S.W.; Holcombe, R.F.; Mahajna, J.; Nowsheen, S. Sustained proliferation in cancer: Mechanisms and novel therapeutic targets. Semin. Cancer Biol. 2015, 35, S25–S54.
  51. von Frieling, J.; Fink, C.; Hamm, J.; Klischies, K.; Forster, M.; Bosch, T.C.G.; Sommer, F. Grow with the Challenge-Microbial Effects on Epithelial Proliferation, Carcinogenesis, and Cancer Therapy. Front. Microbiol. 2018, 9, 2020.
  52. McAllister, F.; Housseau, F.; Sears, C.L. Microbiota and immune responses in colon cancer: More to learn. Cancer J. 2014, 20, 232–236.
  53. Wu, J.; Li, Q.; Fu, X. Fusobacterium nucleatum Contributes to the Carcinogenesis of Colorectal Cancer by Inducing Inflammation and Suppressing Host Immunity. Transl. Oncol. 2019, 12, 846–851.
  54. Han, Y.W. Fusobacterium nucleatum: A commensal-turned pathogen. Curr. Opin. Microbiol. 2015, 23, 141–147.
  55. Bullman, S.; Pedamallu, C.S.; Sicinska, E.; Clancy, T.E.; Zhang, X.; Cai, D.; Meyerson, M. Analysis of Fusobacterium persistence and antibiotic response in colorectal cancer. Science 2017, 358, 1443–1448.
  56. Rubinstein, M.R.; Wang, X.; Liu, W.; Hao, Y.; Cai, G.; Han, Y.W. Fusobacterium nucleatum promotes colorectal carcinogenesis by modulating E-cadherin/beta-catenin signaling via its FadA adhesin. Cell Host Microbe 2013, 14, 195–206.
  57. Pai, S.G.; Carneiro, B.A.; Mota, J.M.; Costa, R.; Leite, C.A.; Barroso-Sousa, R.; Giles, F.J. Wnt/beta-catenin pathway: Modulating anti-cancer immune response. J. Hematol. Oncol. 2017, 10, 101.
  58. Zagato, E.; Pozzi, C.; Bertocchi, A.; Schioppa, T.; Saccheri, F.; Guglietta, S.; Rescigno, M. Endogenous murine microbiota member Faecalibaculum rodentium and its human homologue protect from intestinal tumour growth. Nat. Microbiol. 2020, 5, 511–524.
  59. Hanus, M.; Parada-Venegas, D.; Landskron, G.; Wielandt, A.M.; Hurtado, C.; Alvarez, K.; De la Fuente, M. Immune System, Microbiota, and Microbial Metabolites: The Unresolved Triad in Colorectal Cancer Microenvironment. Front. Immunol. 2021, 12, 612826.
  60. Fares, J.; Fares, M.Y.; Khachfe, H.H.; Salhab, H.A.; Fares, Y. Molecular principles of metastasis: A hallmark of cancer revisited. Signal Transduct. Target. Ther. 2020, 5, 28.
  61. Hunter, K.W.; Crawford, N.P.; Alsarraj, J. Mechanisms of metastasis. Breast Cancer Res. 2008, 10, S2.
  62. McCoy, A.N.; Araujo-Perez, F.; Azcarate-Peril, A.; Yeh, J.J.; Sandler, R.S.; Keku, T.O. Fusobacterium is associated with colorectal adenomas. PLoS ONE 2013, 8, e53653.
  63. Sakamoto, Y.; Mima, K.; Ishimoto, T.; Ogata, Y.; Imai, K.; Miyamoto, Y.; Baba, H. Relationship between Fusobacterium nucleatum and anti-tumor immunity in colorectal cancer liver metastasis. Cancer Sci. 2021, 112, 4470–4477.
  64. Wang, S.; Liu, Y.; Li, J.; Zhao, L.; Yan, W.; Lin, B.; Wei, Y. Fusobacterium nucleatum Acts as a Pro-carcinogenic Bacterium in Colorectal Cancer: From Association to Causality. Front. Cell Dev. Biol. 2021, 9, 710165.
  65. Brennan, C.A.; Garrett, W.S. Fusobacterium nucleatum-symbiont, opportunist and oncobacterium. Nat. Rev. Microbiol. 2019, 17, 156–166.
  66. Kaczanowski, S. Apoptosis: Its origin, history, maintenance and the medical implications for cancer and aging. Phys. Biol. 2016, 13, 031001.
  67. D’Arcy, M.S. Cell death: A review of the major forms of apoptosis, necrosis and autophagy. Cell Biol. Int. 2019, 43, 582–592.
  68. Galluzzi, L.; Vitale, I.; Abrams, J.M.; Alnemri, E.S.; Baehrecke, E.H.; Blagosklonny, M.V. Molecular definitions of cell death subroutines: Recommendations of the Nomenclature Committee on Cell Death 2012. Cell Death Differ. 2012, 19, 107–120.
  69. Jan, R.; Chaudhry, G.E. Understanding Apoptosis and Apoptotic Pathways Targeted Cancer Therapeutics. Adv. Pharm. Bull 2019, 9, 205–218.
  70. Zhang, L.; Yu, J. Role of apoptosis in colon cancer biology, therapy, and prevention. Curr. Colorectal. Cancer Rep. 2013, 9, 331–340.
  71. Woznicki, J.A.; Flood, P.; Bustamante-Garrido, M.; Stamou, P.; Moloney, G.; Fanning, A.; Nally, K. Human BCL-G regulates secretion of inflammatory chemokines but is dispensable for induction of apoptosis by IFN-gamma and TNF-alpha in intestinal epithelial cells. Cell Death Dis. 2020, 11, 68.
  72. Tang, W.; Liu, J.; Ma, Y.; Wei, Y.; Liu, J.; Wang, H. Impairment of Intestinal Barrier Function Induced by Early Weaning via Autophagy and Apoptosis Associated with Gut Microbiome and Metabolites. Front. Immunol. 2021, 12, 804870.
  73. Waldecker, M.; Kautenburger, T.; Daumann, H.; Busch, C.; Schrenk, D. Inhibition of histone-deacetylase activity by short-chain fatty acids and some polyphenol metabolites formed in the colon. J. Nutr. Biochem. 2008, 19, 587–593.
  74. Chen, J.; Zhao, K.N.; Vitetta, L. Effects of Intestinal Microbial(-)Elaborated Butyrate on Oncogenic Signaling Pathways. Nutrients 2019, 11, 1026.
  75. Parada Venegas, D.; De la Fuente, M.K.; Landskron, G.; Gonzalez, M.J.; Quera, R.; Dijkstra, G.; Hermoso, M.A. Short Chain Fatty Acids (SCFAs)-Mediated Gut Epithelial and Immune Regulation and Its Relevance for Inflammatory Bowel Diseases. Front. Immunol. 2019, 10, 277.
  76. Tian, Y.; Xu, Q.; Sun, L.; Ye, Y.; Ji, G. Short-chain fatty acids administration is protective in colitis-associated colorectal cancer development. J. Nutr. Biochem. 2018, 57, 103–109.
  77. Wang, T.; Cai, G.; Qiu, Y.; Fei, N.; Zhang, M.; Pang, X.; Zhao, L. Structural segregation of gut microbiota between colorectal cancer patients and healthy volunteers. ISME J. 2012, 6, 320–329.
  78. Salcedo, R.; Worschech, A.; Cardone, M.; Jones, Y.; Gyulai, Z.; Dai, R.-M.; Wang, E.; Ma, W.; Haines, D.; O’Huigin, C. MyD88-mediated signaling prevents development of adenocarcinomas of the colon: Role of interleukin 18. J. Exp. Med. 2010, 207, 1625–1636.
  79. Tye, H.; Yu, C.H.; Simms, L.A.; de Zoete, M.R.; Kim, M.L.; Zakrzewski, M.; Masters, S.L. NLRP1 restricts butyrate producing commensals to exacerbate inflammatory bowel disease. Nat. Commun. 2018, 9, 3728.
  80. Pan, L.J.; Ma, S.Y.; Wen, J.; Zhang, X.Q.; Xing, H.J.; Jia, C.S. Direct contact moxibustion promotes apoptosis of gastric cancer cells in rats by regulating intestinal flora. J. Tradit. Chin. Med. 2021, 41, 943–952.
  81. Zhang, Y.; Zhou, L.; Bao, Y.L.; Wu, Y.; Yu, C.L.; Huang, Y.X.; Sun, Y.; Zheng, L.H.; Li, Y.X. Butyrate induces cell apoptosis through activation of JNK MAP kinase pathway in human colon cancer RKO cells. Chem. Biol. Interact. 2010, 185, 174–181.
  82. Housman, G.; Byler, S.; Heerboth, S.; Lapinska, K.; Longacre, M.; Snyder, N.; Sarkar, S. Drug resistance in cancer: An overview. Cancers 2014, 6, 1769–1792.
  83. Urruticoechea, A.; Alemany, R.; Balart, J.; Villanueva, A.; Vinals, F.; Capella, G. Recent advances in cancer therapy: An overview. Curr. Pharm. Des. 2010, 16, 3–10.
  84. Seager, R.J.; Hajal, C.; Spill, F.; Kamm, R.D.; Zaman, M.H. Dynamic interplay between tumour, stroma and immune system can drive or prevent tumour progression. Converg. Sci. Phys. Oncol. 2017, 3, 034002.
  85. Inthagard, J.; Edwards, J.; Roseweir, A.K. Immunotherapy: Enhancing the efficacy of this promising therapeutic in multiple cancers. Clin. Sci. 2019, 133, 181–193.
  86. Farkona, S.; Diamandis, E.P.; Blasutig, I.M. Cancer immunotherapy: The beginning of the end of cancer? BMC Med. 2016, 14, 73.
  87. Galluzzi, L.; Vacchelli, E.; Bravo-San Pedro, J.M.; Buque, A.; Senovilla, L.; Baracco, E.E.; Kroemer, G. Classification of current anti-cancer immunotherapies. Oncotarget 2014, 5, 12472–12508.
  88. Dine, J.; Gordon, R.; Shames, Y.; Kasler, M.K.; Barton-Burke, M. Immune Checkpoint Inhibitors: An Innovation in Immunotherapy for the Treatment and Management of Patients with Cancer. Asia Pac. J. Oncol. Nurs. 2017, 4, 127–135.
  89. Koury, J.; Lucero, M.; Cato, C.; Chang, L.; Geiger, J.; Henry, D.; Tran, A. Immunotherapies: Exploiting the Immune System for Cancer Treatment. J. Immunol. Res. 2018, 2018, 9585614.
  90. Topalian, S.L.; Hodi, F.S.; Brahmer, J.R.; Gettinger, S.N.; Smith, D.C.; McDermott, D.F.; Sznol, M. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 2012, 366, 2443–2454.
  91. Manz, S.M.; Losa, M.; Fritsch, R.; Scharl, M. Efficacy and side effects of immune checkpoint inhibitors in the treatment of colorectal cancer. Ther. Adv. Gastroenterol. 2021, 14, 17562848211002018.
  92. Vivarelli, S.; Salemi, R.; Candido, S.; Falzone, L.; Santagati, M.; Stefani, S.; Libra, M. Gut Microbiota and Cancer: From Pathogenesis to Therapy. Cancers 2019, 11, 38.
  93. Shui, L.; Yang, X.; Li, J.; Yi, C.; Sun, Q.; Zhu, H. Gut Microbiome as a Potential Factor for Modulating Resistance to Cancer Immunotherapy. Front. Immunol. 2019, 10, 2989.
  94. Vetizou, M.; Pitt, J.M.; Daillere, R.; Lepage, P.; Waldschmitt, N.; Flament, C.; Zitvogel, L. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 2015, 350, 1079–1084.
  95. Silva, M.; Brunner, V.; Tschurtschenthaler, M. Microbiota and Colorectal Cancer: From Gut to Bedside. Front. Pharmacol. 2021, 12, 760280.
  96. Chen, D.; Wu, J.; Jin, D.; Wang, B.; Cao, H. Fecal microbiota transplantation in cancer management: Current status and perspectives. Int. J. Cancer 2019, 145, 2021–2031.
  97. Van Nood, E.; Vrieze, A.; Nieuwdorp, M. Duodenal infusion of donor feces for recurrent Clostridium difficile. N. Engl. J. Med. 2013, 368, 407–415.
  98. Kim, K.O.; Gluck, M. Fecal Microbiota Transplantation: An Update on Clinical Practice. Clin. Endosc. 2019, 52, 137–143.
  99. Rosshart, S.P.; Vassallo, B.G.; Angeletti, D. Wild mouse gut microbiota promotes host fitness and improves disease resistance. Cell 2017, 171, 1015–1028.e13.
  100. Cassidy, A.; Minihane, A.M. The role of metabolism (and the microbiome) in defining the clinical efficacy of dietary flavonoids. Am. J. Clin. Nutr. 2017, 105, 10–22.
  101. Al-Ishaq, R.K.; Overy, A.J.; Busselberg, D. Phytochemicals and Gastrointestinal Cancer: Cellular Mechanisms and Effects to Change Cancer Progression. Biomolecules 2020, 10, 105.
  102. Gali-Muhtasib, H.U.; Younes, I.H.; Karchesy, J.J.; el-Sabban, M.E. Plant tannins inhibit the induction of aberrant crypt foci and colonic tumors by 1,2-dimethylhydrazine in mice. Nutr. Cancer 2001, 39, 108–116.
  103. Al-Ishaq, R.K.; Liskova, A.; Kubatka, P.; Busselberg, D. Enzymatic Metabolism of Flavonoids by Gut Microbiota and Its Impact on Gastrointestinal Cancer. Cancers 2021, 13, 3934.
More
Upload a video for this entry
Information
Subjects: Immunology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , ,
View Times: 838
Revisions: 2 times (View History)
Update Date: 06 May 2022
1000/1000
Hot Most Recent
Academic Video Service