Mol. Cells 2021; 44(5): 356-362
Published online April 23, 2021
https://doi.org/10.14348/molcells.2021.0032
© The Korean Society for Molecular and Cellular Biology
Correspondence to : heungkyu.lee@kaist.ac.kr
This is an open-access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-sa/3.0/.
An increasing number of studies have revealed an interaction between gut microbiota and tumors. The enrichment of specific bacteria strains in the intestines has been found to modulate tumor growth and influence the mechanisms of tumor treatment. Various bacteria are involved in modulating the effects of chemotherapeutic drugs currently used to treat patients with cancer, and they affect not only gastrointestinal tract tumors but also distant organ tumors. In addition, changes in the gut microbiota are known to be involved in the antitumor immune response as well as the modulation of the intestinal immune system. As a result, the gut microbiota plays an important role in modulating the efficacy of immune checkpoint inhibitors. Therefore, gut microbiota could be considered as an adjuvant treatment option with other cancer treatment or as another marker for predicting treatment response. In this review, we examine how gut microbiota affects cancer treatments.
Keywords cancer, cancer therapy, gut microbiota, immune checkpoint inhibitors
Malignant tumors are among the most fatal diseases in modern society (GBD 2015 Disease and Injury Incidence and Prevalence Collaborators, 2016). Carcinogenesis is the result of the stochastic intracellular accumulation of mutations during DNA replication or environmental exposure of various carcinogens, infectious agents, ultraviolet radiation, and toxic substances (Tomasetti and Vogelstein, 2015). Due to the efforts of many researchers, the overall death rate for cancer has been decreasing (Hashim et al., 2016). In addition to the conventional therapy of surgery, chemotherapy and radiation therapy, new treatment therapies including targeted therapy and immunotherapy such as immune checkpoint inhibitors (ICIs) which have recently attracted attention as novel therapeutic drugs, have been emerging for effective treatment modalities (Waldmann, 2003). As a result, the treatment results of patients with cancer are gradually improving (Hashim et al., 2016). However, patients with advanced stage or unfavorable malignant tumors still face a poor prognosis (Coleman et al., 2013; Miller et al., 2019; Nevala-Plagemann et al., 2020). Recently, attention has been drawn to the role of intestinal microbes to influence the therapeutic efficacy of tumor treatments (Louis et al., 2014; Wang et al., 2012; Zhu et al., 2013; Zitvogel et al., 2015).
The intestines of a healthy person contain a complex network of approximately 39 trillion bacteria (Sekirov et al., 2010; Sender et al., 2016). In the last few decades, many studies have been conducted to examine the effects of intestinal microbes on the host (Jandhyala et al., 2015). Intestinal microbes play important roles in normal physiologic development (Ku et al., 2020). In addition, the dysbiosis of intestinal microflora is associated with various diseases ranging from nervous system diseases to metabolic-related and gastrointestinal diseases (Shui et al., 2019). Some evidences indicate that these diseases are associated with tumors (Kuen et al., 2020; Louis et al., 2014; Wang et al., 2012; Zhu et al., 2013; Zitvogel et al., 2015). In the early 20th century, most studies on the relationship between these intestinal microbes and tumors were limited to the pathogenic aspect of microbes (Shui et al., 2019). As a result, many researchers have focused on microorganisms that cause tumors including
Because antitumor immune responses have been regarded as important in tumor treatment in recent years, attention has also been paid to the effect of intestinal microbes on tumor immunity (Viaud et al., 2013). Compelling evidence indicates that intestinal microbes influence intestinal immune barriers such as intraepithelial lymphocytes development (Jung et al., 2019). Literature has shown that gut microbiota-derived short chain fatty acids play an important role in regulating the adaptive immune response (Kim et al., 2014). Regarding the treatment with ICIs, changes in intestinal microflora affect not only the effects of these ICIs but also their therapeutic toxicity (Choi and Lee, 2020; Viaud et al., 2013). For example, microbial control has been used as a novel therapeutic method to improve the effectiveness of ICIs (Gopalakrishnan et al., 2018; Vetizou et al., 2015). This review discusses the latest studies that have examined the effect of intestinal microbes on tumor growth and antitumor therapy including immunosuppressants.
Antibiotics have saved humanity from infectious diseases and are commonly used drugs in modern medical circumstances (Iizumi et al., 2017). However, they cause dysbiosis of intestinal microbes, which have linked to various disease in recent reports. In addition, gut microbiota depletion due to antibiotics use has various effects on tumor progression and the immune environment.
Recent literature has shown that the loss of intestinal microflora by antibiotics use results in an antitumor effect (Sethi et al., 2018). Several studies have been conducted on gut microbiota depletion using oral antibiotics administration in various mouse tumor models. In one study, the continuous administration of a broad-spectrum antibiotics cocktail of vancomycin, neomycin, metronidazole, ampicillin, and amphotericin B reduced the growth of the tumor burden in pancreatic cancer, colon cancer and melanoma animal models (Sethi et al., 2018). This result was attributed to increased production of interferon-gamma (IFN-γ), interleukin (IL) 17a, and IL-10 in T cells.
Vancomycin-depleted gut microbiota not only affects the antitumor immune response but also increases the antitumor activity of radiation therapy (Uribe-Herranz et al., 2020). In the melanoma mouse model, the vancomycin-depleted group exhibited increased cross-presentation of tumor-associated antigens to cytotoxic T lymphocytes and IFN-γ production (Uribe-Herranz et al., 2020). However, butyrate secreted from vancomycin-sensitive bacteria plays an important role in enhancing antitumor activity by radiotherapy. As a result, the reduction of specific strains caused by these antibiotics decreases the synergistic action of depleted gut microbiota.
However, antibiotics-induced gut microbiota depletion and dysbiosis have also been shown to increase tumor growth and progression. In this regard, reports indicate that certain microbial metabolites promote tumor growth (Laborda-Illanes et al., 2020). In addition, dysbiosis induced through a broad-spectrum antibiotics cocktail in mouse colon cancer model can lead to increases colon tumor susceptibility via the induction of CD8+ IFN-γ+ T cells (Yu et al., 2020). In this study, increased CD8+ IFN-γ+ T cells in the colon lamina propria induced inflammation in the serous membrane, affecting tumor development. After tumor formation, the antitumor immune response was suppressed by inducing the exhaustion of functional CD8+ T cells (Yu et al., 2020). The use of broad-spectrum antibiotics also affects dendritic cells. The depleted gut microbiota by vancomycin increased the number of systemic CD8a+ dendritic cells, thereby enhancing the effect of adoptive T-cell therapy and increasing IL-12 cytokine in mice (Xu et al., 2017).
The effect of gut microbiota depletion by antibiotics has been reported as both antitumoral and protumoral activity. Although there is still no clear mechanism, this varied tumor response seems to be due to differences in affected bacteria strains by antibiotics and tumor types.
The use of an antibiotics-induced dysbiosis model can confirm a variety of reactions to tumor growth. However, the decrease or change in intestinal microflora by antibiotics has limitations in confirming tumor response to specific bacteria species. Several studies have reported that the enrichment of specific bacteria in animal models is associated with tumor growth.
Some microbial-derived short-chain fatty acids may have anticancer effects.
In addition, several studies have shown that
As the link between intestinal microbes and tumor growth has been gradually revealed, studies have examined the synergistic effects of these microbes with antitumor treatments (Roy and Trinchieri, 2017). In addition to existing chemotherapeutic agents, their synergistic effects with radiation therapy and ICIs have been newly discovered (Roy and Trinchieri, 2017). There are some changes in gene expression associated with drug metabolism by the RNA-seq analysis of hepatic drug-processing genes in the germ-free mice (Selwyn et al., 2015). Therefore, intestinal microbes may be involved in the metabolism of antitumor drugs. Continuous exposure to cyclophosphamide-induced changes in intestinal microflora in tumor-bearing mice (Viaud et al., 2013). Table 1 lists the intestinal microbes found to modulate the anticancer effect of chemotherapeutic agents, as described in various papers.
The effect of platinum-based chemotherapeutic agents such as oxaliplatin and CpG oligodeoxynucleotides was poor in mice treated with antibiotics (Iida et al., 2013). In the mice administered antibiotics, cytokine secretion and ROS were reduced, resulting in decreased tumor necrosis and cytotoxicity of the antitumor therapy. Correspondingly, in a study of patients with chronic lymphocytic leukemia, tumor progression was observed and the overall survival rate decreased in patients who were prescribed anti-gram-positive antibiotics (Pflug et al., 2016). As a result, anti-gram-positive antibiotics were suggested to negatively affect the cytotoxicity of cyclophosphamide and cisplatin.
As previously shown, continuous exposure of cyclophosphamide-induced changes in the intestinal microflora, and it was confirmed that gram-positive bacteria including
In a colon cancer model, intratumoral bacteria were found to modulate the effect of chemotherapy. Gemcitabine is converted into an inactivated form by specific gammaproteobacteria present in the tumor, reducing the antitumor effect of the drug. The bacteria contain an enzyme in the form of a long isoform of cytidine deaminase. The suppression of the anticancer effect disappeared when bacteria were killed through antibiotics treatment in colon cancer mouse model (Geller et al., 2017). Moreover, another study, related to gemcitabine, confirmed that the presence of bacteria in an
In the study of
Beta-glucuronidase, an enzyme produced by intestinal microbes, plays an important role in converting inactivated irinotecan into its active form, SN-38. Taking antibiotics consequently inhibits this metabolism, thereby reducing the antitumor efficacy of irinotecan (Kodawara et al., 2016). In addition, beta-glucuronidase is involved in the activation of ciprofloxacin, demonstrating that an important role of intestinal microbes in determining the effectiveness of chemotherapy (Alexander et al., 2017; Wallace et al., 2010).
Among the mechanisms reviewed above, some intestinal microbes contribute to the anticancer effect by improving the immune response. With this background, studies have examined the synergistic effects of intestinal microbes with ICIs and found that they can play an important role in increasing the effects of ICIs (Fig. 1).
Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) inhibitors are one of the ICIs used in clinical practice currently because of its recognized effectiveness.
The development of metagenomics has helped to elucidate the relationship between ICIs and intestinal microbes. In a FMT study of patients with metastatic melanoma, an analysis of 16S rRNA sequencing data performed after transplantation but before ICI treatment showed that patients who responded to ICIs had more strains of
The study of interactions between intestinal microbiota and malignant tumors has a short history. To date, various strains are known to affect tumor treatment, but no studies have clearly described the corresponding mechanism. In addition, methods to use these intestinal microorganisms as therapeutic agents such as probiotics or FMT require further investigation. Studies on the clinical application of gut microbiota are ongoing for the purpose of predicting and diagnosing cancer, as well as the potential for improving the effect or alleviating side effects of antitumor treatment (Wong et al., 2019). In recent studies, FMT can overcomes the resistance to immunotherapy in melanoma patients whose cancers do not respond (Baruch et al., 2021; Davar et al., 2021). These clinical results suggest that gut microbiota modulation through FMT can be effectively used for cancer treatment. Because the intestinal environment is a system in which microorganisms form a huge network, more research needs to be conducted to understand this complex ecosystem and apply it for treatment.
This research was supported by the Bio & Medical Technology Development Program of the National Research Foundation (NRF) & funded by the Korean government (MSIT) (No. 2021M3A9H3015688).
J.K. and H.K.L. conceived and wrote the manuscript. H.K.L. secured funding.
The authors have no potential conflicts of interest to disclose.
Summary of intestinal microorganism modulating the efficacy of chemotherapeutic drugs
Bacteria | Chemotherapeutic agent | Interaction mechanism | Reference |
---|---|---|---|
Lactobacillus species | Cyclophosphamide | Promoted Th17 and Th1 cell response during cyclophosphamide treatment | Viaud et al. (2013) |
Segmented filamentous bacteria | |||
Cyclophosphamide | Associated with increased CD8/ Treg ratio | Daillere et al. (2016) | |
Infiltration of interferon-g-producing gd-T cells | |||
5-FU | Distribution of bacterial deoxynucleotide pools regulates the effect of 5-FU | Scott et al. (2017) | |
5-FU | Modulation autophagy pathway and inhibit tumor cell apoptosis | Yu et al. (2017) | |
Oxaliplatin | |||
Gammaproteobacteria | Gemcitabine | Gemcitabine was converted into an inactivated form by long isoform of cytidine deaminase | Geller et al. (2017) |
β-glucuronidase producers | Irinotecan | Converting inactivated irinotecan into the active form, SN-38 | Kodawara et al. (2016) |
Ciprofloxacin | Involved in the activation of ciprofloxacin | Alexander et al. (2017);Wallace et al. (2010) |
Mol. Cells 2021; 44(5): 356-362
Published online May 31, 2021 https://doi.org/10.14348/molcells.2021.0032
Copyright © The Korean Society for Molecular and Cellular Biology.
Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
Correspondence to:heungkyu.lee@kaist.ac.kr
This is an open-access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-sa/3.0/.
An increasing number of studies have revealed an interaction between gut microbiota and tumors. The enrichment of specific bacteria strains in the intestines has been found to modulate tumor growth and influence the mechanisms of tumor treatment. Various bacteria are involved in modulating the effects of chemotherapeutic drugs currently used to treat patients with cancer, and they affect not only gastrointestinal tract tumors but also distant organ tumors. In addition, changes in the gut microbiota are known to be involved in the antitumor immune response as well as the modulation of the intestinal immune system. As a result, the gut microbiota plays an important role in modulating the efficacy of immune checkpoint inhibitors. Therefore, gut microbiota could be considered as an adjuvant treatment option with other cancer treatment or as another marker for predicting treatment response. In this review, we examine how gut microbiota affects cancer treatments.
Keywords: cancer, cancer therapy, gut microbiota, immune checkpoint inhibitors
Malignant tumors are among the most fatal diseases in modern society (GBD 2015 Disease and Injury Incidence and Prevalence Collaborators, 2016). Carcinogenesis is the result of the stochastic intracellular accumulation of mutations during DNA replication or environmental exposure of various carcinogens, infectious agents, ultraviolet radiation, and toxic substances (Tomasetti and Vogelstein, 2015). Due to the efforts of many researchers, the overall death rate for cancer has been decreasing (Hashim et al., 2016). In addition to the conventional therapy of surgery, chemotherapy and radiation therapy, new treatment therapies including targeted therapy and immunotherapy such as immune checkpoint inhibitors (ICIs) which have recently attracted attention as novel therapeutic drugs, have been emerging for effective treatment modalities (Waldmann, 2003). As a result, the treatment results of patients with cancer are gradually improving (Hashim et al., 2016). However, patients with advanced stage or unfavorable malignant tumors still face a poor prognosis (Coleman et al., 2013; Miller et al., 2019; Nevala-Plagemann et al., 2020). Recently, attention has been drawn to the role of intestinal microbes to influence the therapeutic efficacy of tumor treatments (Louis et al., 2014; Wang et al., 2012; Zhu et al., 2013; Zitvogel et al., 2015).
The intestines of a healthy person contain a complex network of approximately 39 trillion bacteria (Sekirov et al., 2010; Sender et al., 2016). In the last few decades, many studies have been conducted to examine the effects of intestinal microbes on the host (Jandhyala et al., 2015). Intestinal microbes play important roles in normal physiologic development (Ku et al., 2020). In addition, the dysbiosis of intestinal microflora is associated with various diseases ranging from nervous system diseases to metabolic-related and gastrointestinal diseases (Shui et al., 2019). Some evidences indicate that these diseases are associated with tumors (Kuen et al., 2020; Louis et al., 2014; Wang et al., 2012; Zhu et al., 2013; Zitvogel et al., 2015). In the early 20th century, most studies on the relationship between these intestinal microbes and tumors were limited to the pathogenic aspect of microbes (Shui et al., 2019). As a result, many researchers have focused on microorganisms that cause tumors including
Because antitumor immune responses have been regarded as important in tumor treatment in recent years, attention has also been paid to the effect of intestinal microbes on tumor immunity (Viaud et al., 2013). Compelling evidence indicates that intestinal microbes influence intestinal immune barriers such as intraepithelial lymphocytes development (Jung et al., 2019). Literature has shown that gut microbiota-derived short chain fatty acids play an important role in regulating the adaptive immune response (Kim et al., 2014). Regarding the treatment with ICIs, changes in intestinal microflora affect not only the effects of these ICIs but also their therapeutic toxicity (Choi and Lee, 2020; Viaud et al., 2013). For example, microbial control has been used as a novel therapeutic method to improve the effectiveness of ICIs (Gopalakrishnan et al., 2018; Vetizou et al., 2015). This review discusses the latest studies that have examined the effect of intestinal microbes on tumor growth and antitumor therapy including immunosuppressants.
Antibiotics have saved humanity from infectious diseases and are commonly used drugs in modern medical circumstances (Iizumi et al., 2017). However, they cause dysbiosis of intestinal microbes, which have linked to various disease in recent reports. In addition, gut microbiota depletion due to antibiotics use has various effects on tumor progression and the immune environment.
Recent literature has shown that the loss of intestinal microflora by antibiotics use results in an antitumor effect (Sethi et al., 2018). Several studies have been conducted on gut microbiota depletion using oral antibiotics administration in various mouse tumor models. In one study, the continuous administration of a broad-spectrum antibiotics cocktail of vancomycin, neomycin, metronidazole, ampicillin, and amphotericin B reduced the growth of the tumor burden in pancreatic cancer, colon cancer and melanoma animal models (Sethi et al., 2018). This result was attributed to increased production of interferon-gamma (IFN-γ), interleukin (IL) 17a, and IL-10 in T cells.
Vancomycin-depleted gut microbiota not only affects the antitumor immune response but also increases the antitumor activity of radiation therapy (Uribe-Herranz et al., 2020). In the melanoma mouse model, the vancomycin-depleted group exhibited increased cross-presentation of tumor-associated antigens to cytotoxic T lymphocytes and IFN-γ production (Uribe-Herranz et al., 2020). However, butyrate secreted from vancomycin-sensitive bacteria plays an important role in enhancing antitumor activity by radiotherapy. As a result, the reduction of specific strains caused by these antibiotics decreases the synergistic action of depleted gut microbiota.
However, antibiotics-induced gut microbiota depletion and dysbiosis have also been shown to increase tumor growth and progression. In this regard, reports indicate that certain microbial metabolites promote tumor growth (Laborda-Illanes et al., 2020). In addition, dysbiosis induced through a broad-spectrum antibiotics cocktail in mouse colon cancer model can lead to increases colon tumor susceptibility via the induction of CD8+ IFN-γ+ T cells (Yu et al., 2020). In this study, increased CD8+ IFN-γ+ T cells in the colon lamina propria induced inflammation in the serous membrane, affecting tumor development. After tumor formation, the antitumor immune response was suppressed by inducing the exhaustion of functional CD8+ T cells (Yu et al., 2020). The use of broad-spectrum antibiotics also affects dendritic cells. The depleted gut microbiota by vancomycin increased the number of systemic CD8a+ dendritic cells, thereby enhancing the effect of adoptive T-cell therapy and increasing IL-12 cytokine in mice (Xu et al., 2017).
The effect of gut microbiota depletion by antibiotics has been reported as both antitumoral and protumoral activity. Although there is still no clear mechanism, this varied tumor response seems to be due to differences in affected bacteria strains by antibiotics and tumor types.
The use of an antibiotics-induced dysbiosis model can confirm a variety of reactions to tumor growth. However, the decrease or change in intestinal microflora by antibiotics has limitations in confirming tumor response to specific bacteria species. Several studies have reported that the enrichment of specific bacteria in animal models is associated with tumor growth.
Some microbial-derived short-chain fatty acids may have anticancer effects.
In addition, several studies have shown that
As the link between intestinal microbes and tumor growth has been gradually revealed, studies have examined the synergistic effects of these microbes with antitumor treatments (Roy and Trinchieri, 2017). In addition to existing chemotherapeutic agents, their synergistic effects with radiation therapy and ICIs have been newly discovered (Roy and Trinchieri, 2017). There are some changes in gene expression associated with drug metabolism by the RNA-seq analysis of hepatic drug-processing genes in the germ-free mice (Selwyn et al., 2015). Therefore, intestinal microbes may be involved in the metabolism of antitumor drugs. Continuous exposure to cyclophosphamide-induced changes in intestinal microflora in tumor-bearing mice (Viaud et al., 2013). Table 1 lists the intestinal microbes found to modulate the anticancer effect of chemotherapeutic agents, as described in various papers.
The effect of platinum-based chemotherapeutic agents such as oxaliplatin and CpG oligodeoxynucleotides was poor in mice treated with antibiotics (Iida et al., 2013). In the mice administered antibiotics, cytokine secretion and ROS were reduced, resulting in decreased tumor necrosis and cytotoxicity of the antitumor therapy. Correspondingly, in a study of patients with chronic lymphocytic leukemia, tumor progression was observed and the overall survival rate decreased in patients who were prescribed anti-gram-positive antibiotics (Pflug et al., 2016). As a result, anti-gram-positive antibiotics were suggested to negatively affect the cytotoxicity of cyclophosphamide and cisplatin.
As previously shown, continuous exposure of cyclophosphamide-induced changes in the intestinal microflora, and it was confirmed that gram-positive bacteria including
In a colon cancer model, intratumoral bacteria were found to modulate the effect of chemotherapy. Gemcitabine is converted into an inactivated form by specific gammaproteobacteria present in the tumor, reducing the antitumor effect of the drug. The bacteria contain an enzyme in the form of a long isoform of cytidine deaminase. The suppression of the anticancer effect disappeared when bacteria were killed through antibiotics treatment in colon cancer mouse model (Geller et al., 2017). Moreover, another study, related to gemcitabine, confirmed that the presence of bacteria in an
In the study of
Beta-glucuronidase, an enzyme produced by intestinal microbes, plays an important role in converting inactivated irinotecan into its active form, SN-38. Taking antibiotics consequently inhibits this metabolism, thereby reducing the antitumor efficacy of irinotecan (Kodawara et al., 2016). In addition, beta-glucuronidase is involved in the activation of ciprofloxacin, demonstrating that an important role of intestinal microbes in determining the effectiveness of chemotherapy (Alexander et al., 2017; Wallace et al., 2010).
Among the mechanisms reviewed above, some intestinal microbes contribute to the anticancer effect by improving the immune response. With this background, studies have examined the synergistic effects of intestinal microbes with ICIs and found that they can play an important role in increasing the effects of ICIs (Fig. 1).
Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) inhibitors are one of the ICIs used in clinical practice currently because of its recognized effectiveness.
The development of metagenomics has helped to elucidate the relationship between ICIs and intestinal microbes. In a FMT study of patients with metastatic melanoma, an analysis of 16S rRNA sequencing data performed after transplantation but before ICI treatment showed that patients who responded to ICIs had more strains of
The study of interactions between intestinal microbiota and malignant tumors has a short history. To date, various strains are known to affect tumor treatment, but no studies have clearly described the corresponding mechanism. In addition, methods to use these intestinal microorganisms as therapeutic agents such as probiotics or FMT require further investigation. Studies on the clinical application of gut microbiota are ongoing for the purpose of predicting and diagnosing cancer, as well as the potential for improving the effect or alleviating side effects of antitumor treatment (Wong et al., 2019). In recent studies, FMT can overcomes the resistance to immunotherapy in melanoma patients whose cancers do not respond (Baruch et al., 2021; Davar et al., 2021). These clinical results suggest that gut microbiota modulation through FMT can be effectively used for cancer treatment. Because the intestinal environment is a system in which microorganisms form a huge network, more research needs to be conducted to understand this complex ecosystem and apply it for treatment.
This research was supported by the Bio & Medical Technology Development Program of the National Research Foundation (NRF) & funded by the Korean government (MSIT) (No. 2021M3A9H3015688).
J.K. and H.K.L. conceived and wrote the manuscript. H.K.L. secured funding.
The authors have no potential conflicts of interest to disclose.
Summary of intestinal microorganism modulating the efficacy of chemotherapeutic drugs
Bacteria | Chemotherapeutic agent | Interaction mechanism | Reference |
---|---|---|---|
Lactobacillus species | Cyclophosphamide | Promoted Th17 and Th1 cell response during cyclophosphamide treatment |
Viaud et al. (2013) |
Segmented filamentous bacteria | |||
|
Cyclophosphamide | Associated with increased CD8/ Treg ratio |
Daillere et al. (2016) |
|
Infiltration of interferon-g-producing gd-T cells | ||
|
5-FU | Distribution of bacterial deoxynucleotide pools regulates the effect of 5-FU |
Scott et al. (2017) |
|
5-FU | Modulation autophagy pathway and inhibit tumor cell apoptosis |
Yu et al. (2017) |
Oxaliplatin | |||
Gammaproteobacteria | Gemcitabine | Gemcitabine was converted into an inactivated form by long isoform of cytidine deaminase |
Geller et al. (2017) |
β-glucuronidase producers | Irinotecan | Converting inactivated irinotecan into the active form, SN-38 |
Kodawara et al. (2016) |
Ciprofloxacin | Involved in the activation of ciprofloxacin |
Alexander et al. (2017);Wallace et al. (2010) |
. Summary of intestinal microorganism modulating the efficacy of chemotherapeutic drugs.
Bacteria | Chemotherapeutic agent | Interaction mechanism | Reference |
---|---|---|---|
Lactobacillus species | Cyclophosphamide | Promoted Th17 and Th1 cell response during cyclophosphamide treatment | Viaud et al. (2013) |
Segmented filamentous bacteria | |||
Cyclophosphamide | Associated with increased CD8/ Treg ratio | Daillere et al. (2016) | |
Infiltration of interferon-g-producing gd-T cells | |||
5-FU | Distribution of bacterial deoxynucleotide pools regulates the effect of 5-FU | Scott et al. (2017) | |
5-FU | Modulation autophagy pathway and inhibit tumor cell apoptosis | Yu et al. (2017) | |
Oxaliplatin | |||
Gammaproteobacteria | Gemcitabine | Gemcitabine was converted into an inactivated form by long isoform of cytidine deaminase | Geller et al. (2017) |
β-glucuronidase producers | Irinotecan | Converting inactivated irinotecan into the active form, SN-38 | Kodawara et al. (2016) |
Ciprofloxacin | Involved in the activation of ciprofloxacin | Alexander et al. (2017);Wallace et al. (2010) |
Christopher J. Occhiuto, Jessica A. Moerland, Ana S. Leal, Kathleen A. Gallo, and Karen T. Liby
Mol. Cells 2023; 46(3): 176-186 https://doi.org/10.14348/molcells.2023.2191Aryatara Shakya, Nicholas W. McKee, Matthew Dodson, Eli Chapman, and Donna D. Zhang
Mol. Cells 2023; 46(3): 165-175 https://doi.org/10.14348/molcells.2023.0005Takafumi Suzuki, Jun Takahashi, and Masayuki Yamamoto
Mol. Cells 2023; 46(3): 133-141 https://doi.org/10.14348/molcells.2023.0028