Next Article in Journal
Potential Therapeutic Improvements in Prostate Cancer Treatment Using Pencil Beam Scanning Proton Therapy with LETd Optimization and Disease-Specific RBE Models
Next Article in Special Issue
Exosomes—Promising Carriers for Regulatory Therapy in Oncology
Previous Article in Journal
Drug Response of Patient-Derived Lung Cancer Cells Predicts Clinical Outcomes of Targeted Therapy
Previous Article in Special Issue
The Mechanism of Oxymatrine Targeting miR-27a-3p/PPAR-γ Signaling Pathway through m6A Modification to Regulate the Influence on Hemangioma Stem Cells on Propranolol Resistance
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Microbiome Modulates the Immune System to Influence Cancer Therapy

by
Ruchi Roy
1,*,† and
Sunil Kumar Singh
2,*,†
1
UICentre for Drug Discovery, The University of Illinois, Chicago, IL 60612, USA
2
Department of Surgery, Division of Surgical Oncology, The University of Illinois at Chicago, Chicago, IL 60612, USA
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2024, 16(4), 779; https://doi.org/10.3390/cancers16040779
Submission received: 22 January 2024 / Revised: 8 February 2024 / Accepted: 8 February 2024 / Published: 14 February 2024
(This article belongs to the Special Issue Cancer Therapy: Where We Are and Where We Need to Go)

Abstract

:

Simple Summary

This review discussed the importance of the gut microbiome for cancer patients and how disturbances in the microbiome can lead to unwanted side effects during cancer treatment. The way in which a variety of foods can influence the microbiota and its abundance was described. Not only this, the effects of antibiotics can also modulate the abundance of the gut microbiome and its diversity. Alterations in the bacterial population can help achieve better immune responses and maintain the intestinal barrier’s integrity. In this regard, this article also discussed several advanced approaches utilizing the advantages of probiotics along with immunotherapies.

Abstract

The gut microbiota composition can affect the tumor microenvironment and its interaction with the immune system, thereby having implications for treatment predictions. This article reviews the studies available to better understand how the gut microbiome helps the immune system fight cancer. To describe this fact, different mechanisms and approaches utilizing probiotics to improve advancements in cancer treatment will be discussed. Moreover, not only calorie intake but also the variety and quality of diet can influence cancer patients’ immunotherapy treatment because dietary patterns can impair immunological activities either by stimulating or suppressing innate and adaptive immunity. Therefore, it is interesting and critical to understand gut microbiome composition as a biomarker to predict cancer immunotherapy outcomes and responses. Here, more emphasis will be given to the recent development in immunotherapies utilizing microbiota to improve cancer therapies, which is beneficial for cancer patients.

1. Introduction

Cancer is the second leading cause of death worldwide according to the World Health Organization (WHO), affecting over 9 million people worldwide [1]. Epidemiological studies have shown that diet is crucial for influencing the risk of cancer. In 2018, the WHO emphasized the association between diet and lifestyle-related risk factors and the burden of diseases [2]. Evidence supports that several cancers’ development and treatment can be affected by modifying diet and nutrition [3]. However, the association between diet and cancer cannot be completely derived, sometimes due to inherent biases and some other factors. These factors include environmental stress, eating habits, alcohol, obesity, inappropriate physical activity, high blood glucose, cholesterol level, and fruit and vegetable intake, which accounted for 18% and 25% of the world’s death burden in low- and middle-income countries and high-income countries, respectively [4,5]. Therefore, studies are more focused on restricting diet and energy intake to influence cancer development and its treatment [6]. Here, we describe the quality of diet that can influence cancer treatment and the biomarkers associated with digestive tract cancers. Although there are therapeutic strategies available to treat cancer patients, they do not completely enhance their lifestyles and can cause them to suffer associated side effects. These treatments greatly impact their day-to-day lives due to the pain associated with them, particularly chemotherapy and radiotherapy, which cause severe abdominal pain, diarrhea, mucositis, vomiting, constipation, etc. [7]. These therapies alter the composition of the intestinal microbiota, i.e., dysbiosis, leading to immunological dysregulation and sometimes resistance to some bacteria. For example, paclitaxel, a known chemotherapeutic agent, not only affects the gut microbiome, colonic tissue integrity, and microglia activation but also increases some pro-inflammatory agents like matrix metalloproteinase 9 (MMP9) and tumor necrosis factor-alpha (TNF-α) that alter bacterial diversity in the colon [8]. Nonetheless, gut microbiota may accelerate the development of cancer in several ways, such as by causing DNA damage or genetic alteration, affecting cell cycle progression through its products, making pro-tumorigenic microbial niches, including biofilms, or creating immune dysregulation.
Thus, in this article, we discuss how any diseased condition or even cancer development can make changes to the gut microbiota and its influence on immune responses. Moreover, we address how the side effects or symptoms associated with cancer treatment can be cured or alleviated with the administration of probiotics, which can also help or facilitate cancer treatment (Figure 1). In addition, we discuss in detail how various treatment procedures involving the usage of live microorganisms (probiotics) or dietary fiber fermented by intestinal microorganisms (prebiotics) are being studied and used to help the growth and abundance of friendly bacteria modify the gut microbiota, preventing dysbiosis and maintaining their homeostasis condition to control cancer progression.

2. Poor Quality of Food Increases the Risk of Cancer

In 2018, the World Cancer Research Fund reported an inconclusive association between fruits or vegetables in the diet and the risk of any cancer [2]. Usually, the risk of cancer in vegetarians is less compared to nonvegetarians because their diet is more dependent on fruit and vegetables than nonvegetarians, but this cannot be true for each and every individual cancer type [9]. It is evident that the long-term health of vegetarians is comparatively better than that of nonvegetarians due to a lower prevalence of health issues such as weight gain, obesity, diabetes, stroke, and cancer. The intake of processed meat and cheeses could be one of the reasons for increasing cancer risk because of their high content of nitrite/nitrate [10,11]. Also, the International Agency for Research on Cancer (IARC) in 2018 documented the toxic effects of processed or unprocessed meat due to the presence of haem iron, which leads to an enhanced formation of nitroso compounds in the gastrointestinal tract, e.g., S-nitrosothiols and the nitrosyl heme [12,13]. These nitroso compounds can alkylate guanine to form the promutagenic DNA lesions O6-methylguanine and O6-carboxymethylguanine. This mutation can cause cancer if not repaired. Moreover, the cooking of meat at high temperatures can produce mutagenic heterocyclic amines and polycyclic aromatic hydrocarbons, which can also be one of the reasons that increase the risk of cancer [14].
A detailed study by Reynolds in 2019 explained the association between carbohydrate quality and human health by performing extensive meta-analyses of prospective studies and reviewing systematic published reviews from databases. Their findings, based on a thorough search of the literature available in databases such as PubMed, Embase, Ovid MEDLINE, and the Cochrane Central Register of Controlled Trials, suggested that higher intakes of whole grains were associated with a 15 to 30% reduction in the risk for all critical outcomes, including cardiovascular-associated mortality, colorectal cancer, and type 2 diabetes [15]. High dietary folate [16] and high dietary fiber consumption can reduce the rate of colorectal cancer. Especially cereal fiber and wholegrain cereals are more protective than fiber from fruit or vegetables [15,17]. However, dietary fibers are controversial and classified as probable causes of cancer as per the World Cancer Research Fund (WCRF) and the American Institute for Cancer Research (AICR) [10]. Reports from the WCRF and the AICR provide updates on specific cancers and guidance for cancer patients and survivors on diet and lifestyle changes. These reports also benefit individuals who are at risk due to genetic predisposition or carcinogenic exposures [10]. Among the most probable causes associated with cancer risk is alcohol consumption, as it can lead to the development of cirrhosis and alcoholic hepatitis. The second most probable cause of cancer is overweight and obesity [18]. An association between vegetable intake and cancer risk has been established among women but not observed among men. A detailed study, including several parameters such as age, sex, smoking, energy intake, ethnicity, and other potential dietary factors, revealed a higher intake of nonstarchy vegetables, salt, pickled food, and processed meat is inversely associated with the risk of bladder and gastric cancer [19,20,21].
Foods containing vitamin C, calcium supplements [22], and folate supplements help to suppress the development of tumors in normal tissues [23]. Folate is required for DNA synthesis and replication, and its deficiency can result in ineffective DNA synthesis and the transformation of normal cells into neoplastic ones. Despite these components being added as a supplement factor in fortified food as a protective factor that may decrease the risk of prostate cancer, folic acid is listed as a nutrient that may induce prostate cancer risk. However, there are many controversial reports on the association of the intake of folate with cancer; for example, studies have not reached a sufficient significance level to comment on its direct associations in the case of prostate cancer [24,25]. A dose–response meta-analysis-based study by Wang et al. summarized the evidence regarding the associations of folate intake and serum folate levels with prostate cancer risk [25]. They found no significant effect of dietary folate intake on prostate risk; however, increased serum folate levels can be associated with the risk of prostate cancer. In addition, metabolomics studies by Liss et al. identified significantly altered folate and arginine pathways in the gut microbiome, which suggests that gut bacteria might affect the risk of prostate cancer [26]. Therefore, it is difficult to conclude the importance of folate in prostate cancer.
Similarly, the consumption of coffee reduces cancer risk due to the presence of strong antioxidants and anti-inflammatory agents, such as polyphenolic compounds. A bibliographic study by Pauwels et al., 2021, based on a search on PubMed and Embase, identified that intake of coffee is inversely associated with the risk of hepatocellular cancer and breast cancer among postmenopausal women [21]. The reason for this association and the beneficiary effect of coffee was ascribed to the presence of some bioactive components such as caffeine, cafestol, kahweol, and chlorogenic acids. The protective role of coffee was investigated and confirmed by another population-based case–control study, where coffee was proposed as a protective factor for colorectal cancer because it reduces the cancer risk by 26% [27]. However, this search could not comment clearly on this association with other cancer types, including esophagus, pancreas, colorectum, kidneys, bladder, ovaries, and prostate, because the results were inconclusive or the data were insignificant in the reports found. Thus, straightforward evidence is needed before recommending any food or beverage consumption.
A Mediterranean-style dietary pattern (MSDP) also strongly lowers the risk of cancer, as shown in a semi-quantitative food frequency questionnaire-based exam involving 2966 participants of the Framingham Offspring who were free of prevalent cancer. This study showed the beneficial effects of an MSDP on cancer risk. In this study, MSDPs had a beneficial effect on women, showing a lower risk of cancer as compared to men, who have higher adherence to MSDPs [28].
The role of sugar and salt is important for the taste of food, but if they are taken in a disproportionate amount, they can also pose a risk to health. A study by Wu et al., 2021, performed a systematic review and meta-analysis to determine the association of salt intake with gastric cancer morbidity and mortality. This cohort study observed that highly but not moderately pickled food intake increases gastric cancer risk, whereas no effect of moderate or high salted fish intake was associated with gastric cancer risk. Moreover, it is advisable to not consume fermented Cantonese-style salted fish or to have a high intake of processed meat, as they can be associated with a higher risk of gastric cancer [29,30]. Another study by Strumylaite et al., 2006, reported that the consumption of salted mushrooms might increase the risk of gastric cancer for people who like salty food, salt-preserved meat, and fish [31].
Contaminated food with certain fungi that are found on crops may also increase the risk of liver cancer. Toxins produced by Aspergillus flavus and Aspergillus parasiticus, i.e., aflatoxins, which are abundant in warm and humid regions of the world, are known to be associated with an increased risk of liver cancer [32]. Not only can contaminated food be the cause of an increased risk of cancer, but drinking water contaminated with arsenic can also cause solid cancers of the lungs, urinary bladder, skin, kidneys, liver, and prostate. Arsenic is a known carcinogen that can enter the body through exposure to arsenic-contaminated drinking water. This was shown in a recent cohort study by Issanov et al., 2023, and Lin et al., 2022, where they highlighted the relationship between arsenic in drinking water and the incidences of lymphoma and leukemia by sex, exposure category, and time period [33,34]. These studies suggested that exposure to arsenic increases the risk of developing bladder and kidney cancers.

3. Gut Microbiome and Biomarkers for Unfavorable Microbiome

The human gut harbors a large population of microbes such as bacteria, viruses, protozoa, and fungi and has an estimated biomass of 1.5 kg, which is comparable in size to the liver, so it can be considered an organ. Therefore, any changes to this composition can lead to a diseased condition. Gut dynamics can be influenced by several factors, such as age, diet, gender, ethnicity, and environment [35,36]. A study by Brooks et al. showed how differences across ethnicities can potentially affect the microbial composition. In this comprehensive study, including 1673 individuals in the United States, the authors reported twelve microbial genera and families that reproducibly vary by ethnicity [35]. Diet and gut microbiome are the deciding factors of human health. Microbial ecology, genotype, and their proportion affect energy and nutrient harvesting. To understand these interrelations, a study model representing the human gut ecosystem was prepared by Turnbaugh et al. using germ-free C57BL/6J mice by transplanting fresh or frozen adult human fecal microbial communities. They observed altered microbiome gene expression, increased adiposity, and a changed representation of metabolic pathways in the microbiome after switching from a low-fat, plant polysaccharide-rich diet to a high-fat, high-sugar “Western” diet [37].
Disturbance in the microbiome can result in disease conditions, evidence and a discussion of which can be found in this review (Table 1). Dectin-1 is a pattern-recognition receptor and innate immune response modulator protein that potentially serves as a biomarker to identify people with a potentially unfavorable gut microbiome. Dectin-1 gene expression in subcutaneous adipose tissue was investigated in the context of obesity and associated inflammatory markers. The results showed a correlation between the expression of dectin-1 transcripts and various proinflammatory cytokines, chemokines, and their cognate receptors in terms of body mass index, fat percentage, and monocyte/macrophage markers (CD16, CD68, CD86, and CD163). These results suggest that dectin-1 can be used as an adipose tissue biomarker of metabolic inflammation in obesity [38].
Similarly, short-chain fatty acids (SCFAs) like propionate, butyrate, and acetate have been found to be another biomarker for multiple sclerosis pathogenesis. Upon comparing the serum SCFA levels of multiple sclerosis patients with those of healthy ones for immune cell abundance and phenotype as well as with other relevant serum factors, a significant reduction in propionate levels in the serum of the patients was noticed. There is a positive correlation between serum propionate and the frequencies of circulating T follicular regulatory cells and T follicular helper cells. However, butyrate levels are associated positively with the frequencies of IL-10-producing B-cells and negatively with the frequencies of class-switched memory B-cells. In addition, acetate levels are negatively correlated with TNF production by polyclonally activated B-cells. Therefore, serum SCFA levels can be associated with changes in circulating immune cells and biomarkers implicated in the development of multiple sclerosis [51]. Nevertheless, propionate and acetate produced by Propionibacterium during fermentation processes are major cytotoxic components secreted by the bacteria. SCFAs can kill human colorectal carcinoma cell lines in multiple ways, including apoptosis, a loss of mitochondrial transmembrane potential, the generation of reactive oxygen species, caspase-3 processing, influencing extracellular pH, nuclear chromatin condensation, and necrosis. These results suggest that SCFAs produced by propionibacteria could exhibit deleterious effects on cancerous cells and probiotic properties which are efficient in digestive cancer prophylaxis [52].
Due to the availability of advanced techniques like fluorescent oligonucleotide hybridization, it is now easier to figure out the microbiome composition by labeling bacterial cells, which can be easily and readily analyzed by flow cytometry [53] and molecular-phylogenetic methods [54]. The probes labeled with tetramethylrhodamine are used to complement short-sequence elements common to phylogenetically coherent assemblages of microorganisms within 16S rRNA and hybridize to suspensions of fixed cells. Similarly, molecular-phylogenetic methods also help to identify microbes by taking advantage of polymerase chain reaction amplification and phylogenetic analysis of small-subunit ribosomal RNA gene sequences. This technique also helps design sequence-based tools for identifying, tracking, and diagnosing the presence of microbes in complex samples [54].
The method of amplicon sequencing the bacterial 16S rRNA gene has been used to discriminate between pancreatic cancer patients and controls, where fourteen bacterial features were found to be altered [55]. Bacterial 16S rDNA Illumina® sequencing and 16S rDNA quantitative polymerase chain reaction methods help determine the association between the urinary microbiome and prostate cancer. These methodologies were used in a study by Shrestha et al., 2018, to describe the association between the prevalence of pro-inflammatory bacteria and uropathogens in the urinary tract of men with prostate cancer, which is related to chronic inflammation [56]. A study in 2018 by Liss et al. showed that Streptococcus and Bacteroides species were higher in the rectal swab samples of men suffering from prostate cancer [26]. Similarly, another case–control pilot study on 20 men with prostate cancer showed a higher relative abundance of Bacteroides massiliensis in the gut microbiome as compared to the controls [50]. Patients undergoing treatment with androgen receptor axis-targeted therapy (ATT) showed a higher relative abundance of Ruminococcaceae spp. and Akkermansia muciniphila, which are involved in steroid hormone biosynthesis [57]. These studies indicated the importance of specific microbial compositions that can be utilized as a potential biomarker of cancer pathogenesis.
On the other hand, the association between the microbiome and the prognosis of head and neck squamous cell carcinoma (HNSCC) was uncertain. Since then, several studies have highlighted the importance of the microbiota in the development and prognosis of several types of cancer. A recent study on a retrospective cohort of 129 patients with oral cavity squamous cell carcinoma (OSCC) and oropharyngeal squamous cell carcinoma (OPSCC) reported greater abundances of the genera Bacillus, Lactobacillus, and Sphingomonas to be linked with favorable prognoses in head and neck squamous cell carcinoma (HNSCC) [58].
Cancer patients suffering from renal cell carcinoma (RCC) and non-small-cell lung cancer (NSCLC) treated with antiprogrammed cell death ligand-1 (PD-L1) or PD-L1 plus CTLA-4 mAb monotherapy who were also undergoing treatment with antibiotics (β-lactam or quinolones for pneumonia or urinary tract infections) in the beginning were examined for thirty days for gut microbiota diversity and composition leading to dysbiosis. In this study, the results suggested those patients’ receiving antibiotics suffered an increased risk of primary progressive disease, which means the antibiotics were affecting or lowering the effectiveness of immune-checkpoint inhibitor therapy. The antibiotic treatment increased the risk of primary progressive disease in both types of cancer patients, suggesting that antibiotics reduced the clinical benefits of immune-checkpoint inhibitor therapy in the RCC and NSCLC patients, which can be modulated by altering the gut microbiota composition [59]. This fact can be seen in a study where the ablation of the microbiome with immunogenic reprogramming protected against preinvasive and invasive pancreatic ductal adenocarcinoma (PDA). The removal of the microbiome resulted in a reduction in myeloid-derived suppressor cells and increased M1 macrophage and T cell differentiation and activation. This strategy enabled efficacy for checkpoint-targeted immunotherapy by upregulating PD-1 expression via activating Toll-like receptors in monocytic cells [60].
Anticancer drugs alter the intestinal microbiota in such a way they cause an induction of immune responses; for example, cyclophosphamide imposes antitumor immune responses by altering the composition of the small intestine microbiota and induces the translocation of selected species of Gram-positive bacteria (Lactobacillus johnsonii, Lactobacillus murinus, and Enterococcus hirae) into secondary lymphoid organs in mouse model. These bacteria help induce the generation of pathogen-specific T helper 17 (Th17) cells and memory immune responses. This fact was demonstrated by Viaud et al., where the authors showed how tumor-bearing mice treated with antibiotics got rid of Gram-positive bacteria, causing a reduction in the pathogen-specific Th17 responses that made them resistant to cyclophosphamide. Seven days postcyclophosphamide treatment, there was a significant reduction in the frequencies of CD103+CD11b+ dendritic cells and TCRαβ+CD3+ T cells that were expressing the transcription factor RORγt in the lamina propria (LP) of the small intestine. However, the adoptive transfer of pathogen-specific Th17 cells to vancomycin-treated mice partially restored the antitumor efficacy of cyclophosphamide, suggesting the efficacity of the gut microbiome in strengthening anticancer immune responses [61].
An intact gut microbiome is required to obtain favorable patient outcomes during cancer chemotherapies because microbiota have a direct influence on local and systemic inflammation and influence disease progression and treatment. It has been reported that disruption to the microbiota impairs the response of subcutaneous tumors to CpG-oligonucleotide immunotherapy and platinum chemotherapy. This aspect can be seen in detail in a study by Iida et al., where C57Bl/6 mice were pretreated with an antibiotic cocktail containing vancomycin, imipenem, and neomycin before tumor inoculation, and this continued till the end of the study. The treatment of the antibiotic mix reduced the frequency of monocyte-derived Ly6C+ MHC class II+ cells and Ly6Ghigh neutrophils. The authors observed that the production of cytokines and tumor necrosis after the CpG-oligonucleotide treatment was lower in the antibiotic-treated or germ-free mice, suggesting that the optimal response to chemotherapy demands an effective and intact gut microbiome [62]. In a study, T cell infiltration into solid tumors facilitated immunotherapy. Immune cell infiltration into the tumor site makes immunotherapies work, as described by Sivan et al. by using a 16S ribosomal RNA sequencing method to identify functionally relevant bacterial taxa. A comparative analysis between quick-growing-tumor-bearing mice from Jackson Laboratory and slow-growing-tumor-bearing mice from Taconic Farms revealed that 257 taxa were significantly different. A further analysis identified Bifidobacterium’s association with antitumor effects since its combined treatment with PD-L1-specific antibody therapy abolished tumor outgrowth. This resulted in the accumulation of antigen-specific T cells and induced IFN-γ by enhancing dendritic cell function, leading to enhanced CD8+ T cell priming and accumulation in the tumor microenvironment [63].
Research has shown how disrupting β-glucuronidase enzyme activity, which is present in the commensal microbiota, can alleviate toxicity caused by anticancer drugs. There was a study focused not on altering the bacterial population but on targeting symbiotic bacterial β-glucuronidases, which cause severe diarrhea upon colon cancer chemotherapeutic CPT-11 (irinotecan) treatment. CPT-11 is a derivative of a known potent antineoplastic compound, Camptothecin, which is undergoing clinical use but still elicits pronounced side effects that limit its efficacy. The authors screened bacterial β-glucuronidase inhibitors by high-throughput screening using a β-glucuronidase assay. This investigation selected potential inhibitors that had no effect on orthologous mammalian enzymes since crystallographic studies revealed that the E. coli enzyme contains a 17-residue “bacterial loop”, which is not present in the human ortholog, which makes it selective to bacterial enzymes but not to mammalian enzymes. The findings were confirmed in mice, showing that the oral administration of a β-glucuronidase inhibitor protected Balb/cJ mice from CPT-11-induced toxicity without losing the glandular structures of the intestinal tissues and keeping the gastrointestinal epithelium intact, thereby reducing both diarrhea and bloody diarrhea. Therefore, drugs should be designed to inhibit undesirable enzyme activities rather than alter the actual microbiome to enhance chemotherapeutic efficacy [64].

4. Probiotics Improve Cancer Treatment

There are diverse procedures these days to go with surgery, such as chemotherapy, radiotherapy, immunotherapy, and hormonal therapy, which produce a lot of side effects, especially abdominal pain, severe gastric issues, constipations, nausea, vomiting, diarrhea, taste disturbances, mucositis, and swallowing difficulties, thus affecting the quality of life of these patients [1,65]. Thanks to advanced research and therapeutic strategies that started using probiotics to modify the microbiota to manage these complications (Table 2), a number of studies have proposed and confirmed that probiotics can be effective at controlling the growth of cancer cells [1,66,67,68,69].
The intake of fermented dairy products reduces the risk of cancer by inducing immune responses and creating balanced and healthy gut microbiomes to support all therapies. A detailed, 12-year-long study conducted to determine the effect of yogurt consumption on cancer prevention in volunteers of the EPIC-Italy cohort suggested the protective role of yogurt against colorectal cancer (CRC). This finding suggests that yogurt should be part of a diet to prevent the disease [72].
The exopolysaccharides derived from probiotic yeast Kluyveromyces marxianus and Pichia kudriavzevii have shown inhibitory effects on different colon cancer cell lines, hindering the AKT-1, mTOR, and JAK-1 pathways and induced apoptosis [97]. Patients receiving CRC and undergoing a perioperative probiotic administration of combined probiotics containing Bifidobacterium longum (≥1.0 × 107 cfu/g), Lactobacillus acidophilus (≥1.0 × 107 cfu/g), and Enterococcus faecalis (≥1.0 × 107 cfu/g) showed that this significantly influenced the recovery of bowel function [98].
One of the most widely used probiotic strains is Lactobacillus, lactic-acid-producing bacteria that is continuously studied and used to improve the function of the gut system. In this context, a study evaluated Lactobacillus reuteri FLRE5K1 for its antimelanoma activity in cell assays and animal models. The results showed that treatment with Lactobacillus reuteri FLRE5K1 reduced the incidence of tumors as compared to the model group. FLRE5K1 did not inhibit melanoma tumor mass but prolonged the survival of tumor-bearing mice. The uptake of Lactobacillus reuteri FLRE5K1 by immune cells induced elevated levels of TNF-α and INF-γ in serum, suggesting that it stimulated immune responses and limited the infiltration of melanoma cells with blood circulation [99]. In addition to prolonging the survival of tumor-bearing mice, Lactobacillus reuteri helps alleviate liver injury and intestinal inflammation induced by D-galactosamine in Sprague–Dawley (SD) rats. Probiotics work by maintaining redox homeostasis by increasing the expression of the intestinal tight junction protein proteins ZO-1 and occludin, which help the intestinal barrier morphology to remain intact. The supplementation of Lactobacillus reuteri inhibited intestinal apoptosis by downregulating the expression of caspase-3, activating Nrf-2 nuclear translocation, and elevating the induction of HO-1 via activating phosphoinositide 3-kinase/protein kinase B (PI3K/Akt), protein kinase C (PKC), and their phosphorylated forms [100]. Treatment with anticytotoxic T-lymphocyte-associated protein 4 (CTLA-4) or PD-1/PD-L1 triggers severe inflammatory side effects such as colitis due to changes in the composition of the gut microbiota, especially a reduction in the abundance of Lactobacillus. In this case, the oral administration of Lactobacillus reuteri inhibited the development and progression of colitis by restoring the colon structure and reducing leukocyte infiltration. This treatment decreased the levels of inflammatory cytokines (KC, TNF-α, IFN-γ, and IL-6) and reversed the loss of body weight and inflammatory status induced by antiCTLA-4 or antiPD-L1 immunotherapy [101].
Another example, Lactobacillus rhamnosus GG, showed protective benefits against colon cancer by suppressing proliferation and promoting apoptosis in colon cancer cell lines [66]. Consequently, the administration of a probiotic combination could indeed restore immune function and the composition of an altered gut microbiota. The consumption of yogurt containing high counts of viable Streptoccocus thermophilus and Lactobacillus delbrueckii subsp. bulgaricus was inversely associated with the risk of colorectal cancer by lowering the levels of cancer biomarkers, stimulating anticancer effects, and increasing the survival rate [72]. Similarly, a double-blind trial conducted with 138 patients suffering from superficial transitional cell carcinoma of the bladder showed the prophylactic effects of consuming Lactobacillus casei in preparation for treatment [102]. Lactobacillus casei BL23 treatment in C57BL6 mice protected against colitis-associated colorectal cancer by inducing immunomodulatory effects through downregulating IL-22 cytokines and antiproliferative effects through upregulating caspase-7, caspase-9, and Bik RNA expression and downregulating Ki67 levels [82]. Nonetheless, this strain induces IL-17A, a pro-inflammatory cytokine produced by TH17 cells that participates in antitumor effects. In this study, Jacoutan et al. developed a recombinant strain of Lactobacillus casei, BL23, capable of secreting IL17A to understand the involvement of IL17A in cancer. Their results showed that in around 26% of the mice treated intranasally with L. lactis-IL-17A and challenged with a tumor cell line, TC-1 cells derived from lung epithelial cells of C57BL/6 mice remained protective against the tumors, confirming the antitumor effect of IL17 cytokines through inducing IL-6 secretion in splenocytes [89]. Another population-based case–control study based on a questionnaire and interview was conducted to assess the combination effects of the regular consumption of Lactobacillus casei Shirota (BLS) and isoflavones on the incidence of breast cancer in Japanese women, in which 306 cases with breast cancer and 662 controls aged 40–55 were included in the analyses. Their analysis suggested the inverse association between soy consumption and breast cancer incidence could be due to isoflavones’ anticancer properties and their inducing natural killer cell activity [76].
Moreover, Streptococcus thermophilus is a lactic acid bacterium known to be used as a starter culture for a number of dairy products to produce folate during growth. Its cytotoxic effect on cancer cells was tested in a study using eight S. thermophilus strains (1F8CT, MTH17CL396, M17PTZA496, TH982, TH985, TH1435, TH1436, TH1477) isolated from curd, cheese, whey, and raw milk dairy products in Italy [90]. This group evaluated all the strains for their number of health-related traits, such as the production of folic acid and anticancer properties, by evaluating their ability to attach and inhibit the growth of human HT-29 colorectal adenocarcinoma cells. The results revealed that two strains of S. thermophilus, namely M17PTZA496 and TH982, have probiotic properties along with anticancer activity and folate production capability. These strains were superior to Lactobacillus rhamnosus GG for some of these properties, which suggests that they can pose better health benefits if used commercially, but before that, they need to be studied in more detail.
In addition to probiotics, prebiotics can also manage the gut microbiota, alleviate the side effects of cancer therapies, and prevent gastric cancer. Prebiotics promote the growth of beneficial bacteria and possess protective effects against colon carcinogenesis. The fermentation of prebiotics by gut microflora generates short-chain fatty acids that alter the gene expression and differentiation of tumor cells [103]. An in vivo study on azoxymethane/dextran–sodium–sulfate-induced colorectal cancer in C57BL/6 mice has shown prebiotics’ preventive effects against colorectal cancer [104]. A clinical trial among colorectal cancer patients showed that preoperative oral supplementation can alleviate postoperative complications. Patients received a daily oral intake of 30 g of prebiotic supplements, which showed positive effects on immune status [105]. Not only does the intake of prebiotics prevent cancer, but it also increases the prevalence of commensal microbiota (Bacteroides, Bifidobacterium, Escherichia-Shigella, and Enterococcus) in these individuals. Another randomized double-blind trial conducted on 73 colorectal cancer patients with preceding colorectal operations found that patients treated with an oral intake of higher concentrations of lactic acid bacteria did not have severe inflammatory responses after surgery [106].
The gut microbiome impacts the therapeutic outcomes of checkpoint blockade immunotherapy in human cancer patients. An examination of the oral and gut microbiome of melanoma patients under the treatment of antiPD-1 immunotherapy demonstrated a significant difference in the diversity and composition of the gut microbiome. Patients whose fecal samples showed a high diversity and abundance of Ruminococcaceae/Faecalibacterium had significantly better progression-free survival. These changes in the gut microbiome may modulate therapy responses to antiPD-1 immunotherapy in melanoma patients by modulating immune responses. Patients with a higher abundance of these strains had higher levels of CD4+ and CD8+ T cells in their systemic circulation [86]. In line with this, T cell responses during immunotherapies depend on distinct Bacteroides species, such as Bacteroides thetaiotaomicron or Bacteroides fragilis. The CTLA blockade effect on tumors was not enough in the mice treated with antibiotics or the germ-free mice, while this defect was overcome by the adoptive transfer of B. fragilis-specific T cells [107]. A study applied a combination of eleven bacterial strains (Parabacteroides distasonis, Parabacteroides gordonii, Parabacteroides johnsonii, Alistipes senegalensis JC50, Paraprevotella xylaniphila, Bacteroides uniformis, Bacteroides vulgatus, Eubacterium limosum, Ruthenibacterium lactatiformans strain 585-1, Phascolarctobacterium succinatutens YIT 12067, Fusobacterium ulceranus) from healthy human donor feces to syngeneic tumor mouse models. All the strains altogether mediated local and systemic immunomodulation by enhancing the recruitment of IFNγ+ CD8 T cells in the intestine without causing inflammation due to CD103+ dendritic cells and major histocompatibility (MHC) class Ia molecules. This strain mixture not only enhanced host resistance against Listeria monocytogenes infection but also improved anticancer immunity in the tumor models [108]. Similarly, fecal transplantation therapy by low-volume enema helped to overcome side effects from chronic antibiotic (oral vancomycin) therapy [109]. Treatment with antibiotics ameliorates the benefits of immune checkpoint inhibitors. The administration of fecal microbiota from cancer patients who responded to immune checkpoint inhibitors helped to reverse gut microbiota dysbiosis to a healthy gut environment in mouse models. These patients’ stool was highly abundant in Akkermansia muciniphila; therefore, its oral supplementation restored the efficacy of the PD-1 blockade. The adjuvant effect of A. muciniphila induced dendritic cells to secrete IL-12, a Th1 cytokine with importance in the immunogenicity of the PD-1 blockade in eubiotic conditions, which helped recruit CCR9+CXCR3+CD4+ T lymphocytes into mouse tumor beds within 48 h after the first injection in mesenteric lymph nodes [87].
Enterococcus faecium RM11 and Lactobacillus fermentum RM28 isolated from fermented dairy milks trigger the antiproliferation of colon cancer cells at rates of 21–29% and 22–29%, respectively [71]. Colorectal cancer patients treated for six months with probiotics containing a 30 × 109 colony-forming unit mixture of six Lactobacillus and Bifidobacteria strains showed a significant reduction in the level of pro-inflammatory cytokines, TNF-α, IL-6, IL-10, IL-12, IL-17A, IL-17C, and IL-22, and also avoided the requirement for antibiotics and showed no infections [92].
Collectively, all these studies suggest that any change or dysbiosis in the gut microbiota has a significant effect on cancer growth, development, and treatment [110,111]. The challenges faced during cancer therapies, i.e., chemotherapy, surgery, immunotherapy, and radiotherapy, which mainly include diarrhea, mucositis, abdominal pain, and reduced quality of life, or the adverse effects caused by heavy antibiotic treatment can be alleviated by encouraging alternative therapeutic methods along with cancer therapies involving adjuvant therapies like the application of probiotics in clinical management. Including probiotics, synbiotics, or probiotics in a correct manner can help to resolve these cons (Table 3). The administration of yeast probiotics has been shown to have an adjuvant property in treating colorectal cancer by several means, including boosting immunological and antioxidant properties [112]. Antioxidant properties have greater implications for cancer treatment, as can be seen in the study where Lactobacillus plantarum AS1 (AS1) showed its direct impact on colon cancer treatment by inducing activities of antioxidant enzymes, e.g., lipid peroxide, superoxide dismutase, catalase, glutathione-S transferase, alkaline phosphatase, and acid phosphatase, in the colon and plasma of cancer-bearing animals [73].

5. Conclusions

Taken together, this article highlights the quality and type of food that should be consumed and the types of food that can enhance the risk of cancer. Disturbance in the gut microbiome leads to resistance to cancer therapies and can ameliorate their beneficial effects. The gut microbiota can be modified to benefit immune responses and support immunotherapies by using probiotics and prebiotics as adjuvants. Several clinical trials and in vivo and in vitro experiments have proposed the use of these adjuvant therapies, as they can shape the intestinal microbiota, which is disturbed in diseased conditions, and help to promote overall wellness. In this review, we focused on the identification of the microbiome as a biomarker of diseased conditions and how the specific usage of probiotics, or some strains, can be useful to improve the efficacy of treatments and eliminate their side effects.

Author Contributions

Conceptualization, R.R. and S.K.S.; writing—original draft preparation, R.R. and S.K.S.; writing—review and editing, R.R. and S.K.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Rodriguez-Arrastia, M.; Martinez-Ortigosa, A.; Rueda-Ruzafa, L.; Folch Ayora, A.; Ropero-Padilla, C. Probiotic Supplements on Oncology Patients’ Treatment-Related Side Effects: A Systematic Review of Randomized Controlled Trials. Int. J. Environ. Res. Public Health 2021, 18, 4265. [Google Scholar] [CrossRef] [PubMed]
  2. World Cancer Research Fund; American Institute for Cancer Research. Diet, Nutrition, Physical Activity and Cancer: A Global Perspective. Continuous Update Project Expert Report 2018; American Institute for Cancer Research: Arlington, VA, USA, 2018. [Google Scholar]
  3. Papadimitriou, N.; Markozannes, G.; Kanellopoulou, A.; Critselis, E.; Alhardan, S.; Karafousia, V.; Kasimis, J.C.; Katsaraki, C.; Papadopoulou, A.; Zografou, M.; et al. An umbrella review of the evidence associating diet and cancer risk at 11 anatomical sites. Nat. Commun. 2021, 12, 4579. [Google Scholar] [CrossRef] [PubMed]
  4. Reeves, G.K.; Pirie, K.; Beral, V.; Green, J.; Spencer, E.; Bull, D. Cancer incidence and mortality in relation to body mass index in the Million Women Study: Cohort study. Bmj 2007, 335, 1134. [Google Scholar] [CrossRef] [PubMed]
  5. World Health Organization. Global Health Risks: Mortality and Burden of Disease Attributable to Selected Major Risks; World Health Organization: Geneva, Switzerland, 2009. [Google Scholar]
  6. Key, T.J.; Bradbury, K.E.; Perez-Cornago, A.; Sinha, R.; Tsilidis, K.K.; Tsugane, S. Diet, nutrition, and cancer risk: What do we know and what is the way forward? Bmj 2020, 368, m511. [Google Scholar] [CrossRef] [PubMed]
  7. Link, W. Principles of Cancer Treatment and Anticancer Drug Development; Springer: Berlin/Heidelberg, Germany, 2019. [Google Scholar]
  8. Loman, B.R.; Jordan, K.R.; Haynes, B.; Bailey, M.T.; Pyter, L.M. Chemotherapy-induced neuroinflammation is associated with disrupted colonic and bacterial homeostasis in female mice. Sci. Rep. 2019, 9, 16490. [Google Scholar] [CrossRef]
  9. Appleby, P.N.; Key, T.J. The long-term health of vegetarians and vegans. Proc. Nutr. Soc. 2016, 75, 287–293. [Google Scholar] [CrossRef]
  10. Clinton, S.K.; Giovannucci, E.L.; Hursting, S.D. The world cancer research fund/American institute for cancer research third expert report on diet, nutrition, physical activity, and cancer: Impact and future directions. J. Nutr. 2020, 150, 663–671. [Google Scholar] [CrossRef]
  11. Karwowska, M.; Kononiuk, A. Nitrates/Nitrites in Food-Risk for Nitrosative Stress and Benefits. Antioxidants 2020, 9, 241. [Google Scholar] [CrossRef] [PubMed]
  12. WHO. Time to Deliver: Report of the WHO Independent High-Level Commission on Noncommunicable Diseases; World Health Organization: Geneva, Switzerland, 2018. [Google Scholar]
  13. Vergnaud, A.-C.; Romaguera, D.; Peeters, P.H.; Van Gils, C.H.; Chan, D.S.; Romieu, I.; Freisling, H.; Ferrari, P.; Clavel-Chapelon, F.; Fagherazzi, G. Adherence to the World Cancer Research Fund/American Institute for Cancer Research guidelines and risk of death in Europe: Results from the European Prospective Investigation into Nutrition and Cancer cohort study. Am. Clin. Nutr. 2013, 97, 1107–1120. [Google Scholar] [CrossRef] [PubMed]
  14. International Agency for Research on Cancer Red Meat and Processed Meat. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. Vol 114 IARC. 2018. Available online: https://monographs.iarc.who.int/wp-content/uploads/2018/06/Evaluations.pdf (accessed on 25 January 2024).
  15. Reynolds, A.; Mann, J.; Cummings, J.; Winter, N.; Mete, E.; Te Morenga, L. Carbohydrate quality and human health: A series of systematic reviews and meta-analyses. Lancet 2019, 393, 434–445. [Google Scholar] [CrossRef]
  16. Kim, Y.I. Folate: A magic bullet or a double edged sword for colorectal cancer prevention? Gut 2006, 55, 1387–1389. [Google Scholar] [CrossRef] [PubMed]
  17. Burkitt, D.P. Editorial: Large-bowel cancer: An epidemiologic jigsaw puzzle. J. Natl. Cancer Inst. 1975, 54, 3–6. [Google Scholar] [CrossRef] [PubMed]
  18. Inoue, M.; Sawada, N.; Matsuda, T.; Iwasaki, M.; Sasazuki, S.; Shimazu, T.; Shibuya, K.; Tsugane, S. Attributable causes of cancer in Japan in 2005—Systematic assessment to estimate current burden of cancer attributable to known preventable risk factors in Japan. Ann. Oncol. 2012, 23, 1362–1369. [Google Scholar] [CrossRef]
  19. Petimar, J.; Smith-Warner, S.A.; Fung, T.T.; Rosner, B.; Chan, A.T.; Hu, F.B.; Giovannucci, E.L.; Tabung, F.K. Recommendation-based dietary indexes and risk of colorectal cancer in the Nurses’ Health Study and Health Professionals Follow-up Study. Am. J. Clin. Nutr. 2018, 108, 1092–1103. [Google Scholar] [CrossRef] [PubMed]
  20. Yu, E.Y.; Wesselius, A.; Mehrkanoon, S.; Goosens, M.; Brinkman, M.; van den Brandt, P.; Grant, E.J.; White, E.; Weiderpass, E.; Le Calvez-Kelm, F.; et al. Vegetable intake and the risk of bladder cancer in the BLadder Cancer Epidemiology and Nutritional Determinants (BLEND) international study. BMC Med. 2021, 19, 56. [Google Scholar] [CrossRef]
  21. Pauwels, E.K.J.; Volterrani, D. Coffee Consumption and Cancer Risk: An Assessment of the Health Implications Based on Recent Knowledge. Med. Princ. Pract. 2021, 30, 401–411. [Google Scholar] [CrossRef]
  22. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef]
  23. World Cancer Research Fund; American Institute for Cancer Research. Resources and Toolkits. 2018. Available online: https://www.wcrf.org/diet-activity-and-cancer/global-cancer-update-programme/resources-and-toolkits/ (accessed on 25 January 2024).
  24. Figueiredo, J.C.; Grau, M.V.; Haile, R.W.; Sandler, R.S.; Summers, R.W.; Bresalier, R.S.; Burke, C.A.; McKeown-Eyssen, G.E.; Baron, J.A. Folic acid and risk of prostate cancer: Results from a randomized clinical trial. J. Natl. Cancer Inst. 2009, 101, 432–435. [Google Scholar] [CrossRef]
  25. Wang, R.; Zheng, Y.; Huang, J.Y.; Zhang, A.Q.; Zhou, Y.H.; Wang, J.N. Folate intake, serum folate levels, and prostate cancer risk: A meta-analysis of prospective studies. BMC Public Health 2014, 14, 1326. [Google Scholar] [CrossRef]
  26. Liss, M.A.; White, J.R.; Goros, M.; Gelfond, J.; Leach, R.; Johnson-Pais, T.; Lai, Z.; Rourke, E.; Basler, J.; Ankerst, D.; et al. Metabolic Biosynthesis Pathways Identified from Fecal Microbiome Associated with Prostate Cancer. Eur. Urol. 2018, 74, 575–582. [Google Scholar] [CrossRef]
  27. Schmit, S.L.; Rennert, H.S.; Rennert, G.; Gruber, S.B. Coffee Consumption and the Risk of Colorectal Cancer. Cancer Epidemiol. Biomark. Prev. 2016, 25, 634–639. [Google Scholar] [CrossRef] [PubMed]
  28. Yiannakou, I.; Singer, M.R.; Jacques, P.F.; Xanthakis, V.; Ellison, R.C.; Moore, L.L. Adherence to a Mediterranean-Style Dietary Pattern and Cancer Risk in a Prospective Cohort Study. Nutrients 2021, 13, 4064. [Google Scholar] [CrossRef] [PubMed]
  29. World Cancer Research Fund International. Preservation and Processing of Foods and Cancer Risk. Available online: https://www.wcrf.org/wp-content/uploads/2021/02/Preservation-and-processing-of-foods.pdf (accessed on 25 January 2024).
  30. Wu, B.; Yang, D.; Yang, S.; Zhang, G. Dietary Salt Intake and Gastric Cancer Risk: A Systematic Review and Meta-Analysis. Front. Nutr. 2021, 8, 801228. [Google Scholar] [CrossRef]
  31. Strumylaite, L.; Zickute, J.; Dudzevicius, J.; Dregval, L. Salt-preserved foods and risk of gastric cancer. Medicina 2006, 42, 164–170. [Google Scholar] [PubMed]
  32. Kimanya, M.E.; Routledge, M.N.; Mpolya, E.; Ezekiel, C.N.; Shirima, C.P.; Gong, Y.Y. Estimating the risk of aflatoxin-induced liver cancer in Tanzania based on biomarker data. PLoS ONE 2021, 16, e0247281. [Google Scholar] [CrossRef]
  33. Lin, M.H.; Li, C.Y.; Cheng, Y.Y.; Guo, H.R. Arsenic in Drinking Water and Incidences of Leukemia and Lymphoma: Implication for Its Dual Effects in Carcinogenicity. Front. Public Health 2022, 10, 863882. [Google Scholar] [CrossRef]
  34. Issanov, A.; Adewusi, B.; Dummer, T.J.B.; Saint-Jacques, N. Arsenic in Drinking Water and Urinary Tract Cancers: A Systematic Review Update. Water 2023, 15, 2185. [Google Scholar] [CrossRef]
  35. Brooks, A.W.; Priya, S.; Blekhman, R.; Bordenstein, S.R. Gut microbiota diversity across ethnicities in the United States. PLoS Biol. 2018, 16, e2006842. [Google Scholar] [CrossRef]
  36. Santos-Marcos, J.A.; Haro, C.; Vega-Rojas, A.; Alcala-Diaz, J.F.; Molina-Abril, H.; Leon-Acuña, A.; Lopez-Moreno, J.; Landa, B.B.; Tena-Sempere, M.; Perez-Martinez, P.; et al. Sex Differences in the Gut Microbiota as Potential Determinants of Gender Predisposition to Disease. Mol. Nutr. Food Res. 2019, 63, e1800870. [Google Scholar] [CrossRef]
  37. Turnbaugh, P.J.; Ridaura, V.K.; Faith, J.J.; Rey, F.E.; Knight, R.; Gordon, J.I. The effect of diet on the human gut microbiome: A metagenomic analysis in humanized gnotobiotic mice. Sci. Transl. Med. 2009, 1, 6ra14. [Google Scholar] [CrossRef]
  38. Al Madhoun, A.; Kochumon, S.; Al-Rashed, F.; Sindhu, S.; Thomas, R.; Miranda, L.; Al-Mulla, F.; Ahmad, R. Dectin-1 as a Potential Inflammatory Biomarker for Metabolic Inflammation in Adipose Tissue of Individuals with Obesity. Cells 2022, 11, 2879. [Google Scholar] [CrossRef]
  39. Nagy, K.; Sonkodi, I.; Szöke, I.; Nagy, E.; Newman, H. The microflora associated with human oral carcinomas. Oral Oncol. 1998, 34, 304–308. [Google Scholar] [CrossRef]
  40. Mager, D.L.; Haffajee, A.D.; Devlin, P.M.; Norris, C.M.; Posner, M.R.; Goodson, J.M. The salivary microbiota as a diagnostic indicator of oral cancer: A descriptive, non-randomized study of cancer-free and oral squamous cell carcinoma subjects. J. Transl. Med. 2005, 3, 27. [Google Scholar] [CrossRef]
  41. Guerrero-Preston, R.; Godoy-Vitorino, F.; Jedlicka, A.; Rodríguez-Hilario, A.; González, H.; Bondy, J.; Lawson, F.; Folawiyo, O.; Michailidi, C.; Dziedzic, A.; et al. 16S rRNA amplicon sequencing identifies microbiota associated with oral cancer, human papilloma virus infection and surgical treatment. Oncotarget 2016, 7, 51320–51334. [Google Scholar] [CrossRef]
  42. Guerrero-Preston, R.; White, J.R.; Godoy-Vitorino, F.; Rodríguez-Hilario, A.; Navarro, K.; González, H.; Michailidi, C.; Jedlicka, A.; Canapp, S.; Bondy, J.; et al. High-resolution microbiome profiling uncovers Fusobacterium nucleatum, Lactobacillus gasseri/johnsonii, and Lactobacillus vaginalis associated to oral and oropharyngeal cancer in saliva from HPV positive and HPV negative patients treated with surgery and chemo-radiation. Oncotarget 2017, 8, 110931–110948. [Google Scholar] [CrossRef]
  43. Hayes, R.B.; Ahn, J.; Fan, X.; Peters, B.A.; Ma, Y.; Yang, L.; Agalliu, I.; Burk, R.D.; Ganly, I.; Purdue, M.P.; et al. Association of Oral Microbiome With Risk for Incident Head and Neck Squamous Cell Cancer. JAMA Oncol. 2018, 4, 358–365. [Google Scholar] [CrossRef]
  44. Perera, M.; Al-Hebshi, N.N.; Perera, I.; Ipe, D.; Ulett, G.C.; Speicher, D.J.; Chen, T.; Johnson, N.W. Inflammatory Bacteriome and Oral Squamous Cell Carcinoma. J. Dent. Res. 2018, 97, 725–732. [Google Scholar] [CrossRef] [PubMed]
  45. Narikiyo, M.; Tanabe, C.; Yamada, Y.; Igaki, H.; Tachimori, Y.; Kato, H.; Muto, M.; Montesano, R.; Sakamoto, H.; Nakajima, Y.; et al. Frequent and preferential infection of Treponema denticola, Streptococcus mitis, and Streptococcus anginosus in esophageal cancers. Cancer Sci. 2004, 95, 569–574. [Google Scholar] [CrossRef] [PubMed]
  46. Kim, N.H.; Park, J.P.; Jeon, S.H.; Lee, Y.J.; Choi, H.J.; Jeong, K.M.; Lee, J.G.; Choi, S.P.; Lim, J.H.; Kim, Y.H.; et al. Purulent Pericarditis Caused by Group G Streptococcus as an Initial Presentation of Colon Cancer. J. Korean Med. Sci. 2002, 17, 571–573. [Google Scholar] [CrossRef] [PubMed]
  47. Castellarin, M.; Warren, R.L.; Freeman, J.D.; Dreolini, L.; Krzywinski, M.; Strauss, J.; Barnes, R.; Watson, P.; Allen-Vercoe, E.; Moore, R.A.; et al. Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma. Genome Res. 2012, 22, 299–306. [Google Scholar] [CrossRef] [PubMed]
  48. Farrell, J.J.; Zhang, L.; Zhou, H.; Chia, D.; Elashoff, D.; Akin, D.; Paster, B.J.; Joshipura, K.; Wong, D.T. Variations of oral microbiota are associated with pancreatic diseases including pancreatic cancer. Gut 2012, 61, 582–588. [Google Scholar] [CrossRef]
  49. You, W.C.; Zhang, L.; Gail, M.H.; Chang, Y.S.; Liu, W.D.; Ma, J.L.; Li, J.Y.; Jin, M.L.; Hu, Y.R.; Yang, C.S.; et al. Gastric dysplasia and gastric cancer: Helicobacter pylori, serum vitamin C, and other risk factors. J. Natl. Cancer Inst. 2000, 92, 1607–1612. [Google Scholar] [CrossRef]
  50. Golombos, D.M.; Ayangbesan, A.; O’Malley, P.; Lewicki, P.; Barlow, L.; Barbieri, C.E.; Chan, C.; DuLong, C.; Abu-Ali, G.; Huttenhower, C.; et al. The Role of Gut Microbiome in the Pathogenesis of Prostate Cancer: A Prospective, Pilot Study. Urology 2018, 111, 122–128. [Google Scholar] [CrossRef]
  51. Trend, S.; Leffler, J.; Jones, A.P.; Cha, L.; Gorman, S.; Brown, D.A.; Breit, S.N.; Kermode, A.G.; French, M.A.; Ward, N.C.; et al. Associations of serum short-chain fatty acids with circulating immune cells and serum biomarkers in patients with multiple sclerosis. Sci. Rep. 2021, 11, 5244. [Google Scholar] [CrossRef]
  52. Jan, G.; Belzacq, A.; Haouzi, D.; Rouault, A.; Metivier, D.; Kroemer, G.; Brenner, C. Propionibacteria induce apoptosis of colorectal carcinoma cells via short-chain fatty acids acting on mitochondria. Cell Death Differ. 2002, 9, 179–188. [Google Scholar] [CrossRef]
  53. Amann, R.I.; Binder, B.J.; Olson, R.J.; Chisholm, S.W.; Devereux, R.; Stahl, D.A. Combination of 16S rRNA-targeted oligonucleotide probes with flow cytometry for analyzing mixed microbial populations. Appl. Environ. Microbiol. 1990, 56, 1919–1925. [Google Scholar] [CrossRef]
  54. Frank, D.N.; Pace, N.R. Molecular-phylogenetic analyses of human gastrointestinal microbiota. Curr. Opin. Gastroenterol. 2001, 17, 52–57. [Google Scholar] [CrossRef]
  55. Half, E.; Keren, N.; Reshef, L.; Dorfman, T.; Lachter, I.; Kluger, Y.; Reshef, N.; Knobler, H.; Maor, Y.; Stein, A.; et al. Fecal microbiome signatures of pancreatic cancer patients. Sci. Rep. 2019, 9, 16801. [Google Scholar] [CrossRef]
  56. Shrestha, E.; White, J.R.; Yu, S.H.; Kulac, I.; Ertunc, O.; De Marzo, A.M.; Yegnasubramanian, S.; Mangold, L.A.; Partin, A.W.; Sfanos, K.S. Profiling the Urinary Microbiome in Men with Positive versus Negative Biopsies for Prostate Cancer. J. Urol. 2018, 199, 161–171. [Google Scholar] [CrossRef]
  57. Sfanos, K.S.; Markowski, M.C.; Peiffer, L.B.; Ernst, S.E.; White, J.R.; Pienta, K.J.; Antonarakis, E.S.; Ross, A.E. Compositional differences in gastrointestinal microbiota in prostate cancer patients treated with androgen axis-targeted therapies. Prostate Cancer Prostatic Dis. 2018, 21, 539–548. [Google Scholar] [CrossRef]
  58. Dou, Y.; Ma, C.; Wang, K.; Liu, S.; Sun, J.; Tan, W.; Neckenig, M.; Wang, Q.; Dong, Z.; Gao, W.; et al. Dysbiotic tumor microbiota associates with head and neck squamous cell carcinoma outcomes. Oral. Oncol. 2022, 124, 105657. [Google Scholar] [CrossRef]
  59. Derosa, L.; Hellmann, M.D.; Spaziano, M.; Halpenny, D.; Fidelle, M.; Rizvi, H.; Long, N.; Plodkowski, A.J.; Arbour, K.C.; Chaft, J.E.; et al. Negative association of antibiotics on clinical activity of immune checkpoint inhibitors in patients with advanced renal cell and non-small-cell lung cancer. Ann. Oncol. 2018, 29, 1437–1444. [Google Scholar] [CrossRef]
  60. Pushalkar, S.; Hundeyin, M.; Daley, D.; Zambirinis, C.P.; Kurz, E.; Mishra, A.; Mohan, N.; Aykut, B.; Usyk, M.; Torres, L.E. The pancreatic cancer microbiome promotes oncogenesis by induction of innate and adaptive immune suppression. Cancer Discov. 2018, 8, 403–416. [Google Scholar] [CrossRef]
  61. Viaud, S.; Saccheri, F.; Mignot, G.; Yamazaki, T.; Daillère, R.; Hannani, D.; Enot, D.P.; Pfirschke, C.; Engblom, C.; Pittet, M.J. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science 2013, 342, 971–976. [Google Scholar] [CrossRef]
  62. 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. [Google Scholar] [CrossRef]
  63. Sivan, A.; Corrales, L.; Hubert, N.; Williams, J.B.; Aquino-Michaels, K.; Earley, Z.M.; Benyamin, F.W.; Lei, Y.M.; Jabri, B.; Alegre, M.L.; et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 2015, 350, 1084–1089. [Google Scholar] [CrossRef]
  64. Wallace, B.D.; Wang, H.; Lane, K.T.; Scott, J.E.; Orans, J.; Koo, J.S.; Venkatesh, M.; Jobin, C.; Yeh, L.-A.; Mani, S. Alleviating cancer drug toxicity by inhibiting a bacterial enzyme. Science 2010, 330, 831–835. [Google Scholar] [CrossRef]
  65. Mazzotti, E.; Antonini Cappellini, G.C.; Buconovo, S.; Morese, R.; Scoppola, A.; Sebastiani, C.; Marchetti, P. Treatment-related side effects and quality of life in cancer patients. Support Care Cancer 2012, 20, 2553–2557. [Google Scholar] [CrossRef]
  66. Orlando, A.; Refolo, M.; Messa, C.; Amati, L.; Lavermicocca, P.; Guerra, V.; Russo, F. Antiproliferative and proapoptotic effects of viable or heat-killed Lactobacillus paracasei IMPC2. 1 and Lactobacillus rhamnosus GG in HGC-27 gastric and DLD-1 colon cell lines. Nutr. Cancer 2012, 64, 1103–1111. [Google Scholar] [CrossRef]
  67. Rafter, J.J. The role of lactic acid bacteria in colon cancer prevention. Scand. J. Gastroenterol. 1995, 30, 497–502. [Google Scholar] [CrossRef]
  68. Blaut, M. Relationship of prebiotics and food to intestinal microflora. Eur. J. Nutr. 2002, 41, i11–i16. [Google Scholar] [CrossRef]
  69. Kim, S.-K.; Guevarra, R.B.; Kim, Y.-T.; Kwon, J.; Kim, H.; Cho, J.H.; Kim, H.B.; Lee, J.-H. Role of Probiotics in Human Gut Microbiome-Associated Diseases. 2019. Available online: https://www.jmb.or.kr/journal/view.html?uid=5262&vmd=Full (accessed on 25 January 2024).
  70. Lan, A.; Lagadic-Gossmann, D.; Lemaire, C.; Brenner, C.; Jan, G. Acidic extracellular pH shifts colorectal cancer cell death from apoptosis to necrosis upon exposure to propionate and acetate major end-products of the human probiotic propionibacteria. Apoptosis 2007, 12, 573591. [Google Scholar] [CrossRef]
  71. Thirabunyanon, M.; Boonprasom, P.; Niamsup, P. Probiotic potential of lactic acid bacteria isolated from fermented dairy milks on antiproliferation of colon cancer cells. Biotechnol. Lett. 2009, 31, 571–576. [Google Scholar] [CrossRef]
  72. Pala, V.; Sieri, S.; Berrino, F.; Vineis, P.; Sacerdote, C.; Palli, D.; Masala, G.; Panico, S.; Mattiello, A.; Tumino, R.; et al. Yogurt consumption and risk of colorectal cancer in the Italian European prospective investigation into cancer and nutrition cohort. Int. J. Cancer 2011, 129, 2712–2719. [Google Scholar] [CrossRef]
  73. Kumar, R.S.; Kanmani, P.; Yuvaraj, N.; Paari, K.A.; Pattukumar, V.; Thirunavukkarasu, C.; Arul, V. Lactobacillus plantarum AS1 isolated from south Indian fermented food Kallappam suppress 1,2-dimethyl hydrazine (DMH)-induced colorectal cancer in male Wistar rats. Appl. Biochem. Biotechnol. 2012, 166, 620–631. [Google Scholar] [CrossRef]
  74. Escamilla, J.; Lane, M.A.; Maitin, V. Cell-free supernatants from probiotic Lactobacillus casei and Lactobacillus rhamnosus GG decrease colon cancer cell invasion in vitro. Nutr. Cancer 2012, 64, 871–878. [Google Scholar] [CrossRef]
  75. Maroof, H.; Hassan, Z.M.; Mobarez, A.M.; Mohamadabadi, M.A. Lactobacillus acidophilus could modulate the immune response against breast cancer in murine model. J. Clin. Immunol. 2012, 32, 1353–1359. [Google Scholar] [CrossRef]
  76. Toi, M.; Hirota, S.; Tomotaki, A.; Sato, N.; Hozumi, Y.; Anan, K.; Nagashima, T.; Tokuda, Y.; Masuda, N.; Ohsumi, S.; et al. Probiotic Beverage with Soy Isoflavone Consumption for Breast Cancer Prevention: A Case-control Study. Curr. Nutr. Food Sci. 2013, 9, 194–200. [Google Scholar] [CrossRef] [PubMed]
  77. Kahouli, I.; Malhotra, M.; Tomaro-Duchesneau, C.; Rodes, L.S.; Alaoui-Jamali, M.A.; Prakash, S. Identification of lactobacillus fermentum strains with potential against colorectal cancer by characterizing short chain fatty acids production, anti-proliferative activity and survival in an intestinal fluid: In vitro analysis. J. Bioanal. Biomed. 2015, 7, 4. [Google Scholar]
  78. Lee, H.A.; Kim, H.; Lee, K.-W.; Park, K.-Y. Dead nano-sized Lactobacillus plantarum inhibits azoxymethane/dextran sulfate sodium-induced colon cancer in Balb/c mice. J. Med. Food 2015, 18, 1400–1405. [Google Scholar] [CrossRef]
  79. Han, K.J.; Lee, N.-K.; Park, H.; Paik, H.-D. Anticancer and anti-inflammatory activity of probiotic Lactococcus lactis NK34. J. Microbiol. Biotechnol. 2015, 25, 1697–1701. [Google Scholar] [CrossRef] [PubMed]
  80. Konishi, H.; Fujiya, M.; Tanaka, H.; Ueno, N.; Moriichi, K.; Sasajima, J.; Ikuta, K.; Akutsu, H.; Tanabe, H.; Kohgo, Y. Probiotic-derived ferrichrome inhibits colon cancer progression via JNK-mediated apoptosis. Nat. Commun. 2016, 7, 12365. [Google Scholar] [CrossRef] [PubMed]
  81. Tiptiri-Kourpeti, A.; Spyridopoulou, K.; Santarmaki, V.; Aindelis, G.; Tompoulidou, E.; Lamprianidou, E.E.; Saxami, G.; Ypsilantis, P.; Lampri, E.S.; Simopoulos, C. Lactobacillus casei exerts anti-proliferative effects accompanied by apoptotic cell death and up-regulation of TRAIL in colon carcinoma cells. PLoS ONE 2016, 11, e0147960. [Google Scholar] [CrossRef] [PubMed]
  82. Jacouton, E.; Chain, F.; Sokol, H.; Langella, P.; Bermudez-Humaran, L.G. Probiotic strain Lactobacillus casei BL23 prevents colitis-associated colorectal cancer. Front. Immunol. 2017, 8, 1553. [Google Scholar] [CrossRef]
  83. Kahouli, I.; Malhotra, M.; Westfall, S.; Alaoui-Jamali, M.A.; Prakash, S. Design and validation of an orally administrated active L. fermentum-L. acidophilus probiotic formulation using colorectal cancer Apc Min/+ mouse model. Appl. Microbiol. Biotechnol. 2017, 101, 1999–2019. [Google Scholar] [CrossRef]
  84. An, B.C.; Hong, S.; Park, H.J.; Kim, B.-K.; Ahn, J.Y.; Ryu, Y.; An, J.H.; Chung, M.J. Anti-colorectal cancer effects of probiotic-derived p8 protein. Genes 2019, 10, 624. [Google Scholar] [CrossRef]
  85. Aghazadeh, Z.; Pouralibaba, F.; Khosroushahi, A.Y. The prophylactic effect of Acetobacter syzygii probiotic species against squamous cell carcinoma. J. Dent. Res. Dent. Clin. Dent. Prospect. 2017, 11, 208. [Google Scholar]
  86. Gopalakrishnan, V.; Spencer, C.N.; Nezi, L.; Reuben, A.; Andrews, M.C.; Karpinets, T.V.; Prieto, P.A.; Vicente, D.; Hoffman, K.; Wei, S.C.; et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 2018, 359, 97–103. [Google Scholar] [CrossRef]
  87. Routy, B.; Le Chatelier, E.; Derosa, L.; Duong, C.P.M.; Alou, M.T.; Daillère, R.; Fluckiger, A.; Messaoudene, M.; Rauber, C.; Roberti, M.P.; et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 2018, 359, 91–97. [Google Scholar] [CrossRef]
  88. El-Deeb, N.M.; Yassin, A.M.; Al-Madboly, L.A.; El-Hawiet, A. A novel purified Lactobacillus acidophilus 20079 exopolysaccharide, LA-EPS-20079, molecularly regulates both apoptotic and NF-κB inflammatory pathways in human colon cancer. Microb. Cell Factories 2018, 17, 29. [Google Scholar] [CrossRef]
  89. Jacouton, E.; Torres Maravilla, E.; Boucard, A.-S.; Pouderous, N.; Pessoa Vilela, A.P.; Naas, I.; Chain, F.; Azevedo, V.; Langella, P.; Bermúdez-Humarán, L.G. Anti-tumoral Effects of Recombinant Lactococcus lactis Strain Secreting IL-17A Cytokine. Front. Microbiol. 2018, 9, 3355. [Google Scholar] [CrossRef]
  90. Tarrah, A.; Castilhos, J.d.; Rossi, R.C.; Duarte, V.d.S.; Ziegler, D.R.; Corich, V.; Giacomini, A. In vitro probiotic potential and anti-cancer activity of newly isolated folate-producing Streptococcus thermophilus strains. Front. Microbiol. 2018, 9, 2214. [Google Scholar] [CrossRef]
  91. Wang, L.; Wang, Y.; Li, Q.; Tian, K.; Xu, L.; Liu, G.; Guo, C. Exopolysaccharide, isolated from a novel strain Bifidobacterium breve lw01 possess an anticancer effect on head and neck cancer–genetic and biochemical evidences. Front. Microbiol. 2019, 10, 1044. [Google Scholar] [CrossRef] [PubMed]
  92. Zaharuddin, L.; Mokhtar, N.M.; Muhammad Nawawi, K.N.; Raja Ali, R.A. A randomized double-blind placebo-controlled trial of probiotics in post-surgical colorectal cancer. BMC Gastroenterol. 2019, 19, 131. [Google Scholar] [CrossRef]
  93. Chung, I.-C.; OuYang, C.-N.; Yuan, S.-N.; Lin, H.-C.; Huang, K.-Y.; Wu, P.-S.; Liu, C.-Y.; Tsai, K.-J.; Loi, L.-K.; Chen, Y.-J. Pretreatment with a heat-killed probiotic modulates the NLRP3 inflammasome and attenuates colitis-associated colorectal cancer in mice. Nutrients 2019, 11, 516. [Google Scholar] [CrossRef]
  94. Tukenmez, U.; Aktas, B.; Aslim, B.; Yavuz, S. The relationship between the structural characteristics of lactobacilli-EPS and its ability to induce apoptosis in colon cancer cells in vitro. Sci. Rep. 2019, 9, 8268. [Google Scholar] [CrossRef]
  95. Ghanavati, R.; Asadollahi, P.; Shapourabadi, M.B.; Razavi, S.; Talebi, M.; Rohani, M. Inhibitory effects of Lactobacilli cocktail on HT-29 colon carcinoma cells growth and modulation of the Notch and Wnt/β-catenin signaling pathways. Microb. Pathog. 2020, 139, 103829. [Google Scholar] [CrossRef]
  96. Maghsood, F.; Johari, B.; Rohani, M.; Madanchi, H.; Saltanatpour, Z.; Kadivar, M. Anti-proliferative and anti-metastatic potential of high molecular weight secretory molecules from probiotic Lactobacillus reuteri cell-free supernatant against human colon cancer stem-like cells (HT29-ShE). Int. J. Pept. Res. Ther. 2020, 26, 2619–2631. [Google Scholar] [CrossRef]
  97. Saadat, Y.R.; Khosroushahi, A.Y.; Movassaghpour, A.A.; Talebi, M.; Gargari, B.P. Modulatory role of exopolysaccharides of Kluyveromyces marxianus and Pichia kudriavzevii as probiotic yeasts from dairy products in human colon cancer cells. J. Funct. Foods 2020, 64, 103675. [Google Scholar] [CrossRef]
  98. Yang, Y.; Xia, Y.; Chen, H.; Hong, L.; Feng, J.; Yang, J.; Yang, Z.; Shi, C.; Wu, W.; Gao, R.; et al. The effect of perioperative probiotics treatment for colorectal cancer: Short-term outcomes of a randomized controlled trial. Oncotarget 2016, 7, 8432–8440. [Google Scholar] [CrossRef]
  99. Luo, M.; Hu, M.; Feng, X.; Xiao Li, W.; Dong, D.; Wang, W. Preventive effect of Lactobacillus reuteri on melanoma. Biomed. Pharmacother. 2020, 126, 109929. [Google Scholar] [CrossRef]
  100. Zhou, Q.; Wu, F.; Chen, S.; Cen, P.; Yang, Q.; Guan, J.; Cen, L.; Zhang, T.; Zhu, H.; Chen, Z. Lactobacillus reuteri improves function of the intestinal barrier in rats with acute liver failure through Nrf-2/HO-1 pathway. Nutrition 2022, 99–100, 111673. [Google Scholar] [CrossRef]
  101. Wang, T.; Zheng, N.; Luo, Q.; Jiang, L.; He, B.; Yuan, X.; Shen, L. Probiotics Lactobacillus reuteri Abrogates Immune Checkpoint Blockade-Associated Colitis by Inhibiting Group 3 Innate Lymphoid Cells. Front. Immunol. 2019, 10, 1235. [Google Scholar] [CrossRef] [PubMed]
  102. Aso, Y.; Akaza, H.; Kotake, T.; Tsukamoto, T.; Imai, K.; Naito, S.; Group, B.S. Preventive effect of a Lactobacillus casei preparation on the recurrence of superficial bladder cancer in a double-blind trial. Eur. Urol. 1995, 27, 104–109. [Google Scholar] [CrossRef] [PubMed]
  103. Liong, M.T. Roles of probiotics and prebiotics in colon cancer prevention: Postulated mechanisms and in-vivo evidence. Int. J. Mol. Sci. 2008, 9, 854–863. [Google Scholar] [CrossRef]
  104. Ji, X.; Hou, C.; Gao, Y.; Xue, Y.; Yan, Y.; Guo, X. Metagenomic analysis of gut microbiota modulatory effects of jujube (Ziziphus jujuba Mill.) polysaccharides in a colorectal cancer mouse model. Food Funct. 2020, 11, 163–173. [Google Scholar] [CrossRef]
  105. Xie, X.; He, Y.; Li, H.; Yu, D.; Na, L.; Sun, T.; Zhang, D.; Shi, X.; Xia, Y.; Jiang, T. Effects of prebiotics on immunologic indicators and intestinal microbiota structure in perioperative colorectal cancer patients. Nutrition 2019, 61, 132–142. [Google Scholar] [CrossRef]
  106. Krebs, B. Prebiotic and Synbiotic Treatment before Colorectal Surgery--Randomised Double Blind Trial. Coll. Antropol. 2016, 40, 35–40. [Google Scholar]
  107. Vétizou, M.; Pitt, J.M.; Daillère, R.; Lepage, P.; Waldschmitt, N.; Flament, C.; Rusakiewicz, S.; Routy, B.; Roberti, M.P.; Duong, C.P.; et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 2015, 350, 1079–1084. [Google Scholar] [CrossRef]
  108. Tanoue, T.; Morita, S.; Plichta, D.R.; Skelly, A.N.; Suda, W.; Sugiura, Y.; Narushima, S.; Vlamakis, H.; Motoo, I.; Sugita, K.; et al. A defined commensal Consortium elicits CD8 T cells and anti-cancer immunity. Nature 2019, 565, 600–605. [Google Scholar] [CrossRef]
  109. Silverman, M.S.; Davis, I.; Pillai, D.R. Success of self-administered home fecal transplantation for chronic Clostridium difficile infection. Clin. Gastroenterol. Hepatol. 2010, 8, 471–473. [Google Scholar] [CrossRef]
  110. Goedert, J.J.; Jones, G.; Hua, X.; Xu, X.; Yu, G.; Flores, R.; Falk, R.T.; Gail, M.H.; Shi, J.; Ravel, J. Investigation of the association between the fecal microbiota and breast cancer in postmenopausal women: A population-based case-control pilot study. J. Natl. Cancer Inst. 2015, 107, djv147. [Google Scholar] [CrossRef]
  111. Molteni, A.; Brizio-Molteni, L.; Persky, V. In vitro hormonal effects of soybean isoflavones. J. Nutr. 1995, 125, 751S–756S. [Google Scholar] [PubMed]
  112. Sambrani, R.; Abdolalizadeh, J.; Kohan, L.; Jafari, B. Recent advances in the application of probiotic yeasts, particularly Saccharomyces, as an adjuvant therapy in the management of cancer with focus on colorectal cancer. Mol. Biol. Rep. 2021, 48, 951–960. [Google Scholar] [CrossRef] [PubMed]
  113. Masuno, T.; Kishimoto, S.; Ogura, T.; Honma, T.; Niitani, H.; Fukuoka, M.; Ogawa, N. A comparative trial of LC9018 plus doxorubicin and doxorubicin alone for the treatment of malignant pleural effusion secondary to lung cancer. Cancer 1991, 68, 1495–1500. [Google Scholar] [CrossRef] [PubMed]
  114. Österlund, P.; Ruotsalainen, T.; Peuhkuri, K.; Korpela, R.; Ollus, A.; Ikonen, M.; Joensuu, H.; Elomaa, I. Lactose intolerance associated with adjuvant 5-fluorouracil-based chemotherapy for colorectal cancer. Clin. Gastroenterol. Hepatol. 2004, 2, 696–703. [Google Scholar] [CrossRef] [PubMed]
  115. Giralt, J.; Regadera, J.P.; Verges, R.; Romero, J.; de la Fuente, I.; Biete, A.; Villoria, J.; Cobo, J.M.; Guarner, F. Effects of probiotic Lactobacillus casei DN-114 001 in prevention of radiation-induced diarrhea: Results from multicenter, randomized, placebo-controlled nutritional trial. Int. J. Radiat. Oncol. Biol. Phys. 2008, 71, 1213–1219. [Google Scholar] [CrossRef]
  116. Chitapanarux, I.; Chitapanarux, T.; Traisathit, P.; Kudumpee, S.; Tharavichitkul, E.; Lorvidhaya, V. Randomized controlled trial of live lactobacillus acidophilus plus Bifidobacterium bifidum in prophylaxis of diarrhea during radiotherapy in cervical cancer patients. Radiat. Oncol. 2010, 5, 31. [Google Scholar] [CrossRef] [PubMed]
  117. Holma, R.; Korpela, R.; Sairanen, U.; Blom, M.; Rautio, M.; Poussa, T.; Saxelin, M.; Osterlund, P. Colonic methane production modifies gastrointestinal toxicity associated with adjuvant 5-fluorouracil chemotherapy for colorectal cancer. J. Clin. Gastroenterol. 2013, 47, 45–51. [Google Scholar] [CrossRef]
  118. Ki, Y.; Kim, W.; Nam, J.; Kim, D.; Lee, J.; Park, D.; Jeon, H.; Ha, H.; Kim, T.; Kim, D. Probiotics for rectal volume variation during radiation therapy for prostate cancer. Int. J. Radiat. Oncol. Biol. Phys. 2013, 87, 646–650. [Google Scholar] [CrossRef]
  119. Shao, F.; Xin, F.-Z.; Yang, C.-G.; Yang, D.-G.; Mi, Y.-T.; Yu, J.-X.; Li, G.-Y. The impact of microbial immune enteral nutrition on the patients with acute radiation enteritis in bowel function and immune status. Cell Biochem. Biophys. 2014, 69, 357–361. [Google Scholar] [CrossRef] [PubMed]
  120. Demers, M.; Dagnault, A.; Desjardins, J. A randomized double-blind controlled trial: Impact of probiotics on diarrhea in patients treated with pelvic radiation. Clin. Nutr. 2014, 33, 761–767. [Google Scholar] [CrossRef] [PubMed]
  121. Golkhalkhali, B.; Rajandram, R.; Paliany, A.S.; Ho, G.F.; Wan Ishak, W.Z.; Johari, C.S.; Chin, K.F. Strain-specific probiotic (microbial cell preparation) and omega-3 fatty acid in modulating quality of life and inflammatory markers in colorectal cancer patients: A randomized controlled trial. Asia-Pac. J. Clin. Oncol. 2018, 14, 179–191. [Google Scholar] [CrossRef] [PubMed]
  122. Motoori, M.; Yano, M.; Miyata, H.; Sugimura, K.; Saito, T.; Omori, T.; Fujiwara, Y.; Miyoshi, N.; Akita, H.; Gotoh, K.; et al. Randomized study of the effect of synbiotics during neoadjuvant chemotherapy on adverse events in esophageal cancer patients. Clin. Nutr. 2017, 36, 93–99. [Google Scholar] [CrossRef]
  123. Linn, Y.H.; Thu, K.K.; Win, N.H.H. Effect of probiotics for the prevention of acute radiation-induced diarrhoea among cervical cancer patients: A randomized double-blind placebo-controlled study. Probiotics Antimicrob. Proteins 2019, 11, 638–647. [Google Scholar] [CrossRef]
  124. De Sanctis, V.; Belgioia, L.; Cante, D.; La Porta, M.R.; Caspiani, O.; Guarnaccia, R.; Argenone, A.; Muto, P.; Musio, D.; De Felice, F. Lactobacillus brevis CD2 for prevention of oral mucositis in patients with head and neck tumors: A multicentric randomized study. Anticancer. Res. 2019, 39, 1935–1942. [Google Scholar] [CrossRef]
  125. Jiang, C.; Wang, H.; Xia, C.; Dong, Q.; Chen, E.; Qiu, Y.; Su, Y.; Xie, H.; Zeng, L.; Kuang, J. A randomized, double-blind, placebo-controlled trial of probiotics to reduce the severity of oral mucositis induced by chemoradiotherapy for patients with nasopharyngeal carcinoma. Cancer 2019, 125, 1081–1090. [Google Scholar] [CrossRef]
  126. Bajramagic, S.; Hodzic, E.; Mulabdic, A.; Holjan, S.; Smajlovic, S.V.; Rovcanin, A. Usage of Probiotics and its Clinical Significance at Surgically Treated Patients Sufferig from Colorectal Carcinoma. Med. Arch. 2019, 73, 316–320. [Google Scholar] [CrossRef]
  127. Vesty, A.; Gear, K.; Boutell, S.; Taylor, M.W.; Douglas, R.G.; Biswas, K. Randomised, double-blind, placebo-controlled trial of oral probiotic Streptococcus salivarius M18 on head and neck cancer patients post-radiotherapy: A pilot study. Sci. Rep. 2020, 10, 13201. [Google Scholar] [CrossRef]
  128. Doppalapudi, R.; Vundavalli, S.; Prabhat, M. Effect of probiotic bacteria on oral Candida in head-and neck-radiotherapy patients: A randomized clinical trial. J. Cancer Res. Ther. 2020, 16, 470–477. [Google Scholar] [CrossRef] [PubMed]
  129. Zheng, C.; Chen, T.; Lu, J.; Wei, K.; Tian, H.; Liu, W.; Xu, T.; Wang, X.; Wang, S.; Yang, R.; et al. Adjuvant treatment and molecular mechanism of probiotic compounds in patients with gastric cancer after gastrectomy. Food Funct. 2021, 12, 6294–6308. [Google Scholar] [CrossRef]
  130. Juan, Z.; Chen, J.; Ding, B.; Yongping, L.; Liu, K.; Wang, L.; Le, Y.; Liao, Q.; Shi, J.; Huang, J.; et al. Probiotic supplement attenuates chemotherapy-related cognitive impairment in patients with breast cancer: A randomised, double-blind, and placebo-controlled trial. Eur. J. Cancer 2022, 161, 10–22. [Google Scholar] [CrossRef]
  131. Lin, B.; Zhao, F.; Liu, Y.; Wu, X.; Feng, J.; Jin, X.; Yan, W.; Guo, X.; Shi, S.; Li, Z.; et al. Randomized Clinical Trial: Probiotics Alleviated Oral-Gut Microbiota Dysbiosis and Thyroid Hormone Withdrawal-Related Complications in Thyroid Cancer Patients Before Radioiodine Therapy Following Thyroidectomy. Front. Endocrinol. 2022, 13, 834674. [Google Scholar] [CrossRef] [PubMed]
  132. Motoori, M.; Sugimura, K.; Tanaka, K.; Shiraishi, O.; Kimura, Y.; Miyata, H.; Yamasaki, M.; Makino, T.; Miyazaki, Y.; Iwama, M.; et al. Comparison of synbiotics combined with enteral nutrition and prophylactic antibiotics as supportive care in patients with esophageal cancer undergoing neoadjuvant chemotherapy: A multicenter randomized study. Clin. Nutr. 2022, 41, 1112–1121. [Google Scholar] [CrossRef] [PubMed]
  133. Huang, F.; Li, S.; Chen, W.; Han, Y.; Yao, Y.; Yang, L.; Li, Q.; Xiao, Q.; Wei, J.; Liu, Z.; et al. Postoperative Probiotics Administration Attenuates Gastrointestinal Complications and Gut Microbiota Dysbiosis Caused by Chemotherapy in Colorectal Cancer Patients. Nutrients 2023, 15, 356. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Manipulating the gut microbiota to alleviate cancer therapies’ side effects.
Figure 1. Manipulating the gut microbiota to alleviate cancer therapies’ side effects.
Cancers 16 00779 g001
Table 1. Prevalence of specific microbiota for different cancer types.
Table 1. Prevalence of specific microbiota for different cancer types.
BiomarkersCancer TypeAbundanceRef.
Veillonella, Fusobacterium, Prevotella, Porphyromonas, Actinomyces and Clostridium, Haemophilus, Enterobacteriaceae and Streptococcus spp. and Candida albicansOral carcinomaElevated in tumor sites[39]
Capnocytophaga gingivalis, Prevotella melaninogenica and Streptococcus mitisOral squamous cell carcinoma (OSCC)Elevated in the saliva of individuals with OSCC[40]
Fusobacterium nucleatum, S salivarius: Streptococcus vestibularis, Prevotella oris, and Rothia mucilaginosaHead and neck squamous cell carcinoma (HNSCC)HNSCC patients had a significant loss in richness and diversity of microbiota species[41]
Fusobacterium nucleatumHNSCCElevated in the saliva[42]
Corynebacterium and KingellaHNSCCGreater oral abundance of these commensals is associated with decreased risk of HNSCC[43]
Capnocytophaga, Pseudomonas, and AtopobiumOSCCHighly abundant in biopsy tissue[44]
Lautropia, Staphylococcus, and PropionibacteriumFibroepithelial polypHighly abundant in biopsy tissue[44]
Treponema denticola, Streptococcus mitis, and Streptococcus anginosusEsophageal cancerInduction of inflammatory cytokines[45]
Group G streptococciColon cancerLarge amount of pericardial effusion[46]
Fusobacterium nucleatumColorectal carcinomaOver-representation in tumor specimen[47]
Neisseria elongate, Streptococcus mitis and Granulicatella adiacenPancreatic cancer and chronic pancreatitisSalivary microbiota as an informative source for discovering noninvasive biomarkers of systemic diseases[48]
Helicobacter pyloriDysplasia or gastric cancerPresence of H. pylori at baseline was associated with an increased risk of
progression to dysplasia or gastric cancer
[49]
Streptococcus and Bacteroides species (Bacteroides massiliensis)Prostate cancerHigher relative abundance in prostate cancer cases compared to controls.[26,50]
Table 2. Probiotics to support cancer treatment.
Table 2. Probiotics to support cancer treatment.
ProbioticsCancer TypeStudy ModelEffectRef.
Propionibacterium freuden reichiiColorectal cancerHT-29 cellsInduced cell cycle arrest in the G2/M phase[70]
Enterococcus faecium RM11, Lactobacillus fermentum RM28Colon cancerCaco-2 cellsTriggered antiproliferation of colon cancer cells[71]
Yogurt probioticsColorectal cancerClinical trial [72]
Lactobacillus plantarum AS1Colon cancerRatAntioxidant-dependent mechanism[73]
Lactobacillus casei and Lactobacillus rhamnosus GGColorectal cancerHCT-116 cellsDecreased metalloproteinase-9 activity and increased levels of tight junction protein zona occludens-1[74]
Lactobacillus plantarum AS1Colorectal cancerMale Wistar ratsAntioxidant property reduced tumor volume diameter and total number of tumors [73]
Lactobacillus acidophilusBreast cancerBalb/C inbred female miceInduces production of IFNγ, IL-4, and TGF-β[75]
L. casei ShirotaBreast cancerCase –control studyNK cell activation and NK cell-mediated antitumor activity[76]
Lactobacillus casei ShirotaBreast cancerPopulation-based case –control studyEnhanced NK cell activity-mediated antitumor activity[76]
Lactobacillus fermentumColorectal cancerCaco-2 colon cancer cellAntiproliferative activity[77]
Dead nano-sized Lactobacillus plantarumColon cancerBalb/c miceSuppressed inflammation, induced cell cycle arrest and apoptosis, and enhanced IgA secretion[78]
Lactobacillus lactis NK34Lung, colon, gastric adenocarcinoma, breast cancer SK-MES-1, DLD-1, HT-29, LoVo, AGS, MCF-7, and RAW 264.7 cellsReduced production of nitric oxide and proinflammatory cytokines (tumor necrosis factor-α, interleukin-18, and cyclooxygenase-2)[79]
Lactobacillus casei ATCC334Colon cancerHuman colon cancer cell lines (Caco2bbe, SKCO-1, and SW620) and Xenografts (SW620 cells injected into male BALB/c nude mice)Ferrichrome-induced apoptosis by activating C-jun N-terminal kinase and suppressed tumor growth[80]
Lactobacillus casei ATCC 393Colon cancerMurine (CT26) and human (HT29) colon carcinoma cell linesApoptotic cell death and upregulation of TRAIL in colon carcinoma cells[81]
Lactobacillus casei BL23Colitis-associated colorectal cancerC57BL6 miceImmunomodulatory effect, mediated through the downregulation of the IL-22 cytokine, and an antiproliferative effect, mediated through the upregulation of caspase-7, caspase-9, and Bik[82]
Lactobacillus acidophilus ATCC 314 and Lactobacillus fermentum NCIMB 5221Colorectal cancerApc Min/+ CRC mouseDownregulated proliferation markers (Ki-67, E-cadherin, β-catenin)[83]
Lactobacillus reuteri NCIMB 701,359Colorectal cancerDLD-1 cell lineProbiotic-derived protein, p8, inhibits p53-p21-Cyclin B1/Cdk1 signal pathway[84]
Acetobacter syzygiiSquamous cell carcinomaHuman oral cancer (KB) and human normal epithelial (KDR) cell linesInduced apoptosis[85]
Bifidobacterium longum, Collinsella aerofaciens, and Enterococcus faeciumMetastatic melanomaMelanoma patientsEnhanced systemic and antitumor immune responses mediated by increased antigen presentation and improved effector T cell function [86]
Akkermansia muciniphilaNon-small-cell lung cancer, renal cell cancer, and urothelial cancerMiceIncreasing the recruitment of CCR9+CXCR3+CD4+ T lymphocytes[87]
Lactobacillus acidophilus 20079Colon cancerColon cancer (CaCo-2) and human breast cancer (MCF7) cell linesIncreased apoptosis in sub-G0/G1 cell cycle phase, stimulated immune response, and inactivated NF-κB inflammatory pathway[88]
Recombinant Lactococcus lactis Mouse allograft model of human papilloma virus (HPV)-induced cancer and TC-1 cell lineSecreting IL-17 to stimulate the TH17 pathway[89]
Streptococcus thermophilusColorectal cancerHT-29 human colorectal adenocarcinoma cellsHigh production of folic acid, tyramine, and histamine; high cytotoxic to cancer cells[90]
Bifidobacterium breve lw01Head and neck cancerSCC15, CAL 27, and WSU-HN6 cell linesIncreased expression of cell apoptosis protein caspase 3 and PARP and increased proportion of Cl-PARP/PARP[91]
Lactobacillus and Bifidobacteria strainCRCRandomized double-blind placebo-controlled trialInterfered with the signaling pathways to stimulate or suppress the level of cytokine production[92]
Enterococcus faecalisColorectal cancerC57BL/6 miceInhibited NLRP3 inflammasome activation in macrophages[93]
Lactobacillus delbrueckii ssp. bulgaricus B3Colon cancerHT-29 cellsInhibited cell proliferation in HT-29 via apoptosis[94]
Recombinant Lactococcus lactisColorectal cancerMurine fibroblast 3T3 L1 cell lines and mouse allograft model of human papilloma virus-induced cancerEfficiently secretes biologically active IL-17A cytokines[89]
Lactobacilli cocktailColorectal cancerHT-29 colon carcinoma cellsAntitumor effects on HT-29 cells by modulating the Notch and Wnt/β-catenin pathways[95]
Lactobacillus reuteriColon CancerColon cancer stem-like cells (HT29-ShE)Antimetastatic and antiproliferative[96]
Kluyveromyces marxianus and Pichia kudriavzeviiColon cancerColon cancer cell lines (SW-480, HT-29, HCT-116)Hindered AKT-1, mTOR, and JAK-1 pathways and induced apoptosis[97]
Table 3. Clinical trial using probiotics during cancer treatment.
Table 3. Clinical trial using probiotics during cancer treatment.
ProbioticsCancer Type (Year)Effect and ResultsRef.
Lactobacillus casei LC9018Lung cancer (malignant pleural effusions secondary to lung cancer) (1991)
  • Significantly greater improvement in performance status and symptoms (chest pain, chest discomfort, and anorexia).
  • Response rate for treatment with intrapleural doxorubicin plus LC9018 was significantly higher.
[113]
Lactobacillus rhamnosus GG ATCC 53103Colorectal cancer (lactose intolerance associated with adjuvant 5-fluorouracil-based chemotherapy) (2007)
  • Bowel mucosal injury associated with 5-fluorouracil (5-FU) treatment.
  • The frequency of severe 5-FU-based chemotherapy-related diarrhea was reduced.
[114]
Lactobacillus casei DN-114 001Endometrial adenocarcinoma patients (2008)
  • Oral supplementation did not reduce the incidence of radiation-induced diarrhea.
  • Showed a significant effect on stool consistency.
[115]
Lactobacillus acidophilus plus Bifidobacterium bifidumCervical cancer (2010)
  • Reduced the incidence of radiation-induced diarrhea and the need for antidiarrheal medication.
  • Had a significant benefit on stool consistency.
[116]
Lactobacillus rhamnosus GGColorectal cancer (2013)
  • Lactobacillus intervention during 5-fluorouracil chemotherapy.
  • Reduced diarrhea only in nonproducers of methane.
[117]
Lactobacillus acidophilusProstate cancer (2013)
  • Useful in reducing the percentage volume change of the rectum, a crucial factor in prostate movement.
[118]
Bifidobacterium, Lactobacillus and Streptococcus thermophilusAcute radiation enteritis (2014)
  • Treated group showed better food intake than control group.
[119]
Lactobacillus acidophilus, Lactobacillus rhamnosus, Lactobacillus casei DN-114001 and Bifidobacterium bifidumAbdominal or pelvic cancer (2014)
  • Beneficial effects on treatment of radiation-induced diarrhea.
  • Improved the quality of patients’ lives.
[120]
Lactobacillus acidophilus BMC12130, Lactobacillus casei BCMC12313, Lactobacillus lactis BCMC12451, Bifidobacterium bifidum BCMC02290, Bifidobacterium longum BCMC02120 and Bifidobacterium infantis BCMC02129Colorectal cancer (2017)
  • Improved quality of life.
  • Reduced certain inflammatory biomarkers (IL-6) and relieved certain side effects of chemotherapy.
[121]
Bifidobacterium breve strain Yakult,
Lactobacillus casei strain Shirota
Esophageal cancer (2017)
  • Reduced the occurrence of adverse events from chemotherapy through adjustments to the intestinal microbiota.
  • Concentrations of acetic acid and propionic acid were significantly higher in the synbiotics group.
  • The frequencies of severe lymphopenia and diarrhea were significantly less. Febrile neutropenia occurred less in the synbiotics group.
[122]
Lactobacillus acidophilus LA-5 plus Bifidobacterium animalis subsp. lactis BB-12Cervical cancer (2019)
  • Incidence of diarrhea was reduced.
  • Severity of abdominal pain was significantly reduced.
[123]
Lactobacillus brevis CD2Head and neck cancer (2019)
  • Modulated homeostasis of the salivary microbiota.
[124]
Bifidobacterium longum, Lactobacillus lactis and Enterococcus faeciumNasopharyngeal carcinoma (2019)
  • Significantly increased the immune responses.
  • Reduced the severity of oropharyngeal mucositis.
[125]
Lactobacillus acidophilus, Lactobacillus casei, Lactobacillus plantarum, Lactobacillus rhamnosus, Bifidobacterium lactis, Bifidobacterium bifidum, Bifidobacterium breve, Streptococcus thermophilusColorectal cancer (2019)
  • Significant reduction in postoperative complications in the localization of tumors on the rectum −33.3% and the ascending colon −16.7%.
[126]
Streptococcus salivarius M18Head and neck cancer (2020)
  • Improvement in periodontal screening and plaque index scores was observed.
[127]
Lactobacillus acidophilus, Lactobacillus rhamnosus, Bifidobacterium longum and Saccharomyces boulardiiHead and neck cancer (2020)
  • Significant reduction in Candida spp. Counts, specifically Candida glabrata and Candida tropicalis.
[128]
Lactobacillus plantarum MH-301 (CGMCC NO. 18618), L. rhamnosus LGG-18 (CGMCC NO. 14007), Lactobacillus acidophilus and Bifidobacterium animalis subsp. lactis LPL-RH (CGMCC NO. 4599)Gastric cancer (2021)
  • Probiotic compounds can restore gut microbiota homeostasis, reduce inflammation, maintain intestinal mucosal barrier and immunity, and finally promote recovery after gastrectomy.
[129]
Bifidobacterium longum, Lactobacillus acidophilus and Enterococcus faecalisBreast cancer (2022)
  • Probiotic supplement attenuates chemotherapy-related cognitive impairment in patients.
[130]
Bifidobacterium infantis, Lactobacillus acidophilus, Enterococcus faecalis, and Bacillus cereusThyroid cancer (2022)
  • Probiotics alleviated lack of energy, constipation, weight gain, and dry mouth and decreased the levels of fecal/serum LPS and plasma lipid indicators (total cholesterol, triglycerides, low-density lipoproteins, and apolipoprotein A).
[131]
Lacticaseibacillus paracasei strain Shirota (YIT9029), Bifidobacterium breve strain Yakult,Esophageal cancer (2022)
  • Incidences of grade 4 neutropenia and grades 2–4 diarrhea were significantly reduced.
[132]
Bifidobacterium infantis, L. acidophilus, Enterococcus faecalis, and Bacillus cereusColorectal cancer (2023)
  • Reduced chemotherapy-induced gastrointestinal complications, especially in the case of diarrhea.
  • Probiotics also promoted the production of short-chain fatty acids, particularly increasing acetate, butyrate, and propionate.
[133]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Roy, R.; Singh, S.K. The Microbiome Modulates the Immune System to Influence Cancer Therapy. Cancers 2024, 16, 779. https://doi.org/10.3390/cancers16040779

AMA Style

Roy R, Singh SK. The Microbiome Modulates the Immune System to Influence Cancer Therapy. Cancers. 2024; 16(4):779. https://doi.org/10.3390/cancers16040779

Chicago/Turabian Style

Roy, Ruchi, and Sunil Kumar Singh. 2024. "The Microbiome Modulates the Immune System to Influence Cancer Therapy" Cancers 16, no. 4: 779. https://doi.org/10.3390/cancers16040779

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop