Next Article in Journal
Synthetic Small-Molecule Ligands Targeted to Adenosine Receptors: Is There Potential Towards Ischemic Heart Disease?
Previous Article in Journal
When Two Worlds Collide: The Contribution and Association Between Genetics (APOEε4) and Neuroinflammation (IL-1β) in Alzheimer’s Neuropathogenesis
Previous Article in Special Issue
Mirvetuximab Soravtansine Induces Potent Cytotoxicity and Bystander Effect in Cisplatin-Resistant Germ Cell Tumor Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Microbiome in Neuroblastoma: A Virgin Island in the World of Onco-Microbiome

by
Ashwath Keshav Giri
,
Poorvi Subramanian
,
Loganayaki Periyasamy
,
Sivaroopan Aravindan
and
Natarajan Aravindan
*
Department of Physiological Science, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
*
Author to whom correspondence should be addressed.
Cells 2025, 14(15), 1218; https://doi.org/10.3390/cells14151218
Submission received: 25 June 2025 / Revised: 26 July 2025 / Accepted: 5 August 2025 / Published: 7 August 2025
(This article belongs to the Special Issue Signaling Pathways and Mechanisms in Cancer Therapy Resistance)

Abstract

The composition of the gut and/or tumor microbiome has been intricately involved in the onset of carcinogenesis, tumor progression, therapy response, and patient outcomes in diverse solid cancers. The microbiome type, composition, and their metabolome have been functionally implicated in the multifarious cellular processes, transformation, proliferation, tumor immune evasion, cellular migration, etc. Despite such compelling evidence on the role of microbiome interactions in cancer, the realization of their role in neuroblastoma (NB), the deadly extracranial tumor in infants is few and fragmentary. This review comprehends the composition, diversity, and significance of microbiota in human health. Further, this review discusses the microbiota composition, their mode of action, and their signaling flow through and cellular processes in diverse cancers including NB. Precisely, this study for the first time has realized the functional relevance and clinical significance of the gut and tumor microbiome for NB. Interestingly, large cohort clinical and preclinical in vivo models of NB realized the following: gut microbiota predicts the risk for NB; postnatal (and or not maternal transmission) microbiome rearrangements; gut microbial effect on NB pathogenesis; tumor-altering gut microbial composition; microbial composition predicts treatment outcomes in NB; prebiotic remedies for stabilizing NB-associated microbial rearrangements; microbial composition in tumor-infiltrating microbiota predicts NB outcomes.

1. Microbiome

The human body functions as a complex ecosystem, inhabited by trillions of microorganisms collectively known as the microbiota or microbes. In the natural environment, microorganisms establish communities, with each species having a distinct ecological function. Microorganisms, despite their small size and invisible nature, significantly contribute to human health and overall well-being [1]. The microbiome is an entire collection of microorganisms—including bacteria, viruses, fungi, and archaea, along with their genetic material—which interact with their surrounding environment [2]. The definition of “microbiota” refers to microorganisms, while “microbiome” covers the entire habitat, including the microbes, their genomes, and the related environmental factor [3]. Research on the human microbiome has revealed the presence of symbiotic bacteria living on various body surfaces, including the skin, airways, urinary system, teeth, and epithelial lining of the gastrointestinal tract [4].

1.1. Composition of Microbiota

The population of microbiota varies between anatomical regions, such as the lungs, oral, gut, and skin [1] (Figure 1). Its composition is influenced by extrinsic factors, such as age, nutrition, and environmental circumstances, which are essential for maintaining homeostasis [2]. The human gut microbiome, characterized by distinct individual microbial compositions, typically consists of six principal phyla: Actinobacteria (1–10%), Bacteroidetes (20–40%), Firmicutes (40–60%), Fusobacteria (<1%), Proteobacteria (1–5%), and Verrucomicrobia (<1%). Among these, Bacteroidetes and Firmicutes constitute 90% of the gut microbiome [3]. The oral microbiota is regarded as the second biggest microbial community in humans, with the major bacterial phyla being Actinobacteria (5%), Bacteroidetes (18%), Firmicutes (38%), Fusobacteria (9%), and Proteobacteria (30%), collectively constituting about 96% of the oral microbiome, and the remaining 4% is composed of several minor phyla including spirochaetes, chlamydiae, tenericutes, and euryarchaeota [4]. The human skin microbiota comprises a unique assemblage of microorganisms, including Actinobacteria (40–50%), Bacteroidetes (5–10%), Cyanobacteria (3%), Firmicutes (25–30%), and Proteobacteria (15–20%) [5]. The core lung microbiota is primarily composed of microorganisms from the phyla Actinobacteria (5–10%), Bacteroidetes (30–40%), Firmicutes (25–35%), and Proteobacteria (20–30%), while minor phyla such as Fusobacteria, Chloroflexi, Verrucomicrobia, and TM7 (Saccharibacteria) typically comprise less than 5% [6]. Its composition is mainly influenced by three fundamental factors: (1) the invasion of microorganisms, (2) the eradication of microbes, and (3) the replication rates of microorganisms [7]. Invasion of microbes occurs through various mechanisms, including microaspiration, via which oropharyngeal and upper respiratory bacteria, such as Streptococcus pneumoniae, invade the lungs [8]. Airborne inhalation of microorganisms, such as Haemophilus influenzae and Mycobacterium tuberculosis, along with hematogenous dissemination in the bloodstream, can contribute to microbial colonization in the lungs [9]. Direct exposure through medical procedures, including ventilators, can introduce stable microbial populations like Klebsiella pneumoniae [10]. Microbes can easily be eliminated by several types of defenses such as mucociliary clearance, where inhaled microbes get trapped in mucus and are expelled by cilia [11]. The replication of microorganisms in the lungs is mediated by nutrient availability, host immune responses, and environmental conditions, with Firmicutes proliferating in high-fat diets and Bacteroidetes linked to fiber-rich diets [12]. Besides bacteria, the microbiome includes fungi, viruses, archaea, and protozoans, which contribute to intricate ecological interactions with the microbial ecosystem. Fungi, although less studied than bacteria, contributes to the gut microbiome, with prevalent species such as Candida, Saccharomyces, Malassezia, and Cladosporium [13]. Along with bacteria and fungi, the human gut microbiota also comprises viruses (Norovirus and Rotavirus), phages (T4 phage and Lambda phage), and archaea (Methanobrevibacter smithii) [14].

1.2. Traits of Microbial Diversity

Microbial diversity is established at birth and is continuously modulated throughout life, with diet exerting the greatest influence, followed by the mode of delivery and other environmental factors. Diet affects the gut microbiota, with sustained dietary habits determining its composition and diversity. Fiber-rich, plant-based diets and fermented items enhance the proliferation of beneficial bacteria and the synthesis of short-chain fatty acids (SCFAs) [15]. The mode of childbirth significantly influences the newborn’s microbiome. During vaginal birth, infants are exposed to their mother’s vaginal and intestinal microbiota, promoting a diverse and advantageous initial microbiome with necessary bacteria such as Bifidobacterium and Lactobacillus facilitating the development of the immune system and metabolic functions [16]. In contrast, babies delivered via caesarean section are frequently protected with maternal skin-associated bacteria, predominantly Staphylococcus species, resembling skin microbiota more than gut microbiota [17]. This variation in microbial exposure emphasizes the role of the birth mode in forming the infant’s microbiome and consequently increases the risk of metabolic and immune-related illnesses later in life [18]. In the initial phase of life (0–12 months), the microbiome undergoes rapid development, with breastfeeding being crucial as it supplies advantageous bacteria such as Bifidobacterium and human milk oligosaccharides (HMOs), which develop the proliferation of beneficial bacteria, mainly Bifidobacterium and Lactobacillus, enhancing the immune system and digestive health [19]. In infants (1–3 years), a varied diet fosters the development of bacteria essential for immunity and digestion, resulting in a microbiome like that of adults, which is characterized by increased levels of Firmicutes and Bacteroidetes. By the end of this period, the gut microbiome attains greater stability, establishing a foundation for future health [20]. During childhood (4–12 years), the microbiome is more diverse and has an adult-like community structure [21]. A balanced adult microbiome includes Firmicutes, Bacteroidetes, Actinobacteria, and Proteobacteria, essential for metabolism, immune function, and disease resistance [22]. Adolescence is an important stage that features hormonal changes, and dietary and lifestyle modifications, and increased exposure to environmental bacteria influences the microbiota and indicates the continuous maturation of the immune system [23]. (Table 1) illustrates the evolution of the microbial community across the human lifespan, from birth to adulthood [24]. Environmental factors influence the human microbiome via both macro- and microenvironments. Specific exposures, such as smoking and alcohol consumption, can alter the oral and gut microbiomes, thereby increasing health risks. Toxicants in the built environment, such as volatile organic compounds (VOCs) like benzene and formaldehyde found in paints, cleaning products, and air fresheners, can disrupt microbial balance and influence health, including the development of asthma [25].
In addition to chronological age and developmental stages, numerous extrinsic and intrinsic determinants substantially influence the composition of the gut microbiota. Among these, the use of antibiotics and medications can profoundly alter the gut microbiome, reducing microbial diversity and eradicating beneficial taxa [26]. Genetics also play a crucial role in determining the initial microbial colonization and can influence long-term microbial composition, though environmental factors often exert a stronger effect [27]. Geographic location, sanitation practices, and socio-economic factors are also critical variables, as individuals from rural or traditional communities frequently display enhanced microbial diversity compared to their urban counterparts [28]. Regular physical activity is consistently associated with increased gut microbial diversity and metabolic benefits [29], while disruptions to sleep patterns and circadian rhythms may negatively impact microbiome. Environmental exposures such as tobacco use and alcohol intake modify the gut microbiota with potential health ramifications, while exposure to domestic toxicants, including volatile organic compounds from consumer products, can disturb microbial equilibrium and has been associated with conditions such as asthma [30]. Although age and dietary habits are principal determinants, a diverse array of environmental, lifestyle, and physiological factors continually modulate the trajectory and health implications of the human gut microbiome.

1.3. Microbiota in Human Health

A well-balanced microbiome enhances human health by aiding in food digestion and nutrient absorption, regulating immune system functions, influencing mental health via the gut–brain axis, contributing to metabolic activities, and defending the body against harmful pathogens [31]. The microbiome helps in regulating the immune system by interacting with immune cells and maintaining homeostasis. Helpful microbial metabolites such as SCFAs support immunological tolerance and protect against infections [32], while SCFAs generated by gut bacteria also influence cognitive function via the gut–brain axis; however, dysbiosis (an imbalance in the gut microbiota) disrupts the gut–brain connection, ultimately resulting in mental health issues such as anxiety and depression [33]. In addition to its protective and regulatory roles, the microbiome also impacts disease susceptibility, including cancer. Microbial pathogens such as Fusobacterium nucleatum and Escherichia coli (E. coli) facilitate carcinogenesis, whereas beneficial metabolites like butyrate exhibit anti-carcinogenic properties [34,35]. The microbiome further influences cancer susceptibility via modulating inflammation, DNA damage, and metabolic processes, enhancing responses to immunotherapy and chemotherapy [36]. Children with malignancies clearly have different microbiota compositions than healthy children. Children with leukemia, for example, exhibit altered bacterial patterns and reduced microbial diversity [37].
Dysbiosis in the microbiome has been associated with a broad spectrum of disorders. It plays a significant role in the pathogenesis of chronic inflammatory conditions, mainly inflammatory bowel disease (IBD), which is due to an imbalance in the composition of microorganisms, which results in a decrease in the number of beneficial bacteria and an increase in the number of pathogenic microorganisms, thereby promoting chronic inflammation [38]. In IBD, a decrease in the abundance of anti-inflammatory bacteria (such as Faecalibacterium prausnitzii) and an increase in pro-inflammatory taxa aggravate mucosal immune activation and tissue injury [39]. A similar association has been shown between dysbiosis and metabolic syndrome and obesity, all of which are disorders that are defined by altered energy extraction and elevated inflammatory responses, both of which contribute to the course of disease [40]. Gut microbiota in obesity exhibits increased Firmicutes-to-Bacteroidetes ratios and an enhanced capacity for energy harvest from the diet, promoting fat deposition and low-grade inflammation [41]. In type 2 diabetes, there is a reduced abundance of butyrate-producing bacteria and an increased proportion of Gram-negative bacteria, which collectively promote inflammation and impair glucose metabolism [42]. In addition, changes in the microbiome can cause disruptions in immunological tolerance, which can lead to the development of allergic disorders, autoimmune diseases, and insulin resistance in diabetics [43]. Dysbiosis can influence the gut–brain axis, exacerbating neuroinflammation in neurodegenerative disorders such as Alzheimer’s disease, Parkinson’s disease, Multiple Sclerosis, Huntington’s Disease, Autism Spectrum Disorder, and Chronic Fatigue Syndrome [44] (Table 2). Host genetic characteristics, antibiotic use, immunological response, nutrition, drugs, infections, chronic diseases, and environmental exposures are all factors that might lead to microbial dysbiosis [45,46]. The human microbiome has become a critical factor in understanding the progression of diseases and their responses to therapy, shedding light on the broader implications for health [47].
Table 2. Delineates disease-specific gut microbiota compositional shifts, underlying host and environmental factors contributing to microbial dysbiosis, and the resultant pathophysiological effects on neuroinflammatory and neurodegenerative processes via the gut–brain axis.
Table 2. Delineates disease-specific gut microbiota compositional shifts, underlying host and environmental factors contributing to microbial dysbiosis, and the resultant pathophysiological effects on neuroinflammatory and neurodegenerative processes via the gut–brain axis.
DiseaseMicrobiome InvolvedFactors Contributing to DysbiosisEffect of DysbiosisReference
Parkinson’s disease Akkermansia, Lactobacillus
Prevotella, Roseburia
Genetic traits, pesticide exposure, antibiotic use, dietExacerbates neuroinflammation via gut–brain axis[48]
Alzheimer’s disease Escherichia/Shigella, Bacteroides
Faecalibacterium prausnitzii
Aging, poor nutrition, inflammation, chronic diseasePromotes neuroinflammation and amyloid-beta deposition[49]
Multiple Sclerosis Methanobrevibacter smithii, Akkermansia
Clostridia,
Vitamin D deficiency, antibiotics, environmental factorsAlters immune regulation and gut permeability [50]
Huntington’s Disease Proteobacteria,
Lactobacillus
Genetic mutation, oxidative stress, altered nutritionDisrupts metabolic and neuroimmune homeostasis[51]
Autism Spectrum Disorder Clostridia, Desulfovibrio
Bifidobacterium
Cesarean birth, early antibiotic exposure, formula feedingImpairs neurodevelopment via microbial metabolite imbalance[52]
Chronic Fatigue SyndromeEnterobacteriaceae
Faecalibacterium, SCFA-producing bacteria
Viral infections, chronic stress, gut permeabilityDrives systemic inflammation and immune dysfunction[53]

2. Microbiome in Cancer

Recent research highlights the complex interaction between the microbiome and cancer, showing that microbial populations are essential in cancer development and progression [54], influencing carcinogenesis through mechanisms such as immune system modulation, microbial metabolite synthesis, and influence on cancer treatments [55]. About 20% of human cancers are linked to microbes [56] and understanding these connections could pave the way for new prevention strategies and treatments that target microbial influences on cancer. The microbiome aids in cancer development through inflammation and genotoxic agents. A modification in the microbiome elevates pro-inflammatory cytokine levels, thereby disrupting the immune system and increasing the risk of various cancers, such as pancreatic and colorectal cancer, which not only promote tumor growth but also hinder the effectiveness of treatments [57]. Currently, eleven organisms—seven viruses (Hepatitis B, type 1 human T cell lymphotropic virus, Epstein–Barr virus, Hepatitis C, Kaposi sarcoma herpesvirus, human immunodeficiency virus-1, human papilloma viruses), three platyhelminths (Schistosoma haematobium, Clonorchis sinensis, Opisthorchis viverrini), and one bacterium (Helicobacter pylori)—have been explicitly identified as unique causes of cancer in humans [58] (Figure 2). These microorganisms play a significant role in cancer progression including neuroblastoma (NB) through several mechanisms, such as activation of B cell differentiation, disruption of normal cell cycle, inflammation, angiogenesis, cytokine production, cell proliferation and transformation, metabolic shift, invasion, metastasis, stromal reprogramming, desmoplasia, genomic instability, tissue remodeling, DNA damage, immune hyperactivation [59] (Table 3). T-cell dysregulation (in EBV and HTLV infection), and direct oncogenesis induced by Hepatitis virus and KSV in hepatocellular carcinomas and Kaposi sarcoma [60]. The gut microbiome influences the development, progression, and therapeutic outcomes of both gastrointestinal (e.g., colorectal, gastric, esophageal, pancreatic) [61] and non-gastrointestinal cancers (e.g., lung, breast, prostate, ovarian, skin) [62].
Table 3. Overview of microbe-associated signaling pathways implicated in various cancers, detailing specific microbial species, the molecular mechanisms they influence, and their resultant effects on tumorigenesis, immune modulation, and cancer progression.
Table 3. Overview of microbe-associated signaling pathways implicated in various cancers, detailing specific microbial species, the molecular mechanisms they influence, and their resultant effects on tumorigenesis, immune modulation, and cancer progression.
CancerMicrobesPathwayMechanismEffectsReference
Colorectal CancerFusobacterium nucleatumWnt/β-cateninFadA from F. nucleatum activates β-catenin via E-cadherin bindingPromote proliferation and tumor initiation[63]
Enterotoxigenic Bacteroides fragilisNFκB/STAT3BFT toxin and LPS stimulate chronic inflammation,
IL-6 upregulates STAT3
Drives inflammation and immune evasion[64]
E. coli (pks+) islandPI3K/AKT/mTORColibactin and ROS activate PI3K/Akt
Promotes tumorigenesis
Alters tumor suppressor genes TP53 and proto-oncogenes KRAS
Enhance survival, angiogenesis[65]
F. nucleatum, E. coliTLR/MyD88/MAPKMicrobial ligands activate TLRs → MyD88 → MAPK cascadeInflammatory signaling, cytokine production[66]
Gastric CancerH. pylori (CagA)SHP-2/Ras/ERKCagA protein activates NFκB (direct injection into epithelial cells); LPS induces inflammation
loss of polarity and hyperproliferation
Promotes cell proliferation and transformation[67]
H. pylori,
Peptostreptococcus
NFκB/STAT3IL-6, TNFα driven by H. pylori and others → survival signalingChronic inflammation and immune modulation[68]
H. pylori (VacA)PI3K/AKTSCFAs and VacA promote survival and immune suppressionCell survival and metabolic shift[69]
F. nucleatum
Peptostreptococcus,
EMT-related (Snail, Twist)Inflammation triggers EMT programs via NFκB and othersInvasion and metastasis[70]
Esophageal CancerP. gingivalis,
F. nucleatum
NFκB/IL-6/STAT3Chronic exposure to P. gingivalis LPS and F. nucleatum leads to NFκB activation → IL-6 secretion → STAT3 phosphorylation via TLR4 signaling
Drives cell survival, angiogenesis, and immune evasion
Inflammation-driven survival, growth, and angiogenesis[71]
Candida albicansEGFR/STAT3Candida albicans and oral dysbiosis increase EGF, HER2 receptor kinase, TGF-α (EGFR ligand expression) → EGFR activation → STAT3-driven survival and proliferationProliferation, inhibition of apoptosis[72]
Candida albicans, Prevotella, VeillonellaTLR/MyD88/MAPKTLR2/4 recognize PAMPs from fungi and Gram-negative bacteria → activate MyD88 → downstream MAPKs (ERK, JNK, p38) → cytokine storm and inflammationImmune modulation and inflammation[73]
Pancreatic CancerMalasseziaKRAS/MAPKMutant KRAS is central to PDAC. Microbiota-driven inflammation (e.g., IL-1β, TNFα) enhances KRAS downstream signaling (ERK, MEK)
Increased proliferation and survival
Drives oncogenesis[74,75]
E. coli, MalasseziaHedgehog (Shh/Gli)Microbial imbalance can dysregulate Shh/Gli signaling in the tumor microenvironment, leading to excessive stroma formation, reducing drug delivery and enhancing immune exclusionStromal reprogramming and desmoplasia[76]
F. nucleatum, Malassezia, E. coliTLR/MyD88Bacterial and fungal components (LPS, β-glucans) bind TLRs → MyD88-dependent signaling → NFκB/MAPK activation → inflammation
Macrophage reprogramming to pro-tumor M2 phenotype
Inflammation and immune evasion[77]
Breast
Cancer
Firmicutes, ProteobacteriaEstrogen MetabolismAltered microbiome composition increases β-glucuronidase activity → deconjugates estrogen glucuronide → promotes reabsorption of active estrogenIncreased risk of ER+ breast cancer[78]
Bacteroides spp.EstrobolomeProduces β-glucuronidase enzyme that deconjugates estrogens in the gut → estrogen re-enters circulation (enterohepatic recycling)Elevated estrogen levels promote ER+ breast cancer[78,79]
E. coli, S. aureusNFκB, STAT3Induce ROS and inflammatory cytokines
Promote DNA double-strand breaks
Activate survival and proliferation pathways
Promotes tumorigenesis via genomic instability and inflammation[80]
Lung
Cancer
Akkermansia muciniphilaTLR2–IL-10 axisStimulates TLR2 → promotes anti-inflammatory cytokine IL-10
Enhances gut barrier integrity and Treg induction
Protectiv—reduces inflammation and promotes immune surveillance[81]
Prevotella spp, Veillonella,TLR/MyD88/NFκBBacterial PAMPs (e.g., LPS) bind TLRs (TLR2, TLR4), activates MyD88 → NFκB pathway, cytokine release (IL-6, TNFα) → chronic inflammationPromotes inflammation, DNA damage, immune suppression → tumor initiation and progression[82]
Bifidobacterium spp.TLR9/IFN-γ signalingStimulates TLR9 on dendritic cells → Increases IFN-γ, CD8+ T cell activation
Enhances antigen presentation
Enhances anti-tumor immunity, reduces immunosuppressive microenvironment[83]
Haemophilus, Streptococcus,MAPK/ERKTLR signaling and microbial cytokines activate MAPK cascade
Activates ERK, p38 → gene expression for proliferation
Increases proliferation, survival, and tissue remodeling favorable for tumor development[84]
Skin
Cancer
Staphylococcus aureusTLR2/TLR4Lipoteichoic acid and peptidoglycan activate TLR2/TLR4 → MyD88-dependent NFκB activation → IL-6, IL-1β secretion
Produces toxins that induce reactive oxygen species (ROS) → oxidative stress
Enhances tumor progression and immune evasion and DNA damage, genomic instability[85]
Staphylococcus epidermidis6-HAP-mediated InhibitionProduces 6-HAP (6-N-hydroxyaminopurine) → inhibits DNA polymerase activity → reduces DNA synthesis in tumor cellsSuppresses tumor growth and exhibits protective effects[86]
Cutibacterium acnesTLR2/NFκBActivates TLR2 on keratinocytes → NFκB activation → pro-inflammatory cytokine release
Induces chronic inflammation and oxidative stress
Promotes DNA damage and tumor initiation[87]
Brain
Cancer
Bacteroides fragilisKynurenine/AHR PathwayAlters tryptophan metabolism → increases kynurenine → activates aryl hydrocarbon receptor (AHR) in brain tissuesFavors glioblastoma progression[88,89]
Clostridium sppEpigenetic Modulation via SCFAsProduces butyrate → inhibits histone deacetylases (HDACs) → promotes apoptosis and DNA repair in glial cellsOpposes tumor proliferation in gliomas[90,91]
Prevotella spp.Th17/IL-17Promotes IL-17-producing Th17 cells via mucosal stimulation → enhances systemic inflammation and disrupts blood–brain barrier (BBB) integrityFacilitates immune cell infiltration and may promote glioma invasiveness[92]
NeuroblastomaBacteroides fragilisSTAT3/NFκBAccumulations of myeloid-derived suppressor cells and inhibition of dendritic cell differentiationSupports tumor progression[93]

2.1. Gut Microbiome in Cancer Development

While both GI and non-GI cancers are influenced by microbial dysbiosis, the nature of this influence differs significantly. In GI cancers, the microbiome exerts a direct effect on tumorigenesis through physical contact with the mucosal epithelium, promoting inflammation, genotoxicity, and epithelial barrier disruption. In contrast, non-GI cancers are affected more indirectly, with the microbiome modulating systemic immune responses and hormone metabolism and influencing the efficacy of immunotherapies. Additionally, non-GI cancers are influenced by microbial metabolites and immune signaling originating from distant sites like the gut or oral cavity. These mechanistic differences underscore the critical need for precision microbiome-targeted strategies tailored to the anatomical site and tumor microenvironment for effective cancer prevention, diagnosis, and treatment. The direct and indirect (or otherwise the GI and non-GI cancer) effects on the composition, function, and determinants of the microbiome are discussed below.

2.1.1. Gastrointestinal Cancer (GI)

Colorectal cancer (CRC) is one of the most extensively studied malignancies affected by the gut microbiome. The colon harbors a high density of microbial populations, and dysbiosis is strongly associated with CRC initiation, progression, and prognosis. CRC malignancies have been found to harbor pathogenic bacteria, including Fusobacterium nucleatum, which promotes inflammation, alters the immunological microenvironment, and enhances tumor aggressiveness by activating Toll-like receptors (TLRs) [94]. F. nucleatum exerts its oncogenic effects adhesin FadA, which attaches to E-cadherin on epithelial cells, triggering the β-catenin/Wnt signaling pathway [63]. This cell proliferation pathway is often dysregulated in colorectal cancer. F. nucleatum stimulates Wnt signaling by stabilizing β-catenin, which translocates to the nucleus and expresses genes that promote cell growth and survival, promoting tumorigenesis [95]. Simultaneously F. nucleatum develops a pro-tumorigenic microenvironment by recruiting immune cells and promoting the synthesis of pro-inflammatory cytokines like IL-6 and TNFα, hence supporting tumor growth and enabling immune evasion [96]. CRC is marked by distinct microbial alterations, including an overabundance of E. coli, and Bacteroides fragilis, along with a reduction in beneficial bacteria like Roseburia and other butyrate-producing bacteria [97]. E. coli, Enterococcus faecalis, and H. pylori are among pathogenic bacteria that cause persistent inflammation that promote carcinogenesis [98]. E. coli strains with the polyketide synthase (pks) island generate colibactin, a genotoxin causing DNA damage that alters tumor suppressor genes TP53 and proto-oncogenes KRAS [65]. Genomic instability caused by this mutagenic effect favors early adenoma formation and cancer development. Enterotoxigenic Bacteroides fragilis generates Bacteroides fragilis toxin (BFT), which degrades E-cadherin, disturbing the tight junctions [64]. This improves cell permeability and triggers the Wnt/β-catenin and Nuclear factor kappa light chain enhancer of activated B cell (NFκB) signaling pathways, promoting inflammation, epithelial hyperplasia, and tumor development [99]. Beneficial gut bacteria such as Faecalibacterium prausnitzii and Roseburia generate SCFAs such as butyrate, which have anti-cancer activity [100]. By inhibiting histone deacetylases (HDAC), butyrate promotes expression of tumor suppressor genes and preserves intestinal epithelial integrity [101]. Conversely, bacteria like Bifidobacterium and Lactobacillus reduce inflammation and provide cancer-preventive effects [102]. H. pylori significantly contributes to the development of gastric cancer (GC) by affecting the gastric epithelium through the release of toxins like CagA (Cytotoxin-associated gene A) and VacA (Vacuolating cytotoxin A) [103]. CagA disrupts epithelial junctions and modifies cell polarity by interfering with important signaling pathways like SHP-2 and Ras/ERK, thereby affecting cell behavior and promoting uncontrolled growth [67]. Beyond H. pylori, other bacteria, including Lactobacillus, Streptococcus, Peptostreptococcus, Fusobacterium, Prevotella, and Veillonella, become more prominent when H. pylori colonization decreases due to antibiotics, immunological responses, and microbial competition [104]. Patients with esophageal cancer exhibit an increased presence of firmicutes, Actinobacteria, Streptococcus, and Actinomyces in the microbiome, along with a decrease in Bacteroidetes and Prevotella [105]. Higher levels of Firmicutes and Actinobacteria, along with Streptococcus and Actinomyces, cause persistent inflammation and oxidative stress, leading to DNA damage [106]. In contrast, a decrease in Bacteroidetes and Prevotella weakens the defensive mechanism, exacerbating inflammation, leading to the progression towards esophageal adenocarcinoma [107]. Pancreatic cancer has been progressively linked to bacterial translocation from the gastrointestinal tract to pancreatic tissues [108], where specific microorganisms, Fusobacterium nucleatum and Porphyromonas gingivalis, identified in pancreatic tumors and ductal tissues are colonized [109]. These bacteria contribute to immune evasion by inhibiting T cell responses and activating inflammatory pathways through TLRs, which drive oncogenic signaling and tumor growth [110]. Additionally, Malassezia, a fungal component, translocates to the pancreas, triggering the complement cascade and further driving tumor growth [74].

2.1.2. Non-Gastrointestinal (Non-GI) Cancer

Comprising malignancies in organs like the breast, lungs, skin, and brain, non-gastrointestinal cancers constitute a major worldwide health burden and greatly influence cancer-related morbidity and mortality [111]. Changes in the local microbiome—in particular a rise in Firmicutes and Proteobacteria—have been linked to breast carcinogenesis and estrogen metabolism [112]. Evidently the gut microbiome influences breast cancer risk mainly by regulating estrogen metabolism [113]. One major pathway is estrogen metabolism via the ‘estrobolome,’ where the bacterial species (e.g., Bacteroides spp.) generate β-glucuronidase, an enzyme deconjugating estrogen glucuronide discharged into the gut [79]. Particularly in postmenopausal women, this reactivation allows estrogens to be reabsorbed into the bloodstream through enterohepatic circulation, thereby increasing estrogen levels and the risk of estrogen receptor-positive (ER+) [78]. Clostridium spp. helps to offset this mechanism by generating butyrate, which lowers inflammation, improves regulatory T cells, and acts as an HDAC inhibitor to support tumor cell death and DNA repair [114]. By activating TLRs to balance cytokines and increase immunoglobulin A (IgA), Lactobacillus spp. helps immunity by encouraging dendritic and NK cells, playing a protective role against breast cancer development and progression [115]. Alterations in microorganisms—especially a decrease in beneficial bacteria like Akkermansia muciniphila and Bifidobacterium spp., and an increase in inflammation-related genera like Prevotella spp.—have been linked to lung cancer development and compromised immunotherapy response [81]. A decrease in Akkermansia muciniphila was found to strengthen the gut barrier and stimulate Th1 immune responses by promoting dendritic cell maturation and interferon-γ production, thereby improving response to anti-PD-1 immunotherapy [116]. Bifidobacterium spp. activates dendritic cells and secretes pro-immune cytokines to increase T cell priming and immune checkpoint inhibitor activity, helping tumor recognition and clearance [117]. Conversely, an increase in Prevotella spp. is associated with low-grade chronic inflammation and immune dysregulation [118]. It promotes Th17-type immune responses and the synthesis of IL-6 and IL-17, which helps in creating a tumor-supportive microenvironment [119]. Microbial dysbiosis in skin cancer—including melanoma and non-melanoma forms—is found to be a source of persistent inflammation, fostering carcinogenesis. In melanoma, the gut microbiota is essential in determining the reaction to immunological checkpoint blockade (ICB) treatments [120]. Particularly pathogens like Staphylococcus aureus have been accompanied by a higher risk of skin malignancies, underscoring the important function of the skin microbiome in cancer development [121]. Melanoma patients with poor responses to immunotherapy are enriched with Prevotella spp. and Ruminococcus spp., which foster immune suppression by growing regulatory T cells and raising anti-inflammatory cytokines [122]. The microbiome influences brain cancers, such as gliomas (tumors originating from glial cells), by modifying immune responses in the central nervous system (CNS) via gut-derived bacteria, which affects tumor proliferation and therapy resistance [88].

2.2. Direct and Indirect Effects of Microbiome

To understand the unique roles of microbes in tumor growth and progression, it is crucial to distinguish between direct and indirect microbial effects on cancer. Direct effects occur when microbial species or their products interact with cancer cells or their immediate microenvironment, altering processes such as cell signaling, DNA integrity, or immune evasion. For instance, gut bacteria can create metabolites that promote apoptosis or decrease tumor cell development [123]. In contrast, indirect effects arise when the microbiome influences host systems, particularly the immune, metabolic, or neuroendocrine systems, which in turn affect cancer development, diagnosis, or treatment outcomes [124]. The GI microbiome interacts directly through colonization, digestion, and immune regulation [125] (Table 4), while the non-gastrointestinal microbiome (skin, oral, etc.) interacts indirectly via immunological, metabolic, and neuroendocrine mechanisms [126].
Table 4. Microbial metabolites, associated microbes, and their roles in cancer progression and immune modulation.
Table 4. Microbial metabolites, associated microbes, and their roles in cancer progression and immune modulation.
CancerMicrobial MetabolitesMicrobes InvolvedRole of Microbial Metabolites in CancerReference
Colorectal cancerSCFA-butyrateClostridium butyricumButyrate enhances immune responses and inhibits tumor progression by altering T cell differentiation and stemness.[127]
Ferroptosis Inhibitors-LactateE. coli,
Klebsiella
Inhibit ferroptosis to prevent iron-dependent oxidative damage and promote CRC cell survival and growth.[128,129]
Indole-3-propionic acidLactobacillus, BifidobacteriumIndole-3-acetate modulates immunological responses via the gut homeostasis-maintaining aryl hydrocarbon receptor (AhR) and enhances mucosal barrier integrity.[130]
Tryptophan Metabolites- kynurenineE. faecalisInhibit immune responses and contribute to tumor immune evasion.[131]
Phenylacetic Acid (PAA)Bacteroides, LactobacillusAffects tumor microenvironment and metabolic pathways by modulating immune responses for cell proliferation.[123]
Gastric cancerN-nitroso Compounds (NOCs)E. coli,
Enterococcus faecalis
Nitrosamines are mutagenic, generating DNA adducts that induce mutations and genetic instability.[132]
tryptophan, arginineLactobacillus, streptococcusUpregulated in neoplastic tissues; facilitate tumor proliferation and immune evasion through metabolic reprogramming.[133]
Esophageal cancerPerfluorooctanoateClostridium leptumIncreased PFOA levels influenced by C. leptum are linked to heightened EC risk. Endocrine disruptor[134]
SCFAsPhascolarctobacterium, Fusobacterium nucleatumSCFAs can modulate inflammation and support tumor growth by promoting lipid synthesis and maintaining epithelial proliferation[107]
LipopolysaccharidesFusobacterium nucleatumPromotes chronic inflammation, leading to epithelial damage and carcinogenesis and activates TLR4/NFκB signaling pathway, increasing IL-6, TNFα[135]
Pancreatic cancerTrimethylamine N-oxideClostridium sporogenes, Anaerococcus hydrogenalisEnhances anti-tumor immunity; administration in PDAC-bearing mice reduced tumor growth and activated effector T cell responses[136]
Indole-3-acetateEnterococcus faecalis, Lactobacillus spp.Suppress the anti-tumor activity by inducing immunosuppressive tumor-associated macrophages[137]
Breast
cancer
SCFA- butyrate, propionate, acetateEubacterium rectale, Clostridium perfringen, Faecalibacterium prausnitziiSCFA act as HDAC inhibitors, inducing apoptosis, cell cycle arrest, and epigenetic changes and modulate IL-10 and TGF-β.[138]
Trimethylamine N-oxideClostridiales, Faecalibacterium, RuminococcaceaeInduces ferroptosis or pyroptosis in tumor cells and promotes anti-tumor immunity[138]
Lung
cancer
kynurenine, indolesClostridium sporogenes, LactobacillusRegulate pulmonary immune microenvironment via aryl hydrocarbon receptor signaling[139]
Secondary bile acidsClostridium, EubacteriumInfluence lung immunity via gut–lung circulation and improve immunotherapy outcomes[139]
Skin
cancer
Lipoteichoic acidStaphylococcus epidermidisInhibits UV-induced skin tumor formation via TLR2 signaling.[140]
Phenol-soluble modulinsStaphylococcus aureus, Cutibacterium acnesPromote inflammation and immune evasion, contributing to squamous cell carcinoma development[140]
Brain
cancer
Polyunsaturated fatty acidsAlistipes, BacteroidesDysregulated PUFA metabolism leads to neuroinflammation, which is a shared mechanism in glioma and brain tumors[141]
Arachidonic acid, Phenylacetic acidBacteroides, Clostridium scindensPromotes neuroinflammation and amyloid-beta aggregation[142]

2.2.1. Microbial Metabolites (Direct Effect)

One significant class of microbial metabolites involved in cancer progression is the secondary bile acids [143]. Clostridium scindens synthesize deoxycholic acid (DCA) through the 7α-dehydroxylation of primary bile acids in hepatocellular carcinoma (HCC) [144]. DCA generates oxidative stress in the liver, leading to ROS accumulation that damages DNA strand breaks, disrupts chromosomal integrity, and promotes mutations in tumor suppressors genes like TP53 [145]. Additionally, DCA activates inflammatory pathways, such as NFκB and STAT3, that promote cell proliferation, angiogenesis, and immune evasion [146]. Microbial genotoxins underline how direct genomic changes and inflammatory signaling pathways in the microbiome contribute to carcinogenesis. In addition to the genotoxic effects, gut bacteria produce microbial metabolites including SCFAs, folate, and B vitamins. These metabolites function as epigenetic controllers, influencing gene expression and contributing to cancer development [147]. SCFAs, particularly butyrate, modify DNA methylation and histone modifications, leading to changes in the gene expression without changing the DNA structure [148]. This process can either drive cancer progression or prevent its occurrence, depending on whether it activates or suppresses oncogenes and tumor suppressor genes [149].

2.2.2. Reactive Oxygen and Nitrogen Species (Direct Effect)

Highly reactive molecules known as reactive oxygen species (ROS) and reactive nitrogen species (RNS) are very important in the development and progression of cancer [150]. They are produced as natural byproducts of cellular metabolism, particularly through mitochondrial activity and immune responses. However, they have the potential to contribute to cellular damage, inflammation, and the development of cancer when their production becomes excessive or unregulated [151]. Enterococcus faecalis can generate ROS, including H2O2 and O2 which damage DNA [152], proteins, lipids, and biological structures. This damage may result in DNA mutations or strand breaks, undermining the integrity of genetic material and consequently heightening the risk of cancer, especially colorectal cancer [153]. ROS and RNS not only induce DNA damage but also activate oncogenic pathways that promote inflammation, cellular proliferation, and cancer progression. The NFκB signaling pathway is one of the most significant pathways triggered by ROS. Under normal circumstances, NFκB is strongly regulated, but excessive ROS causes continuous NFκB activation, releasing pro-inflammatory cytokines including Tumor Necrosis Factor-alpha (TNFα), Interleukin-6 (IL-6), and Interleukin-1 beta (IL-1β) [154]. To that end, we have shown that amplification of ROS could be a sensing mechanism for the activation of the signaling cascade that influences self-fate, including transformation, death, or survival. Previously our studies have shown that an increase in ROS can trigger NFκB signaling that in turn leads to feedback through transactivation of SOD2 and controls the switch that directs self-fate [155]. Eventually we unearthed how the ROS-activated NFκB could initiate and maintain TNFα, IL-1α, cMYC, and SOD2-dependent autocrine and/or paracrine cellular response. In general, NFκB is believed to be an immediate early response, transcriptional regulator, whose activation upon a cellular insult function is like an umbrella response. However, our sequential studies convey the critical requirements of NFκB sustained (2nd/3rd phase activation) through unique systemic cyclic feedbacks for steering specific functions [156]. For instance, ROS-triggered NFκB-mediated TNFα-dependent cyclic feedback steers cellular survival; NFκB-triggered MMP9-mediated ERK-dependent [156] cyclic feedback sustains NFκB for endorsing cellular migration and NFκB-mTOR signaling feedback for clonal expansions [155]. These cytokines not only promote cancer cell survival and multiplication but also cause chronic inflammation. Activation of NFκB also promotes the expression of genes linked to angiogenesis and metastases, therefore accelerating the growth of tumors [157]. Furthermore, ROS and RNS stimulate the signal transducer and activator of the transcription 3 (STAT3) pathway, an important regulatory mechanism for inflammation, by increasing immune evasion by overexpressing genes that allow malignancies to escape immune surveillance. Chronic activation of STAT3 increases cancer cell survival, provides resistance to cell death, and promotes metastasis [158].

2.2.3. Immune Modulation (Indirect Effect)

The immune system is essential for tumor surveillance, maintaining an essential balance between anti-tumor and pro-tumor responses. Immune function is greatly dependent on the microbiome, which regulates both innate and adaptive immune systems and which helps to eliminate tumors or contribute to cancer progression [159]. The immune system is essential for tumor surveillance, with the innate immune system comprising macrophages, neutrophils, and dendritic cells acting as the primary defense. Among these, macrophages can exhibit dual roles: the M1 type combats tumors, while the M2 type supports tumor growth and progression [160]. However, dysbiosis in the microbiome can disturb this equilibrium, leading to activation of TLRs which drive macrophages toward the M2 phenotype, thereby enhancing tumor growth through the stimulation of angiogenesis and modification of adjacent tissues to aid cancer progression [161]. The adaptive immune system, including T cells, B cells, and NK cells, is critical for the elimination of malignant cells. Dysbiosis leads to a disruption in the equilibrium between effector T cells, which are responsible for promoting tumor rejection. Consistently regulatory T cells (Tregs) lead to the expansion of Tregs in the TME, suppressing the anti-tumor immune response [162].

2.2.4. Cancer Therapy Modulation

Beyond transformation, carcinogenesis, and tumor progression, microbiomes are important for modulating cancer therapy. Particularly, in non-GI cancers, microbial populations found in places like the oral cavity and skin have been proven to influence the effectiveness of immunotherapy, chemotherapy, and radiation treatment among other cancer treatments [163]. Immunotherapy has emerged as a transformative approach in the treatment of cancer, particularly through checkpoint inhibitors such as Programmed Cell Death Protein 1 (PD-1) and Cytotoxic T-Lymphocyte Antigen 4 (CTLA-4), which have altered cancer care by utilizing the immune system of the body to target and destroy tumor cells in diverse cancers such as melanoma, lung cancer, bladder cancer, and other types of cancer [164]. Patients with melanoma who have higher specific skin and gut microbiota species respond better to anti-PD-1 therapy, suggesting that microbiome composition enhances the effectiveness of immunotherapy and immune response [165]. Faecalibacterium prausnitzii and Bacteroides thetaiotaomicron affect the efficacy of immune checkpoint inhibitors like anti-PD-1 by enhancing the anti-tumor immune response by influencing the effector T cell, thereby modulating systemic immune activation [166]. Dysbiosis in the oral, cutaneous, or respiratory microbiome can impair immune responses and diminish the efficacy of chemotherapy [167]. Studies indicate that oral microorganisms alter cytokine production and immunological responses, thereby influencing therapeutic outcomes [168]. Oral dysbiosis in breast cancer patients is correlated with unsatisfactory chemotherapy response, probably due to heightened inflammation and modified drug metabolism [169], which eventually influences the immune system’s response to treatment by modulating inflammation, altering metabolic pathways, and thus impacting the effectiveness of cancer therapies, diminishing their efficacy.

3. The Role of Gut Microbiota in Pediatric Cancer: Implications for Immune Modulation, Dysbiosis, and Therapeutic Interventions

Childhood malignancies are the primary cause of disease-related morbidity and mortality in children in the United States and around the globe, after accidents [170]. Among childhood cancers, acute lymphoblastic leukemia (ALL) accounts for around 25% of all pediatric cancers [171], with leukemias overall being the most prevalent, followed by brain and CNS tumors. Within the pediatric solid tumors, NB and Wilms tumor are the most frequently diagnosed cancers [172]. The gut microbiota plays a pivotal role in the formation and functioning of the immune system [173]. Growth and activity of the immune system depend on the gut microbiota, particularly in childhood cancer, as inadequate immune response can promote cancer growth and progression [174]. The early-life microbiome significantly influences immunological responses through its interaction with gut-associated lymphoid tissue (GALT) [175], which promotes immune tolerance, modulates inflammation, and aids in the development of critical immune cells, including T cells [176], regulatory T cells (Tregs), and antigen-presenting cells, which is essential for differentiating between self- and non-self-antigens [177]. High levels of inflammatory cytokines, such as IL-6, TNFα, and IL-1β, are often observed in pediatric malignancies. These cytokines can alter the composition of the gut microbiota, leading to dysbiosis [178]. Dysbiosis can contribute to gut barrier dysfunction and increased permeability, which facilitates the entry of harmful microbial products, such as lipopolysaccharides (LPS), into the systemic circulation. This mechanism is particularly relevant in pediatric cancers, where the gut barrier and immune system are still developing. Unlike adults, children have a more immature epithelial lining and a microbiome in the process of being established, making them more vulnerable to systemic inflammation caused by microbial translocation. Disruptions during this developmental window, due to chemotherapy, antibiotics, poor nutrition, or hospitalization, can have more profound and lasting effects, promoting tumor progression more significantly than in adult malignancies. Additionally, gut bacteria, which play a crucial role in immune system development from infancy, can influence immune responses. These responses can either promote or hinder oncogenesis [179]. This higher gut permeability triggers systematic inflammation, worsens immune system malfunction, and accelerates cancer progression.
Pediatric brain tumors—especially medulloblastomas and gliomas—represent a considerable share of childhood malignancies, with medulloblastomas the most common malignant brain tumor in children [180] and glioblastoma contributing to notable morbidity and death. Neuroinflammation, tumor growth, and immune responses in pediatric brain tumors are influenced by the gut–brain axis [181], an emerging field of study that indicates a critical interaction between gut microbiota and CNS pathology. In addition to immune signaling and microbial metabolites, recent evidence suggests that the gut microbiota may influence levels of brain-derived neurotrophic factor (BDNF), a key modulator of neurodevelopment, synaptic plasticity, and cognitive function [182]. Alterations in the gut microbiome, especially during early life, can reduce BDNF expression in brain regions such as the hippocampus and cortex, potentially impairing hippocampal neurogenesis and microglial regulation [183]. In pediatric brain tumors like medulloblastoma and glioma, reduced BDNF levels have been associated with tumor-associated neuroinflammation, which may exacerbate disease progression [184]. Thus, gut microbiota-induced modulation of BDNF represents another critical axis through which microbial dysbiosis can influence CNS pathology in pediatric cancers. The gut microbiome can influence the immune system’s capacity to identify and react to tumor cells via microbial metabolites, such as LPS and tryptophan metabolites, which travel the blood–brain barrier (BBB) and impact neuroimmune signaling [185]. SCFAs, such as butyrate and propionate, have shown the ability to modulate neuroinflammation by enhancing the production of anti-inflammatory cytokines, particularly Interleukin-10 (IL-10), and maintaining the integrity of the gut epithelium and BBB [186]. The gut microbiota is crucial for the maturation and activation of microglia, the resident immune cells of the CNS, with microbial signals necessary for development [187]. In addition to this, the gut microbiota profoundly affects the CNS via multiple processes, including microglial activity, neurotransmitter synthesis, and possibly increasing neuroinflammation linked to brain cancers [188]. Therefore, dysbiosis can lead to chronic microglial activation, fostering a pro-tumor environment in gliomas [189]. Similarly, lymphomas, comprising Hodgkin’s lymphoma (HL) and non-Hodgkin’s lymphoma (NHL), are prevalent pediatric malignancies that affect the immune system [190]. The gut microbiome is essential for the modulation of the systemic immune system, as it influences the development of lymphoid tissue, the differentiation of immune cells, and chronic inflammation—key factors in lymphomagenesis [191]. The absence of beneficial bacteria, including Firmicutes and Bacteroidetes, can further exacerbate systemic inflammation and disrupt immune homeostasis, thereby contributing to the development of lymphoma [192]. Although the effect of gut bacteria on solid cancers such NB and Wilms tumor is yet unknown, early-life microbial exposure can be suggested as a preventive measure against NB development [193]. NB, a tumor of the sympathetic nervous system, arises from neural crest cells (NCCs) [194] and its progression is influenced by immune system interactions [195] modulated by gut microbiota. Initial immunological conditioning by the gut microbiota helps the immune system distinguish between self- and non-self-antigens, while breastfeeding supports this process by promoting beneficial bacteria like Bifidobacterium and Lactobacillus [196], which enhance immune development and may reduce NB risk through increased IgA production.
Early infancy is known as the “critical period” due to the increased susceptibility of the microbiome to alteration, where the gut flora eventually becomes stable throughout the course [197]. In pediatric patients with AML, Bacteroides and Bifidobacterium levels were considerably decreased six weeks post-leukemia treatment, when compared to control subjects, despite the recovery of other genera [198]. This reveals that children receiving cancer treatment during this critical period develop microbial dysbiosis even after the last course of therapy. Dysbiosis is primarily caused by the depletion of microbiomes that are beneficial to health or by an increase in bacteria that are harmful to health [45]. Host genetics shape gut microbiota composition, diversity, and stability. In childhood cancer patients, immune responses and microbiota composition are determined by genetic predisposition. Some genetic variants linked to immunological function, such as TLRs, can change microbial interactions and cause dysbiosis by means of mutations in pattern recognition receptors [199]. Genetic causes of reduced microbial diversity could contribute to increased gut permeability, sometimes referred to as “leaky gut,” which allows bacterial components to enter systemic circulation and causes chronic inflammation [200]—a known risk factor for children malignancies including leukemia. Antibiotic administration significantly contributes to gut microbial dysbiosis in the general population by reducing microbial diversity and altering the composition of beneficial bacteria [201]. One of the most important consequences of antibiotic treatment is the reduction of helpful, commensal bacteria such as Bifidobacterium and Lactobacillus, which helps in the regulation of immune responses, including the promotion of anti-inflammatory cytokines, in which the immune system distinguishes between beneficial and harmful pathogens [202]. The immune system’s capacity to regulate inflammation and sustain tolerance declines when antibiotics decreases the bacterial populations [203]. Pediatric cancer patients receiving rigorous therapies frequently suffer from malnutrition because of appetite, sickness, changed metabolism, and gastrointestinal side effects, which can significantly affect gut microbiota and overall immune system function [204]. More specifically, a diet low in prebiotic-rich foods, such as oligosaccharides, can impair the growth of beneficial gut bacteria. In such conditions, Bifidobacterium, Lactobacillus, and other fiber-fermenting bacteria that are essential for producing SCFAs may be destroyed [205]. More precisely, vitamins can drastically change the balance of gut flora and weaken the immune systems in pediatric cancer patients [206]. Vitamin A is important for keeping epithelial integrity, immune cell growth, and mucosal immunity, and its deficiency can impair the gut barrier, allowing pathogenic microbes to invade and causing dysbiosis [207]. Similarly, low vitamin D intake or absorption brought on by adverse effects from medication may aggravate dysbiosis and raise the risk of cancer development [208]. Because of their immunocompromised state and intensive therapies, pediatric cancer patients sometimes suffer hospital-associated infections (HAIs) [209]. While sanitized, hospitals limit exposure to different natural bacteria, therefore lowering gut microbiota diversity and allowing dangerous bacteria to flourish—which cause dysbiosis [210]. This imbalance worsens the immune system, increasing the risk of infections. Exposure to environmental contaminants such as heavy metals, pesticides, and toxins affects the gut microbiota in children receiving cancer therapy, fostering dangerous bacteria, compromising the gut barrier, and promoting immunological dysfunction and inflammation, which delays recovery [211].
Given the significant consequences of dysbiosis, various techniques are being explored to re-establish gut microbial equilibrium in children receiving cancer therapy. Especially in pediatric cancer patients, prebiotics and probiotics have garnered curiosity for their ability to restore gut flora composition and function [212]. Prebiotics are non-digestible dietary fibers that nourish and promote the growth of beneficial gut bacteria, hence promoting a stable microbiome and enhancing overall immune health; probiotics are advantageous living microorganisms that aid in restoring gut microbiota balance and augmenting immune function [213]. Lactobacillus rhamnosus and Bifidobacterium breve probiotic supplements have been demonstrated to restore microbial diversity in leukemia patients enduring intensive chemotherapy [214], thereby rebalancing gut flora and improving overall health outcomes. Additionally, probiotics also lower the gastrointestinal side effects related to cancer treatment. Two prevalent and debilitating side effects of chemotherapy, diarrhea and mucositis, greatly affect the quality of life in childhood cancer [215]. Studies have demonstrated that probiotic supplements may significantly decrease the severity and length of chemotherapy-induced diarrhea, therefore enhancing treatment tolerance and general well-being [216]. Conversely, prebiotic supplementation—especially with galactooligosaccharides (GOS) and fructooligosaccharides (FOS)—has been shown to enhance Bifidobacterium and Lactobacillus, while lowering the intestinal inflammation in pediatric cancer patients [217]. SCFAs which control immune responses, lower systemic inflammation, improve nutrient absorption, and strengthen gut barrier integrity are produced when prebiotics are fermented by gut bacteria [218]. This helps immune recovery following chemotherapy and helps to prevent malnutrition—a problem in pediatric cancer patients suffering appetite loss and gastrointestinal toxicity. Prebiotics and probiotics are a good way to manage gut flora imbalances, reduce therapy-related issues, and boost the immune system in pediatric cancer patients [219]. In addition to prebiotics and probiotics, other emerging strategies restoring the gut microbial balance include synbiotics (a combination of prebiotics and probiotics), postbiotics (non-viable microbial products or metabolites that confer health benefits), and fecal microbiota transplantation (FMT), which has shown promise in re-establishing microbial diversity in immunocompromised patients [220]. Furthermore, dietary interventions rich in fiber, polyphenols, and fermented foods are being investigated for their ability to support a healthy microbiome and enhance treatment outcomes [221]. These approaches, alongside prebiotic and probiotic supplementation, represent a growing toolkit for managing dysbiosis and improving immune resilience in pediatric oncology. Despite the microbiome restoration techniques in pediatric cancer, recent studies indicate an association between gut microbiota and the progression, immune response, and treatment results in particular malignancies, including NB, which is associated with immunological dysregulation and inflammation, affected by the gut microbiome [222]. Further in-depth investigations on such relationships could result in the development of novel medicines that improve immune function and treatment efficacy.

4. Neuroblastoma

NB, an embryonal neuroendocrine tumor arising from neural crest progenitor cells [223], accounts for 9.1% of pediatric cancer deaths. NB originates from sympathoadrenal progenitors generated from the neural crest during embryonic development and possesses distinct clinical and biological features. NCCs, a unique class of pluripotent cells, arise early in the embryonic development, a trait of the origin of NB [224]. Originating from the ectodermal layer, these cells migrate to parts of the body, developing into a wide spectrum of tissues, including neurons, glial cells, and components of the peripheral nervous system [225]. Despite intensive multi-modal clinical therapy (IMCT) [226,227], more than half of high-risk phenotypic patients will relapse with hematogenous metastasis [228]. The treatment of high-risk illness is rare due to the disease’s heterogeneity, resistance, and poor hematological reserve, resulting in less than 10% five-year overall survival and 2% 10-year survival, compared with 38–71% for low-risk disease [229,230]. Typically, high-risk diseases are characterized by numerous genetic and molecular abnormalities [231]. Somatic MYCN amplification occurs in around 20% of NB cases and is independently associated with advanced stage and poor outcomes. MYCN amplification occurs in just 25–35% of high-risk NB, while 65–75% are classified as MYCN non-amplified (MYCN-na) [232,233]. The IMCT for high-risk NB comprises an (i) induction phase alternating high-dose chemotherapy, (ii) consolidation phase intensifying chemotherapy, radiation, and stem cell transplantation, and (iii) maintenance phase utilizing retinoids, immunotherapy, and cytokines. The first relapse takes >18 months and decreases significantly subsequently due to genetic and molecular rearrangements in undifferentiated tumorigenic NCCs that mediate NB progression [227]. Our recent investigations using a mouse model of PD showed that aggressive CSC-like NB cells exhibit reversible and adaptive plasticity, determining the evolution of NB [234]. Given the varying survival outcomes associated with different risk groups, staging plays a crucial role in determining disease severity and guiding appropriate treatment strategies. NB is categorized according to two important staging systems: the International Neuroblastoma Risk Group Staging System (INRGSS) and International Neuroblastoma Staging System (INSS). The INRGSS (Table 5) determines the stage of NB based on the imaging result, while the INSS (Table 6) uses surgical results to determine the cancer’s stage [235]. In NB, epigenetics plays a critical role [236]. Alterations in genes such as TP53 and MYCN [237] contribute to the progression and severity of the disease by playing a key role in cancer growth and propagation.
The microbiome composition varies significantly throughout different stages of NB, potentially affecting disease progression. In patients with early-stage or low-risk NB, the microbiome is associated with immune homeostasis and tumor progression [238]. Bacteroidetes and Firmicutes are two major phyla in the healthy gut that are essential for maintaining homeostasis and regulating inflammatory responses, thereby enhancing anti-tumor activity [239]. A balanced Firmicutes-to-Bacteroidetes ratio contributes to the optimal gut barrier function, preventing the spread of pro-inflammatory bacterial endotoxins [240]. In addition, the presence of Lactobacillus and Bifidobacterium, two well-known probiotic genera, promotes gastrointestinal health and immune regulation by modulating cytokines and causing anti-inflammatory effects [241]. These probiotics increase regulatory T cell (Treg) function, facilitating immunological tolerance and decreasing excessive immune activation linked to tumor growth [242].
Considering the NCCs origin of NB and its intricate connection with the immune system, the microbiome, which significantly modulates both innate and adaptive immunity, affects tumor growth and therapeutic results. Certain beneficial gut bacteria enhance the anti-tumor immune responses by stimulating essential immunological components. Bifidobacterium species enhance the dendritic cell (DC) functionality via the stimulator of interferon genes (STING) signaling pathway [243] thereby promoting antigen presentation and the activation of cytotoxic T cells [244] essential against NB, where vigorous T cell activation is imperative to combat the immunosuppressive TME (Table 7). In addition to T cells, NK cells are also crucial for the immune response against NB by detecting and eliminating cancer cells without prior sensitization. NB cells often evade NK cell-induced apoptosis by downregulating major histocompatibility complex class I (MHC-I) molecules and secreting immunosuppressive cytokines (Transforming growth factor-beta (TGF-β), IL-6, IL-10, and prostaglandin E2 (PGE2)) [245]. The gut microbiome, particularly Lactobacillus species, enhances NK cell cytotoxicity by modulating cytokine synthesis. This results in increased levels of interferon-gamma (IFN-γ), TNFα, and interleukin-2 [246] and upregulating activating NK cell receptors such as natural killer group 2 member D (NKG2D), which is crucial for targeting NB cells, especially in conjugation with monoclonal antibody therapy like dinutuximab [247]. Another beneficial bacterium, Faecalibacterium prausnitzii, produces butyrate, which plays an important role in immune homeostasis and the mitigation of chronic inflammation [248]. This production of butyrate assists in maintaining a balanced immune response and supports immune-mediated tumor suppression in NB. Although beneficial microbes enhance anti-tumor immunity, certain pathogenic bacteria promote immunosuppressive TME, thereby facilitating immune evasion and tumor progression. NB tumors evade immune surveillance by recruiting regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs), which collectively suppress anti-tumor immune responses and foster immunosuppressive TME [249].
Despite the increasing interest in the microbiome’s function in many malignancies, the relationship between NB and the microbiome remains relatively unexplored. A comprehensive examination of the thus far documented studies sheds some, yet significant, light on the association between NB and the microbiome. These limits significantly recognize the gap in the field, especially for these deadly developmental tumors. Owing to the clear demarcation of clinically favorable immune hot NB with commendable survival compared to the progressive, immune cold NB with negligible survival, the understanding of the microbiome, its role in altering the immune status, and tumor progression is critical.

4.1. Microbiome and Neuroblastoma

4.1.1. Gut Microbiome Predicts the Risk for NB

By applying a Mendelian randomization (MR) approach, J. Chu investigated the casual relationship between gut microbiota and NB [251]. The study was based on large-scale genome-wide association study (GWAS) data from two reputable sources: the MiBioGen consortium for gut microbiota composition and the IEU Open GWAS Project for NB cases, which comprised 1627 children with NB and 3254 controls. Employing the MR method, the author discovered that a higher genetically proxied abundance of the genus Oscillospira predicted a higher risk for NB. Notably, the bacterial class Erysipelotrichia was associated with a low risk of NB in children, suggesting a possibly protective effect. Erysipelotrichia, recognized for producing SCFAs such as butyrate, has been associated with human health and lipid metabolism. Specifically, MRx0029—a gut bacterium in this category—induces neuronal differentiation in NB cells by fatty acid production, hence elevating MAP2 and SYP levels. These findings support Erysipelotrichia’s protective role in NB, possibly due to its influence on neuronal differentiation. Conversely, the genus Oscillospira was associated with an increased risk that contributes to NB pathogenesis and according to their composition, gut microbiota may either suppress or promote the proliferation of NB. While this MR-based approach offers a novel, genetically informed perspective, it complements rather than replaces conventional methods such as observational epidemiology, molecular biology, and clinical risk stratification. By leveraging genetic variants as instrumental variables, MR mitigates confounding and reverse causation, thereby enhancing the robustness of etiological insights into NB. To investigate the genetic pathways, Chu identified two significant single nucleotide polymorphisms (SNPs)—MUC4 and PELI2—linked to Erysipelotrichia. These two significant genes were specifically associated with NB regulatory genes and were enriched in various upstream transcription factors and multiple tumor progression-related pathways. MUC4 was found to be negatively correlated with FGFR1, an oncogene in NB. PELI2, an E3 ubiquitin ligase, modulates inflammatory signaling pathways including NFκB and MAPK. Both genes were linked to cancer-associated Wnt/β-catenin and KRAS signaling pathways, known to drive NB progression and resistance (Figure 3a).

4.1.2. Postnatal (And Not Maternal Transmission) Programming of Microbiome in NB Patients

To uncover the microbiological alterations in NB, Valles-Colomer and colleagues (2024) conducted a comprehensive metagenomic study of children diagnosed with NB [252]. A study involving 288 individuals utilized shotgun metagenomic sequencing to analyze stool samples from 63 newly diagnosed NB patients, 94 healthy controls, 13 healthy siblings, and 59 mothers from each of the patient and control groups. The findings revealed that children with NB had a significantly lower microbial richness and decreased relative abundance of 18 species including bacteria with reported anti-inflammatory properties such as P. dorei, Bifidobacterium, and butyrate-producers (Roseburia, Faecalibacterium spp.) (Figure 3b). In contrast, Enterobacter hormaechei, a potentially pathogenic species, was more prevalent. Functionally, the gut microbiome of these patients showed decreased potential to metabolize carbohydrates (including starch and glycogen), amino acid synthesis (tyrosine and phenylalanine), and vitamin B1 production, along with a shift toward enhanced protein fermentation. These microbiome differences were not observed in healthy siblings or in the mothers of patients, and mother-to-child microbial transmission rates were comparable between all groups. This suggests that alterations in the microbiome of NB patients are likely to occur postnatally and are not attributable to maternal microbial transmission. These findings facilitate further investigation into the potential of microbiome-targeted interventions—such as next-generation probiotics (e.g., P. dorei, Bifidobacterium) or fecal microbiota transplantation—as adjunctive therapy for NB.

4.1.3. Function of Gut Microbe on NB Pathogenesis

Zhang and colleagues (2024) explored the potential causal link between the gut microbiota and NB through a comprehensive bidirectional Mendelian randomization (MR) analysis and meta-analysis [253]. Using large-scale GWAS data from the IEU Open GWAS Project, they analyzed genetic variants associated with the abundance of 196 gut microbial taxa across several taxonomic levels and evaluated their possible causal impacts on NB risk in a cohort comprising of 4881 people (1627 NB patients and 3254 controls). The MR analysis identified six gut microbiota that exhibited a significant causal relationship with NB. Based on their functional associations, these taxa can be broadly categorized into two functional groups: risk-promoting microbiota, such as Lachnospiraceae, and protective microbiota, including Actinobacteria, Bifidobacteriaceae, Bifidobacterium, Desulfovibrio, and Howardella. Among them, Lachnospiraceae is the primary risk factor for NB due to the positive correlation with the expression of BDNF, which is essential for the establishment of peripheral sympathetic and neural crest-derived sensory neurons and promotes metastasis via the TrkB receptor and invades NB via PI3K/Akt/mTOR and MAPK pathways. Conversely, five microbial taxa—Actinobacteria, Bifidobacteriaceae, Bifidobacterium, Desulfovibrio, and Howardella—were found to have protective effects (Figure 3c). Actinobacteria and their bioactive molecules’ extracts from 24 of 90 strains isolated from big algae exhibited their ability to decrease NB cell proliferation and reduce cell viability. Bifidobacterium, a well-known probiotic, was shown to inhibit NB-associated pathways potentially through galactose production, modulation of immune cytokine IL-27, and reduction of the PI3K/Akt/mTOR signaling cascade. The genus Desulfovibrio, which stimulates the production of hydrogen sulfide (H2S), may contribute to the prevention or deceleration of NB progression, as N-acetyl-L-cysteine (NAC), a precursor of hydrogen sulfide (H2S), has been shown to inhibit NB cell proliferation. The genus Howardella, although less understood, possess anti-tumor activity. These findings not only deepen the knowledge of gut microbial effects on NB pathogenesis but also provide new possibilities for microbiota-targeted preventative and treatment approaches in NB control.

4.1.4. Presence of NB Alters Gut Microbiome Composition

Castellani and colleagues (2017) examined how NB significantly alters the intestinal microbiome and other gut-associated systems, highlighting a direct connection between cancer and gut microbial composition [254]. To investigate this, the human NB cell MHH-NB11 was subperitoneally implanted into immunocompromised mice and compared with the control groups that received only culture medium. Ten weeks after tumor induction, a comprehensive analysis including tumor growth quantification, adipose tissue assessment, measurement of circulating hormones and cytokines, bile acid profiling in serum and feces, and 16S rDNA sequencing of ileal contents revealed significant alterations in the intestinal microbiota of the treatment group compared to the control. Animals with tumors exhibited a decrease in Firmicutes and an increase in Bacteroidetes, Beferribacteres, and Tenericutes. Although the differences in microbial populations were not significant, a decrease in Ruminococcus, Dehalobacterium, and the S24-7 group (Bacteroidales family) were observed pointing to gut dysbiosis in tumor-bearing animals. This suggests that NB induces a unique microbial signature, possibly driven by tumor-specific metabolic or immunological changes. The research additionally identified reduced levels of microbiota-derived bile acids in fecal and serum samples from NB mice, including the secondary bile acids lithocholic acid (LCA) and deoxycholic acid (DCA), together with the tertiary bile acid ursodeoxycholic acid (UDC). This reduction indicates dysfunction in gut microbiota which exacerbates inflammation and cachexia in NB (Figure 3d). This demonstrates that NB itself can reciprocally alter the gut microbial environment, likely through tumor-induced metabolic and immunological changes. This perspective is critical for understanding the dynamic interplay between host and microbiota in cancer and supports the rationale for microbiome-targeted interventions aimed at mitigating disease-associated dysbiosis, inflammation, and cachexia.

4.1.5. Microbial Composition Predicts Treatment Outcome in NB (Murine Model)

The relationship between NB, chemotherapy, and gut health has been examined by Castellani and colleagues (2019), who investigated the impact of these factors on metabolism, the fecal microbiome, VOCs, and gut barrier function in a murine model [255]. The human NB cells were implanted into athymic mice, which are immune-deficient, and then these animals were treated with cyclophosphamide (CTX), a common chemotherapy agent used for NB. The presence of a tumor, both independently and in conjunction with chemotherapy, modified the gut microbiota makeup, resulting in increased gut permeability and the induction of systemic inflammation. NB-bearing mice exhibited a decrease in the relative abundance of Lactobacillus, which was further diminished by CTX therapy. The decline of Lactobacillus was associated with elevated levels of pro-inflammatory cytokines (TNFα, IL-6) and a concurrent decrease in anti-inflammatory markers (TGF-β1 and TGF-β2), contributing to a catabolic condition and development of cachexia. Additionally, an increased gut permeability, which allowed the harmful substances to leak into the bloodstream, was observed. Both the existence of NB and the chemotherapy treatment in this study seemed to damage this barrier, hence increasing its permeability and elevating the likelihood of systemic inflammation. Furthermore, despite the disruption of gut barrier integrity characterized by increased epithelial apoptosis and FITC-dextran leakage, the tight junction protein claudin 4 was unexpectedly upregulated, suggesting that cell death, rather than tight junction impairment, was likely responsible for the barrier dysfunction. The study also investigated fecal VOCs as indicators of microbial metabolism and identified tumor-associated differences in VOC profiles, characterized by reduced levels of certain aldehydes and elevated ketones, suggesting tumor-induced abnormalities in microbial metabolism (Figure 3e).

4.1.6. Prebiotic Treatment Mitigates NB-Steered Microbial Mayhem

Obermüller and colleagues (2020) demonstrated that prebiotic treatment with OMNi-LOGiC® FIBRE in a mouse model of NB-induced tumor-associated cachexia (TAC) resulted in significant alterations in gut microbiota and metabolic profiles [256]. This prebiotic, composed of dextrin and partially hydrolyzed guar gum, induced a favorable alteration in microbial composition, notably enhancing Clostridial Family XIII AD3011, known for SCFA production, while reducing Muribaculum, associated with intestinal inflammation. The microbial alterations were associated with significant changes in fecal VOCs, indicating that the prebiotic influenced microbial metabolism. In TAC mice, inflammatory-related taxa such as Muribaculum were elevated, whereas crucial butyrate-producing bacteria like Eubacterium and Roseburia were diminished.
In parallel, the study also revealed distinct alterations in microbial community. Esters and ketones are the most predominant class affected by NB-associated TAC, with significantly lower quantities of acetone, methylvinylketone, 3-pentanone, and 2-methyl-3-hexanone. These reductions are closely associated with host metabolic stress, particularly in lipid and energy metabolism, as these substances are typically byproducts of microbial fermentation and host metabolic processes involved in fat and amino acid degradation. The results underscore a complex bidirectional connection between NB and the gut microbiome: the malignancy disrupts gut barrier integrity and microbial equilibrium, while the microbiota subsequently alters metabolic and inflammatory responses. Though the prebiotic by itself did not correct the cachexia condition, its capacity to alter the microbiome positively suggests potential for future therapeutic strategies targeting the gut microbiota to support NB patients (Figure 3f).

4.1.7. Microbial Composition in TME Predicts NB Outcome

Li. X and colleagues (2022) explored the intriguing relationship between the tumor-associated microbiome and patient outcomes in NB [257]. This research analyzed RNA sequencing data from 120 tumor samples to determine if microbial gene expression in the tumor microenvironment could serve as a novel prognostic marker. Utilizing machine learning methodologies, they developed a Microbial Gene Abundance Score (M-score), which reflects the relative activity of microbial genes within the tumor microenvironment. This stratification revealed two distinct subgroups among high-risk patients: M_high (high microbial gene expression) and M_low (low microbial gene expression). Patients in the M_high group exhibited significantly poor overall and event-free survival compared to those in the M_low group, suggesting that microbial gene expression offers prognostic insights beyond the traditional Children’s Oncology Group (COG) risk classification. Molecular research indicated that tumors in the M_high group exhibited an upregulated activity in specific pathways in NB progression. Particularly, the upregulation of the CREB (cAMP response element-binding protein) pathway and its oncogenic targets, including BCL-2, VEGF, NGF, and IGF2, all of which are known to promote tumor cell survival, angiogenesis, and metastasis, was observed (Figure 3g). These data indicate that microbiomes may actively influence tumor biology and disease progression rather than just coexisting inside the tumor.

5. Clinical Research Landscape: Challenges and Ongoing Trials in Neuroblastoma Therapy

Despite the evidence implicating the microbiome in the pathogenesis and therapeutic responsiveness of NB, several critical challenges impede its clinical translation. A foremost concern is the heterogeneity in microbiome composition, influenced by variables such as age, dietary habits, antibiotic exposure, and geographic factors. This variability complicates the identification of consistent microbial biomarkers that could reliably predict NB risk or therapeutic outcomes. Furthermore, the predominance of cross-sectional studies limits the capacity to elucidate temporal dynamics and causal relationships between microbiome alterations and disease progression. The paucity of pediatric-specific microbiome research further constrains translational applicability, as developmental differences in immune system maturation and microbial colonization render extrapolation from adult oncology data problematic. Additionally, the intricate bidirectional interactions among the host, microbiome, and tumor microenvironment encompassing immune modulation, metabolic reprogramming, and epigenetic regulation pose significant analytical challenges in delineating mechanistic pathways. From a clinical implementation standpoint, the absence of standardized methodologies for microbiome sampling, sequencing, and bioinformatic analysis, coupled with ethical and regulatory concerns surrounding interventions such as FMT in pediatric populations, further complicates integration into routine clinical practice (Table 8).

6. Conclusions

The human microbiome plays a pivotal role in cancer biology, influencing tumor initiation, progression, immune response, and treatment outcomes. Across multiple malignancies, microbial communities have been shown to promote or inhibit carcinogenesis by altering inflammation, metabolic pathways, and immune surveillance. Despite considerable advancements in understanding the role of microbial imbalances in inflammation, metabolic dysregulation, and immune evasion across several malignancies, NBs’ diversity, function, and mechanism are relatively less explored. Emerging evidence continues to reveal the diverse mechanisms through which microbiota affect host physiology, including epigenetic regulation, metabolite production, and interactions with signaling pathways involved in NB oncogenesis.
Considering NB’s origin from NCCs and its dynamic interplay with the immune system, the potential for microbial manipulation of its tumor microenvironment is significant. The available data suggests a complex bidirectional relationship—wherein NB influences microbial composition, and in turn, the altered microbiome may modulate immune responses, inflammatory signaling, and systemic metabolism in ways that could affect tumor growth and therapy outcomes. Further investigations on analyzing the temporal and molecular relationships between microbial alterations in NBs’ genesis, specifically how early-life microbiota disruptions may predispose certain children to NB or affect their immune cell type composition, are needed. Innovative methodologies, like longitudinal microbiome monitoring, gnotobiotic animal models, and high-resolution single-cell sequencing of tumor-associated microbial genes, can elucidate these mechanisms. Moreover, the realization of the therapeutic potential of microbiota alteration for NB cure is still in its early stages. Likewise, understanding the mode of action of the gut and/or tumor microbiome during disease progression and NB evolution is critical. Strategies such as targeted probiotic supplementation, dietary modifications, and microbial metabolite modulation offer promising adjuncts to existing treatments. These approaches may help recalibrate the immune milieu, enhance treatment efficacy, and reduce adverse effects in vulnerable pediatric populations. These critical gaps in knowledge not only underscore the unrealized realm of NB biology but also highlight the immense potential for breakthroughs for the cure of this deadly disease. Exploring how microbial communities influence NB initiation, progression, and therapeutic response may uncover transformative opportunities for personalized medicine, enabling more precise prognostic tools and innovative microbiome-based therapies.

Author Contributions

N.A. contributed to the conception and design of the review. A.K.G., S.A. and N.A. drafted the manuscript, and P.S., S.A. and L.P. helped in revising it critically. A.K.G., P.S., L.P., S.A. and N.A. contributed to the design and the preparation of the illustrations. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by Department of Defense, DoD CA-210339; Kerr Foundation Kerr-28-34200; Oklahoma Center for the Advancement of Science and Technology, OCAST-HR19-045; and The National Institutes of Health P20GM103639.

Institutional Review Board Statement

Not applicable. This study did not involve humans or animals.

Informed Consent Statement

Not applicable. This study did not involve human participants, human data, or human tissue.

Data Availability Statement

All data and materials are available in the main text of the manuscript.

Conflicts of Interest

All authors have nothing to disclose. No financial or non-financial benefits have been received or will be received from any party related directly or indirectly to the subject of this article.

List of Abbreviations

SCFAsShort-chain fatty acids
HMOsHuman milk oligosaccharides
VOCsVolatile organic compounds
IBDInflammatory bowel disease
TMETumor microenvironment
EBVEpstein–Barr virus
HTLVHuman T cell lymphotropic virus
CRCColorectal cancer
TLRsToll-like receptors
BFTBacteroides fragilis toxin
NFκBNuclear factor kappa light chain enhancer of activated B cell
HDACHistone deacetylases
GCGastric cancer
CagAACytotoxin-associated gene
VacAVacuolating cytotoxin A
CNSCentral nervous system
ICBImmunological checkpoint blockade
NOCsN-nitroso Compounds
DCADeoxycholic acid
HCCHepatocellular carcinoma
ROSReactive oxygen species
RNSReactive nitrogen species
TNFαNecrosis Factor-alpha
IL-6Interleukin-6
IL-1βInterleukin-1 beta
STAT3Signal transducer and activator of transcription 3
TregsRegulatory T cells
PD-1Programmed Cell Death Protein 1
CTLA-4Cytotoxic T-Lymphocyte Antigen 4
ALLAcute lymphoblastic leukemia
GALTGut-associated lymphoid tissue
BDNFBrain-derived neurotrophic factor
LPSLipopolysaccharides
BBBBlood-brain barrier
IL-10Interleukin-10
HLHodgkin’s lymphoma
NHLNon-Hodgkin’s lymphoma
NCCNeural crest cells
NBNeuroblastoma
IgAimmunoglobulin A
HAIsHospital-associated infections
GOSGalactooligosaccharides
FOSFructooligosaccharides
IMCTIntensive multi-modal clinical therapy
INRGSSInternational Neuroblastoma Risk Group Staging System
INSSInternational Neuroblastoma Staging System
DCDendritic cell
MHC-IMajor histocompatibility complex class I
TGF-βTransforming growth factor-beta
PGE2Prostaglandin E2
IFN-γInterferon-gamma
NKG2DNatural killer group 2 member D
GWASGenome-wide association study
SNPsSingle nucleotide polymorphisms
MRMendelian randomization
LCALithocholic acid
DCADeoxycholic acid
UDCUrsodeoxycholic acid
CTXCyclophosphamide
TACTumor-associated cachexia
COGChildren’s Oncology Group
CREBcAMP response element-binding protein

References

  1. Kennedy, M.S.; Chang, E.B. The microbiome: Composition and locations. Prog. Mol. Biol. Transl. Sci. 2020, 176, 1–42. [Google Scholar]
  2. Hou, K.; Wu, Z.X.; Chen, X.Y.; Wang, J.Q.; Zhang, D.; Xiao, C.; Zhu, D.; Koya, J.B.; Wei, L.; Li, J.; et al. Microbiota in health and diseases. Signal Transduct. Target. Ther. 2022, 7, 135. [Google Scholar] [CrossRef] [PubMed]
  3. Laterza, L.; Rizzatti, G.; Gaetani, E.; Chiusolo, P.; Gasbarrini, A. The Gut Microbiota and Immune System Relationship in Human Graft-versus-Host Disease. Mediterr. J. Hematol. Infect. Dis. 2016, 8, e2016025. [Google Scholar] [CrossRef] [PubMed]
  4. Deo, P.N.; Deshmukh, R. Oral microbiome: Unveiling the fundamentals. J. Oral. Maxillofac. Pathol. 2019, 23, 122–128. [Google Scholar] [CrossRef]
  5. Tong, X.; Xu, J.; Lian, F.; Yu, X.; Zhao, Y.; Xu, L.; Zhang, M.; Zhao, X.; Shen, J.; Wu, S.; et al. Structural Alteration of Gut Microbiota during the Amelioration of Human Type 2 Diabetes with Hyperlipidemia by Metformin and a Traditional Chinese Herbal Formula: A Multicenter, Randomized, Open Label Clinical Trial. mBio 2018, 9, e02392-17. [Google Scholar] [CrossRef] [PubMed]
  6. Natalini, J.G.; Singh, S.; Segal, L.N. The dynamic lung microbiome in health and disease. Nat. Rev. Microbiol. 2023, 21, 222–235. [Google Scholar] [CrossRef]
  7. Dickson, R.P.; Martinez, F.J.; Huffnagle, G.B. The role of the microbiome in exacerbations of chronic lung diseases. Lancet 2014, 384, 691–702. [Google Scholar] [CrossRef]
  8. Weiser, J.N.; Ferreira, D.M.; Paton, J.C. Streptococcus pneumoniae: Transmission, colonization and invasion. Nat. Rev. Microbiol. 2018, 16, 355–367. [Google Scholar] [CrossRef]
  9. Dasaraju, P.V.; Liu, C. Infections of the respiratory system. In Medical Microbiology, 4th ed.; University of Texas Medical Branch at Galveston: Galveston, TX, USA, 1996. [Google Scholar]
  10. Bassis, C.; Erb-Downward, J.; Dickson, R. Analysis of the Upper Respiratory Tract Microbiotas as the Source of the Lung and Gastric Microbiotas in Healthy Individuals. MBio 2015, 6, e37. [Google Scholar] [CrossRef]
  11. Li, Z.; Li, Y.; Sun, Q.; Wei, J.; Li, B.; Qiu, Y.; Liu, K.; Shao, D.; Ma, Z. Targeting the pulmonary microbiota to fight against respiratory diseases. Cells 2022, 11, 916. [Google Scholar] [CrossRef]
  12. Anand, S.; Mande, S.S. Diet, microbiota and gut-lung connection. Front. Microbiol. 2018, 9, 2147. [Google Scholar] [CrossRef]
  13. Auchtung, T.A.; Fofanova, T.Y.; Stewart, C.J.; Nash, A.K.; Wong, M.C.; Gesell, J.R.; Auchtung, J.M.; Ajami, N.J.; Petrosino, J.F. Investigating Colonization of the Healthy Adult Gastrointestinal Tract by Fungi. mSphere 2018, 3, 00092-18. [Google Scholar] [CrossRef]
  14. Lozupone, C.A.; Stombaugh, J.I.; Gordon, J.I.; Jansson, J.K.; Knight, R. Diversity, stability and resilience of the human gut microbiota. Nature 2012, 489, 220–230. [Google Scholar] [CrossRef] [PubMed]
  15. Leeming, E.R.; Johnson, A.J.; Spector, T.D.; Le Roy, C.I. Effect of Diet on the Gut Microbiota: Rethinking Intervention Duration. Nutrients 2019, 11, 2862. [Google Scholar] [CrossRef] [PubMed]
  16. Zhang, C.; Li, L.; Jin, B.; Xu, X.; Zuo, X.; Li, Y.; Li, Z. The Effects of Delivery Mode on the Gut Microbiota and Health: State of Art. Front. Microbiol. 2021, 12, 724449. [Google Scholar] [CrossRef] [PubMed]
  17. Dominguez-Bello, M.G.; Costello, E.K.; Contreras, M.; Magris, M.; Hidalgo, G.; Fierer, N.; Knight, R. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc. Natl. Acad. Sci. USA 2010, 107, 11971–11975. [Google Scholar] [CrossRef] [PubMed]
  18. Dunn, A.B.; Jordan, S.; Baker, B.J.; Carlson, N.S. The Maternal Infant Microbiome: Considerations for Labor and Birth. MCN Am. J. Matern. Child. Nurs. 2017, 42, 318–325. [Google Scholar] [CrossRef]
  19. Wernroth, M.L.; Peura, S.; Hedman, A.M.; Hetty, S.; Vicenzi, S.; Kennedy, B.; Fall, K.; Svennblad, B.; Andolf, E.; Pershagen, G.; et al. Development of gut microbiota during the first 2 years of life. Sci. Rep. 2022, 12, 9080. [Google Scholar] [CrossRef]
  20. Tanaka, M.; Nakayama, J. Development of the gut microbiota in infancy and its impact on health in later life. Allergol. Int. 2017, 66, 515–522. [Google Scholar] [CrossRef]
  21. Roswall, J.; Olsson, L.M.; Kovatcheva-Datchary, P.; Nilsson, S.; Tremaroli, V.; Simon, M.-C.; Kiilerich, P.; Akrami, R.; Krämer, M.; Uhlén, M. Developmental trajectory of the healthy human gut microbiota during the first 5 years of life. Cell Host Microbe 2021, 29, 765–776.e3. [Google Scholar] [CrossRef]
  22. Ronan, V.; Yeasin, R.; Claud, E.C. Childhood Development and the Microbiome-The Intestinal Microbiota in Maintenance of Health and Development of Disease During Childhood Development. Gastroenterology 2021, 160, 495–506. [Google Scholar] [CrossRef]
  23. Yao, Y.; Cai, X.; Ye, Y.; Wang, F.; Chen, F.; Zheng, C. The Role of Microbiota in Infant Health: From Early Life to Adulthood. Front. Immunol. 2021, 12, 708472. [Google Scholar] [CrossRef] [PubMed]
  24. Pantazi, A.C.; Balasa, A.L.; Mihai, C.M.; Chisnoiu, T.; Lupu, V.V.; Kassim, M.A.K.; Mihai, L.; Frecus, C.E.; Chirila, S.I.; Lupu, A.; et al. Development of Gut Microbiota in the First 1000 Days after Birth and Potential Interventions. Nutrients 2023, 15, 3647. [Google Scholar] [CrossRef] [PubMed]
  25. Dong, T.S.; Gupta, A. Influence of Early Life, Diet, and the Environment on the Microbiome. Clin. Gastroenterol. Hepatol. 2019, 17, 231–242. [Google Scholar] [CrossRef] [PubMed]
  26. Dethlefsen, L.; Huse, S.; Sogin, M.L.; Relman, D.A. The pervasive effects of an antibiotic on the human gut microbiota, as revealed by deep 16S rRNA sequencing. PLoS Biol. 2008, 6, e280. [Google Scholar] [CrossRef]
  27. Goodrich, J.K.; Waters, J.L.; Poole, A.C.; Sutter, J.L.; Koren, O.; Blekhman, R.; Beaumont, M.; Van Treuren, W.; Knight, R.; Bell, J.T. Human genetics shape the gut microbiome. Cell 2014, 159, 789–799. [Google Scholar] [CrossRef]
  28. Yatsunenko, T.; Rey, F.E.; Manary, M.J.; Trehan, I.; Dominguez-Bello, M.G.; Contreras, M.; Magris, M.; Hidalgo, G.; Baldassano, R.N.; Anokhin, A.P. Human gut microbiome viewed across age and geography. Nature 2012, 486, 222–227. [Google Scholar] [CrossRef]
  29. Mörkl, S.; Lackner, S.; Meinitzer, A.; Mangge, H.; Lehofer, M.; Halwachs, B.; Gorkiewicz, G.; Kashofer, K.; Painold, A.; Holl, A. Gut microbiota, dietary intakes and intestinal permeability reflected by serum zonulin in women. Eur. J. Nutr. 2018, 57, 2985–2997. [Google Scholar] [CrossRef]
  30. Hartmann, E.M.; Hickey, R.; Hsu, T.; Betancourt Román, C.M.; Chen, J.; Schwager, R.; Kline, J.; Brown, G.; Halden, R.U.; Huttenhower, C. Antimicrobial chemicals are associated with elevated antibiotic resistance genes in the indoor dust microbiome. Environ. Sci. Technol. 2016, 50, 9807–9815. [Google Scholar] [CrossRef]
  31. Gupt, A.; Naudiyal, S.; Rani, A.; Kumar, S. Mental health and the microbiome: A review of psychological impacts of gut microflora. Curr. Pharmacol. Rep. 2024, 10, 223–236. [Google Scholar] [CrossRef]
  32. Oldereid, T.S.; Jiang, X.; Nordhus, K.S.; Ponzetta, A.; Bjornholt, J.V.; Bjorkstrom, N.K.; Melum, E.; Rasmussen, H. Role of bacteria and microbial metabolites in immune modulation during early life. Scand. J. Immunol. 2024, 99, e13336. [Google Scholar] [CrossRef]
  33. O’Riordan, K.J.; Collins, M.K.; Moloney, G.M.; Knox, E.G.; Aburto, M.R.; Fulling, C.; Morley, S.J.; Clarke, G.; Schellekens, H.; Cryan, J.F. Short chain fatty acids: Microbial metabolites for gut-brain axis signalling. Mol. Cell Endocrinol. 2022, 546, 111572. [Google Scholar] [CrossRef]
  34. Louis, P.; Flint, H.J. Formation of propionate and butyrate by the human colonic microbiota. Environ. Microbiol. 2017, 19, 29–41. [Google Scholar] [CrossRef]
  35. Schwabe, R.F.; Jobin, C. The microbiome and cancer. Nat. Rev. Cancer 2013, 13, 800–812. [Google Scholar] [CrossRef]
  36. Dadgar, N.; Edlukudige Keshava, V.; Raj, M.S.; Wagner, P.L. The Influence of the Microbiome on Immunotherapy for Gastroesophageal Cancer. Cancers 2023, 15, 4426. [Google Scholar] [CrossRef] [PubMed]
  37. Todor, S.B.; Ichim, C. Microbiome Modulation in Pediatric Leukemia: Impact on Graft-Versus-Host Disease and Treatment Outcomes: A Narrative Review. Children 2025, 12, 166. [Google Scholar] [CrossRef] [PubMed]
  38. Nishida, A.; Inoue, R.; Inatomi, O.; Bamba, S.; Naito, Y.; Andoh, A. Gut microbiota in the pathogenesis of inflammatory bowel disease. Clin. J. Gastroenterol. 2018, 11, 1–10. [Google Scholar] [CrossRef] [PubMed]
  39. Franzin, M.; Stefančič, K.; Lucafò, M.; Decorti, G.; Stocco, G. Microbiota and drug response in inflammatory bowel disease. Pathogens 2021, 10, 211. [Google Scholar] [CrossRef]
  40. Cuevas-Sierra, A.; Ramos-Lopez, O.; Riezu-Boj, J.I.; Milagro, F.I.; Martinez, J.A. Diet, Gut Microbiota, and Obesity: Links with Host Genetics and Epigenetics and Potential Applications. Adv. Nutr. 2019, 10, S17–S30. [Google Scholar] [CrossRef]
  41. Liu, B.-N.; Liu, X.-T.; Liang, Z.-H.; Wang, J.-H. Gut microbiota in obesity. World J. Gastroenterol. 2021, 27, 3837. [Google Scholar] [CrossRef]
  42. Li, W.-Z.; Stirling, K.; Yang, J.-J.; Zhang, L. Gut microbiota and diabetes: From correlation to causality and mechanism. World J. Diabetes 2020, 11, 293. [Google Scholar] [CrossRef]
  43. Akagawa, S.; Kaneko, K. Gut microbiota and allergic diseases in children. Allergol. Int. 2022, 71, 301–309. [Google Scholar] [CrossRef] [PubMed]
  44. Tan, L.Y.; Yeo, X.Y.; Bae, H.G.; Lee, D.P.S.; Ho, R.C.; Kim, J.E.; Jo, D.G.; Jung, S. Association of Gut Microbiome Dysbiosis with Neurodegeneration: Can Gut Microbe-Modifying Diet Prevent or Alleviate the Symptoms of Neurodegenerative Diseases? Life 2021, 11, 698. [Google Scholar] [CrossRef] [PubMed]
  45. Duvallet, C.; Gibbons, S.M.; Gurry, T.; Irizarry, R.A.; Alm, E.J. Meta-analysis of gut microbiome studies identifies disease-specific and shared responses. Nat. Commun. 2017, 8, 1784. [Google Scholar] [CrossRef] [PubMed]
  46. Nieuwdorp, M.; Gilijamse, P.W.; Pai, N.; Kaplan, L.M. Role of the microbiome in energy regulation and metabolism. Gastroenterology 2014, 146, 1525–1533. [Google Scholar] [CrossRef]
  47. Sepich-Poore, G.D.; Zitvogel, L.; Straussman, R.; Hasty, J.; Wargo, J.A.; Knight, R. The microbiome and human cancer. Science 2021, 371, eabc4552. [Google Scholar] [CrossRef]
  48. Klann, E.M.; Dissanayake, U.; Gurrala, A.; Farrer, M.; Shukla, A.W.; Ramirez-Zamora, A.; Mai, V.; Vedam-Mai, V. The gut–brain axis and its relation to Parkinson’s disease: A review. Front. Aging Neurosci. 2022, 13, 782082. [Google Scholar] [CrossRef]
  49. Shabbir, U.; Arshad, M.S.; Sameen, A.; Oh, D.-H. Crosstalk between gut and brain in Alzheimer’s disease: The role of gut microbiota modulation strategies. Nutrients 2021, 13, 690. [Google Scholar] [CrossRef]
  50. Campagnoli, L.I.M.; Marchesi, N.; Varesi, A.; Morozzi, M.; Mascione, L.; Ricevuti, G.; Esposito, C.; Galeotti, N.; Pascale, A. New therapeutic avenues in multiple sclerosis: Is there a place for gut microbiota-based treatments? Pharmacol. Res. 2024, 209, 107456. [Google Scholar] [CrossRef]
  51. Ekwudo, M.N.; Gubert, C.; Hannan, A.J. The microbiota–gut–brain axis in Huntington’s disease: Pathogenic mechanisms and therapeutic targets. FEBS J. 2025, 292, 1282–1315. [Google Scholar] [CrossRef]
  52. De Sales-Millán, A.; Aguirre-Garrido, J.F.; González-Cervantes, R.M.; Velázquez-Aragón, J.A. Microbiome–Gut–Mucosal–Immune–Brain Axis and Autism Spectrum Disorder (ASD): A Novel Proposal of the Role of the Gut Microbiome in ASD Aetiology. Behav. Sci. 2023, 13, 548. [Google Scholar] [CrossRef]
  53. Varesi, A.; Deumer, U.-S.; Ananth, S.; Ricevuti, G. The emerging role of gut microbiota in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS): Current evidence and potential therapeutic applications. J. Clin. Med. 2021, 10, 5077. [Google Scholar] [CrossRef]
  54. Bultman, S.J. The microbiome and its potential as a cancer preventive intervention. Semin. Oncol. 2016, 43, 97–106. [Google Scholar] [CrossRef] [PubMed]
  55. Cullin, N.; Azevedo Antunes, C.; Straussman, R.; Stein-Thoeringer, C.K.; Elinav, E. Microbiome and cancer. Cancer Cell 2021, 39, 1317–1341. [Google Scholar] [CrossRef] [PubMed]
  56. Boccellato, F.; Meyer, T.F. Bacteria Moving into Focus of Human Cancer. Cell Host Microbe 2015, 17, 728–730. [Google Scholar] [CrossRef] [PubMed]
  57. Ge, Y.; Wang, X.; Guo, Y.; Yan, J.; Abuduwaili, A.; Aximujiang, K.; Yan, J.; Wu, M. Gut microbiota influence tumor development and Alter interactions with the human immune system. J. Exp. Clin. Cancer Res. 2021, 40, 42. [Google Scholar] [CrossRef]
  58. IARC Working Group on the Evaluation of Carcinogenic Risks to Humans. IARC monographs on the evaluation of carcinogenic risks to humans. Volume 97. 1, 3-butadiene, ethylene oxide and vinyl halides (vinyl fluoride, vinyl chloride and vinyl bromide). IARC Monogr. Eval. Carcinog. Risks Hum. 2008, 97, 3. [Google Scholar]
  59. Bagga, S.; Bouchard, M.J. Cell cycle regulation during viral infection. In Cell Cycle Control: Mechanisms and Protocols; Springer: Berlin/Heidelberg, Germany, 2014; pp. 165–227. [Google Scholar]
  60. Zhang, Y.; Guo, W.; Zhan, Z.; Bai, O. Carcinogenic mechanisms of virus-associated lymphoma. Front. Immunol. 2024, 15, 1361009. [Google Scholar] [CrossRef]
  61. Liu, Y.; Baba, Y.; Ishimoto, T.; Gu, X.; Zhang, J.; Nomoto, D.; Okadome, K.; Baba, H.; Qiu, P. Gut microbiome in gastrointestinal cancer: A friend or foe? Int. J. Biol. Sci. 2022, 18, 4101. [Google Scholar] [CrossRef]
  62. Pevsner-Fischer, M.; Tuganbaev, T.; Meijer, M.; Zhang, S.H.; Zeng, Z.R.; Chen, M.H.; Elinav, E. Role of the microbiome in non-gastrointestinal cancers. World J. Clin. Oncol. 2016, 7, 200–213. [Google Scholar] [CrossRef]
  63. Rubinstein, M.R.; Wang, X.; Liu, W.; Hao, Y.; Cai, G.; Han, Y.W. Fusobacterium nucleatum promotes colorectal carcinogenesis by modulating E-cadherin/β-catenin signaling via its FadA adhesin. Cell Host Microbe 2013, 14, 195–206. [Google Scholar] [CrossRef]
  64. Yang, J.; Wang, X.; Hu, T.; Huang, H.; Chen, G.; Jin, B.; Zeng, G.; Liu, J. Entero-toxigenic Bacteroides fragilis contributes to intestinal barrier injury and colorectal cancer progression by mediating the BFT/STAT3/ZEB2 pathway. Cell Cycle 2024, 23, 70–82. [Google Scholar] [CrossRef]
  65. Arthur, J.C.; Perez-Chanona, E.; Muhlbauer, M.; Tomkovich, S.; Uronis, J.M.; Fan, T.J.; Campbell, B.J.; Abujamel, T.; Dogan, B.; Rogers, A.B.; et al. Intestinal inflammation targets cancer-inducing activity of the microbiota. Science 2012, 338, 120–123. [Google Scholar] [CrossRef]
  66. Yang, Y.; Weng, W.; Peng, J.; Hong, L.; Yang, L.; Toiyama, Y.; Gao, R.; Liu, M.; Yin, M.; Pan, C. Fusobacterium nucleatum increases proliferation of colorectal cancer cells and tumor development in mice by activating toll-like receptor 4 signaling to nuclear factor−κB, and up-regulating expression of microRNA-21. Gastroenterology 2017, 152, 851–866.e24. [Google Scholar] [CrossRef]
  67. Duan, Y.; Xu, Y.; Dou, Y.; Xu, D. Helicobacter pylori and gastric cancer: Mechanisms and new perspectives. J. Hematol. Oncol. 2025, 18, 10. [Google Scholar] [CrossRef] [PubMed]
  68. Lamb, A.; Chen, L.F. Role of the Helicobacter pylori-Induced inflammatory response in the development of gastric cancer. J. Cell. Biochem. 2013, 114, 491–497. [Google Scholar] [CrossRef] [PubMed]
  69. Foegeding, N.J.; Caston, R.R.; McClain, M.S.; Ohi, M.D.; Cover, T.L. An overview of Helicobacter pylori VacA toxin biology. Toxins 2016, 8, 173. [Google Scholar] [CrossRef] [PubMed]
  70. Zhang, S.; Li, C.; Liu, J.; Geng, F.; Shi, X.; Li, Q.; Lu, Z.; Pan, Y. Fusobacterium nucleatum promotes epithelial-mesenchymal transiton through regulation of the lncRNA MIR4435-2HG/miR-296-5p/Akt2/SNAI1 signaling pathway. FEBS J. 2020, 287, 4032–4047. [Google Scholar] [CrossRef]
  71. Yáñez, L.; Soto, C.; Tapia, H.; Pacheco, M.; Tapia, J.; Osses, G.; Salinas, D.; Rojas-Celis, V.; Hoare, A.; Quest, A.F. Co-culture of P. gingivalis and F. nucleatum synergistically elevates IL-6 expression via TLR4 signaling in oral keratinocytes. Int. J. Mol. Sci. 2024, 25, 3611. [Google Scholar] [CrossRef]
  72. Zhu, W.; Phan, Q.T.; Boontheung, P.; Solis, N.V.; Loo, J.A.; Filler, S.G. EGFR and HER2 receptor kinase signaling mediate epithelial cell invasion by Candida albicans during oropharyngeal infection. Proc. Natl. Acad. Sci. USA 2012, 109, 14194–14199. [Google Scholar] [CrossRef]
  73. Liu, J.; Geng, F.; Sun, H.; Wang, X.; Zhang, H.; Yang, Q.; Zhang, J. Candida albicans induces TLR2/MyD88/NF-κB signaling and inflammation in oral lichen planus-derived keratinocytes. J. Infect. Dev. Ctries 2018, 12, 780–786. [Google Scholar] [CrossRef]
  74. Elaskandrany, M.; Patel, R.; Patel, M.; Miller, G.; Saxena, D.; Saxena, A. Fungi, host immune response, and tumorigenesis. Am. J. Physiol.-Gastrointest. Liver Physiol. 2021, 321, G213–G222. [Google Scholar] [CrossRef] [PubMed]
  75. Bansod, S.; Dodhiawala, P.B.; Lim, K.-H. Oncogenic KRAS-induced feedback inflammatory signaling in pancreatic cancer: An overview and new therapeutic opportunities. Cancers 2021, 13, 5481. [Google Scholar] [CrossRef] [PubMed]
  76. Iriana, S.; Asha, K.; Repak, M.; Sharma-Walia, N. Hedgehog signaling: Implications in cancers and viral infections. Int. J. Mol. Sci. 2021, 22, 1042. [Google Scholar] [CrossRef] [PubMed]
  77. Hu, L.; Liu, Y.; Kong, X.; Wu, R.; Peng, Q.; Zhang, Y.; Zhou, L.; Duan, L. Fusobacterium nucleatum Facilitates M2 Macrophage Polarization and Colorectal Carcinoma Progression by Activating TLR4/NF-κ B/S100A9 Cascade. Front. Immunol. 2021, 12, 658681. [Google Scholar] [CrossRef]
  78. Hu, S.; Ding, Q.; Zhang, W.; Kang, M.; Ma, J.; Zhao, L. Gut microbial beta-glucuronidase: A vital regulator in female estrogen metabolism. Gut Microbes 2023, 15, 2236749. [Google Scholar] [CrossRef]
  79. Sui, Y.; Wu, J.; Chen, J. The role of gut microbial β-glucuronidase in estrogen reactivation and breast cancer. Front. Cell Dev. Biol. 2021, 9, 631552. [Google Scholar] [CrossRef]
  80. Zhang, S.; Huang, J.; Jiang, Z.; Tong, H.; Ma, X.; Liu, Y. Tumor microbiome: Roles in tumor initiation, progression, and therapy. Mol. Biomed. 2025, 6, 1–43. [Google Scholar] [CrossRef]
  81. Souza, V.G.P.; Forder, A.; Pewarchuk, M.E.; Telkar, N.; de Araujo, R.P.; Stewart, G.L.; Vieira, J.; Reis, P.P.; Lam, W.L. The Complex Role of the Microbiome in Non-Small Cell Lung Cancer Development and Progression. Cells 2023, 12, 2801. [Google Scholar] [CrossRef]
  82. Paudel, K.R.; Dharwal, V.; Patel, V.K.; Galvao, I.; Wadhwa, R.; Malyla, V.; Shen, S.S.; Budden, K.F.; Hansbro, N.G.; Vaughan, A. Role of lung microbiome in innate immune response associated with chronic lung diseases. Front. Med. 2020, 7, 554. [Google Scholar] [CrossRef]
  83. Xuan, S.; Ma, Y.; Zhou, H.; Gu, S.; Yao, X.; Zeng, X. The implication of dendritic cells in lung diseases: Immunological role of toll-like receptor 4. Genes Dis. 2023, 11, 101007. [Google Scholar] [CrossRef] [PubMed]
  84. Saleem, S. Targeting MAPK signaling: A promising approach for treating inflammatory lung disease. Pathol.-Res. Pract. 2024, 254, 155122. [Google Scholar] [CrossRef] [PubMed]
  85. Hu, Z. Bacterial and Host Factors in Staphylococcus aureus Septic Arthritis. Master’s Thesis, University of Gothenburg, Gothenburg, Sweden, 2024. [Google Scholar]
  86. Nakatsuji, T.; Chen, T.H.; Butcher, A.M.; Trzoss, L.L.; Nam, S.-J.; Shirakawa, K.T.; Zhou, W.; Oh, J.; Otto, M.; Fenical, W. A commensal strain of Staphylococcus epidermidis protects against skin neoplasia. Sci. Adv. 2018, 4, eaao4502. [Google Scholar] [CrossRef]
  87. Lee, E.; Min, K.; Ahn, H.; Jeon, B.-N.; Park, S.; Yun, C.; Jeon, H.; Yeon, J.-S.; Kim, H.; Park, H. Potential therapeutic skin microbiomes suppressing Staphylococcus aureus-derived immune responses and upregulating skin barrier function-related genes via the AhR signaling pathway. Int. J. Mol. Sci. 2022, 23, 9551. [Google Scholar] [CrossRef]
  88. D’Alessandro, G.; Lauro, C.; Quaglio, D.; Ghirga, F.; Botta, B.; Trettel, F.; Limatola, C. Neuro-Signals from Gut Microbiota: Perspectives for Brain Glioma. Cancers 2021, 13, 2810. [Google Scholar] [CrossRef]
  89. Dehhaghi, M.; Kazemi Shariat Panahi, H.; Heng, B.; Guillemin, G.J. The gut microbiota, kynurenine pathway, and immune system interaction in the development of brain cancer. Front. Cell Dev. Biol. 2020, 8, 562812. [Google Scholar] [CrossRef]
  90. Peng, F.; Li, S.; Wang, L.; Han, M.; Fan, H.; Tang, H.; Peng, C.; Du, J.; Zhou, Z. Combination of Hdac Inhibitor Sodium Butyrate and Immunotherapy in Glioma: Regulation of Immunologically Hot and Cold Tumors Via Gut Microbiota and Metabolites. Front Immunol. 2025, 16, 1532528. [Google Scholar] [CrossRef]
  91. Stein, R.A.; Riber, L. Epigenetic effects of short-chain fatty acids from the large intestine on host cells. Microlife 2023, 4, uqad032. [Google Scholar] [CrossRef]
  92. Bellone, M.; Brevi, A.; Huber, S. Microbiota-propelled T helper 17 cells in inflammatory diseases and cancer. Microbiol. Mol. Biol. Rev. 2020, 84, e00064-19. [Google Scholar] [CrossRef]
  93. Vinay, D.S.; Ryan, E.P.; Pawelec, G.; Talib, W.H.; Stagg, J.; Elkord, E.; Lichtor, T.; Decker, W.K.; Whelan, R.L.; Kumara, H.S. Immune evasion in cancer: Mechanistic basis and therapeutic strategies. In Seminars in Cancer Biology; Elsevier: Amsterdam, The Netherlands, 2015. [Google Scholar]
  94. Yu, M.R.; Kim, H.J.; Park, H.R. Fusobacterium nucleatum Accelerates the Progression of Colitis-Associated Colorectal Cancer by Promoting EMT. Cancers 2020, 12, 2728. [Google Scholar] [CrossRef] [PubMed]
  95. Zhu, Y.; Li, X. Advances of Wnt signalling pathway in colorectal cancer. Cells 2023, 12, 447. [Google Scholar] [CrossRef] [PubMed]
  96. Galasso, L.; Termite, F.; Mignini, I.; Esposto, G.; Borriello, R.; Vitale, F.; Nicoletti, A.; Paratore, M.; Ainora, M.E.; Gasbarrini, A. Unraveling the Role of Fusobacterium nucleatum in Colorectal Cancer: Molecular Mechanisms and Pathogenic Insights. Cancers 2025, 17, 368. [Google Scholar] [CrossRef] [PubMed]
  97. Kim, J.; Lee, H.K. Potential Role of the Gut Microbiome in Colorectal Cancer Progression. Front. Immunol. 2021, 12, 807648. [Google Scholar] [CrossRef] [PubMed]
  98. Hartl, K.; Sigal, M. Microbe-Driven Genotoxicity in Gastrointestinal Carcinogenesis. Int. J. Mol. Sci. 2020, 21, 7439. [Google Scholar] [CrossRef]
  99. Jeon, J.I.; Ko, S.H.; Kim, J.M. Intestinal epithelial cells exposed to Bacteroides fragilis enterotoxin regulate NF-κB activation and inflammatory responses through β-catenin expression. Infect. Immun. 2019, 87, e00312-19. [Google Scholar] [CrossRef]
  100. Nakkarach, A.; Foo, H.L.; Song, A.A.-L.; Mutalib, N.E.A.; Nitisinprasert, S.; Withayagiat, U. Anti-cancer and anti-inflammatory effects elicited by short chain fatty acids produced by Escherichia coli isolated from healthy human gut microbiota. Microb. Cell Factories 2021, 20, 36. [Google Scholar] [CrossRef]
  101. Mirzaei, R.; Afaghi, A.; Babakhani, S.; Sohrabi, M.R.; Hosseini-Fard, S.R.; Babolhavaeji, K.; Akbari, S.K.A.; Yousefimashouf, R.; Karampoor, S. Role of microbiota-derived short-chain fatty acids in cancer development and prevention. Biomed. Pharmacother. 2021, 139, 111619. [Google Scholar] [CrossRef]
  102. Javanmard, A.; Ashtari, S.; Sabet, B.; Davoodi, S.H.; Rostami-Nejad, M.; Esmaeil Akbari, M.; Niaz, A.; Mortazavian, A.M. Probiotics and their role in gastrointestinal cancers prevention and treatment; an overview. Gastroenterol. Hepatol. Bed Bench 2018, 11, 284–295. [Google Scholar]
  103. Ansari, S.; Yamaoka, Y. Role of vacuolating cytotoxin A in Helicobacter pylori infection and its impact on gastric pathogenesis. Expert. Rev. Anti-Infect. Ther. 2020, 18, 987–996. [Google Scholar] [CrossRef]
  104. Fakharian, F.; Asgari, B.; Nabavi-Rad, A.; Sadeghi, A.; Soleimani, N.; Yadegar, A.; Zali, M.R. The interplay between Helicobacter pylori and the gut microbiota: An emerging driver influencing the immune system homeostasis and gastric carcinogenesis. Front. Cell. Infect. Microbiol. 2022, 12, 953718. [Google Scholar] [CrossRef]
  105. Muszynski, D.; Kudra, A.; Sobocki, B.K.; Folwarski, M.; Vitale, E.; Filetti, V.; Dudzic, W.; Kazmierczak-Siedlecka, K.; Polom, K. Esophageal cancer and bacterial part of gut microbiota—A multidisciplinary point of view. Front. Cell Infect. Microbiol. 2022, 12, 1057668. [Google Scholar] [CrossRef]
  106. Lv, J.; Guo, L.; Liu, J.-J.; Zhao, H.-P.; Zhang, J.; Wang, J.-H. Alteration of the esophageal microbiota in Barrett’s esophagus and esophageal adenocarcinoma. World J. Gastroenterol. 2019, 25, 2149. [Google Scholar] [CrossRef]
  107. Moe, K.T.; Tan, K.S.-W. Mechanistic Insights on Microbiota-Mediated Development and Progression of Esophageal Cancer. Cancers 2024, 16, 3305. [Google Scholar] [CrossRef]
  108. Kouzu, K.; Tsujimoto, H.; Kishi, Y.; Ueno, H.; Shinomiya, N. Bacterial translocation in gastrointestinal cancers and cancer treatment. Biomedicines 2022, 10, 380. [Google Scholar] [CrossRef] [PubMed]
  109. D’Antonio, D.L.; Zenoniani, A.; Umme, S.; Piattelli, A.; Curia, M.C. Intratumoral Fusobacterium nucleatum in Pancreatic Cancer: Current and Future Perspectives. Pathogens 2024, 14, 2. [Google Scholar] [CrossRef] [PubMed]
  110. Orlacchio, A.; Mazzone, P. The role of toll-like receptors (TLRs) mediated inflammation in pancreatic cancer pathophysiology. Int. J. Mol. Sci. 2021, 22, 12743. [Google Scholar] [CrossRef] [PubMed]
  111. Mladosievičová, B.; Čierniková, S.; Mego, M. Molecular Mechanisms of Cancer Pathogenesis; Comenius University Bratislava: Bratislava, Slovakia, 2024. [Google Scholar]
  112. Fernandez, M.F.; Reina-Perez, I.; Astorga, J.M.; Rodriguez-Carrillo, A.; Plaza-Diaz, J.; Fontana, L. Breast Cancer and Its Relationship with the Microbiota. Int. J. Environ. Res. Public. Health 2018, 15, 1747. [Google Scholar] [CrossRef]
  113. Parida, S.; Sharma, D. The microbiome–estrogen connection and breast cancer risk. Cells 2019, 8, 1642. [Google Scholar] [CrossRef]
  114. Sun, J.; Chen, S.; Zang, D.; Sun, H.; Sun, Y.; Chen, J. Butyrate as a promising therapeutic target in cancer: From pathogenesis to clinic. Int. J. Oncol. 2024, 64, 44. [Google Scholar] [CrossRef]
  115. Summer, M.; Ali, S.; Fiaz, U.; Tahir, H.M.; Ijaz, M.; Mumtaz, S.; Mushtaq, R.; Khan, R.; Shahzad, H.; Fiaz, H. Therapeutic and immunomodulatory role of probiotics in breast cancer: A mechanistic review. Arch. Microbiol. 2023, 205, 296. [Google Scholar] [CrossRef]
  116. Fan, S.; Jiang, Z.; Zhang, Z.; Xing, J.; Wang, D.; Tang, D. Akkermansia muciniphila: A potential booster to improve the effectiveness of cancer immunotherapy. J. Cancer Res. Clin. Oncol. 2023, 149, 13477–13494. [Google Scholar] [CrossRef]
  117. Pei, B.; Peng, S.; Huang, C.; Zhou, F. Bifidobacterium modulation of tumor immunotherapy and its mechanism. Cancer Immunol. Immunother. 2024, 73, 94. [Google Scholar] [CrossRef]
  118. Larsen, J.M. The immune response to Prevotella bacteria in chronic inflammatory disease. Immunology 2017, 151, 363–374. [Google Scholar] [CrossRef] [PubMed]
  119. Maeda, Y.; Kurakawa, T.; Umemoto, E.; Motooka, D.; Ito, Y.; Gotoh, K.; Hirota, K.; Matsushita, M.; Furuta, Y.; Narazaki, M. Dysbiosis contributes to arthritis development via activation of autoreactive T cells in the intestine. Arthritis Rheumatol. 2016, 68, 2646–2661. [Google Scholar] [CrossRef] [PubMed]
  120. Tomela, K.; Pietrzak, B.; Schmidt, M.; Mackiewicz, A. The tumor and host immune signature, and the gut microbiota as predictive biomarkers for immune checkpoint inhibitor response in melanoma patients. Life 2020, 10, 219. [Google Scholar] [CrossRef] [PubMed]
  121. Coggshall, K.; Brooks, L.; Nagarajan, P.; Arron, S.T. The microbiome and its contribution to skin cancer. Microbiome Cancer 2019, 87–106. [Google Scholar] [CrossRef]
  122. Pietrzak, B.; Tomela, K.; Olejnik-Schmidt, A.; Galus, Ł.; Mackiewicz, J.; Kaczmarek, M.; Mackiewicz, A.; Schmidt, M. A clinical outcome of the anti-PD-1 therapy of melanoma in polish patients is mediated by population-specific gut microbiome composition. Cancers 2022, 14, 5369. [Google Scholar] [CrossRef]
  123. Jaye, K.; Li, C.G.; Chang, D.; Bhuyan, D.J. The role of key gut microbial metabolites in the development and treatment of cancer. Gut Microbes 2022, 14, 2038865. [Google Scholar] [CrossRef]
  124. Xavier, J.B.; Young, V.B.; Skufca, J.; Ginty, F.; Testerman, T.; Pearson, A.T.; Macklin, P.; Mitchell, A.; Shmulevich, I.; Xie, L.; et al. The Cancer Microbiome: Distinguishing Direct and Indirect Effects Requires a Systemic View. Trends Cancer 2020, 6, 192–204. [Google Scholar] [CrossRef]
  125. Pickard, J.M.; Zeng, M.Y.; Caruso, R.; Nunez, G. Gut microbiota: Role in pathogen colonization, immune responses, and inflammatory disease. Immunol. Rev. 2017, 279, 70–89. [Google Scholar] [CrossRef]
  126. Kasarello, K.; Cudnoch-Jedrzejewska, A.; Czarzasta, K. Communication of gut microbiota and brain via immune and neuroendocrine signaling. Front. Microbiol. 2023, 14, 1118529. [Google Scholar] [CrossRef]
  127. Jia, D.; Wang, Q.; Qi, Y.; Jiang, Y.; He, J.; Lin, Y.; Sun, Y.; Xu, J.; Chen, W.; Fan, L. Microbial metabolite enhances immunotherapy efficacy by modulating T cell stemness in pan-cancer. Cell 2024, 187, 1651–1665.e21. [Google Scholar] [CrossRef]
  128. Cui, W.; Guo, M.; Liu, D.; Xiao, P.; Yang, C.; Huang, H.; Liang, C.; Yang, Y.; Fu, X.; Zhang, Y. Gut microbial metabolite facilitates colorectal cancer development via ferroptosis inhibition. Nat. Cell Biol. 2024, 26, 124–137. [Google Scholar] [CrossRef]
  129. Song, Y.-Q.; Yan, X.-D.; Wang, Y.; Wang, Z.-Z.; Mao, X.-L.; Ye, L.-P.; Li, S.-W. Role of ferroptosis in colorectal cancer. World J. Gastrointest. Oncol. 2023, 15, 225. [Google Scholar] [CrossRef]
  130. Niekamp, P.; Kim, C.H. Microbial metabolite dysbiosis and colorectal cancer. Gut Liver 2023, 17, 190. [Google Scholar] [CrossRef] [PubMed]
  131. Ala, M. Tryptophan metabolites modulate inflammatory bowel disease and colorectal cancer by affecting immune system. Int. Rev. Immunol. 2022, 41, 326–345. [Google Scholar] [CrossRef]
  132. Phakathi, M. The Production of N-Nitrosamines by Gut Bacteria. Master’s Thesis, University of the Witwatersrand, Johannesburg, South Africa, 2020. [Google Scholar]
  133. Dai, D.; Yang, Y.; Yu, J.; Dang, T.; Qin, W.; Teng, L.; Ye, J.; Jiang, H. Interactions between gastric microbiota and metabolites in gastric cancer. Cell Death Dis. 2021, 12, 1104. [Google Scholar] [CrossRef] [PubMed]
  134. Li, X.; Xu, B.; Yang, H.; Zhu, Z. Gut Microbiota, Human Blood Metabolites, and Esophageal Cancer: A Mendelian Randomization Study. Genes 2024, 15, 729. [Google Scholar] [CrossRef]
  135. Chiang, H.C.; Hughes, M.; Chang, W.L. The role of microbiota in esophageal squamous cell carcinoma: A review of the literature. Thorac. Cancer 2023, 14, 2821–2829. [Google Scholar] [CrossRef] [PubMed]
  136. Mirji, G.; Worth, A.; Bhat, S.A.; El Sayed, M.; Kannan, T.; Goldman, A.R.; Tang, H.-Y.; Liu, Q.; Auslander, N.; Dang, C.V. The microbiome-derived metabolite TMAO drives immune activation and boosts responses to immune checkpoint blockade in pancreatic cancer. Sci. Immunol. 2022, 7, eabn0704. [Google Scholar] [CrossRef]
  137. Hezaveh, K.; Shinde, R.S.; Klötgen, A.; Halaby, M.J.; Lamorte, S.; Quevedo, R.; Neufeld, L.; Liu, Z.Q.; Jin, R.; Grünwald, B.T. Tryptophan-derived microbial metabolites activate the aryl hydrocarbon receptor in tumor-associated macrophages to suppress anti-tumor immunity. Immunity 2022, 55, 324–340.e8. [Google Scholar] [CrossRef]
  138. Filippou, C.; Themistocleous, S.C.; Marangos, G.; Panayiotou, Y.; Fyrilla, M.; Kousparou, C.A.; Pana, Z.-D.; Tsioutis, C.; Johnson, E.O.; Yiallouris, A. Microbial therapy and breast cancer management: Exploring mechanisms, clinical efficacy, and integration within the one health approach. Int. J. Mol. Sci. 2024, 25, 1110. [Google Scholar] [CrossRef] [PubMed]
  139. Li, X.; Shang, S.; Wu, M.; Song, Q.; Chen, D. Gut microbial metabolites in lung cancer development and immunotherapy: Novel insights into gut-lung axis. Cancer Lett. 2024, 598, 217096. [Google Scholar] [CrossRef] [PubMed]
  140. De Pessemier, B.; Grine, L.; Debaere, M.; Maes, A.; Paetzold, B.; Callewaert, C. Gut–skin axis: Current knowledge of the interrelationship between microbial dysbiosis and skin conditions. Microorganisms 2021, 9, 353. [Google Scholar] [CrossRef] [PubMed]
  141. Qiu, Y.; Hou, Y.; Gohel, D.; Zhou, Y.; Xu, J.; Bykova, M.; Yang, Y.; Leverenz, J.B.; Pieper, A.A.; Nussinov, R. Systematic characterization of multi-omics landscape between gut microbial metabolites and GPCRome in Alzheimer’s disease. Cell Rep. 2024, 43, 114128. [Google Scholar] [CrossRef]
  142. Chen, C.; Liao, J.; Xia, Y.; Liu, X.; Jones, R.; Haran, J.; McCormick, B.; Sampson, T.R.; Alam, A.; Ye, K. Gut microbiota regulate Alzheimer’s disease pathologies and cognitive disorders via PUFA-associated neuroinflammation. Gut 2022, 71, 2233–2252. [Google Scholar] [CrossRef]
  143. Yang, R.; Qian, L. Research on gut microbiota-derived secondary bile acids in cancer progression. Integr. Cancer Ther. 2022, 21, 15347354221114100. [Google Scholar] [CrossRef]
  144. Song, Y.; Lau, H.C.; Zhang, X.; Yu, J. Bile acids, gut microbiota, and therapeutic insights in hepatocellular carcinoma. Cancer Biol. Med. 2024, 21, 144–162. [Google Scholar] [CrossRef]
  145. Mahdi, A.K. Molecular pathology: The Roles of P53 in the Oxidative Stress and DNA Damage Responses in Chronic Liver Disease and Hepatocellular Carcinoma. Master’s Thesis, Newcastle University, Newcastle upon Tyne, UK, 2018. [Google Scholar]
  146. DCA Dichloroacetate Breakthrough Anticancer Agent. Available online: https://truemedmd.com/2019/11/dca-dichloroacetate-breakthrough-anticancer-agent/ (accessed on 25 March 2025).
  147. Liu, J.; Tan, Y.; Cheng, H.; Zhang, D.; Feng, W.; Peng, C. Functions of Gut Microbiota Metabolites, Current Status and Future Perspectives. Aging Dis. 2022, 13, 1106–1126. [Google Scholar] [CrossRef]
  148. Ye, J.; Wu, W.; Li, Y.; Li, L. Influences of the gut microbiota on DNA methylation and histone modification. Dig. Dis. Sci. 2017, 62, 1155–1164. [Google Scholar] [CrossRef]
  149. Chen, X.; Kong, Q.; Zhao, X.; Zhao, C.; Hao, P.; Irshad, I.; Lei, H.; Kulyar, M.F.; Bhutta, Z.A.; Ashfaq, H.; et al. Sodium acetate/sodium butyrate alleviates lipopolysaccharide-induced diarrhea in mice via regulating the gut microbiota, inflammatory cytokines, antioxidant levels, and NLRP3/Caspase-1 signaling. Front. Microbiol. 2022, 13, 1036042. [Google Scholar] [CrossRef] [PubMed]
  150. Kruk, J.; Y Aboul-Enein, H. Reactive oxygen and nitrogen species in carcinogenesis: Implications of oxidative stress on the progression and development of several cancer types. Mini Rev. Med. Chem. 2017, 17, 904–919. [Google Scholar] [CrossRef] [PubMed]
  151. Aggarwal, V.; Tuli, H.S.; Varol, A.; Thakral, F.; Yerer, M.B.; Sak, K.; Varol, M.; Jain, A.; Khan, M.A.; Sethi, G. Role of reactive oxygen species in cancer progression: Molecular mechanisms and recent advancements. Biomolecules 2019, 9, 735. [Google Scholar] [CrossRef]
  152. Huycke, M.M.; Abrams, V.; Moore, D.R. Enterococcus faecalis produces extracellular superoxide and hydrogen peroxide that damages colonic epithelial cell DNA. Carcinogenesis 2002, 23, 529–536. [Google Scholar] [CrossRef]
  153. Gagniere, J.; Raisch, J.; Veziant, J.; Barnich, N.; Bonnet, R.; Buc, E.; Bringer, M.A.; Pezet, D.; Bonnet, M. Gut microbiota imbalance and colorectal cancer. World J. Gastroenterol. 2016, 22, 501–518. [Google Scholar] [CrossRef]
  154. Morgan, M.J.; Liu, Z.G. Crosstalk of reactive oxygen species and NF-kappaB signaling. Cell Res. 2011, 21, 103–115. [Google Scholar] [CrossRef]
  155. Veeraraghavan, J.; Natarajan, M.; Herman, T.S.; Aravindan, N. Low-dose γ-radiation-induced oxidative stress response in mouse brain and gut: Regulation by NFκB–MnSOD cross-signaling. Mutat. Res./Genet. Toxicol. Environ. Mutagen. 2011, 718, 44–55. [Google Scholar] [CrossRef]
  156. Aravindan, N.; Aravindan, S.; Pandian, V.; Khan, F.H.; Ramraj, S.K.; Natt, P.; Natarajan, M. Acquired tumor cell radiation resistance at the treatment site is mediated through radiation-orchestrated intercellular communication. Int. J. Radiat. Oncol. Biol. Phys. 2014, 88, 677–685. [Google Scholar] [CrossRef]
  157. Bahrami, A.; Khalaji, A.; Bahri Najafi, M.; Sadati, S.; Raisi, A.; Abolhassani, A.; Eshraghi, R.; Khaksary Mahabady, M.; Rahimian, N.; Mirzaei, H. NF-kappaB pathway and angiogenesis: Insights into colorectal cancer development and therapeutic targets. Eur. J. Med. Res. 2024, 29, 610. [Google Scholar] [CrossRef]
  158. Wang, Y.; Shen, Y.; Wang, S.; Shen, Q.; Zhou, X. The role of STAT3 in leading the crosstalk between human cancers and the immune system. Cancer Lett. 2018, 415, 117–128. [Google Scholar] [CrossRef]
  159. Charles, A.; Thomas, R.M. The Influence of the microbiome on the innate immune microenvironment of solid tumors. Neoplasia 2023, 37, 100878. [Google Scholar] [CrossRef]
  160. Shen, M.; Du, Y.; Ye, Y. Tumor-associated macrophages, dendritic cells, and neutrophils: Biological roles, crosstalk, and therapeutic relevance. Med. Rev. 2021, 1, 222–243. [Google Scholar] [CrossRef] [PubMed]
  161. Boutilier, A.J.; Elsawa, S.F. Macrophage Polarization States in the Tumor Microenvironment. Int. J. Mol. Sci. 2021, 22, 6995. [Google Scholar] [CrossRef] [PubMed]
  162. Geis, A.L.; Fan, H.; Wu, X.; Wu, S.; Huso, D.L.; Wolfe, J.L.; Sears, C.L.; Pardoll, D.M.; Housseau, F. Regulatory T-cell Response to Enterotoxigenic Bacteroides fragilis Colonization Triggers IL17-Dependent Colon Carcinogenesis. Cancer Discov. 2015, 5, 1098–1109. [Google Scholar] [CrossRef] [PubMed]
  163. Suraya, R.; Nagano, T.; Kobayashi, K.; Nishimura, Y. Microbiome as a Target for Cancer Therapy. Integr. Cancer Ther. 2020, 19, 1534735420920721. [Google Scholar] [CrossRef]
  164. Bouferraa, Y.; Fares, C.; Bou Zerdan, M.; Boyce Kennedy, L. Microbial Influences on Immune Checkpoint Inhibitor Response in Melanoma: The Interplay between Skin and Gut Microbiota. Int. J. Mol. Sci. 2023, 24, 9702. [Google Scholar] [CrossRef]
  165. 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]
  166. Wang, W.; Fan, J.; Zhang, C.; Huang, Y.; Chen, Y.; Fu, S.; Wu, J. Targeted modulation of gut and intra-tumor microbiota to improve the quality of immune checkpoint inhibitor responses. Microbiol. Res. 2024, 282, 127668. [Google Scholar] [CrossRef]
  167. Gopalakrishnan, V.; Helmink, B.A.; Spencer, C.N.; Reuben, A.; Wargo, J.A. The Influence of the Gut Microbiome on Cancer, Immunity, and Cancer Immunotherapy. Cancer Cell 2018, 33, 570–580. [Google Scholar] [CrossRef]
  168. Moreno, C.M.; Boeree, E.; Freitas, C.M.T.; Weber, K.S. Immunomodulatory role of oral microbiota in inflammatory diseases and allergic conditions. Front. Allergy 2023, 4, 1067483. [Google Scholar] [CrossRef]
  169. Lan, Z.; Liu, W.J.; Cui, H.; Zou, K.L.; Chen, H.; Zhao, Y.Y.; Yu, G.T. The role of oral microbiota in cancer. Front. Microbiol. 2023, 14, 1253025. [Google Scholar] [CrossRef]
  170. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2018. CA Cancer J. Clin. 2018, 68, 7–30. [Google Scholar] [CrossRef]
  171. Tai, E.W.; Ward, K.C.; Bonaventure, A.; Siegel, D.A.; Coleman, M.P. Survival among children diagnosed with acute lymphoblastic leukemia in the United States, by race and age, 2001 to 2009: Findings from the CONCORD-2 study. Cancer 2017, 123 (Suppl. S24), 5178–5189. [Google Scholar] [CrossRef]
  172. Ottaviani, G.; Jaffe, N. The epidemiology of osteosarcoma. Cancer Treat. Res. 2009, 152, 3–13. [Google Scholar] [CrossRef] [PubMed]
  173. Wu, H.J.; Wu, E. The role of gut microbiota in immune homeostasis and autoimmunity. Gut Microbes 2012, 3, 4–14. [Google Scholar] [CrossRef] [PubMed]
  174. Lazar, V.; Ditu, L.-M.; Pircalabioru, G.G.; Gheorghe, I.; Curutiu, C.; Holban, A.M.; Picu, A.; Petcu, L.; Chifiriuc, M.C. Aspects of gut microbiota and immune system interactions in infectious diseases, immunopathology, and cancer. Front. Immunol. 2018, 9, 1830. [Google Scholar] [CrossRef] [PubMed]
  175. Bemark, M.; Pitcher, M.J.; Dionisi, C.; Spencer, J. Gut-associated lymphoid tissue: A microbiota-driven hub of B cell immunity. Trends Immunol. 2024, 45, 211–223. [Google Scholar] [CrossRef]
  176. Ignacio, A.; Czyz, S.; McCoy, K.D. Early life microbiome influences on development of the mucosal innate immune system. Semin. Immunol. 2024, 73, 101885. [Google Scholar] [CrossRef]
  177. Zheng, D.; Liwinski, T.; Elinav, E. Interaction between microbiota and immunity in health and disease. Cell Res. 2020, 30, 492–506. [Google Scholar] [CrossRef]
  178. Goswami, M.; Bose, P.D. Gut microbial dysbiosis in the pathogenesis of leukemia: An immune-based perspective. Exp. Hematol. 2024, 133, 104211. [Google Scholar] [CrossRef]
  179. Belkaid, Y.; Hand, T.W. Role of the microbiota in immunity and inflammation. Cell 2014, 157, 121–141. [Google Scholar] [CrossRef]
  180. Thorbinson, C.; Kilday, J.P. Childhood Malignant Brain Tumors: Balancing the Bench and Bedside. Cancers 2021, 13, 6099. [Google Scholar] [CrossRef] [PubMed]
  181. Lin, B.; Ye, Z.; Ye, Z.; Wang, M.; Cao, Z.; Gao, R.; Zhang, Y. Gut microbiota in brain tumors: An emerging crucial player. CNS Neurosci. Ther. 2023, 29 (Suppl. S1), 84–97. [Google Scholar] [CrossRef] [PubMed]
  182. Molska, M.; Mruczyk, K.; Cisek-Woźniak, A.; Prokopowicz, W.; Szydełko, P.; Jakuszewska, Z.; Marzec, K.; Trocholepsza, M. The influence of intestinal microbiota on BDNF levels. Nutrients 2024, 16, 2891. [Google Scholar] [CrossRef] [PubMed]
  183. Sharon, G.; Sampson, T.R.; Geschwind, D.H.; Mazmanian, S.K. The central nervous system and the gut microbiome. Cell 2016, 167, 915–932. [Google Scholar] [CrossRef]
  184. du Chatinier, A.; Velilla, I.Q.; Meel, M.H.; Hoving, E.W.; Hulleman, E.; Metselaar, D.S. Microglia in pediatric brain tumors: The missing link to successful immunotherapy. Cell Rep. Med. 2023, 4, 127668. [Google Scholar] [CrossRef]
  185. Li, D.; Lan, X.; Xu, L.; Zhou, S.; Luo, H.; Zhang, X.; Yu, W.; Yang, Y.; Fang, X. Influence of gut microbial metabolites on tumor immunotherapy: Mechanisms and potential natural products. Front. Immunol. 2025, 16, 1552010. [Google Scholar] [CrossRef]
  186. Silva, Y.P.; Bernardi, A.; Frozza, R.L. The Role of Short-Chain Fatty Acids From Gut Microbiota in Gut-Brain Communication. Front. Endocrinol. 2020, 11, 25. [Google Scholar] [CrossRef]
  187. Abdel-Haq, R.; Schlachetzki, J.C.M.; Glass, C.K.; Mazmanian, S.K. Microbiome-microglia connections via the gut-brain axis. J. Exp. Med. 2019, 216, 41–59. [Google Scholar] [CrossRef]
  188. Camberos-Barraza, J.; Guadrón-Llanos, A.M.; De la Herrán-Arita, A.K. The gut microbiome-neuroglia axis: Implications for brain health, inflammation, and disease. Neuroglia 2024, 5, 254–273. [Google Scholar] [CrossRef]
  189. Keane, L.; Cryan, J.F.; Gleeson, J.P. Exploiting the gut microbiome for brain tumour treatment. Trends Mol. Med. 2025, 31, 213–223. [Google Scholar] [CrossRef]
  190. Marcotte, E.L.; Ritz, B.; Cockburn, M.; Clarke, C.A.; Heck, J.E. Birth characteristics and risk of lymphoma in young children. Cancer Epidemiol. 2014, 38, 48–55. [Google Scholar] [CrossRef] [PubMed]
  191. Jones, G.W.; Hill, D.G.; Jones, S.A. Understanding Immune Cells in Tertiary Lymphoid Organ Development: It Is All Starting to Come Together. Front. Immunol. 2016, 7, 401. [Google Scholar] [CrossRef] [PubMed]
  192. Li, B.; Han, Y.; Fu, Z.; Chai, Y.; Guo, X.; Du, S.; Li, C.; Wang, D. The causal relationship between gut microbiota and lymphoma: A two-sample Mendelian randomization study. Front. Immunol. 2024, 15, 1397485. [Google Scholar] [CrossRef] [PubMed]
  193. Ward, E.; Evans, J. Childhood cancers and immunodeficiency syndromes. Clin. Paediatr. Diet. 2020, 371–392. [Google Scholar] [CrossRef]
  194. Cheung, N.K.; Dyer, M.A. Neuroblastoma: Developmental biology, cancer genomics and immunotherapy. Nat. Rev. Cancer 2013, 13, 397–411. [Google Scholar] [CrossRef]
  195. Subramanian, P.; Mohanvelu, S.; Somasundaram, D.B.; Aravindan, S.; Aravindan, N. Acquired RD3 loss regulates immune surveillance in high-risk and therapy defying progressive neuroblastoma. Cancer Commun. 2024, 44, 1427. [Google Scholar] [CrossRef]
  196. Davis, E.C.; Castagna, V.P.; Sela, D.A.; Hillard, M.A.; Lindberg, S.; Mantis, N.J.; Seppo, A.E.; Jarvinen, K.M. Gut microbiome and breast-feeding: Implications for early immune development. J. Allergy Clin. Immunol. 2022, 150, 523–534. [Google Scholar] [CrossRef]
  197. Rodriguez, J.M.; Murphy, K.; Stanton, C.; Ross, R.P.; Kober, O.I.; Juge, N.; Avershina, E.; Rudi, K.; Narbad, A.; Jenmalm, M.C.; et al. The composition of the gut microbiota throughout life, with an emphasis on early life. Microb. Ecol. Health Dis. 2015, 26, 26050. [Google Scholar] [CrossRef]
  198. van Vliet, M.J.; Tissing, W.J.; Dun, C.A.; Meessen, N.E.; Kamps, W.A.; de Bont, E.S.; Harmsen, H.J. Chemotherapy treatment in pediatric patients with acute myeloid leukemia receiving antimicrobial prophylaxis leads to a relative increase of colonization with potentially pathogenic bacteria in the gut. Clin. Infect. Dis. 2009, 49, 262–270. [Google Scholar] [CrossRef]
  199. Taur, Y.; Jenq, R.R.; Perales, M.A.; Littmann, E.R.; Morjaria, S.; Ling, L.; No, D.; Gobourne, A.; Viale, A.; Dahi, P.B.; et al. The effects of intestinal tract bacterial diversity on mortality following allogeneic hematopoietic stem cell transplantation. Blood 2014, 124, 1174–1182. [Google Scholar] [CrossRef]
  200. Wilde, J.; Slack, E.; Foster, K.R. Host control of the microbiome: Mechanisms, evolution, and disease. Science 2024, 385, eadi3338. [Google Scholar] [CrossRef] [PubMed]
  201. Dethlefsen, L.; Relman, D.A. Incomplete recovery and individualized responses of the human distal gut microbiota to repeated antibiotic perturbation. Proc. Natl. Acad. Sci. USA 2011, 108 (Suppl. S1), 4554–4561. [Google Scholar] [CrossRef] [PubMed]
  202. Gavzy, S.J.; Kensiski, A.; Lee, Z.L.; Mongodin, E.F.; Ma, B.; Bromberg, J.S. Bifidobacterium mechanisms of immune modulation and tolerance. Gut Microbes 2023, 15, 2291164. [Google Scholar] [CrossRef]
  203. Nurgaziyev, M.; Sergazy, S.; Chulenbayeva, L.; Nurgozhina, A.; Gulyayev, A.; Kozhakhmetov, S.; Kartbayeva, G.; Kushugulova, A. The effects of antibiotics on the gut microbiome and the immune system (Review). Georgian Med. News 2020, 167–173. [Google Scholar] [PubMed]
  204. Pedretti, L.; Massa, S.; Leardini, D.; Muratore, E.; Rahman, S.; Pession, A.; Esposito, S.; Masetti, R. Role of Nutrition in Pediatric Patients with Cancer. Nutrients 2023, 15, 710. [Google Scholar] [CrossRef]
  205. Joffe, L.; Ladas, E.J. Nutrition during childhood cancer treatment: Current understanding and a path for future research. Lancet Child. Adolesc. Health 2020, 4, 465–475. [Google Scholar] [CrossRef]
  206. Pham, V.T.; Dold, S.; Rehman, A.; Bird, J.K.; Steinert, R.E. Vitamins, the gut microbiome and gastrointestinal health in humans. Nutr. Res. 2021, 95, 35–53. [Google Scholar] [CrossRef]
  207. Iyer, N.; Vaishnava, S. Vitamin A at the interface of host-commensal-pathogen interactions. PLoS Pathog. 2019, 15, e1007750. [Google Scholar] [CrossRef]
  208. Zhai, Z.; Dong, W.; Sun, Y.; Gu, Y.; Ma, J.; Wang, B.; Cao, H. Vitamin-Microbiota Crosstalk in Intestinal Inflammation and Carcinogenesis. Nutrients 2022, 14, 3383. [Google Scholar] [CrossRef]
  209. Dutta, A.; Flores, R. Infection prevention in pediatric oncology and hematopoietic stem cell transplant recipients. In Healthcare-Associated Infections in Children: A Guide to Prevention and Management; Springer: Berlin/Heidelberg, Germany, 2018; pp. 281–299. [Google Scholar]
  210. Lax, S.; Gilbert, J.A. Hospital-associated microbiota and implications for nosocomial infections. Trends Mol. Med. 2015, 21, 427–432. [Google Scholar] [CrossRef]
  211. Thompson, K.N.; Oulhote, Y.; Weihe, P.; Wilkinson, J.E.; Ma, S.; Zhong, H.; Li, J.; Kristiansen, K.; Huttenhower, C.; Grandjean, P. Effects of Lifetime Exposures to Environmental Contaminants on the Adult Gut Microbiome. Environ. Sci. Technol. 2022, 56, 16985–16995. [Google Scholar] [CrossRef]
  212. Simiakova, M.; Bielik, V. The pros and cons of probiotic use in pediatric oncology patients following treatment for acute lymphoblastic leukemia. Front. Pediatr. 2024, 12, 1427185. [Google Scholar] [CrossRef]
  213. Zolkiewicz, J.; Marzec, A.; Ruszczynski, M.; Feleszko, W. Postbiotics-A Step Beyond Pre- and Probiotics. Nutrients 2020, 12, 2189. [Google Scholar] [CrossRef]
  214. Masetti, R.; Muratore, E.; Leardini, D.; Zama, D.; Turroni, S.; Brigidi, P.; Esposito, S.; Pession, A. Gut microbiome in pediatric acute leukemia: From predisposition to cure. Blood Adv. 2021, 5, 4619–4629. [Google Scholar] [CrossRef] [PubMed]
  215. Montassier, E.; Gastinne, T.; Vangay, P.; Al-Ghalith, G.A.; Bruley des Varannes, S.; Massart, S.; Moreau, P.; Potel, G.; de La Cochetiere, M.F.; Batard, E.; et al. Chemotherapy-driven dysbiosis in the intestinal microbiome. Aliment. Pharmacol. Ther. 2015, 42, 515–528. [Google Scholar] [CrossRef] [PubMed]
  216. Banna, G.L.; Torino, F.; Marletta, F.; Santagati, M.; Salemi, R.; Cannarozzo, E.; Falzone, L.; Ferrau, F.; Libra, M. Lactobacillus rhamnosus GG: An Overview to Explore the Rationale of Its Use in Cancer. Front. Pharmacol. 2017, 8, 603. [Google Scholar] [CrossRef] [PubMed]
  217. Bruno-Barcena, J.M.; Azcarate-Peril, M.A. Galacto-oligosaccharides and Colorectal Cancer: Feeding our Intestinal Probiome. J. Funct. Foods 2015, 12, 92–108. [Google Scholar] [CrossRef]
  218. Ohno, H. The impact of metabolites derived from the gut microbiota on immune regulation and diseases. Int. Immunol. 2020, 32, 629–636. [Google Scholar] [CrossRef]
  219. Slizewska, K.; Markowiak-Kopec, P.; Slizewska, W. The Role of Probiotics in Cancer Prevention. Cancers 2020, 13, 20. [Google Scholar] [CrossRef]
  220. Al-Habsi, N.; Al-Khalili, M.; Haque, S.A.; Elias, M.; Olqi, N.A.; Al Uraimi, T. Health benefits of prebiotics, probiotics, synbiotics, and postbiotics. Nutrients 2024, 16, 3955. [Google Scholar] [CrossRef]
  221. Cardona, F.; Andrés-Lacueva, C.; Tulipani, S.; Tinahones, F.J.; Queipo-Ortuño, M.I. Benefits of polyphenols on gut microbiota and implications in human health. J. Nutr. Biochem. 2013, 24, 1415–1422. [Google Scholar] [CrossRef] [PubMed]
  222. Chen, S.; Zhang, P.; Duan, H.; Wang, J.; Qiu, Y.; Cui, Z.; Yin, Y.; Wan, D.; Xie, L. Gut microbiota in muscular atrophy development, progression, and treatment: New therapeutic targets and opportunities. Innovation 2023, 4, 100479. [Google Scholar] [CrossRef] [PubMed]
  223. Aravindan, N.; Terence, H.; and Aravindan, S. Emerging therapeutic targets for neuroblastoma. Expert. Opin. Ther. Targets 2020, 24, 899–914. [Google Scholar] [CrossRef]
  224. Trainor, P. Neural Crest Cells: Evolution, Development and Disease; Academic Press: Cambridge, MA, USA, 2013. [Google Scholar]
  225. Rubenstein, J.; Puelles, L. Development of the nervous system. In Epstein’s Inborn Errors of Development: The Molecular Basis of Clinical Disorders of Morphogenesis; Oxford University Press: Oxford, UK, 2016; p. 63. [Google Scholar]
  226. Morgenstern, D.A.; Baruchel, S.; Irwin, M.S. Current and future strategies for relapsed neuroblastoma: Challenges on the road to precision therapy. J. Pediatr. Hematol./Oncol. 2013, 35, 337–347. [Google Scholar] [CrossRef]
  227. Simon, T.; Berthold, F.; Borkhardt, A.; Kremens, B.; De Carolis, B.; Hero, B. Treatment and outcomes of patients with relapsed, high-risk neuroblastoma: Results of German trials. Pediatr. Blood Cancer 2011, 56, 578–583. [Google Scholar] [CrossRef]
  228. Maris, J.M.; Hogarty, M.D.; Bagatell, R.; Cohn, S.L. Neuroblastoma. Lancet 2007, 369, 2106–2120. [Google Scholar] [CrossRef]
  229. Garaventa, A.; Parodi, S.; De Bernardi, B.; Dau, D.; Manzitti, C.; Conte, M.; Casale, F.; Viscardi, E.; Bianchi, M.; D’Angelo, P. Outcome of children with neuroblastoma after progression or relapse. A retrospective study of the Italian neuroblastoma registry. Eur. J. Cancer 2009, 45, 2835–2842. [Google Scholar] [CrossRef]
  230. London, W.B.; Castel, V.; Monclair, T.; Ambros, P.F.; Pearson, A.D.; Cohn, S.L.; Berthold, F.; Nakagawara, A.; Ladenstein, R.L.; Iehara, T. Clinical and biologic features predictive of survival after relapse of neuroblastoma: A report from the International Neuroblastoma Risk Group project. J. Clin. Oncol. 2011, 29, 3286–3292. [Google Scholar] [CrossRef]
  231. Caldas, C. Cancer sequencing unravels clonal evolution. Nat. Biotechnol. 2012, 30, 408–410. [Google Scholar] [CrossRef]
  232. Brodeur, G.M. Neuroblastoma: Biological insights into a clinical enigma. Nat. Rev. Cancer 2003, 3, 203–216. [Google Scholar] [CrossRef]
  233. Suita, S.; Tajiri, T.; Kaneko, M.; Hirai, M.; Mugishima, H.; Sugimoto, T.; Tsuchida, Y. Implications of MYCN amplification in patients with stage 4 neuroblastoma who undergo intensive chemotherapy. J. Pediatr. Surg. 2007, 42, 489–493. [Google Scholar] [CrossRef]
  234. Pandian, V.; Ramraj, S.; Khan, F.H.; Azim, T.; Aravindan, N. Metastatic neuroblastoma cancer stem cells exhibit flexible plasticity and adaptive stemness signaling. Stem Cell Res. Ther. 2015, 6, 400. [Google Scholar] [CrossRef] [PubMed]
  235. UT Southwestern Medical Center. Neuroblastoma Diagnosis and Stages. Available online: https://utswmed.org/conditions-treatments/neuroblastoma/neuroblastoma-diagnosis-and-stages/ (accessed on 25 March 2025).
  236. Domingo-Fernandez, R.; Watters, K.; Piskareva, O.; Stallings, R.L.; Bray, I. The role of genetic and epigenetic alterations in neuroblastoma disease pathogenesis. Pediatr. Surg. Int. 2013, 29, 101–119. [Google Scholar] [CrossRef] [PubMed]
  237. Aravindan, N.; Subramanian, K.; Somasundaram, D.B.; Herman, T.S.; Aravindan, S. MicroRNAs in neuroblastoma tumorigenesis, therapy resistance, and disease evolution. Cancer Drug Resist. 2019, 2, 1086. [Google Scholar] [CrossRef]
  238. Sánchez-Alcoholado, L.; Ramos-Molina, B.; Otero, A.; Laborda-Illanes, A.; Ordóñez, R.; Medina, J.A.; Gómez-Millán, J.; Queipo-Ortuño, M.I. The role of the gut microbiome in colorectal cancer development and therapy response. Cancers 2020, 12, 1406. [Google Scholar] [CrossRef]
  239. Stojanov, S.; Berlec, A.; Štrukelj, B. The influence of probiotics on the firmicutes/bacteroidetes ratio in the treatment of obesity and inflammatory bowel disease. Microorganisms 2020, 8, 1715. [Google Scholar] [CrossRef]
  240. Magne, F.; Gotteland, M.; Gauthier, L.; Zazueta, A.; Pesoa, S.; Navarrete, P.; Balamurugan, R. The firmicutes/bacteroidetes ratio: A relevant marker of gut dysbiosis in obese patients? Nutrients 2020, 12, 1474. [Google Scholar] [CrossRef]
  241. Mazziotta, C.; Tognon, M.; Martini, F.; Torreggiani, E.; Rotondo, J.C. Probiotics mechanism of action on immune cells and beneficial effects on human health. Cells 2023, 12, 184. [Google Scholar] [CrossRef]
  242. Zitvogel, L.; Ma, Y.; Raoult, D.; Kroemer, G.; Gajewski, T.F. The microbiome in cancer immunotherapy: Diagnostic tools and therapeutic strategies. Science 2018, 359, 1366–1370. [Google Scholar] [CrossRef]
  243. Yang, L.; Li, A.; Wang, Y.; Zhang, Y. Intratumoral microbiota: Roles in cancer initiation, development and therapeutic efficacy. Signal Transduct. Target. Ther. 2023, 8, 35. [Google Scholar] [CrossRef]
  244. Ma, K.; Wang, L.; Li, W.; Tang, T.; Ma, B.; Zhang, L.; Zhang, L. Turning cold into hot: Emerging strategies to fire up the tumor microenvironment. Trends Cancer 2024, 11, 117–134. [Google Scholar] [CrossRef]
  245. Vanichapol, T.; Chutipongtanate, S.; Anurathapan, U.; Hongeng, S. Immune escape mechanisms and future prospects for immunotherapy in neuroblastoma. BioMed Res. Int. 2018, 2018, 1812535. [Google Scholar] [CrossRef] [PubMed]
  246. Aziz, N.; Bonavida, B. Activation of natural killer cells by probiotics. Onco Ther. 2016, 7, 41–55. [Google Scholar] [CrossRef] [PubMed]
  247. Keyel, M.E.; Reynolds, C.P. Spotlight on dinutuximab in the treatment of high-risk neuroblastoma: Development and place in therapy. Biologics 2018, 13, 1–12. [Google Scholar] [CrossRef] [PubMed]
  248. Martín, R.; Rios-Covian, D.; Huillet, E.; Auger, S.; Khazaal, S.; Bermúdez-Humarán, L.G.; Sokol, H.; Chatel, J.-M.; Langella, P. Faecalibacterium: A bacterial genus with promising human health applications. FEMS Microbiol. Rev. 2023, 47, fuad039. [Google Scholar] [CrossRef]
  249. Pathania, A.S.; Prathipati, P.; Murakonda, S.P.; Murakonda, A.B.; Srivastava, A.; Byrareddy, S.N.; Coulter, D.W.; Gupta, S.C.; Challagundla, K.B. Immune checkpoint molecules in neuroblastoma: A clinical perspective. In Seminars in Cancer Biology; Elsevier: Amsterdam, The Netherlands, 2022; pp. 247–258. [Google Scholar]
  250. Bao, R.; Spranger, S.; Hernandez, K.; Zha, Y.; Pytel, P.; Luke, J.J.; Gajewski, T.F.; Volchenboum, S.L.; Cohn, S.L.; Desai, A.V. Immunogenomic determinants of tumor microenvironment correlate with superior survival in high-risk neuroblastoma. J. Immunother. Cancer 2021, 9, e002417. [Google Scholar] [CrossRef]
  251. Chu, J. Potential cause-and-effect relationship between gut microbiota and childhood neuroblastoma: A mendelian randomization analysis. Indian J. Pediatr. 2024, 92, 717–724. [Google Scholar] [CrossRef]
  252. Valles-Colomer, M.; Manghi, P.; Cumbo, F.; Masetti, G.; Armanini, F.; Asnicar, F.; Blanco-Miguez, A.; Pinto, F.; Punčochář, M.; Garaventa, A. Neuroblastoma is associated with alterations in gut microbiome composition subsequent to maternal microbial seeding. EBioMedicine 2024, 99, 104917. [Google Scholar] [CrossRef]
  253. Zhang, Z.; Li, D.; Xie, F.; Zhang, H. The causal relationship between gut microbiota and neuroblastoma: A bidirectional Mendelian randomization analysis and meta-analysis. Microbiol. Spectr. 2024, 12, e03656-23. [Google Scholar] [CrossRef]
  254. Castellani, C.; Singer, G.; Kaiser, M.; Kaiser, T.; Huang, J.; Sperl, D.; Kashofer, K.; Fauler, G.; Guertl-Lackner, B.; Höfler, G. Neuroblastoma causes alterations of the intestinal microbiome, gut hormones, inflammatory cytokines, and bile acid composition. Pediatr. Blood Cancer 2017, 64, e26425. [Google Scholar] [CrossRef]
  255. Castellani, C.; Singer, G.; Eibisberger, M.; Obermüller, B.; Warncke, G.; Miekisch, W.; Kolb-Lenz, D.; Summer, G.; Pauer, T.M.; ElHaddad, A. The effects of neuroblastoma and chemotherapy on metabolism, fecal microbiome, volatile organic compounds, and gut barrier function in a murine model. Pediatr. Res. 2019, 85, 546–555. [Google Scholar] [CrossRef]
  256. Obermüller, B.; Singer, G.; Kienesberger, B.; Klymiuk, I.; Sperl, D.; Stadlbauer, V.; Horvath, A.; Miekisch, W.; Gierschner, P.; Grabherr, R. The effects of prebiotic supplementation with OMNi-LOGiC® FIBRE on fecal microbiome, fecal volatile organic compounds, and gut permeability in murine neuroblastoma-induced tumor-associated cachexia. Nutrients 2020, 12, 2029. [Google Scholar] [CrossRef]
  257. Li, X.; Wang, X.; Huang, R.; Stucky, A.; Chen, X.; Sun, L.; Wen, Q.; Zeng, Y.; Fletcher, H.; Wang, C. The machine-learning-mediated interface of microbiome and genetic risk stratification in neuroblastoma reveals molecular pathways related to patient survival. Cancers 2022, 14, 2874. [Google Scholar] [CrossRef]
Figure 1. Visual Representation of the Human Microbiome Across Body Sites. Human-associated microbial communities exhibit remarkable spatial diversity, with distinct bacterial populations colonizing specific anatomical sites. The lung microbiome, though less dense, primarily contains Firmicutes, Bacteroidetes, Proteobacteria, and Actinobacteria. The oral microbiome, the second largest microbial community, mainly includes Firmicutes, Proteobacteria, Bacteroidetes, Actinobacteria, and Fusobacteria. The human gut microbiome is primarily composed of six dominant phyla—Firmicutes, Bacteroidetes, Proteobacteria, Actinobacteria, Verrucomicrobia, and Fusobacteria—with Firmicutes and Bacteroidetes being the most abundant. The skin microbiome harbors site-specific microbes from Cyanobacteria, Proteobacteria, Actinobacteria, Bacteroidetes, and Firmicutes.
Figure 1. Visual Representation of the Human Microbiome Across Body Sites. Human-associated microbial communities exhibit remarkable spatial diversity, with distinct bacterial populations colonizing specific anatomical sites. The lung microbiome, though less dense, primarily contains Firmicutes, Bacteroidetes, Proteobacteria, and Actinobacteria. The oral microbiome, the second largest microbial community, mainly includes Firmicutes, Proteobacteria, Bacteroidetes, Actinobacteria, and Fusobacteria. The human gut microbiome is primarily composed of six dominant phyla—Firmicutes, Bacteroidetes, Proteobacteria, Actinobacteria, Verrucomicrobia, and Fusobacteria—with Firmicutes and Bacteroidetes being the most abundant. The skin microbiome harbors site-specific microbes from Cyanobacteria, Proteobacteria, Actinobacteria, Bacteroidetes, and Firmicutes.
Cells 14 01218 g001
Figure 2. Interaction of Microbial Pathogens with Cancer Cells. This schematic illustration depicts various microbial pathogens, including viruses, bacteria, and fungi, that may interact with cancer cells. The viral pathogens include Epstein–Barr Virus, Respiratory Syncytial Virus, Ebola virus, and Rift Valley Fever Virus, which may influence the tumor microenvironment through immune modulation and oncogenic properties. The bacterial pathogens consist of F. nucleatum, Veillonella, Staphylococcus epidermidis, Bacillus subtilis, Micrococcus, H. pylori, Escherichia coli, Salmonella, and Bifidobacteria, Peptostreptococcus, Cutibacterium acnes, Akkermansia muciniphila, Staphylococcus aureus, Prevotella, which may contribute to tumor progression and inflammation, or possess probiotic and therapeutic roles. The fungal pathogens, including Aspergillus, C.neoformans, Candida albicans, and Malassezia, can alter immune responses and affect tumor dynamics. These microbial interactions with cancer cells may have significant implications in tumor development, immune evasion, and microbiome-based therapeutic strategies.
Figure 2. Interaction of Microbial Pathogens with Cancer Cells. This schematic illustration depicts various microbial pathogens, including viruses, bacteria, and fungi, that may interact with cancer cells. The viral pathogens include Epstein–Barr Virus, Respiratory Syncytial Virus, Ebola virus, and Rift Valley Fever Virus, which may influence the tumor microenvironment through immune modulation and oncogenic properties. The bacterial pathogens consist of F. nucleatum, Veillonella, Staphylococcus epidermidis, Bacillus subtilis, Micrococcus, H. pylori, Escherichia coli, Salmonella, and Bifidobacteria, Peptostreptococcus, Cutibacterium acnes, Akkermansia muciniphila, Staphylococcus aureus, Prevotella, which may contribute to tumor progression and inflammation, or possess probiotic and therapeutic roles. The fungal pathogens, including Aspergillus, C.neoformans, Candida albicans, and Malassezia, can alter immune responses and affect tumor dynamics. These microbial interactions with cancer cells may have significant implications in tumor development, immune evasion, and microbiome-based therapeutic strategies.
Cells 14 01218 g002
Figure 3. Illustrations depict the following: (a) Gut microbiome predicts the risk for NB; (b) Postnatal (and not maternal transmission) programming of microbiome in NB patients; (c) Function of gut microbe on NB pathogenesis; (d) Presence of NB alters gut microbiome composition; (e) Microbial composition predicts treatment outcome in NB (murine model); (f) Prebiotic treatment mitigate NB-induced microbial mayhem; (g) Microbial composition in TME predicts NB outcome.
Figure 3. Illustrations depict the following: (a) Gut microbiome predicts the risk for NB; (b) Postnatal (and not maternal transmission) programming of microbiome in NB patients; (c) Function of gut microbe on NB pathogenesis; (d) Presence of NB alters gut microbiome composition; (e) Microbial composition predicts treatment outcome in NB (murine model); (f) Prebiotic treatment mitigate NB-induced microbial mayhem; (g) Microbial composition in TME predicts NB outcome.
Cells 14 01218 g003
Table 1. Evolution of different microbial communities across the human lifespan, from birth to adulthood.
Table 1. Evolution of different microbial communities across the human lifespan, from birth to adulthood.
StageMicrobiome DevelopmentInfluencing Factors
BirthColonization commences with germs originating from the maternal body (vagina, feces, skin).Method of delivery (vaginal versus caesarean)
Newborn infant
(1–4 weeks)
Initially dominated by Staphylococcus and Enterobacteriaceae, followed by subsequent succession by Bifidobacterium.Feeding technique (breast milk versus formula), gestational age, antibiotic administration
Infancy
(2 years)
Enrichment of Bifidobacterium and incorporation of lactic acid bacteriaDuration of breastfeeding, adoption of solid meals
Childhood
(2–4 years)
Change to mature microbes, rise of BacteroidesEnvironmental exposures
Adulthood
(above 18 years)
Firmicutes and BacteroidetesLifestyle, nutrition, environmental conditions
Table 5. International Neuroblastoma Risk Group Staging System (INRGSS).
Table 5. International Neuroblastoma Risk Group Staging System (INRGSS).
INRGSSFeaturesRisk GroupsEvent-Free Survival
L1Locoregional tumor without any identified risk factors based on imagingVery low—low risk5-year—>75–85%
L2Tumor cells have metastasized to adjacent tissuesLow risk5-year—75–85%
MNB cells spread to distant organsLow risk—high risk5-year—50–75%
MSMetastatic disease localized to the skin, liver, or bone marrow.High risk5-year—<50%
Table 6. International Neuroblastoma Staging System (INSS).
Table 6. International Neuroblastoma Staging System (INSS).
INSS StageDescriptionRisk Group5-Year Survival Rate (%)
Stage 1Localized tumor, completely resected by surgeryLow Risk90–95%
Stage 2ATumor localized but cannot be completely removed by surgeryLow/Intermediate Risk80–90%
Stage 2BTumors on one side may not always be fully resectableIntermediate Risk75–85%
Stage 3Unresectable tumor that may involve lymph nodes but has not spread distantlyHigh Risk50–70%
Stage 4Cancer has spread to distant sites (e.g., bone, liver, bone marrow)High Risk20–40%
Stage 4SIn children <1 year, cancer has spread to liver, skin, and/or bone marrow (≤10% involvement)Low/Intermediate Risk80–95%
Table 7. Categorizes tumor microenvironment (TME) subtypes in high-risk NB based on immune activity and gene expression profiles [250]. Each subtype is associated with distinct biological features, such as neoantigen load and oncogenic signaling, which influence patient survival outcomes.
Table 7. Categorizes tumor microenvironment (TME) subtypes in high-risk NB based on immune activity and gene expression profiles [250]. Each subtype is associated with distinct biological features, such as neoantigen load and oncogenic signaling, which influence patient survival outcomes.
TME SubtypeImmune CharacteristicsGenomic FeaturesPathway EnrichmentClinical Implication
T cell-inflamed High CD8+ T cell infiltration, IFN- γ signature, immune checkpoint moleculesHigh neoantigen load, diverse TCR repertoireImmune-related pathways (e.g., IFN signaling)Best overall and event-free survival
IntermediateModerate immune cell markersVariable neoantigen burdenModerate immune and oncogenic signalingIntermediate prognosis
Non-T cell-inflamedLow immune gene expression, T cell exclusion signaturesActivation of MYCN, ASCL1, SOX11, KMT2A, even without MYCN amplificationNeurodevelopmental and cell cycle pathwaysPoor survival; resistant to immunotherapy
Table 8. Comprehensive summary of ongoing clinical trials in high-risk NB.
Table 8. Comprehensive summary of ongoing clinical trials in high-risk NB.
Trial TitleDescriptionEligibility CriteriaObjectiveLead OrganizationPhase
Dinutuximab with Chemo-therapy, Surgery and Stem Cell Transplantation for the Treatment of Children with Newly Diagnosed High Risk NBTests the addition of dinutuximab to induction chemotherapy and standard care in high-risk NB≤30 years, newly diagnosed high-risk NB, specific renal/liver/cardiac function criteriaTo determine if early chemoimmunotherapy improves event-free survivalChildren’s Oncology GroupPhase III
Eflornithine (DFMO) and Etoposide for Relapsed/Refractory NBDFMO + etoposide in relapsed/refractory NB≤30.99 years, relapsed/refractory NB, prior multi-drug chemotherapyEvaluate safety and efficacy of DFMO + etoposideGiselle ShollerPhase I/II
A Study of Therapeutic Iobenguane (131-I) and Vorinostat for Recurrent or Progressive High-Risk NB Subjects131I-MIBG + Vorinostat for recurrent/progressive NBIobenguane-avid high-risk NB, prior induction therapy, stem cell availabilityEvaluate efficacy and safety of combination therapyDRAXIMAGEPhase II
A Study of a Vaccine in Combination with Beta-glucan in People with NBOPT-821 vaccine + beta-glucan for high-risk NBHR-NB in CR, ≥21 and ≤180 days post systemic therapy, adequate organ functionAssess anti-GD2 antibody titersMemorial Sloan Kettering Cancer CenterPhase II
Naxitamab Added to Induction for Newly Diagnosed High-Risk NBNaxitamab added to 5 cycles of induction chemotherapy≤21 years, newly diagnosed high-risk NB, specific INSS stagesEvaluate efficacy and safety of naxitamab in inductionGiselle ShollerPhase II
Autologous hALK. Chimeric Antigen Receptor T Cells (hALK.CAR T) for the Treatment of Relapsed or Refractory High-Risk NBhALK.CAR T cell therapy for relapsed/refractory NB≥12 months and <30 years, relapsed/refractory high-risk NBIdentify MTD and assess safety and efficacyDana-Farber Harvard Cancer CenterPhase I/II
67Cu-SARTATE™ Peptide Receptor Radionuclide Therapy Administered to Pediatric Patients With High-Risk, Relapsed, Refractory NBAdaptive trial of 67Cu-SARTATE in pediatric high-risk NBHigh-risk NB, adequate organ function, stem cell product availableEvaluate safety and efficacy of 67Cu-SARTATEClarity PharmaceuticalsPhase I/II
Donor Immune Cells (Allogenic Ex Vivo Expanded Gamma Delta T Cells), Dinutuximab, Temozolomide, Irinotecan and Zoledronate for the Treatment of Refractory, Relapsed, or Progressive NB or Osteosarcoma in ChildrenGamma delta T cells + dinutuximab + chemo for refractory/relapsed NB≥12 months, high-risk NB or osteosarcoma, measurable diseaseDetermine MTD and define toxicitiesEmory University HospitalPhase I
Reduced Chemotherapy (N10) for the Treatment of High-Risk NB in ChildrenN10 chemo regimen for high-risk NB<19 years, HR-NB, ≤1 prior HR-NB chemo cycleAssess early CR rate and survival outcomesMemorial Sloan Kettering Cancer CentePhase II
High Risk NB, a Study 1.8 of SIOP-Europe (SIOPEN)Multimodal treatment protocol with randomized immunotherapy armsHigh-risk NB (stages 2–4s, MYCN+ or >12 months)Improve EFS with BuMel MAT and immunotherapy, including immunotherapy (e.g., IL-2) which may interact with gut microbiome. St. Anna KinderkrebsforschungPhase I/II
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

Giri, A.K.; Subramanian, P.; Periyasamy, L.; Aravindan, S.; Aravindan, N. Microbiome in Neuroblastoma: A Virgin Island in the World of Onco-Microbiome. Cells 2025, 14, 1218. https://doi.org/10.3390/cells14151218

AMA Style

Giri AK, Subramanian P, Periyasamy L, Aravindan S, Aravindan N. Microbiome in Neuroblastoma: A Virgin Island in the World of Onco-Microbiome. Cells. 2025; 14(15):1218. https://doi.org/10.3390/cells14151218

Chicago/Turabian Style

Giri, Ashwath Keshav, Poorvi Subramanian, Loganayaki Periyasamy, Sivaroopan Aravindan, and Natarajan Aravindan. 2025. "Microbiome in Neuroblastoma: A Virgin Island in the World of Onco-Microbiome" Cells 14, no. 15: 1218. https://doi.org/10.3390/cells14151218

APA Style

Giri, A. K., Subramanian, P., Periyasamy, L., Aravindan, S., & Aravindan, N. (2025). Microbiome in Neuroblastoma: A Virgin Island in the World of Onco-Microbiome. Cells, 14(15), 1218. https://doi.org/10.3390/cells14151218

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