Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (68)

Search Parameters:
Keywords = tumor-associated microbes

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
16 pages, 1267 KB  
Review
The Role of Microbiota in Ovarian Cancer: Implications for Treatment Response and Therapeutic Strategies
by Jose-Ramon Blanco, Rosa del Campo, José Avendaño-Ortiz, Mariano Laguna-Olmos and Amancio Carnero
Cells 2025, 14(22), 1813; https://doi.org/10.3390/cells14221813 - 19 Nov 2025
Viewed by 823
Abstract
Cancer remains a global health challenge (18.1 million new cases in 2020), with incidence projected to reach 28 million within two decades. Ovarian cancer (OC) is the deadliest gynecologic malignancy, usually diagnosed at advanced stages and with poorly understood etiology. Emerging evidence implicates [...] Read more.
Cancer remains a global health challenge (18.1 million new cases in 2020), with incidence projected to reach 28 million within two decades. Ovarian cancer (OC) is the deadliest gynecologic malignancy, usually diagnosed at advanced stages and with poorly understood etiology. Emerging evidence implicates reproductive tract and gut microbiota in OC biology. Microbiota shape carcinogenesis via turnover, immunity, and metabolism; dysbiosis promotes DNA damage, inflammation, and carcinogenic metabolites, engaging multiple hallmarks of cancer. In OC, microbes may reach tumors by local ascent, translocation, or hematogenous spread, originating from vagina, upper reproductive tract, peritoneal fluid, or gut. Lactobacillus-dominant vaginal communities support mucosal integrity, whereas anaerobes disrupt barriers, increase inflammation, and correlate with OC risk; mouse models show vaginal dysbiosis accelerates tumor progression. Distinct microbial profiles in upper reproductive sites and peritoneal fluid associated with immune remodeling. Gut dysbiosis drives barrier loss, immune imbalance, and estrogen reactivation. Microbial metabolites (lipopolysaccharides, short-chain fatty acids) modulate oncogenic pathways, altering epithelial–mesenchymal transition, immune evasion, and drug resistance. Across cohorts, OC tissues and fluids show Pseudomonadota/Bacteroidota enrichment and Akkermansia depletion; fecal microbiota from OC patients accelerates tumor growth in mice, whereas Akkermansia supplementation restores antitumor immunity. Antibiotic exposure and platinum resistance associate with reduced diversity and expansion of lactate-producing taxa. Microbiome-informed interventions–diet, probiotics/postbiotics, fecal microbiota transfer, and selective antibiotics–may augment chemotherapy and immunotherapy. Overall, the microbiome is a modifiable determinant of OC risk, progression, and treatment response, warranting rigorous, standardized, multi-omics studies. Full article
Show Figures

Figure 1

44 pages, 3780 KB  
Review
Lactobacilli-Derived Microbe-Associated Molecular Patterns (MAMPs) in Host Immune Modulation
by Salvatore Furnari, Ruben Ciantia, Adriana Garozzo, Pio Maria Furneri and Virginia Fuochi
Biomolecules 2025, 15(11), 1609; https://doi.org/10.3390/biom15111609 - 17 Nov 2025
Viewed by 1036
Abstract
Although traditionally sidelined by live probiotic effects, Lactobacilli-derived Microbe-Associated Molecular Patterns (MAMPs) are emerging as potent modulators of innate and adaptive immune responses, capable of acting independently of bacterial viability. However, the underlying mechanisms remain incompletely understood. These MAMPs, such as peptidoglycan (PGN), [...] Read more.
Although traditionally sidelined by live probiotic effects, Lactobacilli-derived Microbe-Associated Molecular Patterns (MAMPs) are emerging as potent modulators of innate and adaptive immune responses, capable of acting independently of bacterial viability. However, the underlying mechanisms remain incompletely understood. These MAMPs, such as peptidoglycan (PGN), lipoteichoic acid (LTA), and exopolysaccharides (EPSs), interact with pattern recognition receptors (PRRs) like Toll-like receptors (TLRs), initiating immune-signaling cascades that regulate cytokine production and inflammation. Lactobacilli-derived MAMPs exhibit dual immunomodulatory effects: they can enhance pro-inflammatory responses, e.g., interleukin-1β (IL-1β), IL-6, and tumor necrosis factor alpha (TNF-α) under inflammatory contexts, while enhancing regulatory pathways via IL-10 and regulatory T-cell (Tregs) induction in anti-inflammatory settings. Importantly, these immunomodulatory properties persist in the absence of bacterial viability, making MAMPs promising candidates for postbiotic therapies. This opens new avenues for MAMP-based strategies to target inflammation, overcoming the risks associated with live bacterial administration. This review examines the therapeutic relevance of non-viable MAMPs, particularly in inflammatory diseases where they have demonstrated benefits in reducing tissue damage, enhancing gut barrier function, and alleviating disease symptoms. Additionally, we discuss regulatory and translational challenges hindering their clinical implementation, highlighting the need for standardized characterization, a clear safety framework, and strain-specific profiling. Given their ability to fine-tune immune responses, MAMPs represent an emerging strategy for innovative treatments aimed at restoring immune balance and reinforcing host–microbe interactions. Full article
(This article belongs to the Section Biomacromolecules: Proteins, Nucleic Acids and Carbohydrates)
Show Figures

Figure 1

17 pages, 4925 KB  
Article
Bacterial Infections and Their Cell Wall Ligands Differentially Modulate Doxorubicin Sensitivity in Triple-Negative Breast Cancer Cells
by Sima Kianpour Rad, Runhao Li, Kenny K. L. Yeo, Fangmeinuo Wu, Yoko Tomita, Timothy J. Price, Wendy V. Ingman, Amanda R. Townsend and Eric Smith
Microorganisms 2025, 13(10), 2317; https://doi.org/10.3390/microorganisms13102317 - 7 Oct 2025
Viewed by 718
Abstract
Background: Triple-negative breast cancer (TNBC) is an aggressive subtype with limited treatment options and poor clinical outcomes. Emerging evidence suggests that the tumor-associated microbiome may influence disease progression and therapy response. Methods: We investigated how the Gram-negative bacterium Pseudomonas aeruginosa and Gram-positive bacterium [...] Read more.
Background: Triple-negative breast cancer (TNBC) is an aggressive subtype with limited treatment options and poor clinical outcomes. Emerging evidence suggests that the tumor-associated microbiome may influence disease progression and therapy response. Methods: We investigated how the Gram-negative bacterium Pseudomonas aeruginosa and Gram-positive bacterium Staphylococcus aureus, together with their cell wall components lipopolysaccharide (LPS) and lipoteichoic acid (LTA), modulate doxorubicin (DOX) efficacy in TNBC cells. Using gentamicin protection combined with flow cytometry of eFluor 450-labeled bacteria and CFU quantification, we assessed bacterial uptake, persistence, and effects on drug response in MDA-MB-468, MDA-MB-231, and MDA-MB-453 cells. Results: Both bacteria entered TNBC cells and survived for several days in a cell line-dependent manner. Notably, bacterial infection and purified cell wall ligands (LPS and LTA) significantly increased DOX accumulation and enhanced cytotoxicity in MDA-MB-468 and MDA-MB-231, but not in MDA-MB-453. The similar effects of LPS and LTA implicate Toll-like receptor signaling (TLR2 and TLR4) in modulating drug uptake. Conclusions: These findings demonstrate that bacterial infection and associated ligands can enhance doxorubicin uptake and cytotoxicity in TNBC cells, implicating TLR signaling as a potential contributor. Our results highlight the importance of host–microbe interactions in shaping chemotherapy response and warrant further investigation into their therapeutic relevance. Full article
(This article belongs to the Collection Microbiomes and Cancer: A New Era in Diagnosis and Therapy)
Show Figures

Figure 1

20 pages, 2560 KB  
Article
Fusobacterium nucleatum and Its Impact on Colorectal Cancer Chemoresistance: A Meta-Analysis of In Vitro Co-Culture Infections
by Katie R. Risoen, Claire A. Shaw, Jeremy Chien and Bart C. Weimer
Cancers 2025, 17(19), 3247; https://doi.org/10.3390/cancers17193247 - 7 Oct 2025
Cited by 1 | Viewed by 2172
Abstract
Introduction: Fusobacterium nucleatum, a common oral microbe associated with periodontal disease, has emerged as a significant prognostic indicator in colorectal cancer (CRC). This organism is notably enriched in CRC tissues and is associated with reduced survival times and relapse. Fusobacterium is implicated [...] Read more.
Introduction: Fusobacterium nucleatum, a common oral microbe associated with periodontal disease, has emerged as a significant prognostic indicator in colorectal cancer (CRC). This organism is notably enriched in CRC tissues and is associated with reduced survival times and relapse. Fusobacterium is implicated in encouraging the development of chemoresistance through diverse tumor-promoting pathways that are increasingly being elucidated across molecular domains. Methods: This work uses a combined analysis of public data examining the role of F. nucleatum in CRC by investigating multiple transcriptomic datasets derived from co-culture infections in vitro. Results: In tandem with previously identified mechanisms known to be influenced by F. nucleatum, this analysis revealed that the bacterium activates multiple chemoresistance-associated pathways, including those driving inflammation, immune evasion, DNA damage, and metastasis. Notably, this study uncovered a novel induction of type I and type II interferon signaling, suggesting activation of a pseudo-antiviral state. Furthermore, pathway analysis (IPA) predicted altered regulation of several therapeutic agents, suggesting that F. nucleatum may compromise drug efficacy through transcriptional reprogramming. Conclusions: These findings reinforce the role of F. nucleatum in modulating host cellular pathways and support the hypothesis that bacterial association potentiates chemoresistance. Full article
(This article belongs to the Special Issue Infectious Agents and Cancer in Children and Adolescents)
Show Figures

Figure 1

24 pages, 1761 KB  
Review
Emerging Understanding of Gut Microbiome in Colorectal Cancer and Food-Related Intervention Strategies
by Jie Zhang, Zhao-Jun Wei and Guangsen Fan
Foods 2025, 14(17), 3040; https://doi.org/10.3390/foods14173040 - 29 Aug 2025
Viewed by 3454
Abstract
Colorectal cancer (CRC) is one of the most common cancers, accounting for approximately 10% of all new cancer cases globally. An increasing number of studies have revealed that the gut microbiome is strongly associated with the pathogenesis and progression of CRC. Based on [...] Read more.
Colorectal cancer (CRC) is one of the most common cancers, accounting for approximately 10% of all new cancer cases globally. An increasing number of studies have revealed that the gut microbiome is strongly associated with the pathogenesis and progression of CRC. Based on these advances, this review delineates the mechanistic links between specific microbes and CRC, as well as emerging food-related nutritional intervention strategies. In vivo and in vitro studies have pinpointed the implications of key microbes such as Fusobacterium nucleatum, certain strains of Escherichia coli, enterotoxigenic Bacteroides fragilis, and Enterococcus faecalis, among others, and metabolite involvement and immune responses. Particular attention is paid to the roles of intratumoral microbiota in the development and treatment of CRC, given their direct interaction with tumor cells. Various food-related nutritional intervention strategies have been developed to mitigate CRC risk, including probiotics, antibiotics, or the administration of bioactive compounds such as luteoloside. Finally, we outline critical research directions regarding the influence of animal lineage, carcinoma location, population demographics, the application of advanced in vitro models, and the mediatory roles of gut-associated epithelial cells. In summary, this review might consolidate our current knowledge on the contribution of gut microbiota to CRC and highlights the microbe-based strategies to enhance nutritional interventions for this disease. Full article
Show Figures

Figure 1

14 pages, 908 KB  
Article
Fusobacterium nucleatum Infection Drives Glutathione Depletion in Gastric Cancer: Integrated Multi-Omics and Experimental Validation
by Siru Nie, Yuehua Gong, Ang Wang, Rui Guo, Xiaohui Chen and Yuan Yuan
Microorganisms 2025, 13(8), 1907; https://doi.org/10.3390/microorganisms13081907 - 15 Aug 2025
Cited by 1 | Viewed by 999
Abstract
The colonization of Fusobacterium nucleatum (F. nucleatum) in the microenvironment of gastric cancer (GC) is closely associated with tumor progression, but its impact on host metabolic remodeling remains unclear. This study aims to elucidate the mechanistic link between F. nucleatum infection [...] Read more.
The colonization of Fusobacterium nucleatum (F. nucleatum) in the microenvironment of gastric cancer (GC) is closely associated with tumor progression, but its impact on host metabolic remodeling remains unclear. This study aims to elucidate the mechanistic link between F. nucleatum infection and metabolic changes in GC tissue. By integrating 16S rRNA microbiome sequencing and LC-MS/MS metabolomics, the differences in microbial composition and metabolic profiles between Fusobacterium sp.-positive and -negative GC tissues were systematically compared, and the correlation of differential microbes and differential metabolites was analyzed. The impact of F. nucleatum on the glutathione (GSH) metabolic pathway was validated through in vitro tissue testing and the use of the infection model of GC cell lines (such as AGS and HGC27). Integrative omics analysis showed a strong negative correlation between Fusobacterium sp. infection and antioxidant metabolite GSH levels in GCs (p < 0.001). Metabolic reprogramming features: Eleven differentially expressed metabolites were identified using LC-MS/MS metabolomics screening (p < 0.05). GSH was significantly depleted in the Fusobacterium sp.-positive group. Experimental validation: At the histological level, the abundance of F. nucleatum in GC tissues was higher than that in the paired adjacent non-cancerous (NC) tissues; at the cellular level, after F. nucleatum infection of GC cells, the intracellular GSH level decreased (p < 0.01), accompanied by a decrease in glutathione synthetase (GSS) mRNA expression and reactive oxygen species (ROS). This study is the first to demonstrate that F. nucleatum suppresses the GSH synthesis pathway, leading to the breakdown of antioxidant capacity and the formation of an oxidative stress microenvironment in GC cells. These findings provide new insights into the metabolic mechanism of F. nucleatum in promoting GC progression and suggest that targeting the F. nucleatum-GSH axis could offer a novel strategy for GC therapeutic intervention. Full article
(This article belongs to the Section Medical Microbiology)
Show Figures

Figure 1

15 pages, 860 KB  
Review
Gut Microbiome Alterations in Colorectal Cancer: Mechanisms, Therapeutic Strategies, and Precision Oncology Perspectives
by Miriam Tudorache, Andreea-Ramona Treteanu, Gratiela Gradisteanu Pircalabioru, Irina-Oana Lixandru-Petre, Alexandra Bolocan and Octavian Andronic
Cancers 2025, 17(14), 2294; https://doi.org/10.3390/cancers17142294 - 10 Jul 2025
Cited by 2 | Viewed by 3304
Abstract
Colorectal cancer (CRC) is one of the most prevalent and lethal oncological diseases worldwide, with a concerning rise in incidence, particularly in developing countries. Recent advances in genetic sequencing have revealed that the gut microbiome plays a crucial role in CRC development. Mechanisms [...] Read more.
Colorectal cancer (CRC) is one of the most prevalent and lethal oncological diseases worldwide, with a concerning rise in incidence, particularly in developing countries. Recent advances in genetic sequencing have revealed that the gut microbiome plays a crucial role in CRC development. Mechanisms such as chronic inflammation, metabolic alterations, and oncogenic pathways have demonstrated that dysbiosis, a disruption of the gut microbiome, is linked to CRC. Associations have been found between tumor progression, treatment resistance, and pathogenic microbes such as Fusobacterium nucleatum and Escherichia coli. A promising approach for CRC prevention and treatment is microbiome manipulation through interventions such as probiotics, prebiotics, fecal microbiota transplantation, and selective antibiotics. This article explores how gut microbiome alterations influence CRC pathogenesis and examines microbiome modulation strategies currently used as adjuncts to traditional treatments. Advances in artificial intelligence, single-cell and spatial transcriptomics, and large-scale initiatives such as the ONCOBIOME Project are paving the way for the identification of microbiome-derived biomarkers for early CRC detection and personalized treatment. Despite promising progress, challenges such as interindividual variability, causal inference, and regulatory hurdles must be addressed. Future integration of microbiome analysis into multi-omics frameworks holds great potential to revolutionize precision oncology in CRC management. Full article
Show Figures

Figure 1

23 pages, 6207 KB  
Review
Bacteria and Carcinogenesis and the Management of Cancer: A Narrative Review
by Paulina Plewa, Kajetan Kiełbowski, Oliwia Mentel, Karolina Figiel, Estera Bakinowska, Rafał Becht, Bolesław Banach and Andrzej Pawlik
Pathogens 2025, 14(5), 509; https://doi.org/10.3390/pathogens14050509 - 21 May 2025
Cited by 2 | Viewed by 2965
Abstract
There is a widely known relationship between certain microbes and cancer progression. For instance, Helicobacter pylori is associated with the occurrence of gastric cancer, while HPV is associated with cervical and head and neck cancers. Recent studies have uncovered novel and important associations [...] Read more.
There is a widely known relationship between certain microbes and cancer progression. For instance, Helicobacter pylori is associated with the occurrence of gastric cancer, while HPV is associated with cervical and head and neck cancers. Recent studies have uncovered novel and important associations between bacterial presence and tumor formation and treatment response. Apart from the influence of the intestinal microbiome on cancer, the local activity of bacteria affects disease properties as well. Bacteria can localize within tumors in less vascularized niches. Their presence mediates the activity of signaling pathways, which contribute to tumorigenesis. Furthermore, they affect the composition of the tumor microenvironment, a highly complex structure composed of immunoregulatory cells and secreted inflammatory mediators. Recently, researchers have analyzed the properties of bacteria to develop novel anticancer strategies. The aim of this review is to discuss the latest findings regarding the relationships between bacteria and cancer and the properties of bacteria that could be used to kill cancer cells. Full article
Show Figures

Figure 1

9 pages, 1073 KB  
Brief Report
Association of SEPT9 Gene Methylation with the Clinicopathologic Features and Fusobacterium nucleatum Infection in Colorectal Cancer Patients
by Siew-Wai Pang, Subasri Armon, Jack-Bee Chook, Kaik-Boo Peh, Suat-Cheng Peh and Sin-Yeang Teow
J. Mol. Pathol. 2025, 6(2), 8; https://doi.org/10.3390/jmp6020008 - 23 Apr 2025
Viewed by 1534
Abstract
Background/Objectives: Colorectal cancer (CRC) is a significant global health issue. The identification of methylated Septin 9 (mSEPT9) as a biomarker for CRC represents a significant advancement in cancer diagnostics. On the other hand, Fusobacterium nucleatum (FN) is one of the [...] Read more.
Background/Objectives: Colorectal cancer (CRC) is a significant global health issue. The identification of methylated Septin 9 (mSEPT9) as a biomarker for CRC represents a significant advancement in cancer diagnostics. On the other hand, Fusobacterium nucleatum (FN) is one of the most studied cancer-related microbes in CRC. This study provided cohort evidence on the association of mSEPT9 with clinicopathologic characteristics and FN infection in CRC patients. Methods: Paired formalin-fixed paraffin-embedded (FFPE) tissue DNA (cancerous and adjacent non-cancer tissues) of eighty-three CRC patients was collected. Methylation-specific qPCR targeting the v2 promoter region of mSEPT9 was carried out on bisulfite-converted FFPE DNA. For FN detection, a TaqMan probe-based method targeting the 16S rRNA gene was used. The differences in mSEPT9 levels and FN expression between cancer and non-cancer tissues were evaluated. Association studies between mSEPT9 in the tumor and relative mSEPT9 levels with FN infection and available clinical data were conducted. Results: Higher mSEPT9 levels were found in the cancerous tissue compared to non-cancerous tissue (p < 0.0001). High mSEPT9 levels in the tumor were significantly associated with older patients (p < 0.001) and larger tumor size (p = 0.048) but not with other clinicopathologic variables. In double-positive patients where mSEPT9 was detected in both cancerous and non-cancerous tissue, the expression fold-change in mSEPT9, calculated using the 2−ΔΔCT formula, was significantly higher in patients with tumor size equal to or greater than 5 cm (p = 0.042). High levels of mSEPT9 in tumor were not associated with FN infection. However, high levels of FN infection were associated with mSEPT9 (p < 0.021). Conclusions: High levels of mSEPT9 are found in CRC tumor tissue and are associated with older age and larger tumor size, while high levels of FN infection are associated with mSEPT9 in this single-center cohort study. Full article
Show Figures

Figure 1

30 pages, 1075 KB  
Review
Decoding the Tumor-Associated Microbiota: From Origins to Nanomedicine Applications in Cancer Therapy
by Ruiqi Wang, Weizheng Li, Hongqian Cao and Lei Zhang
Biology 2025, 14(3), 243; https://doi.org/10.3390/biology14030243 - 27 Feb 2025
Cited by 3 | Viewed by 2951
Abstract
Growing evidence reveals that the tumor microbiome—comprising distinct microbial communities within neoplastic tissues—exerts a profound influence on cancer initiation, progression, and therapeutic response. These microbes actively reshape the tumor microenvironment (TME) through metabolite secretion, the modulation of immune pathways, and direct interactions with [...] Read more.
Growing evidence reveals that the tumor microbiome—comprising distinct microbial communities within neoplastic tissues—exerts a profound influence on cancer initiation, progression, and therapeutic response. These microbes actively reshape the tumor microenvironment (TME) through metabolite secretion, the modulation of immune pathways, and direct interactions with host cells, thereby affecting tumor biology and therapeutic outcomes. Despite substantial heterogeneity among cancer types, recent insights underscore the tumor microbiome’s potential as both a diagnostic/prognostic biomarker and a targetable component for innovative treatments. In this review, we synthesize emerging knowledge on the mechanistic roles of tumor-associated microbiota in shaping the TME, with a focus on how these discoveries can guide novel therapeutic strategies. We further explore interdisciplinary advances, including the convergence of microbiomics and nanotechnology, to enhance drug delivery, circumvent resistance, and foster TME remodeling. By highlighting these cutting-edge developments, our review underscores the transformative potential of integrating tumor microbiome research into precision oncology and advancing more personalized cancer therapies. Full article
(This article belongs to the Section Cancer Biology)
Show Figures

Figure 1

19 pages, 4650 KB  
Article
Exploring the Interplay of Gut Microbiota and Systemic Inflammation in Pediatric Obstructive Sleep Apnea Syndrome and Its Impact on Blood Pressure Status: A Cross-Sectional Study
by Chung-Guei Huang, Wan-Ni Lin, Li-Jen Hsin, Tuan-Jen Fang, Hsueh-Yu Li, Chin-Chia Lee and Li-Ang Lee
Int. J. Mol. Sci. 2024, 25(24), 13344; https://doi.org/10.3390/ijms252413344 - 12 Dec 2024
Cited by 2 | Viewed by 2184
Abstract
Obstructive sleep apnea syndrome (OSAS) is prevalent among children and is associated with elevated blood pressure (BP), posing a risk for future hypertension and cardiovascular diseases. While the roles of gut microbiota and systemic inflammation in OSAS pathogenesis are recognized in adults and [...] Read more.
Obstructive sleep apnea syndrome (OSAS) is prevalent among children and is associated with elevated blood pressure (BP), posing a risk for future hypertension and cardiovascular diseases. While the roles of gut microbiota and systemic inflammation in OSAS pathogenesis are recognized in adults and animal models, their impact on pediatric BP remains less understood. This cross-sectional study explored the relationships between polysomnographic parameters, gut microbiota, systemic inflammation, and BP in 60 children with OSAS. Significant associations between specific microbial profiles—including beta diversity and 31 marker microbes—and BP variations were observed. These microbial profiles correlated with significant alterations in systemic inflammation markers like interleukin-17 and tumor necrosis factor-α. Notably, the relative abundance of Acinetobacter was related to fluctuations in these inflammatory markers and BP levels. The research further highlighted the unique microbial and cytokine profiles exhibited by children with different BP levels, indicating a substantial role of gut microbiota and systemic inflammation in influencing pediatric cardiovascular health. The findings suggest integrating gut microbiota management into comprehensive cardiovascular risk strategies for children with OSAS. This initiative underscores the need for further investigations to decode the mechanisms behind these associations, which could lead to innovative treatments for pediatric OSAS. Full article
(This article belongs to the Special Issue Current Advances in Gut Microbiota in Human Diseases and Health)
Show Figures

Figure 1

23 pages, 1756 KB  
Review
Metabolites in the Dance: Deciphering Gut-Microbiota-Mediated Metabolic Reprogramming of the Breast Tumor Microenvironment
by Afaf Altrawy, Maye M. Khalifa, Asmaa Abdelmaksoud, Yomna Khaled, Zeinab M. Saleh, Hager Sobhy, Shaimaa Abdel-Ghany, Amany Alqosaibi, Afnan Al-Muhanna, Jawaher Almulhim, Ahmed El-Hashash, Hussein Sabit and Borros Arneth
Cancers 2024, 16(24), 4132; https://doi.org/10.3390/cancers16244132 - 11 Dec 2024
Cited by 6 | Viewed by 2592
Abstract
Breast cancer (BC), a major cause of death among women worldwide, has traditionally been linked to genetic and environmental factors. However, emerging research highlights the gut microbiome’s significant role in shaping BC development, progression, and treatment outcomes. This review explores the intricate relationship [...] Read more.
Breast cancer (BC), a major cause of death among women worldwide, has traditionally been linked to genetic and environmental factors. However, emerging research highlights the gut microbiome’s significant role in shaping BC development, progression, and treatment outcomes. This review explores the intricate relationship between the gut microbiota and the breast tumor microenvironment, emphasizing how these microbes influence immune responses, inflammation, and metabolic pathways. Certain bacterial species in the gut either contribute to or hinder BC progression by producing metabolites that affect hormone metabolism, immune system pathways, and cellular signaling. An imbalance in gut bacteria, known as dysbiosis, has been associated with a heightened risk of BC, with metabolites like short-chain fatty acids (SCFAs) and enzymes such as β-glucuronidase playing key roles in this process. Additionally, the gut microbiota can impact the effectiveness of chemotherapy, as certain bacteria can degrade drugs like gemcitabine and irinotecan, leading to reduced treatment efficacy. Understanding the complex interactions between gut bacteria and BC may pave the way for innovative treatment approaches, including personalized microbiome-targeted therapies, such as probiotics and fecal microbiota transplants, offering new hope for more effective prevention, diagnosis, and treatment of BC. Full article
(This article belongs to the Special Issue Targeting Tumor Microenvironment in Cancer: Promises and Challenges)
Show Figures

Figure 1

13 pages, 3203 KB  
Article
Fecal Microbiome Composition Correlates with Pathologic Complete Response in Patients with Operable Esophageal Cancer Treated with Combined Chemoradiotherapy and Immunotherapy
by Fyza Y. Shaikh, Seoho Lee, James R. White, Yujie Zhao, Jacqueline T. Ferri, Gavin Pereira, Blair V. Landon, Suqi Ke, Chen Hu, Josephine L. Feliciano, Russell K. Hales, K. Ranh Voong, Richard J. Battafarano, Stephen C. Yang, Stephen Broderick, Jinny Ha, Elizabeth Thompson, Eun J. Shin, David L. Bartlett, Benny Weksler, Drew M. Pardoll, Valsamo Anagnostou, Vincent K. Lam, Ali H. Zaidi, Ronan J. Kelly and Cynthia L. Searsadd Show full author list remove Hide full author list
Cancers 2024, 16(21), 3644; https://doi.org/10.3390/cancers16213644 - 29 Oct 2024
Cited by 7 | Viewed by 2362
Abstract
Background: Preclinical and clinical data indicate that chemoradiotherapy (CRT) in combination with checkpoint inhibitors may prime an anti-tumor immunological response in esophageal cancer. However, responses to neoadjuvant therapy can vary widely and the key biomarkers to determine response remain poorly understood. The [...] Read more.
Background: Preclinical and clinical data indicate that chemoradiotherapy (CRT) in combination with checkpoint inhibitors may prime an anti-tumor immunological response in esophageal cancer. However, responses to neoadjuvant therapy can vary widely and the key biomarkers to determine response remain poorly understood. The fecal microbiome is a novel and potentially modifiable biomarker of immunotherapy response, and both fecal and tumor microbes have been found to associate with outcomes in esophageal cancer. Methods: Fecal and tumor samples were collected from patients with stage II–III resectable esophageal or gastroesophageal junction carcinoma treated with neoadjuvant immune checkpoint inhibitors (ICIs) plus CRT prior to surgical resection. Microbiome profiles were analyzed by 16S rRNA amplicon sequencing and taxonomic data were integrated with fecal metabolite analysis to assess microbial function. Results: The fecal microbiome of patients with pathological complete response (PCR) grouped in distinct clusters compared to patients with residual viable tumor (RVT) by Bray–Curtis diversity metric. Integrated taxonomic and metabolomic analysis of fecal samples identified a sphingolipid and primary bile acid as enriched in the PCR, the levels of which correlated with several bacterial species: Roseburis inulinivorans, Ruminococcus callidus, and Fusicantenibacter saccharivorans. Analysis of the tumor microbiome profiles identified several bacterial genera previously associated with esophageal tumors, including Streptococcus and Veillonella. Conclusions: These results further characterize the fecal and tumor microbiome of patients with operable esophageal cancer and identify specific microbes and metabolites that may help elucidate how microbes contribute to tumor response with neoadjuvant CRT combined with ICI. Full article
(This article belongs to the Special Issue Circulating Cancer Biomarkers: Progress, Challenges and Opportunities)
Show Figures

Figure 1

7 pages, 430 KB  
Brief Report
Insights into Porphyromonas somerae in Bladder Cancer Patients: Urinary Detection by ddPCR
by Filippo Russo, Speranza Esposito, Lorella Tripodi, Savio Domenico Pandolfo, Achille Aveta, Felice Amato, Carmela Nardelli, Ciro Imbimbo, Lucio Pastore and Giuseppe Castaldo
Microorganisms 2024, 12(10), 2049; https://doi.org/10.3390/microorganisms12102049 - 10 Oct 2024
Cited by 12 | Viewed by 1756
Abstract
To date, the increased awareness of the impact of microbes on human health has promoted scientific interest in microbiome studies for diagnostic and therapeutic purposes, revealing correlations between specific taxa and cancer. In particular, numerous species of Porphyromonas have been associated with several [...] Read more.
To date, the increased awareness of the impact of microbes on human health has promoted scientific interest in microbiome studies for diagnostic and therapeutic purposes, revealing correlations between specific taxa and cancer. In particular, numerous species of Porphyromonas have been associated with several types of tumors. Previously, we studied the urobiome using Next-Generation Sequencing (NGS), and found an increase in Porphyromonas somerae in first morning urine of subjects affected by bladder cancer (BCa). Here, we aimed to confirm the presence of P. somerae in BCa patients by using droplet digital Polymerase Chain Reaction (ddPCR), testing a cohort of 102 male subjects over 50 years. Our findings showed a significant increase in P. somerae in the urine of the BCa group within both ddPCR and NGS, and a correlation between the two methods was observed at a statistical level. Moreover, P. somerae’s identification with ddPCR confirmed a significant association between this bacterium and the presence of BCa, highlighting its potential role as a biomarker. This allows us to propose the ddPCR as a suitable method for first-stage BCa screening and follow-up. Full article
(This article belongs to the Special Issue Microbiome and Genitourinary Diseases)
Show Figures

Figure 1

19 pages, 1774 KB  
Review
Bile Acids in Pancreatic Carcinogenesis
by Bharti Sharma, Kate Twelker, Cecilia Nguyen, Scott Ellis, Navin D. Bhatia, Zachary Kuschner, Andrew Agriantonis, George Agriantonis, Monique Arnold, Jasmine Dave, Juan Mestre, Zahra Shafaee, Shalini Arora, Hima Ghanta and Jennifer Whittington
Metabolites 2024, 14(7), 348; https://doi.org/10.3390/metabo14070348 - 21 Jun 2024
Cited by 9 | Viewed by 3494
Abstract
Pancreatic cancer (PC) is a dangerous digestive tract tumor that is becoming increasingly common and fatal. The most common form of PC is pancreatic ductal adenocarcinoma (PDAC). Bile acids (BAs) are closely linked to the growth and progression of PC. They can change [...] Read more.
Pancreatic cancer (PC) is a dangerous digestive tract tumor that is becoming increasingly common and fatal. The most common form of PC is pancreatic ductal adenocarcinoma (PDAC). Bile acids (BAs) are closely linked to the growth and progression of PC. They can change the intestinal flora, increasing intestinal permeability and allowing gut microbes to enter the bloodstream, leading to chronic inflammation. High dietary lipids can increase BA secretion into the duodenum and fecal BA levels. BAs can cause genetic mutations, mitochondrial dysfunction, abnormal activation of intracellular trypsin, cytoskeletal damage, activation of NF-κB, acute pancreatitis, cell injury, and cell necrosis. They can act on different types of pancreatic cells and receptors, altering Ca2+ and iron levels, and related signals. Elevated levels of Ca2+ and iron are associated with cell necrosis and ferroptosis. Bile reflux into the pancreatic ducts can speed up the kinetics of epithelial cells, promoting the development of pancreatic intraductal papillary carcinoma. BAs can cause the enormous secretion of Glucagon-like peptide-1 (GLP-1), leading to the proliferation of pancreatic β-cells. Using Glucagon-like peptide-1 receptor agonist (GLP-1RA) increases the risk of pancreatitis and PC. Therefore, our objective was to explore various studies and thoroughly examine the role of BAs in PC. Full article
Show Figures

Figure 1

Back to TopTop